Hair cell regeneration

毛细胞再生
  • 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
    哺乳动物耳蜗毛细胞(HCs)对听力至关重要,对HCs的损害会导致严重的听力损害。受损的HC可以通过邻近的支持细胞(SCs)再生,因此,HCs的功能再生是恢复体内听觉功能的主要目标。这里,通过诱导关键转录因子Atoh1及其共调节因子Gfi1,Pou4f3和Six1(GPAS)的表达,耳蜗SC转分化为外部和内部HC,成功实现了通过AAV-ie靶向内耳干细胞而注定用于HC发育和成熟的SC所必需的。单细胞核测序和谱系追踪结果显示,大多数新Atoh1来源的HCs处于起始分化状态,而GP(Gfi1,Pou4f3)和GPS(Gfi1,Pou4f3和Six1)将Atoh1诱导的新HC增强为内部和外部HC。此外,膜片钳分析表明,由GPAS强制表达诱导的新生内层HCs具有与天然内层HCs相似的电生理特征.此外,在HC损伤小鼠模型中,GPAS可以诱导HC再生。总之,这项研究表明,AAV介导的多基因共调控,例如GPAS,是实现小鼠耳蜗HC功能再生的有效手段。
    Mammalian cochlear hair cells (HCs) are essential for hearing, and damage to HCs results in severe hearing impairment. Damaged HCs can be regenerated by neighboring supporting cells (SCs), thus the functional regeneration of HCs is the main goal for the restoration of auditory function in vivo. Here, cochlear SC trans-differentiation into outer and inner HC by the induced expression of the key transcription factors Atoh1 and its co-regulators Gfi1, Pou4f3, and Six1 (GPAS), which are necessary for SCs that are destined for HC development and maturation via the AAV-ie targeting the inner ear stem cells are successfully achieved. Single-cell nuclear sequencing and lineaging tracing results showed that the majority of new Atoh1-derived HCs are in a state of initiating differentiation, while GP (Gfi1, Pou4f3) and GPS (Gfi1, Pou4f3, and Six1) enhanced the Atoh1-induced new HCs into inner and outer HCs. Moreover, the patch-clamp analysis indicated that newborn inner HCs induced by GPAS forced expression have similar electrophysiological characteristics to those of native inner HCs. Also, GPAS can induce HC regeneration in the HC-damaged mice model. In summary, the study demonstrates that AAV-mediated co-regulation of multiple genes, such as GPAS, is an effective means to achieve functional HC regeneration in the mouse cochlea.
    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
    听证会开始,在细胞水平上,通过感觉毛细胞进行机电传导。然后通过与听觉神经元的传入突触连接来传输声音信息。频率信息由沿着耳蜗导管的毛细胞的位置编码。毛细胞的损失,突触,或听觉神经元导致哺乳动物永久性听力损失。鸟,相比之下,再生听觉毛细胞并从听力损失中恢复功能。这里,我们对sisomicin耳聋鸡的再生和神经支配进行了表征,发现传入神经元在基底突起的尖端接触再生毛细胞。与发展相比,突触专业化是在远离毛细胞\'身体的这些位置建立的。然后,当再生毛细胞成熟并在耳聋后2周变得有功能时,突起收缩。我们发现听觉阈值在4-5周后恢复。我们将再生特异性突触重建解释为保持定位保真度可能需要的位置保留过程。
    Hearing starts, at the cellular level, with mechanoelectrical transduction by sensory hair cells. Sound information is then transmitted via afferent synaptic connections with auditory neurons. Frequency information is encoded by the location of hair cells along the cochlear duct. Loss of hair cells, synapses, or auditory neurons leads to permanent hearing loss in mammals. Birds, in contrast, regenerate auditory hair cells and functionally recover from hearing loss. Here, we characterized regeneration and reinnervation in sisomicin-deafened chickens and found that afferent neurons contact regenerated hair cells at the tips of basal projections. In contrast to development, synaptic specializations are established at these locations distant from the hair cells\' bodies. The protrusions then contracted as regenerated hair cells matured and became functional 2 weeks post-deafening. We found that auditory thresholds recovered after 4-5 weeks. We interpret the regeneration-specific synaptic reestablishment as a location-preserving process that might be needed to maintain tonotopic fidelity.
