retinal neovascularization

视网膜新生血管形成
  • 文章类型: Journal Article
    视网膜新生血管形成是几种眼部新生血管疾病的共同特征。这是世界上导致失明的主要原因。目前的治疗是通过侵入性玻璃体内注射进行的,导致患者依从性差,严重的眼部并发症和沉重的经济负担。因此,需要一种替代性的非侵入性或非侵入性治疗策略。这里,一种非侵入性口服酪氨酸激酶抑制剂,CM082在斑马鱼幼虫缺氧诱导的视网膜新生血管模型中进行了评估。我们发现CM082能有效抑制视网膜新生血管,拯救了视网膜神经节细胞层中的细胞损失,并挽救了视觉功能缺陷。我们的结果阐明了CM082主要通过抑制Vegfr2磷酸化来介导其治疗功效。研究结果表明,CM082具有很强的抗血管生成作用,可作为眼部新生血管性疾病血管生成的潜在治疗方法。
    Retinal neovascularization is a common feature of several ocular neovascular diseases, which are the leading cause of blindness in the world. Current treatments are administered through invasive intravitreal injections, leading to poor patient compliance, serious ocular complications and heavy economic burdens. Thus, an alternative less or non-invasive therapeutic strategy is in demand. Here, a non-invasive oral tyrosine kinase inhibitor, CM082, was evaluated in a retinal neovascularization model induced by hypoxia in zebrafish larvae. We found that CM082 effectively suppressed retinal neovascularization, rescued cell loss in the retinal ganglion cell layer, and rescued the visual function deficiency. Our results elucidated that CM082 mediated its therapeutic efficacy primarily through the inhibition of Vegfr2 phosphorylation. The findings demonstrated that CM082 possessed strong antiangiogenic effects and may serve as a potential treatment for angiogenesis in ocular neovascular diseases.
    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
    视网膜新生血管形成是晚期年龄相关性黄斑变性(AMD)的重要特征,也是AMD患者失明的主要原因。然而,这种病理性新生血管形成的潜在机制尚不清楚.铁代谢涉及各种生物过程。本研究旨在研究铁代谢对新生血管性AMD(nAMD)视网膜新生血管形成的影响。
    C57BL/6J和极低密度脂蛋白受体(VLDLR)敲除(Vldlr-/-)小鼠,nAMD的鼠模型,在这项研究中使用。Bulk-RNA测序用于鉴定差异表达的基因。进行蛋白质印迹分析以测试蛋白质的表达。通过口服管饲法向小鼠施用铁螯合剂去铁酮(DFP)。荧光素眼底血管造影用于评估视网膜血管渗漏。免疫荧光染色用于检测巨噬细胞和铁相关蛋白。
    RNA测序(RNA-seq)结果显示Vldlr-/-小鼠的视网膜和RPE中转铁蛋白表达改变。在Vldlr-/-小鼠的视网膜和RPE中观察到铁稳态中断。DFP减轻了铁过载,并显着减少了视网膜新生血管形成和血管渗漏。此外,DFP抑制了Vldlr-/-视网膜的炎症。在DFP处理后,在Vldlr-/-小鼠的视网膜和RPE中的新生血管形成部位观察到巨噬细胞的信号降低。Further,IL-6/JAK2/STAT3信号通路在Vldlr-/-小鼠的视网膜和RPE中被激活,并被DFP治疗逆转。
    铁代谢紊乱可能导致nAMD视网膜新生血管形成。通过DFP恢复铁稳态可能是nAMD的潜在治疗方法。
    UNASSIGNED: Retinal neovascularization is a significant feature of advanced age-related macular degeneration (AMD) and a major cause of blindness in patients with AMD. However, the underlying mechanism of this pathological neovascularization remains unknown. Iron metabolism has been implicated in various biological processes. This study was conducted to investigate the effects of iron metabolism on retinal neovascularization in neovascular AMD (nAMD).
    UNASSIGNED: C57BL/6J and very low-density lipoprotein receptor (VLDLR) knockout (Vldlr-/-) mice, a murine model of nAMD, were used in this study. Bulk-RNA sequencing was used to identify differentially expressed genes. Western blot analysis was performed to test the expression of proteins. Iron chelator deferiprone (DFP) was administrated to the mice by oral gavage. Fundus fluorescein angiography was used to evaluate retinal vascular leakage. Immunofluorescence staining was used to detect macrophages and iron-related proteins.
