MyoD

MyoD
  • 文章类型: Journal Article
    心力衰竭(HF)的预后取决于各种调节因素;microRNA-128(miR-128)被鉴定为心脏纤维化的调节因子。有助于HF。MyoD家族抑制剂(MDFI),据报道,这与Wnt/β-catenin通路有关,应该由miR-128调节。本研究探讨了miR-128与MDFI在心肌细胞发育中的相互作用,并阐明了其在心脏损伤中的作用。基因表达谱分析使用qPCR和Western印迹分析评估miR-128对HF中MDFI表达的影响。荧光素酶测定研究了miR-128和MDFI之间的直接相互作用。MTT,transwell,和免疫组织化学评估miR-128和MDFI对HF小鼠心肌细胞的影响。基因扫描和荧光素酶测定验证了miR-128和MDFI序列之间的相互作用。miR-128模拟物显著降低mRNA和蛋白水平的MDFI表达,降低率为55%。miR-128过表达促进细胞凋亡,增加65%,抑制心肌细胞增殖,而MDFI上调显著增强增殖。miR-128水平上调Wnt1和β-连环蛋白表达,而增加的MDFI水平抑制了这些表达。苏木精和伊红染色的组织学分析显示,miR-128吸收降低了MDFI表达,阻碍细胞增殖和心脏修复,超声心动图显示心脏功能相应改善。我们的研究结果表明miR-128与MDFI相互作用,通过调节Wnt1/β-catenin通路在HF管理中发挥关键作用。抑制miR-128可促进心肌细胞增殖,强调miR-128/MDFI相互作用在HF治疗中的潜在价值。
    Heart failure (HF) prognosis depends on various regulatory factors; microRNA-128 (miR-128) is identified as a regulator of cardiac fibrosis, contributing to HF. MyoD family inhibitor (MDFI), which is reported to be related with Wnt/β-catenin pathway, is supposed to be regulated by miR-128. This study investigates the interaction between miR-128 and MDFI in cardiomyocyte development and elucidates its role in heart injury. Gene expression profiling assessed miR-128\'s effect on MDFI expression in HF using qPCR and Western blot analysis. Luciferase assays studied the direct interaction between miR-128 and MDFI. MTT, transwell, and immunohistochemistry evaluated the effects of miR-128 and MDFI on myocardial cells in mice HF. Genescan and luciferase assays validated the interaction between miR-128 and MDFI sequences. miR-128 mimics significantly reduced MDFI expression at mRNA and protein levels with decrease rate of 55%. Overexpression of miR-128 promoted apoptosis with the increase rate 65% and attenuated cardiomyocyte proliferation, while MDFI upregulation significantly enhanced proliferation. Elevated miR-128 levels upregulated Wnt1 and β-catenin expression, whereas increased MDFI levels inhibited these expressions. Histological analysis with haematoxylin and eosin staining revealed that miR-128 absorption reduced MDFI expression, hindering cell proliferation and cardiac repair, with echocardiography showing corresponding improvements in cardiac function. Our findings suggest miR-128 interacts with MDFI, playing a crucial role in HF management by modulating the Wnt1/β-catenin pathway. Suppression of miR-128 could promote cardiomyocyte proliferation, highlighting the potential value of the miR-128/MDFI interplay in HF treatment.
    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:大量研究表明,肌体发育是肌体软骨形成和成骨形成的必要阶段。我们先前的研究已经在体外建立了稳定的前生中胚层祖细胞系(UiPSM)。自然,我们想探讨UiPSM细胞是否可以发展成骨和成肌分化。
    结果:选择性培养条件从UiPSM细胞产生PAX3和PAX7阳性骨骼肌前体。骨骼肌前体经历体外成熟,导致肌管形成。MYOD在短时间内有效促进了骨骼肌细胞的成熟。我们发现UiPSM和MYOD介导的UiPSM细胞来源的骨骼肌细胞在移植到MITRG小鼠胫骨前肌后是有活力的,通过生物发光成像和scRNA-seq评估。缺乏畸胎瘤形成和长期肌细胞植入的证据表明,未来治疗应用的潜力很大。此外,UiPSM细胞可在体外分化为成骨细胞和成软骨细胞。
    结论:UiPSM分化具有作为肌肉骨骼发育研究和治疗肌肉骨骼疾病的发育模型的潜力。
    BACKGROUND: Numerous studies have shown that somite development is a necessary stage of myogenesis chondrogenesis and osteogenesis. Our previous study has established a stable presomitic mesoderm progenitor cell line (UiPSM) in vitro. Naturally, we wanted to explore whether UiPSM cell can develop bone and myogenic differentiation.
