RGCs

RGC
  • 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
    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
    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

  • 文章类型: Journal Article
    青光眼是全球不可逆失明的最常见原因。升高的眼内压(IOP)和视网膜中的相对缺氧刺激了活性氧(ROS)的产生,which,反过来,视网膜和视神经处于慢性氧化应激状态.新的证据表明,氧化应激可以引发PARP-1过度激活,线粒体相关内质网膜(MAM)失调,和NLRP3激活。氧化损伤可以触发炎性体激活,NLRP3是唯一与MAM失调相关的炎性小体。此外,多个转录因子位于MAM上。本研究旨在探讨PARP-1抑制剂(奥拉帕尼)对慢性高眼压相关视网膜细胞损伤的保护作用和潜在机制。我们还通过将细胞暴露于体外0.2%O2来模拟视网膜前体细胞系的缺氧刺激。我们发现慢性高眼压(COH)诱导视网膜神经节细胞(RGC)的氧化损伤和MAM失调。切割的PARP和NLRP3的蛋白质水平在COH大鼠的视网膜中上调。Olaparib,PARP-1抑制剂,减轻了COH引起的RGC损失,视网膜形态学改变,和明视负反应幅度降低。奥拉帕尼还缓解了低氧刺激诱导的细胞活力丧失和MAM失调。此外,线粒体表现的一些指标,比如活性氧的积累,线粒体Ca2+内流,线粒体膜电位崩溃,奥拉帕尼治疗后下降。奥拉帕尼抑制了缺氧诱导的NLRP3蛋白水平的上调以及ERK1/2和组蛋白H2A.X的磷酸化。这些结果表明,奥拉帕尼在体内保护RGCs免受慢性眼内压升高的影响,并减轻了体外缺氧引起的异常MAM失调和线粒体功能障碍。这种保护可以通过抑制PARP-1过度激活来实现,NLRP3上调,和ERK1/2的磷酸化。
    Glaucoma is the most common cause of irreversible blindness worldwide. Elevated intraocular pressure (IOP) and relative hypoxia in the retina stimulate the production of reactive oxygen species (ROS), which, in turn, puts the retina and optic nerve under chronic oxidative stress. Emerging evidence has shown that oxidative stress can trigger PARP-1 overactivation, mitochondrial-associated endoplasmic reticulum membrane (MAM) dysregulation, and NLRP3 activation. Oxidative damage can trigger inflammasome activation, and NLRP3 is the only inflammasome associated with MAM dysregulation. In addition, multiple transcription factors are located on the MAM. This study aimed to investigate the protective effects and underlying mechanisms of a PARP-1 inhibitor (olaparib) against chronic ocular hypertension-associated retinal cell damage. We also mimicked hypoxic stimulation of a retinal precursor cell line by exposing the cells to 0.2% O2 in vitro. We discovered that chronic ocular hypertension (COH) induces oxidative damage and MAM dysregulation in the retinal ganglion cells (RGCs). The protein levels of cleaved-PARP and NLRP3 were upregulated in the retinas of the COH rats. Olaparib, a PARP-1 inhibitor, alleviated COH-induced RGC loss, retinal morphological alterations, and photopic negative response amplitude reduction. Olaparib also relieved hypoxic stimulation-induced loss of cell viability and MAM dysregulation. Additionally, some indicators of mitochondrial performance, such as reactive oxygen species accumulation, mitochondrial Ca2+ influx, and mitochondrial membrane potential collapse, decreased after olaparib treatment. Olaparib attenuated the hypoxia-induced upregulation of NLRP3 protein levels as well as the phosphorylation of ERK1/2 and histone H2A.X. These results suggest that olaparib protects RGCs from chronic intraocular pressure elevation in vivo and alleviates the abnormal MAM dysregulation and mitochondrial dysfunction caused by hypoxia in vitro. This protection may be achieved by inhibiting PARP-1 overactivation, NLRP3 upregulation, and phosphorylation of ERK1/2.