    UNASSIGNED: RNA sequencing (RNA-seq) results showed altered transferrin expression in the retina and RPE of Vldlr-/- mice. Disrupted iron homeostasis was observed in the retina and RPE of Vldlr-/- mice. DFP mitigated iron overload and significantly reduced retinal neovascularization and vascular leakage. In addition, DFP suppressed the inflammation in Vldlr-/- retinas. The reduced signals of macrophages were observed at sites of neovascularization in the retina and RPE of Vldlr-/- mice after DFP treatment. Further, the IL-6/JAK2/STAT3 signaling pathway was activated in the retina and RPE of Vldlr-/- mice and reversed by DFP treatment.
    UNASSIGNED: Disrupted iron metabolism may contribute to retinal neovascularization in nAMD. Restoring iron homeostasis by DFP could be a potential therapeutic approach for nAMD.
    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:眼部新生血管性疾病,这显著导致了视力丧失,缺乏有效的预防治疗。最近的研究强调了免疫细胞在新生血管性视网膜病变中的重要参与。骨髓来源的抑制细胞(MDSCs)促进新生血管的发展,但尚不清楚它们是否参与病理性新生血管形成,以及它们是否有望成为治疗靶点。
    方法:我们使用氧诱导的视网膜病变(OIR)模型研究了MDSCs在促进病理性血管生成中的作用,采用流式细胞术,免疫荧光,和smart-seq分析。然后,我们使用流式细胞术评估了患者血液样本中MDSCs的比例.此外,我们使用抗Gr-1单克隆抗体评估了MDSC耗竭对视网膜血管病变和视网膜小胶质细胞改变的影响.
    结果:在OIR模型中,在II期(新生血管形成)期间,血液和视网膜组织中的MDSCs比例均升高.MDSCs的耗竭导致视网膜新生血管和血管闭塞减少,随着新生血管区域内小胶质细胞的减少。此外,与MDSCs相关的基因转录本分析显示血管内皮生长因子(VEGF)调节和炎症的激活。重要的是,ROP患儿血液样本中MDSC的比例较高.
    结论:我们的结果表明,过多的MDSCs代表了眼部新生血管疾病的一个未被识别的特征,并负责视网膜血管炎症和血管生成,为眼部新生血管性疾病的新治疗方法提供了机会。
    BACKGROUND: Ocular neovascular diseases, which contribute significantly to vision loss, lack effective preventive treatments. Recent studies have highlighted the significant involvement of immune cells in neovascular retinopathy. Myeloid-derived suppressor cells (MDSCs) promote the development of neovascularization, but it is unknown whether they participate in pathological neovascularization and whether they are expected to be a therapeutic target.
    METHODS: We investigated the role of MDSCs in promoting pathological angiogenesis using an oxygen-induced retinopathy (OIR) model, employing flow cytometry, immunofluorescence, and smart-seq analysis. Then, we evaluated the proportion of MDSCs in patient blood samples using flow cytometry. Additionally, we assessed the effect of MDSC depletion using an anti-Gr-1 monoclonal antibody on retinal vasculopathy and alterations in retinal microglia.
    RESULTS: In the OIR model, an elevated ratio of MDSCs was observed in both blood and retinal tissue during phase II (Neovascularization). The depletion of MDSCs resulted in reduced retinal neovascularization and vaso-obliteration, along with a decrease in microglia within the neovascularization area. Furthermore, analysis of gene transcripts associated with MDSCs indicated activation of vascular endothelial growth factor (VEGF) regulation and inflammation. Importantly, infants with ROP exhibited a higher proportion of MDSCs in their blood samples.
    CONCLUSIONS: Our results suggested that excessive MDSCs represent an unrecognized feature of ocular neovascular diseases and be responsible for the retinal vascular inflammation and angiogenesis, providing opportunities for new therapeutic approaches to ocular neovascular disease.
