RGCs

RGC
  • 文章类型: Journal Article
    常染色体显性视神经萎缩(ADOA)是一种罕见的进行性疾病,主要由OPA1突变引起,OPA1是一种编码线粒体蛋白的核基因,在线粒体动力学中起着至关重要的作用。细胞存活,氧化磷酸化,和mtDNA维护。ADOA的特征在于视网膜神经节细胞(RGCs)的变性。这会导致视力丧失,这在许多情况下可能导致法律失明。如今,ADOA没有有效的治疗方法。在这篇文章中,我们使用iPSC技术和基因组编辑工具CRISPR/Cas9从先前生成的具有致病性变异NM_015560.3:c.1861C>T的ADOA加患者的iPSC细胞系中建立了ADOA的等基因人类RGC模型(p.Gln621Ter)在OPA1中的杂合。为此,已经采用了基于向iPSC培养基补充几种小分子和试图模拟胚胎发育的定义因子的方案。随后,创建的模型经过验证,确认基因组间通信缺陷的存在,线粒体呼吸受损,以及细胞凋亡和ROS产生的增加。最后,我们建议通过qPCR分析OPA1表达作为一种简单的读出方法,用于使用创建的RGC模型进行未来的药物筛选研究。总之,该模型为进一步研究ADOAplus的潜在病理生理机制以及测试具有潜在药理作用的化合物提供了有用的平台.
    Autosomal dominant optic atrophy (ADOA) is a rare progressive disease mainly caused by mutations in OPA1, a nuclear gene encoding for a mitochondrial protein that plays an essential role in mitochondrial dynamics, cell survival, oxidative phosphorylation, and mtDNA maintenance. ADOA is characterized by the degeneration of retinal ganglion cells (RGCs). This causes visual loss, which can lead to legal blindness in many cases. Nowadays, there is no effective treatment for ADOA. In this article, we have established an isogenic human RGC model for ADOA using iPSC technology and the genome editing tool CRISPR/Cas9 from a previously generated iPSC line of an ADOA plus patient harboring the pathogenic variant NM_015560.3: c.1861C>T (p.Gln621Ter) in heterozygosis in OPA1. To this end, a protocol based on supplementing the iPSC culture media with several small molecules and defined factors trying to mimic embryonic development has been employed. Subsequently, the created model was validated, confirming the presence of a defect of intergenomic communication, impaired mitochondrial respiration, and an increase in apoptosis and ROS generation. Finally, we propose the analysis of OPA1 expression by qPCR as an easy read-out method to carry out future drug screening studies using the created RGC model. In summary, this model provides a useful platform for further investigation of the underlying pathophysiological mechanisms of ADOA plus and for testing compounds with potential pharmacological action.
    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
    遗传性视神经病变(HON)如显性视神经萎缩(DOA)和Leber遗传性视神经病变(LHON)是线粒体疾病,其特征在于视网膜神经节细胞(RGC)的退行性丧失,并且是世界范围内失明的原因。迄今为止,这些疾病只有有限的改善疾病的治疗方法。诱导多能干细胞(iPSC)技术的发现在HON研究和寻找治疗方法领域开辟了几个有希望的机会。本系统综述集中于两种最常见的HON(LHON和DOA),以及与人类iPSC技术与生物材料技术结合应用相关的最新研究,这些研究在RGC替代疗法的开发中具有潜在用途,最终目的是改善甚至恢复HON患者的视力。为此,用肽修饰的天然和合成生物材料的组合,神经营养因子,和其他中低分子量化合物,模仿眼部细胞外基质,人类iPSC或iPSC衍生的细胞视网膜祖细胞具有巨大的潜力,可以在不久的将来开发可移植的RGC群体。
    Hereditary optic neuropathies (HONs) such as dominant optic atrophy (DOA) and Leber Hereditary Optic Neuropathy (LHON) are mitochondrial diseases characterized by a degenerative loss of retinal ganglion cells (RGCs) and are a cause of blindness worldwide. To date, there are only limited disease-modifying treatments for these disorders. The discovery of induced pluripotent stem cell (iPSC) technology has opened several promising opportunities in the field of HON research and the search for therapeutic approaches. This systematic review is focused on the two most frequent HONs (LHON and DOA) and on the recent studies related to the application of human iPSC technology in combination with biomaterials technology for their potential use in the development of RGC replacement therapies with the final aim of the improvement or even the restoration of the vision of HON patients. To this purpose, the combination of natural and synthetic biomaterials modified with peptides, neurotrophic factors, and other low- to medium-molecular weight compounds, mimicking the ocular extracellular matrices, with human iPSC or iPSC-derived cell retinal progenitors holds enormous potential to be exploited in the near future for the generation of transplantable RGC populations.