    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
    巢蛋白表达与多能性相关。越来越多的证据表明巢蛋白参与毛细胞发育。这项研究的目的是研究出生后早期鼠内耳中巢蛋白表达细胞的形态和作用。使用谱系追踪巢蛋白报告小鼠系来进一步表征这些细胞。对他们的耳蜗和前庭器官进行免疫染色,并进行整体安装以进行细胞计数。我们发现在整个内耳中存在少量Nestin表达细胞。观察到三种形态类型:双极,单极,和球状。在耳蜗中的巢蛋白表达细胞中观察到有丝分裂活性,utricle,囊,还有Crista.然后在两个小鼠模型中去除毛细胞后观察巢蛋白表达细胞特征。首先,报告模型显示,消融毛细胞后,nestin在毛细胞中的表达比例明显高于对照耳蜗。然而,在追踪巢蛋白记者老鼠的血统中,在毛细胞消融后重新填充Corti器官的新毛细胞均未表达巢蛋白,巢蛋白表达细胞的形态也没有改变。总之,在耳蜗和前庭器官中鉴定了巢蛋白表达细胞。毛细胞消融后,巢蛋白表达细胞对损伤没有反应。然而,所有内耳组织中少量的巢蛋白表达细胞表现出有丝分裂活性,支持祖细胞潜能,虽然可能不参与毛细胞再生。
    Nestin expression is associated with pluripotency. Growing evidence suggests nestin is involved in hair cell development. The objective of this study was to investigate the morphology and role of nestin-expressing cells residing in the early postnatal murine inner ear. A lineage-tracing nestin reporter mouse line was used to further characterize these cells. Their cochleae and vestibular organs were immunostained and whole-mounted for cell counting. We found Nestin-expressing cells present in low numbers throughout the inner ear. Three morphotypes were observed: bipolar, unipolar, and globular. Mitotic activity was noted in nestin-expressing cells in the cochlea, utricle, saccule, and crista. Nestin-expressing cell characteristics were then observed after hair cell ablation in two mouse models. First, a reporter model demonstrated nestin expression in a significantly higher proportion of hair cells after hair cell ablation than in control cochleae. However, in a lineage tracing nestin reporter mouse, none of the new hair cells which repopulated the organ of Corti after hair cell ablation expressed nestin, nor did the nestin-expressing cells change in morphotype. In conclusion, Nestin-expressing cells were identified in the cochlea and vestibular organs. After hair cell ablation, nestin-expressing cells did not react to the insult. However, a small number of nestin-expressing cells in all inner ear tissues exhibited mitotic activity, supporting progenitor cell potential, though perhaps not involved in 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
    耳蜗植入学的最新进展通过新颖的功能策略例如双峰电声刺激,其中患者在术前具有完整的低频听力和深刻的高频听力。因此,功能失调的耳蜗毛细胞的协同恢复和保护毛细胞免受耳毒性损伤已成为这种混合系统追求的持久目标。在这项研究中,我们开发了一种复合GelMA/PEDOT:PSS导电水凝胶,适合作为耳蜗植入电极的涂层,用于潜在的局部递送耳再生和耳保护药物。各种材料表征方法(例如,1HNMR光谱,FT-IR,EIS,和SEM),实验模型(例如,鼠耳蜗类器官和氨基糖苷诱导的耳毒性HEI-OC1细胞模型),和生物学分析(例如,共聚焦激光扫描显微镜,实时qPCR,流式细胞术,和生物信息学测序)。结果表明,水凝胶的材料性能良好,如机械(例如,高拉伸应力和杨氏模量),电化学(例如,低阻抗和高电导率),生物相容性(例如,令人满意的耳蜗细胞相互作用和无全身毒性),和生物安全(例如,最小溶血和细胞死亡)特征。此外,由水凝胶可持续释放的CDR药物混合物不仅促进了耳蜗干细胞的扩增,而且促进了从耳蜗支持细胞向毛细胞的转分化。此外,基于水凝胶的药物递送保护毛细胞免受氧化应激和各种形式的程序性细胞死亡(例如,凋亡和铁凋亡)。最后,使用大规模测序,我们丰富了一个复杂的信号通路网络,这些信号通路可能是各种代谢过程和丰富代谢产物的下游.总之,我们提出了一种基于导电水凝胶的双功能药物混合物的局部递送,从而作为双峰听觉康复和超越疾病的耳蜗内治疗的潜在解决方案。
    Recent advances in cochlear implantology are exemplified by novel functional strategies such as bimodal electroacoustic stimulation, in which the patient has intact low-frequency hearing and profound high-frequency hearing pre-operatively. Therefore, the synergistic restoration of dysfunctional cochlear hair cells and the protection of hair cells from ototoxic insults have become a persistent target pursued for this hybrid system. In this study, we developed a composite GelMA/PEDOT:PSS conductive hydrogel that is suitable as a coating for the cochlear implant electrode for the potential local delivery of otoregenerative and otoprotective drugs. Various material characterization methods (e.g., 1H NMR spectroscopy, FT-IR, EIS, and SEM), experimental models (e.g., murine cochlear organoid and aminoglycoside-induced ototoxic HEI-OC1 cellular model), and biological analyses (e.g., confocal laser scanning microscopy, real time qPCR, flow cytometry, and