    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
    这项工作的目的是确定hsa_circ_0004776在糖尿病视网膜病变(DR)进展中的调节功能。hsa_circ_0004776与hsa-miR-382-5p以及hsa-miR-382-5p与BDNF之间的直接相互作用,通过双荧光素酶报告基因测定证实。实时定量PCR分析表明,在高糖环境下,DR患者的房水样本和人视网膜微血管上皮细胞(hRECs)中hsa_circ_0004776高表达,而hsa-miR-382-5p显示相反的趋势。过表达hsa_circ_0004776显著增强DNA合成,扩散,迁移,和高血糖的hRECs中的管形成,而hsa-miR-382-5p模拟逆转了这些变化。此外,在链脲佐菌素诱导的SD大鼠DR模型中,玻璃体显微注射rno-miR-382-5pagomir逆转了DR进展的病理特征,包括视网膜血管渗漏,毛细管去细胞化,周细胞的损失,纤维化,和神经胶质增生。我们的结果表明,在高血糖条件下,hsa_circ_0004776通过hsa-miR-382-5p影响DR的进展,因此代表潜在的治疗靶标。
    The aim of this work was to identify the regulatory function of hsa_circ_0004776 in the progression of diabetic retinopathy (DR). The direct interactions between hsa_circ_0004776 and hsa-miR-382-5p and between hsa-miR-382-5p and BDNF, were confirmed via dual-luciferase reporter assays. Quantitative Real-Time PCR analysis indicated that hsa_circ_0004776 was highly expressed in aqueous humour samples of DR patients and human retinal microvascular epithelial cells (hRECs) under a high-glucose environment, whereas hsa-miR-382-5p showed the opposite trend. Overexpressed hsa_circ_0004776 significantly enhanced DNA synthesis, proliferation, migration, and tube formation in hRECs in hyperglycaemia, while hsa-miR-382-5p mimics reversed these changes. Additionally, in a streptozotocin-induced Sprague-Dawley rat model of DR, vitreous microinjection of rno-miR-382-5p agomir reversed the pathologic features in the progression of DR, including retinal vascular leakage, capillary decellularization, loss of pericytes, fibrosis, and gliosis. Our results indicated that under hyperglycaemic conditions, hsa_circ_0004776 influences the progression of DR via hsa-miR-382-5p and thus represents a potential therapeutic target.
    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:早产儿视网膜病变(ROP)是一种发生在早产儿视网膜上的增生性视网膜血管疾病,是导致儿童失明的主要原因。抗VEGF和视网膜光凝是目前ROP的主流治疗方法,但是它们会出现各种并发症。氢气(H2)被广泛认为是缺氧缺血性疾病的有用的神经保护和抗氧化治疗方法,而没有毒性作用。然而,H2是否提供生理血管生成促进,ROP进展中的新生血管抑制和神经胶质保护作用在很大程度上是未知的。本研究旨在探讨H2对视网膜血管生成的影响,氧诱导的视网膜病变(OIR)小鼠视网膜的新生血管形成和神经胶质功能障碍。
    方法:在本研究中,将7日龄和野生型(WT)或Nrf2缺陷(Nrf2-/-)的小鼠暴露于75%的氧气中5天,然后恢复到正常的空气条件。给予不同阶段的氢气(H2)吸入。血管闭塞,新生血管形成,并对血管渗漏进行分析比较。计算新生血管内皮细胞核的数量,行视网膜切片常规HE染色。使用DyLight594标记的GSLI-isolectinB4(IB4)进行免疫组织化学,以及针对增殖细胞核抗原(PCNA)的一级抗体,胶质纤维酸性蛋白(GFAP),Iba-1蛋白质印迹法检测NF-E2相关因子2(Nrf2)的表达,血管内皮生长因子(VEGF),Notch1、Dll4和HIF-1α。此外,测定靶基因如NQO1、HO-1、Notch1、Hey1、Hey2和Dll4的表达。在低氧条件下用H2处理的人脐静脉内皮细胞(HUVECs)用作体外模型。RT-PCR检测Nrf2、Notch/Dll4和靶基因的mRNA表达。使用免疫荧光染色观察活性氧(ROS)的表达。
    结果:我们的结果表明,3-4%H2不会干扰视网膜生理性血管生成,但改善OIR小鼠的血管闭塞和新生血管形成。此外,H2可防止密度降低,并逆转氧诱导损伤引起的视网膜星形胶质细胞的形态和功能变化。此外,H2吸入减少小胶质细胞活化,特别是在OIR小鼠的新生血管形成领域。H2通过在体内促进Nrf2活化和抑制Dll4诱导的Notch信号通路在血管再生中起保护作用。此外,H2通过负调节Dll4/Notch途径并通过Nrf2途径降低ROS水平来促进低氧下HUVECs的增殖,这与我们在体内的发现一致。此外,视网膜氧敏感机制(HIF-1α/VEGF)也参与氢介导的视网膜血管再生和新生血管抑制.
