Atoh1

Atoh1
  • 文章类型: Journal Article
    感觉神经性听力损失(SNHL)是耳鼻喉科的常见病。一个关键的障碍是找到再生成年动物受损耳蜗毛细胞的有效策略。已开发出一种实用可靠的方法,为内耳干细胞移植治疗SNHL创造了优越的细胞源。Atoh1参与神经元的分化,肠道分泌细胞,和包括听觉毛细胞在内的机械感受器,因此在神经发生中起着重要作用。慢病毒介导的骨髓间充质干细胞(BMSCs)的转染被用来实现必需转录因子Atoh1的稳定表达,这对于发育听觉毛细胞而不损害细胞存活至关重要。通过使用抗贴壁培养改变细胞生长环境来操纵诱导条件,碱性成纤维细胞生长因子(bFGF)和表皮生长因子(EGF)的协同作用可显著提高Atoh1转染后骨髓间充质干细胞(BMSC)向神经干细胞(NSC)的分化效率,从而减少诱导时间。研究表明,新提出的转分化方法在受控环境中有效地将BMSCs转化为NSCs,提供干细胞移植促进毛细胞再生的潜在途径。
    Sensorineural hearing loss (SNHL) is a prevalent condition in otolaryngology. A key obstacle is finding effective strategies for regenerating damaged cochlear hair cells in adult animals. A practical and reliable approach has been developed to create a superior cell source for stem cell transplantation in the inner ear to treat SNHL. Atoh1 is involved in the differentiation of neurons, intestinal secretory cells, and mechanoreceptors including auditory hair cells, and thus plays an important role in neurogenesis. Lentivirus-mediated transfection of bone marrow mesenchymal stem cells (BMSCs) was utilized to achieve stable expression of the essential transcription factor Atoh1, which is crucial for developing auditory hair cells without compromising cell survival. By manipulating the induction conditions through altering the cell growth environment using anti-adherent culture, the synergistic impact of basic fibroblast growth factor (bFGF) and epidermal growth factor (EGF) was effectively applied to significantly improve the differentiation efficiency of bone marrow-derived mesenchymal stem cells (BMSC) into neural stem cells (NSCs) following Atoh1 transfection, thereby reducing the induction time. The study indicated that the newly proposed transdifferentiation method effectively transformed BMSCs into NSCs in a controlled environment, presenting a potential approach for stem cell transplantation to promote hair cell regeneration.
    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
    耳蜗内毛细胞(IHC)是主要的声音受体,因此是开发听力障碍治疗的目标。体内IHC再生已被广泛尝试,虽然尚未在IHC损伤的耳蜗中。此外,新的IHC与野生型IHC相似的程度尚不清楚,新的IHC改善听力的能力也是如此。这里,我们开发了一种体内小鼠模型,其中野生型IHC被预损伤,非感觉支持细胞通过瞬时异位表达Atoh1和永久表达Tbx2转化为IHC.值得注意的是,新的IHC表达功能标记vGlut3,并呈现与野生型IHC相似的转录组和电生理特性。此外,新IHC的形成效率和成熟度高于先前报道的,虽然听力没有显著改善,至少部分是由于新IHC中的机电转换(MET)缺陷。因此,在受损的耳蜗中,我们已经成功地再生了在许多方面类似于野生型IHC的新IHC。我们的发现表明,有缺陷的MET是阻止听力恢复的关键障碍,因此应促进未来的IHC再生研究。
    Cochlear inner hair cells (IHCs) are primary sound receptors, and are therefore a target for developing treatments for hearing impairment. IHC regeneration in vivo has been widely attempted, although not yet in the IHC-damaged cochlea. Moreover, the extent to which new IHCs resemble wild-type IHCs remains unclear, as is the ability of new IHCs to improve hearing. Here, we have developed an in vivo mouse model wherein wild-type IHCs were pre-damaged and nonsensory supporting cells were transformed