    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
    青光眼是由视网膜神经节细胞(RGC)的进行性丧失引起的慢性致盲眼病。目前,没有临床批准的治疗方法可以直接提高RGCs的生存率。载脂蛋白E(APOE)基因与众多神经退行性疾病的遗传风险密切相关,近年来已成为神经退行性疾病研究领域的热点。视神经和视网膜是大脑神经系统的延伸部分。视网膜退行性疾病的发病机制与脑神经退行性疾病密切相关。APOE由三个等位基因组成,ε4,ε3和ε2,在单个基因座中。他们有不同程度的青光眼风险。APOE4和APOE基因缺失(APOE-/-)可以减少RGC损失。相比之下,APOE3和APOE基因的整体存在(APOE+/+)导致RGC体和轴突的显著损失,增加青光眼RGC死亡的风险。目前,没有明确的文献表明APOE2对青光眼有益或有害.这项研究总结了不同APOE基因在青光眼中的作用机制,并推测APOE靶向干预可能是一种有前途的预防青光眼RGCs损失的方法。
    Glaucoma is a chronic blinding eye disease caused by the progressive loss of retinal ganglion cells (RGCs). Currently, no clinically approved treatment can directly improve the survival rate of RGCs. The Apolipoprotein E (APOE) gene is closely related to the genetic risk of numerous neurodegenerative diseases and has become a hot topic in the field of neurodegenerative disease research in recent years. The optic nerve and retina are extensions of the brain\'s nervous system. The pathogenesis of retinal degenerative diseases is closely related to the degenerative diseases of the nerves in the brain. APOE consists of three alleles, ε4, ε3, and ε2, in a single locus. They have varying degrees of risk for glaucoma. APOE4 and the APOE gene deletion (APOE-/-) can reduce RGC loss. By contrast, APOE3 and the overall presence of APOE genes (APOE+/+) result in significant loss of RGC bodies and axons, increasing the risk of glaucoma RGCs death. Currently, there is no clear literature indicating that APOE2 is beneficial or harmful to glaucoma. This study summarises the mechanism of different APOE genes in glaucoma and speculates that APOE targeted intervention may be a promising method for protecting against RGCs loss in glaucoma.
    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
    视网膜缺血,脑缺血后,是一个容易被忽视的病理生理问题,其中炎症被认为起着重要作用。焦亡是一种伴随炎症的细胞死亡模式。Homer支架蛋白1(Homer1)具有抗炎特性,可防止缺血性损伤。然而,关于大脑中动脉阻塞(MCAO)诱导的视网膜缺血后的焦亡以及Homer1参与焦亡发展的调节机制知之甚少。在本研究中,小鼠体内永久性MCAO诱发视网膜缺血性损伤,和视网膜神经节细胞(RGCs)进行氧糖剥夺(OGD)以建立体外模型。研究表明,TXNIP/NLRP3介导的焦亡主要位于RGC中,在视网膜缺血后逐渐增加,并在视网膜缺血后24h达到峰值。有趣的是,RGC的焦亡不仅发生在细胞体中,也发生在轴突中。值得注意的是,焦亡的发生与视网膜缺血后视网膜中Homer1表达的变化同时发生,并且Homer1也与RGCs共定位。研究表明,Homer1的过表达不仅减轻了RGCs的细胞凋亡,抑制了促炎因子的释放,而且导致AMPK的磷酸化增加,抑制ER应激,视网膜缺血后视觉功能的保护。总之,提示Homer1可能通过抑制MCAO诱导的视网膜缺血后的内质网应激相关的TXNIP/NLRP3炎性体激活来保护MCAO诱导的视网膜缺血和RGCs的焦亡。
    Retinal ischemia, after cerebral ischemia, is an easily overlooked pathophysiological problem in which inflammation is considered to play an important role. Pyroptosis is a kind of cell death pattern accompanied by inflammation. Homer scaffold protein 1 (Homer1) has anti-inflammation properties and protects against ischemic injury. However, little is known about pyroptosis following middle cerebral artery occlusion (MCAO)-induced retinal ischemia and the regulatory mechanisms involved by Homer1 for the development of pyroptosis. In the present study, retinal ischemic injury was induced in mice by permanent MCAO in vivo, and retinal ganglion cells (RGCs) were subjected to Oxygen and Glucose Deprivation (OGD) to establish an in vitro model. It was shown that TXNIP/NLRP3-mediated pyroptosis was located predominantly in RGCs, which gradually increased after retinal ischemia and peaked at 24 h after retinal ischemia. Interestingly, the RGCs pyroptosis occurred not only in the cell body but also in the axon. Notably, the occurrence of pyroptosis coincided with the change of Homer1 expression in the retina after retinal ischemia and Homer1 also co-localized with RGCs. It was demonstrated that overexpression of Homer1 not only alleviated RGCs pyroptosis and inhibited the release of pro-inflammatory factors but also led to the increase in phosphorylation of AMPK, inhibition of ER stress, and preservation of visual function after retinal ischemia. In conclusion, it was suggested that Homer1 may protect against MCAO-induced retinal ischemia and RGCs pyroptosis by inhibiting endoplasmic reticulum stress-associated TXNIP/NLRP3 inflammasome activation after MCAO-induced retinal ischemia.