    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
    视神经的退化缩小了视野,最终导致整体视力丧失。这项研究旨在确定视神经挤压(ONC)小鼠视网膜中的整体蛋白质变化,并确定在视神经变性过程中与损伤诱导的细胞死亡有关的关键调节因子和途径。对ONC和假手术小鼠的视网膜蛋白提取物进行无标记定量蛋白质组学结合生物信息学分析。在检测到的1433种蛋白质中,121种蛋白质在ONC小鼠的视网膜中差异表达。进一步的生物信息学分析表明,各种代谢途径,包括谷氨酸代谢和γ-氨基丁酸(GABA)合成,在受损的小鼠视网膜中显著失调。谷氨酸脱羧酶1(GAD1)是将谷氨酸转化为GABA的酶,在ONC损伤期间显著上调。外源性GAD1治疗增加了ONC损伤的视网膜中的视网膜神经节细胞(RGC)存活。此外,在其他几种眼科疾病中也观察到GAD1表达的变化.血管内皮生长因子B(VEGF-B)先前已报道可保护RGCs免受凋亡并正调节视网膜中GAD1的表达。值得注意的是,GAD1和VEGF-B的联合治疗也提供了对损伤诱导的RGC凋亡的强保护作用.这些结果表明,GAD1表达可能是一种内在的保护机制,通常在视网膜损伤期间被激活。靶向GAD1可能是治疗视神经退行性疾病的潜在策略。
    The degeneration of the optic nerve narrows the visual field, eventually causing overall vision loss. This study aimed to identify global protein changes in the retina of optic nerve crushing (ONC) mice and to identify key regulators and pathways involved in injury-induced cell death during the progression of optic neurodegeneration. Label-free quantitative proteomics combined with bioinformatic analysis was performed on retinal protein extracts from ONC and sham-operated mice. Among the 1433 proteins detected, 121 proteins were differentially expressed in the retina of ONC mice. Further bioinformatic analysis showed that various metabolic pathways, including glutamate metabolism and γ-aminobutyric acid (GABA) synthesis, were significantly dysregulated in the injured mouse retinas. Glutamate decarboxylase 1 (GAD1) is the enzyme that converts glutamate into GABA, which was significantly up-regulated during ONC injury. Exogenous GAD1 treatment increased retinal ganglion cell (RGC) survival in the ONC-injured retina. In addition, changes in GAD1 expression were also observed in several other ophthalmic diseases. Vascular endothelial growth factor B (VEGF-B) has previously been reported to protect RGCs from apoptosis and positively regulated the expression of GAD1 in the retina. Notably, combination treatment with GAD1 and VEGF-B also provided strong protection against injury-induced RGC apoptosis. These results suggest that GAD1 expression may serve as an intrinsic protective mechanism that is commonly activated during retinal injury. Targeting GAD1 may serve as a potential strategy to treat optic neurodegenerative diseases.
    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
    外伤性视神经病变(TON)是世界范围内视力丧失和不可逆失明的重要原因。它被定义为由损伤引起的视网膜神经节细胞死亡和轴突变性。视神经挤压(ONC),一个经过充分验证的TON模型,激活视网膜小胶质细胞并引发神经炎症。高移动性组盒1(HMGB1),真核细胞核中的非组蛋白染色体结合蛋白,是小胶质细胞活化和促炎细胞因子释放的重要诱导剂。目的研究HMGB1抑制剂BoxA对创伤性视神经病变中神经炎症诱导的视网膜神经节细胞(RGCs)损伤的保护作用及机制。为此,建立C57BL/6J小鼠10~12周视神经挤压模型。模型小鼠接受PBS和HMGB1抑制剂BoxA的玻璃体内注射。我们的数据表明,视神经挤压后HMGB1表达增加。视网膜神经节细胞功能和形态均受损,视神经挤压后视网膜神经节细胞数量减少。玻璃体内注射BoxA后ONC可减轻损伤。此外,BoxA降低了小胶质细胞的活化和核因子κB(NF-kB)的表达水平,核苷酸结合域,含亮氨酸重复序列的蛋白3(NLRP3),和实验ONC小鼠中含有CARD(ASC)的凋亡相关斑点样蛋白。总之,HMGB1通过NF-kB介导NLRP3炎性体参与ONC后视网膜炎症损伤。因此,玻璃体内注射BoxA对于有效治疗RGC死亡以预防TON具有潜在的治疗益处。
    Traumatic optic neuropathy (TON) is a significant cause of vision loss and irreversible blindness worldwide. It is defined as retinal ganglion cell death and axon degeneration caused by injury. Optic nerve crush (ONC), a well-validated model of TON, activates retinal microglia and initiates neuroinflammation. High-mobility group box 1 (HMGB1), a non-histone chromosomal binding protein in the nucleus of eukaryotic cells, is an important inducer of microglial activation and pro-inflammatory cytokine release. The purpose of this study was to examine the protective effects and mechanism of the HMGB1 inhibitor BoxA to neuroinflammation-induced retinal ganglion cells (RGCs) damage in traumatic optic neuropathy. For that purpose, an optic nerve crush model was established in C57BL/6J mice at 10-12 weeks. Model mice received an intravitreal injection of PBS and the HMGB1 inhibitor BoxA. Our data demonstrated that HMGB1 expression increased after optic nerve crush. Retinal ganglion cell function and morphology were damaged, and retinal ganglion cell numbers were reduced after optic nerve crush. Intravitreal injection of BoxA after ONC can alleviate damage. Furthermore, BoxA reduced microglial activation and expression levels of nuclear factor κB (NF-kB), nucleotide-binding domain, leucine-rich repeat containing protein 3 (NLRP3), and apoptosis-associated speck-like protein containing a CARD (ASC) in experimental ONC mice. In summary, HMGB1 mediates NLRP3 inflammasome via NF-kB to participate in retinal inflammatory injury after ONC. Thus, intravitreal injection of BoxA has potential therapeutic benefits for the effective treatment of RGC death to prevent TON.