into IHCs by ectopically expressing Atoh1 transiently and Tbx2 permanently. Notably, the new IHCs expressed the functional marker vGlut3 and presented similar transcriptomic and electrophysiological properties to wild-type IHCs. Furthermore, the formation efficiency and maturity of new IHCs were higher than those previously reported, although marked hearing improvement was not achieved, at least partly due to defective mechanoelectrical transduction (MET) in new IHCs. Thus, we have successfully regenerated new IHCs resembling wild-type IHCs in many respects in the damaged cochlea. Our findings suggest that the defective MET is a critical barrier that prevents the restoration of hearing capacity and should thus facilitate future IHC regeneration 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
    Atoh1过表达对于哺乳动物听觉和前庭器官的感觉上皮中的毛细胞(HC)再生至关重要。然而,单独的Atoh1过表达不能在哺乳动物内耳中诱导完全成熟和有功能的HC。在目前的研究中,我们通过操纵Atoh1在不同发育阶段的表达来研究Atoh1组成型过表达在天然HC中的作用。我们证明,天然前庭HC中Atoh1的组成型过表达不会影响细胞存活,但会通过干扰HC的亚型分化和发束发育而损害前庭功能。相比之下,Atoh1在耳蜗HC中的过表达阻碍了它们的成熟,最终导致耳蜗HC逐渐丢失和听力功能障碍。我们的研究表明,时间限制的Atoh1表达对于内耳HCs的分化和存活至关重要,这对于通过Atoh1过表达诱导的HC再生策略重建听力和前庭功能至关重要。
    Atoh1 overexpression is essential for hair cell (HC) regeneration in the sensory epithelium of mammalian auditory and vestibular organs. However, Atoh1 overexpression alone cannot induce fully mature and functional HCs in the mammalian inner ear. In the current study, we investigated the effect of Atoh1 constitutive overexpression in native HCs by manipulating Atoh1 expression at different developmental stages. We demonstrated that constitutive overexpression of Atoh1 in native vestibular HCs did not affect cell survival but did impair vestibular function by interfering with the subtype differentiation of HCs and hair bundle development. In contrast, Atoh1 overexpression in cochlear HCs impeded their maturation, eventually leading to gradual HC loss in the cochlea and hearing dysfunction. Our study suggests that time-restricted Atoh1 expression is essential for the differentiation and survival of HCs in the inner ear, and this is pivotal for both hearing and vestibular function re-establishment through Atoh1 overexpression-induced HC regeneration strategies.
    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
    胃癌干细胞(GCSC)是一种自我更新的肿瘤细胞,可控制胃腺癌(GAC)的化学抗性,而它们的调节机制仍然难以捉摸。这里,本研究旨在阐明ATOH1在GCSCs维持中的作用.临床前模型和GAC样本分析表明,ATOH1缺乏与不良GAC预后和化疗耐药相关。ScRNA-seq显示,与癌旁样本相比,ATOH1在GAC的凹坑细胞中下调。谱系追踪表明,Atoh1缺失强烈赋予坑细胞干性。ATOH1耗竭显着加速了Tff1-CreERT2的癌症干性和化学抗性;Rosa26Tdtomato和Tff1-CreERT2;Apcfl/fl;p53fl/fl(TcPP)小鼠模型和类器官。ATOH1缺乏通过抑制GAS1启动子转录下调生长停滞特异性蛋白1(GAS1)。GAS1与RET形成一个复合体,抑制Tyr1062磷酸化,并因此通过ATOH1缺乏激活RET/AKT/mTOR信号通路。联合化疗和靶向AKT/mTOR信号的药物可以克服ATOH1缺乏诱导的化疗耐药。此外,证实DNA异常甲基化诱导ATOH1缺乏。一起来看,结果表明,ATOH1缺失通过GAC中的ATOH1/GAS1/RET/AKT/mTOR信号通路促进肿瘤干性,从而为AKT/mTOR抑制剂治疗ATOH1缺乏症的GAC患者提供了一个潜在的治疗策略.