    RESULTS: Selective culture conditions yielded PAX3 and PAX7 positive skeletal muscle precursors from UiPSM cells. The skeletal muscle precursors undergo in vitro maturation resulting in myotube formation. MYOD effectively promoted the maturity of the skeletal myocytes in a short time. We found that UiPSM and MYOD mediated UiPSM cell-derived skeletal myocytes were viable after transplantation into the tibialis anterior muscle of MITRG mice, as assessed by bioluminescence imaging and scRNA-seq. Lack of teratoma formation and evidence of long-term myocytes engraftment suggests considerable potential for future therapeutic applications. Moreover, UiPSM cells can differentiate into osteoblast and chondroblast cells in vitro.
    CONCLUSIONS: UiPSM differentiation has potential as a developmental model for musculoskeletal development research and treatment of musculoskeletal 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
    细胞疗法具有使受损心肌再肌化的巨大希望,但实际上受到移植后稳定移植在受体心脏中的心脏定向细胞的有限同种异体来源的阻碍。这里,我们证明,心包组织含有肌源性干细胞(pSCs),这些细胞在心肌梗死(MI)后响应炎症信号而被激活.源自MI大鼠的pSC(MI-pSC)显示体内和体外心脏定型,其特征在于心脏特异性Tnnt2表达和在培养物中形成节律性收缩。BulkRNA-seq分析揭示了一组与心脏/肌源性分化相关的基因的显着上调,旁分泌因子,和活化pSC中的细胞外基质与对照pSC(Sham-pSC)相比。值得注意的是,我们将MyoD定义为控制心脏承诺过程的关键因素,siRNA介导的MyoD基因沉默导致生肌潜能显著降低。将心脏定向细胞注射到梗塞的大鼠心脏中导致长期存活和在受体心肌中的稳定植入。因此,这些发现表明心包肌源性祖细胞是基于心脏细胞的治疗的一个有吸引力的候选者,可以使受损的心肌再肌化.
    Cellular therapy holds immense promise to remuscularize the damaged myocardium but is practically hindered by limited allogeneic sources of cardiac-committed cells that engraft stably in the recipient heart after transplantation. Here, we demonstrate that the pericardial tissue harbors myogenic stem cells (pSCs) that are activated in response to inflammatory signaling after myocardial infarction (MI). The pSCs derived from the MI rats (MI-pSCs) show in vivo and in vitro cardiac commitment characterized by cardiac-specific Tnnt2 expression and formation of rhythmic contraction in culture. Bulk RNA-seq analysis reveals significant upregulation of a panel of genes related to cardiac/myogenic differentiation, paracrine factors, and extracellular matrix in the activated pSCs compared to the control pSCs (Sham-pSCs). Notably, we define MyoD as a key factor that governs the process of cardiac commitment, as siRNA-mediated MyoD gene silencing results in a significant reduction of myogenic potential. Injection of the cardiac-committed cells into the infarcted rat heart leads to long-term survival and stable engraftment in the recipient myocardium. Therefore, these findings point to pericardial myogenic progenitors as an attractive candidate for cardiac cell-based therapy to remuscularize the damaged myocardium.