    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: Review
    目的:青光眼病理过程中发生视网膜神经节细胞死亡,由于视网膜神经节细胞的再生能力较差,因此具有重要意义。随着对视网膜细胞死亡机制的不断了解,我们现在知道,仅仅阻断细胞死亡的特定机制可能并不能阻止视网膜神经节细胞死亡。本文旨在总结青光眼模型中视网膜细胞死亡的机制,并讨论这些研究中恢复视觉功能的注意事项。方法:在PubMed上使用包括青光眼在内的关键词进行文献检索,高眼压,视网膜神经节细胞,细胞死亡,凋亡,坏死,焦亡,铁性凋亡,自噬性细胞死亡,还有Parthanatos.通过文献综述,总结了青光眼样动物模型中视网膜神经节细胞的致死途径。结果:根据目的,本综述选择并讨论了100项研究。结论:青光眼动物神经节细胞的损伤可通过多种致死途径发生,其分子机制尚不完全清楚。对不同细胞死亡途径与常见上游调节剂之间的串扰的进一步研究可以促进用于青光眼治愈性治疗的新型靶向剂的开发。
    Purpose: Retinal ganglion cell death occurs during the glaucoma pathological process, and it is significant because of the poor regeneration capacity of retinal ganglion cells. With a constantly increasing understanding of retinal cell death mechanisms, we now know that simply blocking a specific mechanism of cell death might not prevent retinal ganglion cell death. This review aimed to summarize the mechanisms of retinal cell death in glaucoma models and discuss the caveats in restoring visual function in these studies.Methods: A literature search was done on PubMed using key words including glaucoma, ocular hypertension, retinal ganglion cell, cell death, apoptosis, necroptosis, pyroptosis, ferroptosis, autophagic cell death, and parthanatos. The literature was reviewed to summarize the information about the lethal pathways of retinal ganglion cell in the glaucoma-like animal models.Results: Based on the purpose, 100 studies were selected and discussed in this review.Conclusions: The damage to ganglion cells in glaucoma-like animals can occur via multiple lethal pathways and the molecular mechanisms are still incompletely understood. Further investigations on the crosstalk between different cell death pathways and the common upstream regulators could augment the development of novel targeting agents for the curative treatment 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
    DNA甲基化被认为在糖尿病视网膜病变的发生发展中起重要作用。这里,我们的目的是研究甲基-CpG结合域蛋白2(Mbd2)在早期糖尿病视网膜视网膜神经节细胞(RGCs)凋亡中的确切作用.在高糖(HG)治疗后,Mbd2显着上调,并在HG诱导的凋亡过程中在RGC中发挥促凋亡作用。结合ChIP和基因微阵列数据集,结果显示Mbd2具有miR-345-5p的潜在结合位点,从而通过增强启动子去甲基化提高miR-345-5p的表达水平。激活转录因子1(Atf1)在RGCs凋亡过程中发挥抗凋亡作用,并作为miR-345-5p的靶基因。此外,与野生型小鼠相比,Mbd2基因敲除小鼠的糖尿病视网膜中存活的RGC数量增加,视觉功能也相应改善.总的来说,我们的数据表明,HG诱导的Mbd2在视网膜中的过表达是通过miR-345-5p/Atf1轴导致视网膜神经元细胞凋亡的部分原因.因此,Mbd2的靶向可能代表了治疗早期糖尿病视网膜神经变性的新治疗策略.
    DNA methylation is considered to play an important role in the development of diabetic retinopathy. Here, our goal was to investigate the precise role of methyl-CpG binding domain protein 2 (Mbd2) in the apoptosis of retinal ganglion cells (RGCs) in the early diabetic retina. Mbd2 was significantly upregulated after high glucose (HG) treatment and played a proapoptotic role in RGCs during HG-induced apoptosis. Combining ChIP and gene microarray datasets, the results showed that Mbd2 possessed potential binding sites for miR-345-5p, thereby elevating the expression levels of miR-345-5p via the enhancement of promoter demethylation. Activating transcription factor 1 (Atf1) played an anti-apoptotic role during the process of apoptosis in RGCs and acted as the target gene for miR-345-5p. Furthermore, the number of surviving RGCs in the diabetic retina was increased in Mbd2-knockout mice when compared with wild-type mice and the visual function became better accordingly. Collectively, our data demonstrated that the HG-induced overexpression of Mbd2 in the retina was partly responsible for the apoptosis of retinal neuronal cells through the miR-345-5p/Atf1 axis. Therefore, the targeting of Mbd2 might represent a novel therapeutic strategy for the treatment of neurodegeneration in the early diabetic retina.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号