    Gastric cancer stem cells (GCSCs) are self-renewing tumor cells that govern chemoresistance in gastric adenocarcinoma (GAC), whereas their regulatory mechanisms remain elusive. Here, the study aims to elucidate the role of ATOH1 in the maintenance of GCSCs. The preclinical model and GAC sample analysis indicate that ATOH1 deficiency is correlated with poor GAC prognosis and chemoresistance. ScRNA-seq reveals that ATOH1 is downregulated in the pit cells of GAC compared with those in paracarcinoma samples. Lineage tracing reveals that Atoh1 deletion strongly confers pit cell stemness. ATOH1 depletion significantly accelerates cancer stemness and chemoresistance in Tff1-CreERT2; Rosa26Tdtomato and Tff1-CreERT2; Apcfl/fl ; p53fl/fl (TcPP) mouse models and organoids. ATOH1 deficiency downregulates growth arrest-specific protein 1 (GAS1) by suppressing GAS1 promoter transcription. GAS1 forms a complex with RET, which inhibits Tyr1062 phosphorylation, and consequently activates the RET/AKT/mTOR signaling pathway by ATOH1 deficiency. Combining chemotherapy with drugs targeting AKT/mTOR signaling can overcome ATOH1 deficiency-induced chemoresistance. Moreover, it is confirmed that abnormal DNA hypermethylation induces ATOH1 deficiency. Taken together, the results demonstrate that ATOH1 loss promotes cancer stemness through the ATOH1/GAS1/RET/AKT/mTOR signaling pathway in GAC, thus providing a potential therapeutic strategy for AKT/mTOR inhibitors in GAC patients with ATOH1 deficiency.
    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
    感觉神经性听力损失通常是由于外部刺激或遗传因素以及无法将声音机械能转化为神经冲动对耳蜗毛细胞(HCs)的损害引起的。成年哺乳动物耳蜗HC不能自发再生;因此,这种类型的耳聋通常被认为是不可逆转的。对HC分化发育机制的研究表明,耳蜗中的非感觉细胞在过表达特定基因后获得分化为HC的能力,例如Atoh1,这使得HC再生成为可能。基因治疗,通过体外选择和编辑靶基因,将外源基因片段转化到靶细胞中,并改变靶细胞中基因的表达,以激活靶细胞中相应的分化发育程序。本文综述了近年来与耳蜗HCs生长发育相关的基因,并对HC再生领域的基因治疗方法进行了综述。最后讨论了当前治疗方法的局限性,以促进该疗法在临床环境中的早期实施。
    Sensorineural hearing loss is typically caused by damage to the cochlear hair cells (HCs) due to external stimuli or because of one\'s genetic factors and the inability to convert sound mechanical energy into nerve impulses. Adult mammalian cochlear HCs cannot regenerate spontaneously; therefore, this type of deafness is usually considered irreversible. Studies on the developmental mechanisms of HC differentiation have revealed that nonsensory cells in the cochlea acquire the ability to differentiate into HCs after the overexpression of specific genes, such as Atoh1, which makes HC regeneration possible. Gene therapy, through in vitro selection and editing of target genes, transforms exogenous gene fragments into target cells and alters the expression of genes in target cells to activate the corresponding differentiation developmental program in target cells. This review summarizes the genes that have been associated with the growth and development of cochlear HCs in recent years and provides an overview of gene therapy approaches in the field of HC regeneration. It concludes with a discussion of the limitations of the current therapeutic approaches to facilitate the early implementation of this therapy in a clinical setting.