    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
    运动训练可以刺激脂肪酸氧化性慢抽搐的骨骼肌纤维的形成,与肥胖呈负相关,但是这种转化的分子机制需要进一步阐明。这里,我们报告说,通过运动训练下调线粒体二硫化物中继载体CHCHD4降低了TP53调节的凋亡抑制剂1(TRIAP1)向线粒体的导入,可以降低心磷脂水平,促进骨骼肌VDAC寡聚化。VDAC低聚,已知促进mtDNA释放,可以激活cGAS-STING/NFKB先天免疫信号并下调骨骼肌中的MyoD,从而促进氧化慢抽搐纤维的形成。在老鼠身上,CHCHD4单倍体不足足以激活该途径,导致氧化性肌纤维增加,脂肪积累随着年龄的增长而减少。调节肌纤维转化的特定介质的鉴定提供了进一步理解复杂代谢病症如肥胖的分子基础的机会,并且可能具有治疗意义。
    Exercise training can stimulate the formation of fatty-acid-oxidizing slow-twitch skeletal muscle fibers, which are inversely correlated with obesity, but the molecular mechanism underlying this transformation requires further elucidation. Here, we report that the downregulation of the mitochondrial disulfide relay carrier CHCHD4 by exercise training decreases the import of TP53-regulated inhibitor of apoptosis 1 (TRIAP1) into mitochondria, which can reduce cardiolipin levels and promote VDAC oligomerization in skeletal muscle. VDAC oligomerization, known to facilitate mtDNA release, can activate cGAS-STING/NFKB innate immune signaling and downregulate MyoD in skeletal muscle, thereby promoting the formation of oxidative slow-twitch fibers. In mice, CHCHD4 haploinsufficiency is sufficient to activate this pathway, leading to increased oxidative muscle fibers and decreased fat accumulation with aging. The identification of a specific mediator regulating muscle fiber transformation provides an opportunity to understand further the molecular underpinnings of complex metabolic conditions such as obesity and could have therapeutic implications.
    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
    CCCTC结合因子(CTCF),一种广泛表达的建筑蛋白,已经成为细胞身份基因转录的关键调节因子。然而,CTCF特定功能的精确分子机制仍然难以捉摸。这里,我们通过对原代肝细胞的综合分析来研究其机制,肌细胞,来自小鼠和人类的B细胞。我们证明CTCF与谱系特异性先驱转录因子(TFs)合作,包括MyoD,FOXA,和PU.1,以控制细胞身份在1D和3D水平。在1D级别,先锋TFs通过开放染色质促进谱系特异性CTCF占用。在3D层面,CTCF和先驱TFs形成调控中心来管理细胞身份基因的表达。使用MyoD-null小鼠验证了这种机制,CTCF基因敲除小鼠,和肌源性分化过程中的CRISPR编辑。总的来说,这些发现揭示了CTCF作为细胞身份辅助因子通过与先驱TF协调调控中心来控制细胞身份基因的一般机制.
    CCCTC-binding factor (CTCF), a ubiquitously expressed architectural protein, has emerged as a key regulator of cell identity gene transcription. However, the precise molecular mechanism underlying specialized functions of CTCF remains elusive. Here, we investigate the mechanism through integrative analyses of primary hepatocytes, myocytes, and B cells from mouse and human. We demonstrate that CTCF cooperates with lineage-specific pioneer transcription factors (TFs), including MyoD, FOXA, and PU.1, to control cell identity at 1D and 3D levels. At the 1D level, pioneer TFs facilitate lineage-specific CTCF occupancy via opening chromatin. At the 3D level, CTCF and pioneer TFs form regulatory hubs to govern the expression of cell identity genes. This mechanism is validated using MyoD-null mice, CTCF knockout mice, and CRISPR editing during myogenic differentiation. Collectively, these findings uncover a general mechanism whereby CTCF acts as a cell identity cofactor to control cell identity genes via orchestrating regulatory hubs with pioneer TFs.