    结论:总的来说,我们的结果表明,H2可能是一种有前景的POR治疗药物,其在人ROP中的有益作用可能涉及Nrf2-Notch轴以及HIF-1α/VEGF通路的激活.
    BACKGROUND: Retinopathy of Prematurity (ROP) is a proliferative retinal vascular disease occurring in the retina of premature infants and is the main cause of childhood blindness. Nowadays anti-VEGF and retinal photocoagulation are mainstream treatments for ROP, but they develop a variety of complications. Hydrogen (H2) is widely considered as a useful neuroprotective and antioxidative therapeutic method for hypoxic-ischemic disease without toxic effects. However, whether H2 provides physiological angiogenesis promotion, neovascularization suppression and glial protection in the progression of ROP is largely unknown.This study aims to investigate the effects of H2 on retinal angiogenesis, neovascularization and neuroglial dysfunction in the retinas of oxygen-induced retinopathy (OIR) mice.
    METHODS: In this study, mice that were seven days old and either wild-type (WT) or Nrf2-deficient (Nrf2-/-) were exposed to 75% oxygen for 5 days and then returned to normal air conditions. Different stages of hydrogen gas (H2) inhalation were administered. Vascular obliteration, neovascularization, and blood vessel leakage were analyzed and compared. To count the number of neovascularization endothelial nuclei, routine HE staining of retinal sections was conducted. Immunohistochemistry was performed using DyLight 594 labeled GSL I-isolectin B4 (IB4), as well as primary antibodies against proliferating cell nuclear antigen (PCNA), glial fibrillary acidic protein (GFAP), and Iba-1. Western blots were used to measure the expression of NF-E2-related factor 2 (Nrf2), vascular endothelial growth factor (VEGF), Notch1, Dll4, and HIF-1α. Additionally, the expression of target genes such as NQO1, HO-1, Notch1, Hey1, Hey2, and Dll4 was measured. Human umbilical vein endothelial cells (HUVECs) treated with H2 under hypoxia were used as an in vitro model. RT-PCR was used to evaluate the mRNA expression of Nrf2, Notch/Dll4, and the target genes. The expression of reactive oxygen species (ROS) was observed using immunofluorescence staining.
    RESULTS: Our results indicate that 3-4% H2 does not disturb retinal physiological angiogenesis, but ameliorates vaso-obliteration and neovascularization in OIR mice. Moreover, H2 prevents the decreased density and reverses the morphologic and functional changes in retinal astrocytes caused by oxygen-induced injury. In addition, H2 inhalation reduces microglial activation, especially in the area of neovascularization in OIR mice. H2 plays a protective role in vascular regeneration by promoting Nrf2 activation and suppressing the Dll4-induced Notch signaling pathway in vivo. Also, H2 promotes the proliferation of HUVECs under hypoxia by negatively regulating the Dll4/Notch pathway and reducing ROS levels through Nrf2 pathway aligning with our findings in vivo.Moreover, the retinal oxygen-sensing mechanisms (HIF-1α/VEGF) are also involved in hydrogen-mediated retinal revascularization and neovascularization suppression.
    CONCLUSIONS: Collectively, our results indicate that H2 could be a promising therapeutic agent for POR treatment and that its beneficial effect in human ROP might involve the activation of the Nrf2-Notch axis as well as HIF-1α/VEGF pathways.