bioinformatic sequencing) were used. The results demonstrated decent material properties of the hydrogel, such as mechanical (e.g., high tensile stress and Young\'s modulus), electrochemical (e.g., low impedance and high conductivity), biocompatibility (e.g., satisfactory cochlear cell interaction and free of systemic toxicity), and biosafety (e.g., minimal hemolysis and cell death) features. In addition, the CDR medicinal cocktail sustainably released by the hydrogel not only promoted the expansion of the cochlear stem cells but also boosted the trans-differentiation from cochlear supporting cells into hair cells. Furthermore, hydrogel-based drug delivery protected the hair cells from oxidative stress and various forms of programmed cell death (e.g., apoptosis and ferroptosis). Finally, using large-scale sequencing, we enriched a complex network of signaling pathways that are potentially downstream to various metabolic processes and abundant metabolites. In conclusion, we present a conductive hydrogel-based local delivery of bifunctional drug cocktails, thereby serving as a potential solution to intracochlear therapy of bimodal auditory rehabilitation and diseases beyond.
    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
    扁平上皮(FE)的特征是毛细胞(HC)和支持细胞的丢失以及Corti器官转化为简单的扁平或立方体上皮,这可能发生在严重的耳蜗侮辱之后。转录因子Gfi1,Atoh1,Pou4f3和Six1(GAPS)在正常耳的HC分化和存活中起关键作用。以前使用单一转录因子的工作,Atoh1,在体内诱导成熟耳朵中的HC再生通常产生很少的细胞,并且在Corti的严重受损器官中未能产生HC,尤其是那些有FE的。体外研究表明,转录因子的组合可能比任何单一因子都更有效。因此,本研究旨在研究GAPS基因共同过表达在耳聋的成熟豚鼠耳蜗中的作用。耳聋是通过将新霉素注入外淋巴来实现的,导致FE的形成和神经纤维的大量变性。新霉素治疗后七天,将携带GAPS的腺病毒载体注射到scala培养基中,并在FE中成功表达。接种GAPS后一两个月,在FE下的区域(位于基底膜的鼓阶侧)观察到表达肌球蛋白VIIa的细胞,而不是在FE内。细胞的数量,我们将其定义为诱导性HCs(iHCs),在一两个月之间没有显着差异,但是在两个月时更大的N使GAPS治疗的动物中的iHCs明显多于对照组。此外,定性观察表明,未经治疗,FE中GAPS基因表达的耳朵比FE有更多的神经纤维。总之,我们的结果表明,GAPS的共同过表达增强了FE严重病变模型中HC再生的潜力.
    Flat epithelium (FE) is a condition characterized by the loss of both hair cells (HCs) and supporting cells and the transformation of the organ of Corti into a simple flat or cuboidal epithelium, which can occur after severe cochlear insults. The transcription factors Gfi1, Atoh1, Pou4f3, and Six1 (GAPS) play key roles in HC differentiation and survival in normal ears. Previous work using a single transcription factor, Atoh1, to induce HC regeneration in mature ears in vivo usually produced very few cells and failed to produce HCs in severely damaged organs of Corti, especially those with FE. Studies in vitro suggested combinations of transcription factors may be more effective than any single factor, thus the current study aims to examine the effect of co-overexpressing GAPS genes in deafened mature guinea pig cochleae with FE. Deafening was achieved through the infusion of neomycin into the perilymph, leading to the formation of FE and substantial degeneration of nerve fibers. Seven days post neomycin treatment, adenovirus vectors carrying GAPS were injected into the scala media and successfully expressed in the FE. One or two months following GAPS inoculation, cells expressing Myosin VIIa were observed in regions under the FE (located at the scala tympani side of the basilar membrane), rather than within the FE. The number of cells, which we define as induced HCs (iHCs), was not significantly different between one and two months, but the larger N at two months made it more apparent that there were significantly more iHCs in GAPS treated animals than in controls. Additionally, qualitative observations indicated that ears with GAPS gene expression in the FE had more nerve fibers than FE without the treatment. In summary, our results showed that co-overexpression of GAPS enhances the potential for HC regeneration in a severe lesion model of FE.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Editorial