    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
    脊髓发育是由时空基因调控程序精确协调的。然而,潜在的表观遗传机制在很大程度上仍然难以捉摸。这里,我们分析了小鼠神经管中跨胚胎9.5-13.5天的单细胞染色质可及性景观。我们在神经祖细胞和神经元中鉴定了神经元细胞簇特异性顺式调节元件。此外,我们应用了一种新的计算方法,eNet,通过整合单细胞染色质可及性和基因表达数据来构建增强子网络,并识别增强子网络中的中心增强子。Atoh1的体内实验验证了hub增强子的敲除,但不是非中枢增强剂,Atoh1表达明显降低,dp1/dI1细胞减少。一起,我们的工作提供了对脊髓发育的表观遗传调控的见解,以及增强子网络作为转录调控的一般机制的概念验证.
    Spinal cord development is precisely orchestrated by spatiotemporal gene regulatory programs. However, the underlying epigenetic mechanisms remain largely elusive. Here, we profiled single-cell chromatin accessibility landscapes in mouse neural tubes spanning embryonic days 9.5-13.5. We identified neuronal-cell-cluster-specific cis-regulatory elements in neural progenitors and neurons. Furthermore, we applied a novel computational method, eNet, to build enhancer networks by integrating single-cell chromatin accessibility and gene expression data and identify the hub enhancers within enhancer networks. It was experimentally validated in vivo for Atoh1 that knockout of the hub enhancers, but not the non-hub enhancers, markedly decreased Atoh1 expression and reduced dp1/dI1 cells. Together, our work provides insights into the epigenetic regulation of spinal cord development and a proof-of-concept demonstration of enhancer networks as a general mechanism in transcriptional regulation.
    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
    内耳中的耳蜗毛细胞(HCs)负责声音检测。对于HC命运规范,主转录因子Atoh1是必要和充分的。Atoh1的表达是动态的,在发育过程中受到严格调控,但是调解这一规定的顺式监管因素仍未解决。出乎意料的是,我们发现删除唯一识别的Atoh1增强子,此处定义为Eh1,未能损害HC的发展。通过使用高通量测序(ATAC-seq)的转座酶可接近的染色质测定法,我们发现了另外两种Atoh1增强剂:Eh2和Eh3。值得注意的是,Eh2缺失足以损害HC发育,Eh1和Eh2或所有三种增强子的同时缺失导致HCs几乎完全缺失。最后,我们表明Atoh1与所有三种增强剂结合,与其自动调节功能一致。我们的发现表明,在HC发育过程中,三种不同增强剂的协同作用支持有效的Atoh1调节,表明HC再生的潜在治疗方法。
    Cochlear hair cells (HCs) in the inner ear are responsible for sound detection. For HC fate specification, the master transcription factor Atoh1 is both necessary and sufficient. Atoh1 expression is dynamic and tightly regulated during development, but the cis-regulatory elements mediating this regulation remain unresolved. Unexpectedly, we found that deleting the only recognized Atoh1 enhancer, defined here as Eh1, failed to impair HC development. By using the assay for transposase-accessible chromatin with high-throughput sequencing (ATAC-seq), we discovered two additional Atoh1 enhancers: Eh2 and Eh3. Notably, Eh2 deletion was sufficient for impairing HC development, and concurrent deletion of Eh1 and Eh2 or all three enhancers resulted in nearly complete absence of HCs. Lastly, we showed that Atoh1 binds to all three enhancers, consistent with its autoregulatory function. Our findings reveal that the cooperative action of three distinct enhancers underpins effective Atoh1 regulation during HC development, indicating potential therapeutic approaches for HC regeneration.