    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
    骨骼肌成肌细胞系可以为探索控制骨骼肌发育及其相关分子调控的机制提供有价值的新的体外模型。在这项研究中,用胰蛋白酶和胶原酶I消化草鱼的骨骼肌组织,获得原代成肌细胞培养物。通过差异粘附纯化获得成肌细胞,并通过冷冻保存和复苏进行进一步分析,染色体分析,免疫组织化学,和免疫荧光。从草鱼(Ctenopharyngodonidellus)肌肉中建立了连续的草鱼成肌细胞细胞系(称为CIM),并且在一年或更长时间内传代超过100次。TheCIM细胞在冷冻保存1-6个月后以79.78-95.06%的活力恢复,并共享27.24h的种群倍增时间。CIM细胞的模态染色体数为48,CIM细胞系的线粒体12SrRNA序列与GenBank注册的草鱼具有99%的同一性。没有微生物(细菌,真菌,或支原体)在整个研究过程中检测到。通过特异性肌源性蛋白标记的免疫组织化学,证明CIM细胞的细胞类型是成肌细胞,包括CD34,desmin,MyoD,MyHC,以及关键基因的相对表达。诱导分化10天后,该细胞系的成肌率和融合指数分别为8.96~9.42%和3~24%,分别。CIM细胞系的端粒酶活性和转染效率分别为0.027IU/mgprot和23~24%,分别。这些结果表明,已成功建立了具有正常生物学功能的成肌细胞细胞系,名为CIM。这可能为相关的体外研究提供有价值的工具。
    Skeletal muscle myoblastic cell lines can provide a valuable new in vitro model for the exploration of the mechanisms that control skeletal muscle development and its associated molecular regulation. In this study, the skeletal muscle tissues of grass carp were digested with trypsin and collagenase I to obtain the primary myoblast cell culture. Myoblast cells were obtained by differential adherence purification and further analyzed by cryopreservation and resuscitation, chromosome analysis, immunohistochemistry, and immunofluorescence. A continuous grass carp myoblast cell line (named CIM) was established from grass carp (Ctenopharyngodon idellus) muscle and has been subcultured > 100 passages in a year and more. The CIM cells revived at 79.78-95.06% viability after 1-6 months of cryopreservation, and shared a population doubling time of 27.24 h. The number of modal chromosomes of CIM cells was 48, and the mitochondrial 12S rRNA sequence of the CIM cell line shared 99% identity with those of grass carp registered in GenBank. No microorganisms (bacteria, fungi, or mycoplasma) were detected during the whole study. The cell type of CIM cells was proven to be myoblast by immunohistochemistry of specific myogenic protein markers, including CD34, desmin, MyoD, and MyHC, as well as relative expression of key genes. And the myogenic rate and fusion index of this cell line after 10 days of induced differentiation were 8.96 ~ 9.42% and 3-24%, respectively. The telomerase activity and transfection efficiency of CIM cell line were 0.027 IU/mgprot and 23 ~ 24%, respectively. These results suggest that a myoblast cell line named CIM with normal biological function has been successfully established, which may provide a valuable tool for related in vitro studies.
    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
    长链非编码RNA(lncRNAs)参与肌细胞分化过程并发挥重要作用。先前的研究表明,lncRNA-MEG3促进猪骨骼肌卫星细胞(PSC)的分化,但MEG3与靶蛋白相互作用的调控机制尚未得到很好的研究。我们证明MEG3可以通过RNA下拉和RIP-qPCR结合二氢硫酰胺琥珀酰转移酶(DLST)。随后,敲低和过表达实验显示DLST促进PSC分化。挽救实验表明,DLST蛋白的表达随着MEG3的过表达而显著增加,随着MEG3的敲除而降低,而其mRNA表达没有改变。此外,通过染色质免疫沉淀(CHIP)和荧光素酶报告基因检测,我们成功预测并验证了转录因子肌源性分化(MYOD)与MEG3核心启动子的结合.结果表明,MYOD作为MEG3的转录因子促进MEG3的转录。体内MEG3的敲低表明MEG3参与骨骼肌再生。结论MYOD作为转录因子诱导MEG3表达。MEG3作为分子支架结合并促进DLST蛋白表达。本文为MEG3促进PSCs分化提供了新的分子机制。
    Long non-coding RNAs (lncRNAs) are involved in the process of muscle cell differentiation and play an important role. Previous studies have shown that lncRNA-MEG3 promotes the differentiation of porcine skeletal muscle satellite cells (PSCs), but the regulatory mechanism of MEG3 interaction with target protein has not been well studied. We demonstrated that MEG3 can bind dihydrolipoamide succinyltransferase (DLST) by RNA pull down and RIP-qPCR. Subsequently, knockdown and overexpression experiments showed that DLST promotes PSCs differentiation. Rescue experiments showed that the expression of DLST protein was significantly increased with MEG3 overexpression and decreased with MEG3 knockdown, while its mRNA expression was not changed. Furthermore, we have successfully predicted and validated that the transcription factor myogenic differentiation (MYOD) binds to the MEG3 core promoter though utilizing chromatin immunoprecipitation (CHIP) and luciferase reporter assays. The results indicated that MYOD acts as a transcription factor of MEG3 to promote MEG3 transcription. Knockdown of MEG3 in vivo indicated that MEG3 is involved in skeletal muscle regeneration. It is concluded that MYOD acts as a transcription factor to induce MEG3 expression. MEG3 acts as a molecular scaffold to bind and promote DLST protein expression. This paper provides a new molecular mechanism for MEG3 to promote the differentiation of PSCs.