    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
    青光眼是一种致盲率高、发病机制复杂的眼病。眼部高血压(OHT)是一个关键的危险因素,视网膜缺血/再灌注(I/R)是视网膜缺血/再灌注的重要病理生理基础。本研究旨在探讨口服柚皮素在急性视网膜I/R模型和慢性OHT模型中的视网膜神经保护作用及其可能的机制。在建立I/R和OHT模型后,给小鼠服用赋形剂或柚皮素(100mg/kg或300mg/kg)。采用苏木精-伊红(HE)染色、RBPMS和胶质纤维酸性蛋白(GFAP)免疫染色评价视网膜损伤。GFAP,CD38,Sirtuin1(SIRT1),用蛋白质印迹法测定NOD样受体蛋白3(NLRP3)的表达水平。在OHT模型中,损伤后眼内压(IOP)动态维持在约20~25mmHg.视网膜结构受损,和视网膜神经节细胞(RGC)在两个模型中都丢失。柚皮素改善了上述适应症,但也证明了高浓度的柚皮素显著抑制视网膜星形胶质细胞活化,抑制损伤诱导的GFAP表达增加,NLRP3和CD38蛋白,而SIRT1蛋白表达上调。这项研究首次表明柚皮素可以降低OHT模型中微珠诱导的IOP升高,为柚皮素在青光眼中的应用提供了新的证据。柚皮素可能介导CD38/SIRT1信号通路,抑制星形胶质细胞激活,并最终发挥抗炎作用实现视网膜神经保护。
    Glaucoma is an eye disease with a high rate of blindness and a complex pathogenesis. Ocular hypertension (OHT) is a critical risk factor, and retinal ischemia/reperfusion (I/R) is an important pathophysiological basis. This study was designed to investigate the retinal neuroprotective effect of oral naringenin in an acute retinal I/R model and a chronic OHT model and the possible mechanism involved. After the I/R and OHT models were established, mice were given vehicle or naringenin (100 mg/kg or 300 mg/kg). Hematoxylin-eosin (HE) staining and immunostaining of RBPMS and glial fibrillary acidic protein (GFAP) were used to evaluate retinal injury. GFAP, CD38, Sirtuin1 (SIRT1), and NOD-like receptor protein 3 (NLRP3) expression levels were measured by Western blotting. In the OHT model, intraocular pressure (IOP) was dynamically maintained at approximately 20-25 mmHg after injury. The retinal structure was damaged, and retinal ganglion cells (RGCs) were lost in both models. Naringenin ameliorated the abovementioned indications but also demonstrated that high concentrations of naringenin significantly inhibited retinal astrocyte activation and inhibited damage-induced increases in the expression of GFAP, NLRP3, and CD38 proteins, while SIRT1 protein expression was upregulated. This study showed for the first time that naringenin can reduce microbead-induced IOP elevation in the OHT model, providing new evidence for the application of naringenin in glaucoma. Naringenin may mediate the CD38/SIRT1 signaling pathway, inhibit astrocyte activation, and ultimately exert an anti-inflammatory effect to achieve retinal neuroprotection.