    暂无摘要。
    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
    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
    听力损失变得越来越普遍,并导致相当大的残疾,从而给全球经济带来沉重负担。毛细胞的不可逆损失是感音神经性听力损失的主要原因,目前,唯一相对有效的临床治疗仅限于数字听力设备,如人工耳蜗和助听器,但这些对患者的益处有限。因此,迫切需要了解损伤修复的机制,以开发新的神经保护策略。目前,如何促进功能性毛细胞的再生是听力研究领域的一个关键科学问题。多种信号通路和转录因子触发毛细胞祖细胞的激活,并确保新生毛细胞的成熟,在这篇文章中,我们首先回顾了毛细胞繁殖的主要机制。然后,我们进一步讨论涉及多个信号通路共同调节的治疗策略,以诱导变性后有效的功能性毛细胞再生。我们总结了目前在毛细胞再生方面的成就。最后,我们讨论潜在的未来方法,如小分子药物和基因治疗,这可能是应用于再生功能性毛细胞在临床上。
    Hearing loss has become increasingly prevalent and causes considerable disability, thus gravely burdening the global economy. Irreversible loss of hair cells is a main cause of sensorineural hearing loss, and currently, the only relatively effective clinical treatments are limited to digital hearing equipment like cochlear implants and hearing aids, but these are of limited benefit in patients. It is therefore urgent to understand the mechanisms of damage repair in order to develop new neuroprotective strategies. At present, how to promote the regeneration of functional hair cells is a key scientific question in the field of hearing research. Multiple signaling pathways and transcriptional factors trigger the activation of hair cell progenitors and ensure the maturation of newborn hair cells, and in this article, we first review the principal mechanisms underlying hair cell reproduction. We then further discuss therapeutic strategies involving the co-regulation of multiple signaling pathways in order to induce effective functional hair cell regeneration after degeneration, and we summarize current achievements in hair cell regeneration. Lastly, we discuss potential future approaches, such as small molecule drugs and gene therapy, which might be applied for regenerating functional hair cells in the clinic.
    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
    耳蜗毛细胞损失是人类耳聋的主要原因。相邻的支持细胞有一些再生毛细胞的能力。然而,随着支持细胞的成熟(小鼠出生后第5天),它们的再生潜力急剧下降。我们最近报道,RNA结合蛋白LIN28B的再激活可恢复P5耳蜗支持细胞的毛细胞再生潜能。这里,我们将LIN28B靶标Trim71鉴定为支持细胞可塑性的新型且同样有效的增强子。TRIM71是干细胞行为和细胞重编程的关键调节因子;然而,它在细胞再生中的作用知之甚少。采用基于类器官的检测方法,我们显示TRIM71的再表达通过促进P5耳蜗支持细胞去分化为祖细胞样细胞而增加了其有丝分裂和毛细胞形成潜力。我们的机械工作表明,TRIM71的RNA结合活性对于这种能力至关重要,我们的转录组学分析确定了与TRIM71和LIN28B介导的支持细胞重编程相关的基因模块。此外,我们的研究发现TRIM71-LIN28B靶标Hmga2对于支持细胞自我更新和毛细胞形成至关重要.
    Cochlear hair cell loss is a leading cause of deafness in humans. Neighboring supporting cells have some capacity to regenerate hair cells. However, their regenerative potential sharply declines as supporting cells undergo maturation (postnatal day 5 in mice). We recently reported that reactivation of the RNA-binding protein LIN28B restores the hair cell-regenerative potential of P5 cochlear supporting cells. Here, we identify the LIN28B target Trim71 as a novel and equally potent enhancer of supporting cell plasticity. TRIM71 is a critical regulator of stem cell behavior and cell reprogramming; however, its role in cell regeneration is poorly understood. Employing an organoid-based assay, we show that TRIM71 re-expression increases the mitotic and hair cell-forming potential of P5 cochlear supporting cells by facilitating their de-differentiation into progenitor-like cells. Our mechanistic work indicates that TRIM71\'s RNA-binding activity is essential for such ability, and our transcriptomic analysis identifies gene modules that are linked to TRIM71 and LIN28B-mediated supporting cell reprogramming. Furthermore, our study uncovers that the TRIM71-LIN28B target Hmga2 is essential for supporting cell self-renewal and hair cell formation.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号