    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:糖尿病视网膜病变(DR)是糖尿病的常见并发症,和最近的研究结果表明,长链非编码RNA(lncRNAs)可能参与其发病机制。通过生物信息学分析,我们发现lncRNAATP2B2-IT2可能参与了这一过程。这项研究主要研究了高糖条件下lncRNAATP2B2-IT2在人视网膜微血管内皮细胞(HRMEC)中的表达及其对HRMEC增殖的影响。迁移,和新血管形成。
    方法:我们使用RT-PCR评估了正常葡萄糖(5.5mmol/L)和高糖(30mmol/L)条件下HRMEC中lncRNAATP2B2-IT2和血管内皮生长因子(VEGF)的表达水平。随后将HRMEC分为四组:正常葡萄糖(NG),高葡萄糖(HG),高葡萄糖与lncRNAATP2B2-IT2沉默(HGsi-lncRNAATP2B2-IT2),和高糖沉默对照组(HG+si-NC)。使用RT-PCR测定各组中lncRNAATP2B2-IT2和VEGF的表达水平。此后,细胞增殖,迁移,使用CCK-8,Transwell,和试管形成测定,分别。
    结果:RT-PCR显示,HG组lncRNAATP2B2-IT2和VEGF的表达水平高于NG组(P<0.05)。lncRNAATP2B2-IT2沉默后,VEGF的表达显著降低(P<0.05)。随后的CCK-8,Transwell,和试管形成试验表明,与NG组相比,HG组的HRMEC表现出显著增加的增殖,迁移,新生血管形成(P<0.05)。然而,lncRNAATP2B2-IT2沉默后,增殖,迁移,与HG组相比,HGsi-lncRNAATP2B2-IT2组的HRMECs新生血管明显减少(P<0.05)。
    结论:LncRNAATP2B2-IT2可能促进细胞增殖,高葡萄糖条件下HRMEC的迁移和新生血管形成。
    OBJECTIVE: Diabetic retinopathy (DR) is a common complication of diabetes, and recent findings have shown that long noncoding RNAs (lncRNAs) may be involved in its pathogenesis. Through bioinformatics analysis, we found that lncRNA ATP2B2-IT2 may be involved in this process. This study primarily investigated the expression of the lncRNA ATP2B2-IT2 in human retinal microvascular endothelial cells (HRMECs) under high-glucose conditions and its effects on HRMEC proliferation, migration, and neovascularization.
    METHODS: We used RT‒PCR to assess the expression levels of lncRNA ATP2B2-IT2 and vascular endothelial growth factor (VEGF) in HRMECs under normal glucose (5.5 mmol/L) and high glucose (30 mmol/L) conditions. HRMECs were subsequently divided into four groups: the normal glucose (NG), high glucose (HG), high glucose with lncRNA ATP2B2-IT2 silencing (HG + si-lncRNA ATP2B2-IT2), and high glucose with silencing control (HG + si-NC) groups. The expression levels of the lncRNA ATP2B2-IT2 and VEGF in each group were determined using RT‒PCR. Thereafter, cell proliferation, migration, and neovascularization were assessed using CCK-8, Transwell, and tube formation assays, respectively.
    RESULTS: RT‒PCR revealed that the expression levels of the lncRNA ATP2B2-IT2 and VEGF were greater in the HG group than in the NG group (P < 0.05). After silencing of the lncRNA ATP2B2-IT2, the expression of VEGF decreased significantly (P < 0.05). Subsequent CCK-8, Transwell, and tube formation assays demonstrated that compared to those in the NG group, the HRMECs in the HG group exhibited significantly increased proliferation, migration, and neovascularization (P < 0.05). However, after silencing of the lncRNA ATP2B2-IT2, the proliferation, migration, and neovascularization of HRMECs were significantly decreased in the HG + si-lncRNA ATP2B2-IT2 group compared to those in the HG group (P < 0.05).
    CONCLUSIONS: LncRNA ATP2B2-IT2 may promote the proliferation, migration and neovascularization of HRMECs under high-glucose conditions.