    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
    青光眼是一种复杂的多因素眼病,表现为视网膜神经节细胞(RGC)死亡和视神经变性,最终导致不可逆的视力丧失。近年来的研究大大增强了我们对青光眼RGC变性机制的理解。很明显,高眼内压(IOP)不是青光眼发病机理的唯一因素。视网膜中促存活和促死亡信号通路的平衡强烈影响青光眼中RGC和视神经轴突的功能和存活。来自人类视网膜组织分析和一系列青光眼实验模型的分子证据对揭示这些机制做出了重大贡献。越来越多的证据表明,广泛的分子信号通路可以单独或通过复杂的网络来诱导神经变性。几种分子的作用,包括神经营养因子,细胞内激酶和磷酸盐的相互作用,Caveolae和衔接蛋白,丝氨酸蛋白酶及其抑制剂,核受体,淀粉样β和tau,以及它们的功能障碍如何影响视网膜神经元在这篇综述中进行了讨论。我们进一步强调了表现出RGC变性和对青光眼相关神经元损伤易感性的各种动物模型的解剖学改变如何帮助表征青光眼的分子机制。此外,我们还提出了不同的调节细胞死亡途径,这些途径在青光眼的RGC变性中起关键作用。
    Glaucoma is a complex multifactorial eye disease manifesting in retinal ganglion cell (RGC) death and optic nerve degeneration, ultimately causing irreversible vision loss. Research in recent years has significantly enhanced our understanding of RGC degenerative mechanisms in glaucoma. It is evident that high intraocular pressure (IOP) is not the only contributing factor to glaucoma pathogenesis. The equilibrium of pro-survival and pro-death signalling pathways in the retina strongly influences the function and survival of RGCs and optic nerve axons in glaucoma. Molecular evidence from human retinal tissue analysis and a range of experimental models of glaucoma have significantly contributed to unravelling these mechanisms. Accumulating evidence reveals a wide range of molecular signalling pathways that can operate -either alone or via intricate networks - to induce neurodegeneration. The roles of several molecules, including neurotrophins, interplay of intracellular kinases and phosphates, caveolae and adapter proteins, serine proteases and their inhibitors, nuclear receptors, amyloid beta and tau, and how their dysfunction affects retinal neurons are discussed in this review. We further underscore how anatomical alterations in various animal models exhibiting RGC degeneration and susceptibility to glaucoma-related neuronal damage have helped to characterise molecular mechanisms in glaucoma. In addition, we also present different regulated cell death pathways that play a critical role in RGC degeneration in glaucoma.
    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
    这项研究的目的是评估浅层(ODD-S)或深层(ODD-D)视盘玻璃疣患者的视网膜神经节细胞(RGC)和视觉通路的形态功能参与。这项研究招募了17名ODD患者(平均年龄为59.10±12.68岁),提供19只眼和20名对照受试者(平均年龄58.62±8.77岁),提供20只眼。我们评估了以下内容:最佳矫正视力,视野平均偏差(MD),模式视网膜电图(PERG)的振幅(A),隐含时间(IT)和视觉诱发电位(VEP)的A,视网膜神经纤维层厚度(RNFL-T)和神经节细胞厚度(GC-T)。在ODD-S眼中,测量玻璃疣的可见高度。在26.3%和73.7%的ODD眼中检测到ODD-D和ODD-S,分别。显著(p<0.01)降低MD,A级,VEP振幅,与对照相比,在ODD组中发现RNFL-T和GC-T值以及显著(p<0.01)增加的VEPIT值。在ODD组中,PERGAs与VEPITs之间没有发现显着相关性(p>0.01)。在ODD-S中,可见身高与MD降低显著相关(p<0.01),PERGAs和RNFL-T以及增加的PSD和VEPIT值。我们的发现表明,ODD可能会引起RGC及其纤维的形态功能变化,以及导致或不导致视野缺陷的无关视觉通路功能障碍。观察到的形态功能损害应归因于逆行(从轴突到RGC)和顺行(从RGC到视觉皮层)轴质运输的改变。在ODD-S眼中,300微米的最小可见高度代表异常的阈值,这表明“奇数越高,减值越严重。“
    The aim of this study was to assess the morpho-functional involvement of the retinal ganglion cells (RGCs) and of the visual pathways in patients with superficial (ODD-S) or deep (ODD-D) optic disc drusen. This study enrolled 17 patients with ODD (mean age of 59.10 ± 12.68 years) providing 19 eyes and 20 control subjects (mean age 58.62 ± 8.77 years) providing 20 eyes. We evaluated the following: best-corrected visual acuity, visual field mean deviation (MD), the amplitude (A) of Pattern Electroretinogram (PERG), the implicit time (IT) and A of Visual Evoked Potentials (VEPs), retinal nerve fiber layer thickness (RNFL-T) and ganglion cell thickness (GC-T). In ODD-S eyes, the drusen visible height was measured. ODD-D and ODD-S were detected in 26.3% and 73.7% of ODD eyes, respectively. Significantly (p < 0.01) reduced MD, PERG A, VEP amplitude, RNFL-T and GC-T values and significantly (p < 0.01) increased VEP IT values were found in the ODD Group as compared to the Control one. In the ODD Group, no significant correlation (p > 0.01) between PERG As and VEP ITs was found. In ODD-S, the visible height was significantly correlated (p < 0.01) with reduced MD, PERG As and RNFL-T and with increased PSD and VEP IT values. Our findings suggest that ODD might induce morpho-functional changes in RGCs and their fibers and an unrelated visual pathway dysfunction leading or not leading to visual field defects. The observed morpho-functional impairment should be ascribed to an alteration in retrograde (from the axons to the RGCs) and anterograde (from the RGCs up to the visual cortex) axoplasmic transport. In ODD-S eyes, a minimum visible height of 300 microns represented the threshold for the abnormalities, suggesting that \"the higher the ODD, the worse the impairment\".