    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
    本研究旨在探讨Myoz2在成肌细胞分化中的功能作用,并阐明在启动子转录调控中与Myoz2相互作用的潜在因素。时空表达结果表明,牛Myoz2基因在背最长肌表达量最高,在个体生长阶段和成肌细胞分化阶段。敲除Myoz2抑制成肌细胞分化,和MyoD的负面影响,MyoG,MyH和MEF2A在mRNA水平上的表达。随后,提取1.7Kb序列的牛Myoz2基因启动子区,然后它被设置为八个系列的删除片段,将其连接到pGL3-basic中以检测成肌细胞和肌管中Myoz2基因的核心启动子区域。转录因子MyoD和MyoG被鉴定为核心启动子区域中的重要顺式作用元件(-159/+1)。此外,根据双荧光素酶分析和多序列比对分析,它在不同物种中高度保守,分别。此外,染色质免疫沉淀(ChIP)分析结合定点突变和siRNA干扰和过表达证实,MyoD和MyoG的组合发生在-159/+1区,在牛Myoz2基因的调控中起重要作用。这些发现探索了Myoz2基因在牛骨骼肌发育过程中的调控网络机制。
    This study aims to explore the functional role of Myoz2 in myoblast differentiation, and elucidate the potential factors interact with Myoz2 in promoter transcriptional regulation. The temporal-spatial expression results showed that the bovine Myoz2 gene was highest expressed in longissimus dorsi, and in individual growth stages and myoblast differentiation stages. Knockdown of Myoz2 inhibited the differentiation of myoblast, and negative effect of MyoD, MyoG, MyH and MEF2A expression on mRNA levels. Subsequently, the promoter region of bovine Myoz2 gene with 1.7 Kb sequence was extracted, and then it was set as eight series of deleted fragments, which were ligated into pGL3-basic to detect core promoter regions of Myoz2 gene in myoblasts and myotubes. Transcription factors MyoD and MyoG were identified as important cis-acting elements in the core promoter region (-159/+1). Also, it was highly conserved in different species based on dual-luciferase analysis and multiple sequence alignment analysis, respectively. Furthermore, a chromatin immunoprecipitation (ChIP) analysis combined with site-directed mutation and siRNA interference and overexpression confirmed that the combination of MyoD and MyoG occurred in region -159/+1, and played an important role in the regulation of bovine Myoz2 gene. These findings explored the regulatory network mechanism of Myoz2 gene during the development of bovine skeletal muscle.
    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
    4-辛基衣康酸(OI)是一种细胞可渗透的衣康酸衍生物。研究表明,在系统性硬化症中具有抗纤维化作用,OI也影响破骨细胞分化。本研究的目的是通过RNA-seq分析探索OI对成肌细胞分化影响的分子机制。
    成肌细胞增殖,分化,在用各种浓度(2.5、10、25、50和100μmol/L)的OI处理的C2C12成肌细胞中检查肌肉调节因子。用PI3K-Akt激活剂IGF-1处理细胞以探索PI3K-Akt途径在肌源性分化的OI抑制中的作用。通过RNA-seq和随后的基因本体论(GO)进一步研究了OI在肌生成中的调控机制。京都基因和基因组百科全书(KEGG)和,基因集富集分析(GSEA)。
    各种浓度的OI在细胞增殖过程中没有显示任何作用。在分化过程中,OI以剂量依赖性方式抑制成熟肌管肌球蛋白重链(MHC)和肌原蛋白标记的表达。OI通过抑制AKT1磷酸化影响MyoD调节的活性来抑制肌肉分化。KEGG富集分析和GSEA结果表明,OI在成肌分化过程中影响了多种代谢途径,包括PI3K-Akt信号,钙信号,和PPAR信号。
    我们的研究拓宽了对肌源性分化的OI抑制的理解。OI通过靶向多种分子和途径发挥其功能,提供对OI整体效应的理解的新颖见解。
    The 4-octyl itaconate (OI) is a type of cell-permeable itaconate derivative. Studies have shown that with an anti-fibrotic effect in systemic sclerosis, the OI also affects osteoclast differentiation. The aim of this study was to explore the molecular mechanisms underlying the effects of OI on myoblast differentiation by RNA-seq analysis.