    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
    眼底新生血管疾病是一系列严重损害视力的致盲眼病。目前,在临床实践中治疗这些疾病的手段在不断发展,并迅速彻底改变了治疗意见。然而,诸如治疗有效性不足等关键问题,高复发率,和不良的患者依从性仍然需要紧急解决。多功能纳米药物可以对内源性和外源性微环境做出特异性反应,有效地将药物递送到特定的靶标,并参与生物成像和小分子检测等活动。纳米在微(NIM)输送系统,如金属,金属氧化物和上转换纳米颗粒(NPs),量子点,和碳材料,在克服眼球内生理屏障的存在方面显示出一定的优势,并被广泛用于眼科疾病的治疗。很少有研究,然而,已经评估了NIM递送系统治疗眼底新生血管性疾病(FND)的疗效。本研究描述了与使用NIM递送系统治疗FND相关的主要临床治疗策略和不良事件,并总结了必须克服的解剖学障碍。在这次审查中,我们希望强调眼内微环境正常化的原理,旨在为设计新的NIM输送系统以治疗特定FND提供更合理的方法。
    Fundus neovascularization diseases are a series of blinding eye diseases that seriously impair vision worldwide. Currently, the means of treating these diseases in clinical practice are continuously evolving and have rapidly revolutionized treatment opinions. However, key issues such as inadequate treatment effectiveness, high rates of recurrence, and poor patient compliance still need to be urgently addressed. Multifunctional nanomedicine can specifically respond to both endogenous and exogenous microenvironments, effectively deliver drugs to specific targets and participate in activities such as biological imaging and the detection of small molecules. Nano-in-micro (NIM) delivery systems such as metal, metal oxide and up-conversion nanoparticles (NPs), quantum dots, and carbon materials, have shown certain advantages in overcoming the presence of physiological barriers within the eyeball and are widely used in the treatment of ophthalmic diseases. Few studies, however, have evaluated the efficacy of NIM delivery systems in treating fundus neovascular diseases (FNDs). The present study describes the main clinical treatment strategies and the adverse events associated with the treatment of FNDs with NIM delivery systems and summarizes the anatomical obstacles that must be overcome. In this review, we wish to highlight the principle of intraocular microenvironment normalization, aiming to provide a more rational approach for designing new NIM delivery systems to treat specific FNDs.
    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
    视网膜新生血管(RNV),大多数新生血管性疾病的典型病理表现,导致视网膜脱离,视力丧失,最终不可逆转的失明。针对RNV开发了抗VEGF药物的重复玻璃体内注射,具有不完全反应和不利影响的局限性。因此,需要一种疗效更好、剂量更长的新疗法。这里,我们通过用拮抗剂C176抑制cGAS-STING信号诱导巨噬细胞极化为抗炎M2表型,从而了解cGAS-STING信号在视网膜中在促炎M1极化中的作用.C176负载和磷脂酰丝氨酸修饰的树枝状介孔二氧化硅纳米颗粒通过单次玻璃体内注射构建和检查。生物安全的纳米颗粒被视网膜巨噬细胞通过磷脂酰丝氨酸介导的“吃我”信号吞噬,持续释放C176以抑制STING信号传导,从而特异性促进巨噬细胞M2极化。单一剂量可有效缓解小鼠氧诱导视网膜病变模型中的病理性血管生成表型。总之,这些C176负载的纳米颗粒具有增强的细胞摄取和持久的STING抑制作用,可能是治疗RNV的有希望的方法。
    Retinal neovascularization (RNV), a typical pathological manifestation involved in most neovascular diseases, causes retinal detachment, vision loss, and ultimately irreversible blindness. Repeated intravitreal injections of anti-VEGF drugs were developed against RNV, with limitations of incomplete responses and adverse effects. Therefore, a new treatment with a better curative effect and more prolonged dosage is demanding. Here, we induced macrophage polarization to anti-inflammatory M2 phenotype by inhibiting cGAS-STING signaling with an antagonist C176, appreciating the role of cGAS-STING signaling in the retina in pro-inflammatory M1 polarization. C176-loaded and phosphatidylserine-modified dendritic mesoporous silica nanoparticles were constructed and examined by a single intravitreal injection. The biosafe nanoparticles were phagocytosed by retinal macrophages through a phosphatidylserine-mediated \"eat me\" signal, which persistently release C176 to suppress STING signaling and thereby promote macrophage M2 polarization specifically. A single dosage can effectively alleviate pathological angiogenesis phenotypes in murine oxygen-induced retinopathy models. In conclusion, these C176-loaded nanoparticles with enhanced cell uptake and long-lasting STING inhibition effects might serve as a promising way for treating RNV.