    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
    目的:使用明视负反应(PhNR)和谱域光学相干断层扫描(SD-OCT)分析Leber遗传性视神经病变(LHON)期间视网膜神经节细胞(RGCs)及其轴突的功能和结构变化。
    方法:被诊断为LHON的个体及其家庭成员被邀请参加这项横断面研究。使用PhNR和OCT。比较3组患者的PhNR波幅和乳头周围视网膜神经纤维层(pRNFL)厚度。此外,受影响的个体分为亚急性,基于疾病持续时间的动态和慢性期,以评估RGC功能和结构的衰减。
    结果:包括73个受影响的携带者和30个具有m.11778G>A突变的携带者。与对照组相比,受累个体和携带者的PhNR振幅和pRNFL厚度显着降低(P<0.001)。然而,携带者与对照组比较差异无统计学意义(P>0.05)。不同相位的PhNR振幅无差异(P=0.464)。在亚急性期,仅颞叶pRNFL厚度明显下降(P<0.001)。PRNFL厚度在动态阶段明显下降(P<0.001)。在慢性期,pRNFL的时间厚度持续下降(P=0.042)。
    结论:在亚急性期,RGC功能严重受损.在疾病进展过程中,四个象限的pRNFL厚度显着降低。在慢性期,pRNFL厚度略有下降。在病理变化发生之前,载体已经显示出RGC功能障碍,提示亚临床异常.
    To analyze the functional and structural changes in retinal ganglion cells (RGCs) and their axons that occur during Leber\'s hereditary optic neuropathy (LHON) using photopic negative response (PhNR) and spectral domain optical coherence tomography (SD-OCT).
    Individuals diagnosed with LHON and their family members were invited to participate in this cross-sectional study. PhNR and OCT were used. The PhNR amplitude and peripapillary retinal nerve fiber layer (pRNFL) thicknesses were compared among the three groups. In addition, affected individuals were divided into subacute, dynamic and chronic phases based on disease duration in order to evaluate the decay in RGCs function and structure.
    73 affected and 30 carriers with a m.11778G > A mutation were included. PhNR amplitude and the thickness of pRNFL significantly decreased in affected individuals and carriers compared to that of the controls (P<0.001). However, there was no difference between the carriers and the controls (P>0.05). There was no difference in the PhNR amplitude of different phases (P = 0.464). In the subacute phase, only temporal pRNFL thickness decreased significantly (P<0.001). PRNFL thickness decreased significantly in dynamic phase (P<0.001). Temporal pRNFL thickness continued to decrease in the chronic phase (P = 0.042).
    In the subacute phase, the function of RGCs was severely impaired. Thickness of pRNFL decreased significantly in four quadrants during disease progression. In the chronic phase, pRNFL thickness decreased slightly. Carriers have shown RGCs dysfunction before pathological changes occur, suggesting subclinical abnormalities.