    Myoblast proliferation, differentiation, and muscle regulatory factors were examined in C2C12 myoblasts treated with OI of various concentrations (2.5, 10, 25, 50, and 100 μmol/L). Cells were treated with the PI3K-Akt activator IGF-1 to explore the role of the PI3K-Akt pathway in OI inhibition of myogenic differentiation. The regulatory mechanisms of OI in myogenesis were further investigated by RNA-seq and subsequent gene ontology (GO), kyoto encyclopedia of genes and genomes (KEGG) and, gene set enrichment analysis (GSEA).
    OI of various concentrations did not show any effect during cell proliferation. During differentiation, OI inhibited the expressions of the marker of mature myotubes myosin heavy chain (MHC) and myogenin in a dose-dependent manner. OI inhibited muscle differentiation by affecting MyoD-regulated activity through inhibition of AKT1 phosphorylation. The results of the KEGG enrichment analysis and GSEA showed that OI affected multiple metabolic pathways during myogenic differentiation, including PI3K-Akt signaling, calcium signaling, and PPAR signaling.
    Our study broadens the understanding of the OI inhibition of myogenic differentiation. OI plays its functions by targeting multiple molecules and pathways, providing novel insights into the understanding of the overall effect of OI.
    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
    关键转录因子的表达可以诱导体细胞转分化;然而,由于未定义的内在障碍,这种转换通常是不完整的。这里,我们使用MyoD诱导的成纤维细胞转分化作为模型来说明阻碍细胞命运转变的染色质结构。专注于三维(3D)染色质相互作用,我们证明MyoD直接建立染色质环来激活生肌转录程序。同样,CTCF介导的染色质相互作用的动态变化有利于成纤维细胞向成肌细胞的转化.然而,大部分CTCF介导的染色质相互作用保持稳定,并且相关基因稳定表达并富集成纤维细胞功能,这可能会限制细胞同一性转化。时间CTCF耗竭可以中断抗性染色质环,以增强小鼠的肌源性转分化,猪,和鸡的成纤维细胞。因此,在诱导转分化过程中,转录因子可以直接重组3D染色质相互作用,CTCF介导的基因组拓扑结构的扰动可以解决细胞命运转变的局限性。
    Expression of key transcription factors can induce transdifferentiation in somatic cells; however, this conversion is usually incomplete due to undefined intrinsic barriers. Here, we employ MyoD-induced transdifferentiation of fibroblasts as a model to illustrate the chromatin structures that impede the cell-fate transition. Focusing on the three-dimensional (3D) chromatin interactions, we show that MyoD directly establishes chromatin loops to activate myogenic transcriptional program. Similarly, dynamic changes of CTCF-mediated chromatin interactions are favorable for fibroblast-to-myoblast conversion. However, a substantial portion of CTCF-mediated chromatin interactions remain stable, and the associated genes are steady in expression and enriched for fibroblast function that may restrict cell-identity transformation. Temporal CTCF depletion can interrupt the resistant chromatin loops to enhance myogenic transdifferentiation in mice, pig, and chicken fibroblasts. Therefore, during induced transdifferentiation, the transcription factor can directly reorganize the 3D chromatin interactions, and perturbation of CTCF-mediated genome topology may resolve the limitations of cell fate transitions.
    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