    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:结肠腺癌(COAD)是当今威胁人类健康的主要恶性肿瘤之一。免疫检查点抑制剂(ICIs)最近开始成为治疗COAD患者的有效选择。但并非所有患者都能从ICI治疗中获益。先前的研究表明,ICI对微卫星不稳定性高(MSI-H)患者具有显着的临床效果,而相反,微卫星稳定/微卫星不稳定性低(MSS/MSI-L)患者的反应有限。
    方法:我们使用ATAC-seq,RNA-seq,和来自癌症基因组图谱结肠腺癌(TCGA-COAD)队列的突变数据,以对具有不同MSI状态的COAD样品进行多组学差异分析,然后通过将这些结果与生存分析相结合来进一步筛选基因。我们分析了筛选的基因对COAD患者肿瘤微环境和免疫原性的影响,并随后使用一系列预测指标确定其对COAD患者ICIs疗效的影响。
    结果:在TCGA-COAD队列中筛选了12个基因,合并生存分析后,我们确定ATOH1具有显著作用.ATOH1的特点是高染色质可及性,高表达,MSI-H组中COAD患者的高突变。ATOH1高表达的COAD患者预后较好,独特的免疫微环境,在ICI治疗中的疗效较高。富集分析显示,ATOH1高表达的COAD患者体液免疫及其他相关通路显著上调。
    结论:我们推测ATOH1可能通过影响肿瘤的免疫微环境和免疫原性来影响COAD患者ICIs治疗的疗效。
    BACKGROUND: Colon adenocarcinoma (COAD) is one of the major varieties of malignant tumors threatening human health today. Immune checkpoint inhibitors (ICIs) have recently begun to emerge as an effective option for the treatment of COAD patients, but not all patients can benefit from ICI treatment. Previous studies have suggested that ICIs boast significant clinical effects on patients with microsatellite instability-high (MSI-H), while conversely patients with microsatellite-stable/microsatellite instability-low (MSS/MSI-L) have shown limited response.
    METHODS: We used ATAC-seq, RNA-seq, and mutation data from The Cancer Genome Atlas Colon adenocarcinoma (TCGA-COAD) cohort to perform multi-omics differential analysis on COAD samples with different MSI statuses, then further screened genes by additionally combining these results with survival analysis. We analyzed the effects of the screened genes on the tumor microenvironment and immunogenicity of COAD patients, and subsequently determined their influence on the efficacy of ICIs in COAD patients using a series of predictive indexes.
    RESULTS: Twelve genes were screened in the TCGA-COAD cohort, and after the combined survival analysis, we identified ATOH1 as having significant effects. ATOH1 is characterized by high chromatin accessibility, high expression, and high mutation in COAD patients in the MSI-H group. COAD patients with high ATOH1 expression are associated with a better prognosis, unique immune microenvironment, and higher efficacy in ICI treatment. Enrichment analysis showed that COAD patients with high ATOH1 expression displayed significant upregulation in their humoral immunity and other related pathways.
    CONCLUSIONS: We speculate that ATOH1 may influence the efficacy of ICIs therapy in patients with COAD by affecting the immune microenvironment and immunogenicity of the tumor.
    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
    内耳毛细胞的丧失导致哺乳动物不可逆的声损伤,和再生内耳毛细胞恢复听力损失是具有挑战性的。ATOH1是毛细胞发育和再生的症结基因。小激活RNA(saRNA)可以靶向基因以特异性上调其表达。本研究旨在探讨小活化RNA是否能在体外联合生长因子诱导人脂肪间充质干细胞分化为毛细胞样细胞,从而为毛细胞再生和感音神经性耳聋的治疗提供新的策略。在293T和人脂肪间充质干细胞中设计并筛选了15种靶向人ATOH1基因的小活化RNA,发现这些候选物中的3个能够有效且稳定地激活ATOH1基因表达。然后将选择的小活化RNA转染到毛细胞祖细胞中,转染后10天检查毛细胞标记物。转染选定的小活化RNA后,内耳毛细胞特征性标志物的表达,POU类4homeobox3(POU4F3)和肌球蛋白VIIA(MYO7A),被检测到。人脂肪间充质干细胞具有向人毛细胞祖细胞分化的潜能。体外,小活化RNA能够诱导毛细胞祖细胞分化为毛细胞样细胞.因此,RNA激活技术有潜力为毛细胞的再生提供新的策略。