    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
    星形胶质细胞,神经系统中的非神经元神经胶质细胞类型,对调节中枢神经系统的生理功能至关重要。在中枢神经系统的各种损伤和疾病中,星形胶质细胞通常将其表型转变为参与促炎反应的神经毒性型(以下称为“免疫功能”)。这种星形细胞免疫功能不仅限于脑部疾病,而且在青光眼等眼部神经退行性疾病中也有发现。一种视网膜神经退行性疾病,是全球失明的主要原因。眼睛有两个星形胶质细胞谱系细胞:星形胶质细胞和Müller细胞。它们维持视网膜和视神经的生理环境,从而控制视觉功能。星形胶质细胞谱系细胞的功能障碍可能与青光眼的发病和进展有关。这些细胞在青光眼患者中变得反应,动物研究表明,它们的免疫反应可能与青光眼相关事件有关:组织重塑,神经元死亡,和周围免疫细胞的浸润。在这次审查中,探讨星形胶质细胞免疫功能在青光眼发病机制中的作用。
    Astrocytes, a non-neuronal glial cell type in the nervous system, are essential for regulating physiological functions of the central nervous system. In various injuries and diseases of the central nervous system, astrocytes often change their phenotypes into neurotoxic ones that participate in pro-inflammatory responses (hereafter referred to as \"immune functions\"). Such astrocytic immune functions are not only limited to brain diseases but are also found in ocular neurodegenerative diseases such as glaucoma, a retinal neurodegenerative disease that is the leading cause of blindness worldwide. The eye has two astrocyte-lineage cells: astrocytes and Müller cells. They maintain the physiological environment of the retina and optic nerve, thereby controlling visual function. Dysfunction of astrocyte-lineage cells may be involved in the onset and progression of glaucoma. These cells become reactive in glaucoma patients, and animal studies have suggested that their immune responses may be linked to glaucoma-related events: tissue remodeling, neuronal death, and infiltration of peripheral immune cells. In this review, we discuss the role of the immune functions of astrocyte-lineage cells in the pathogenesis of glaucoma.
    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:视网膜神经节细胞(RGCs)凋亡是视网膜缺血/再灌注(I/R)损伤的重要表现,然而,潜在的机制还没有得到很好的理解。目前正在探索长链非编码RNA(lncRNA)对该细胞过程的贡献。基于lncRNA芯片检测,我们旨在研究lncRNAuc007nnj.1在缺血诱导的RGCs凋亡的病理过程中的作用。
    方法:汉克的平衡盐溶液含有10µM抗霉素A和2µM钙离子载体2h,在RGC中构建缺血模型,并将眼压升高至120mmHg1h,以构建小鼠视网膜I/R损伤模型。
    结果:在这项研究中,lncRNAuc007nnj.1响应于RGC和小鼠视网膜中的I/R损伤而高度上调。此外,lncRNAuc007nnj.1敲除在体外和体内减少视网膜神经元细胞凋亡,并显着改善视网膜功能。
    结论:机械上,结果表明,lncRNAuc007nnj.1作为CERNA竞争性结合miR-155-5p,从而增强Tle4的表达水平,从而加重RGCs中缺血相关的凋亡。
    结论:最后,我们的研究将lncRNAuc007nnj.1/miR-155-5p/Tle4轴确定为预防I/R诱导的视网膜神经元死亡的潜在靶标。
    Retinal ganglion cells (RGCs) apoptosis is a vital manifestation of retinal ischemia/reperfusion (I/R) injury, yet the underlying mechanisms are not well understood. The contribution of long noncoding RNAs (lncRNAs) to this cellular process is currently being explored. Based on a lncRNA chip assay, we aimed to investigate the role of lncRNA uc007nnj.1 in the pathological process of ischemia-induced RGCs apoptosis.
    Hank\'s balanced salt solution containing 10 µM antimycin A and 2 µM calcium ionophore for 2 h to construct an ischemic model in RGCs, and elevation of intraocular pressure to 120 mm Hg for 1 h was used to construct a mouse model of retinal I/R injury.
    In this study, lncRNA uc007nnj.1 was highly upregulated in response to I/R injury in RGCs and mouse retinas. In addition, lncRNA uc007nnj.1 knockdown reduced retinal neuronal cell apoptosis in vitro and in vivo and significantly improved retinal function.
    Mechanistically, the results demonstrated that lncRNA uc007nnj.1 acts as ceRNA competitively binding miR-155-5p, thereby enhancing the expression levels of Tle4, thus aggravating ischemia-related apoptosis in RGCs.
    Finally, our study identifies the lncRNA uc007nnj.1/miR-155-5p/Tle4 axis as a potential target for the prevention of I/R-induced retinal neuronal death.
    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