    The loss of inner ear hair cells leads to irreversible acoustic injury in mammals, and regeneration of inner ear hair cells to restore hearing loss is challenging. ATOH1 is a key gene in the development and regeneration of hair cells. Small activating RNAs (saRNAs) can target a gene to specifically upregulate its expression. This study aimed to explore whether small activating RNAs could induce the differentiation of human adipose-derived mesenchymal stem cells into hair cell-like cells with a combination of growth factors in vitro and thus provide a new strategy for hair cell regeneration and the treatment of sensorineural hearing loss. Fifteen small activating RNAs targeting the human ATOH1 gene were designed and screened in 293 T and human adipose-derived mesenchymal stem cells, and 3 of these candidates were found to be capable of effectively and stably activating ATOH1 gene expression. The selected small activating RNAs were then transfected into hair cell progenitor cells, and hair cell markers were examined 10 days after transfection. After transfection of the selected small activating RNAs, the expression of the characteristic markers of inner ear hair cells, POU class 4 homeobox 3 (POU4F3) and myosin VIIA (MYO7A), was detected. Human adipose-derived mesenchymal stem cells have the potential to differentiate into human hair cell progenitor cells. In vitro, small activating RNAs were able to induce the differentiation of hair cell progenitor cells into hair cell-like cells. Therefore, RNA activation technology has the potential to provide a new strategy for the regeneration of hair cells.
    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
    Atoh1被认为是耳蜗毛细胞(HC)发育所必需的。然而,Atoh1的时间和空间表达模式仍然存在广泛争议。这里,我们旨在通过产生新的敲入小鼠品系Atoh13*HA-P2A-Cre来获得证据以解决有关Atoh1表达的争议。
    Atoh13*HA-P2A-Cre/;Rosa26-CAG-LSL-tdTomato(Ai9)/小鼠的命运定位分析使我们能够同时表征Atoh1蛋白的时间表达(通过HA标签免疫染色)并可视化表达Atoh1mRNA的细胞(作为tdTomato细胞)。我们的发现表明,尽管Atoh1mRNA表达在早期耳蜗祖细胞中迅速开启,Atoh1蛋白仅在分化HC或仅致力于HC命运的祖细胞中检测到。Atoh13*HA-P2A-Cre/+中的Cre活性也比以前的小鼠模型更强,因为这里几乎所有的耳蜗HC和附近的支持细胞都是tdTomato+。此外,tdTomato,但不是HA,在中部和顶端螺旋神经节神经元中表达。
    集体,我们的发现表明,Atoh13*HA-P2A-Cre可以作为发育生物学领域的一个强大的遗传模型。
    Atoh1 is recognized to be essential for cochlear hair cell (HC) development. However, Atoh1 temporal and spatial expression patterns remain widely debated. Here, we aimed to obtain evidence to resolve the controversies regarding Atoh1 expression by generating a new knockin mouse strain: Atoh13*HA-P2A-Cre .
    Fate-mapping analysis of Atoh13*HA-P2A-Cre/+ ; Rosa26-CAG-LSL-tdTomato (Ai9)/+ mice enabled us to concurrently characterize the temporal expression of Atoh1 protein (through HA-tag immunostaining) and visualize the cells expressing Atoh1 mRNA (as tdTomato+ cells). Our findings show that whereas Atoh1 mRNA expression is rapidly turned on in early cochlear progenitors, Atoh1 protein is only detected in differentiating HCs or progenitors just committed to the HC fate. Cre activity is also stronger in Atoh13*HA-P2A-Cre/+ than in previous mouse models, because almost all cochlear HCs and nearby supporting cells here are tdTomato+. Furthermore, tdTomato, but not HA, is expressed in middle and apical spiral ganglion neurons.
    Collectively, our findings indicate that Atoh13*HA-P2A-Cre can serve as a powerful genetic model in the developmental biology field.
    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