    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
    由视网膜缺血引起的病理性眼部新生血管形成构成视力丧失的主要原因。当前的抗VEGF疗法依赖于贝伐单抗(Beva)的繁重的玻璃体内注射。本文开发了包封Beva(P@Beva)的超小聚合物胶束,用于非侵入性局部递送至眼后部组织。Beva通过超支化两亲共聚物的自组装有效地加载到11nm胶束中。中立的,刷状胶束表现出优异的药物包封和胶体稳定性。体外,与游离药物相比,P@Beva增强眼细胞中Beva的细胞内递送。眼药水后的离体角膜和结膜-巩膜-脉络膜组织运输改善了23倍和7.9倍,分别。与单独的Beva相比,P@Beva保留了抗血管生成生物活性,从而引起对内皮管形成和脉络膜发芽的更大抑制。值得注意的是,在氧诱导的视网膜病变(OIR)模型中,玻璃体内注射Beva的局部P@Beva匹配疗效,临床标准。全面的生物相容性验证了安全性。总的来说,这个开创性的蛋白质递送平台有望将范例从侵入性玻璃体内注射转向简化,针对后眼疾病的生物治疗的非侵入性给药。本文受版权保护。保留所有权利。
    Pathological ocular neovascularization resulting from retinal ischemia constitutes a major cause of vision loss. Current anti-VEGF therapies rely on burdensome intravitreal injections of Bevacizumab (Beva). Herein ultrasmall polymeric micelles encapsulating Beva (P@Beva) are developed for noninvasive topical delivery to posterior eye tissues. Beva is efficiently loaded into 11 nm micelles fabricated via self-assembly of hyperbranched amphiphilic copolymers. The neutral, brush-like micelles demonstrate excellent drug encapsulation and colloidal stability. In vitro, P@Beva enhances intracellular delivery of Beva in ocular cells versus free drug. Ex vivo corneal and conjunctival-sclera-choroidal tissues transport after eye drops are improved 23-fold and 7.9-fold, respectively. Anti-angiogenic bioactivity is retained with P@Beva eliciting greater inhibition of endothelial tube formation and choroid sprouting over Beva alone. Remarkably, in an oxygen-induced retinopathy (OIR) model, topical P@Beva matching efficacy of intravitreal Beva injection, is the clinical standard. Comprehensive biocompatibility verifies safety. Overall, this pioneering protein delivery platform holds promise to shift paradigms from invasive intravitreal injections toward simplified, noninvasive administration of biotherapeutics targeting posterior eye 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
    早产儿视网膜病(ROP)是一种引起视网膜疾病的视网膜新生血管形成,可导致失明。氧诱导的视网膜病变(OIR)是广泛使用的ROP动物模型。淫羊藿苷(ICA)具有抗氧化和抗炎特性;然而,ICA是否对OIR有调节作用尚不清楚。在这项研究中,ICA减轻病理性新生血管形成,体内小胶质细胞活化和血视网膜屏障(BRB)损伤。进一步的结果表明,内皮细胞管的形成,ICA处理可在体外恢复迁移和增殖。蛋白质组学芯片和分子模拟显示,ICA可以直接结合己糖激酶2(HK2)并在体内和体外降低HK2蛋白的表达。此外,ICA抑制AKT/mTOR/HIF1α通路的激活。ICA对病理性新生血管的影响,体内HK2过表达后,小胶质细胞活化和BRB损伤消失。同样,HK2过表达后,内皮细胞功能得到改善。HK2过表达在体内和体外可逆转ICA诱导的AKT/mTOR/HIF1α途径抑制。因此,ICA以HK2依赖性方式预防OIR中的病理性血管生成,暗示ICA是ROP的潜在治疗剂。
    Retinopathy of prematurity (ROP) is a retinal disease-causing retinal neovascularization that can lead to blindness. Oxygen-induced retinopathy (OIR) is a widely used ROP animal model. Icariin (ICA) has anti-oxidative and anti-inflammation properties; however, whether ICA has a regulatory effect on OIR remains unclear. In this study, ICA alleviated pathological neovascularization, microglial activation and blood-retina barrier (BRB) damage in vivo. Further results indicated that endothelial cell tube formation, migration and proliferation were restored by ICA treatment in vitro. Proteomic microarrays and molecular mimicry revealed that ICA can directly bind to hexokinase 2 (HK2) and decrease HK2 protein expression in vivo and in vitro. In addition, ICA inhibited the AKT/mTOR/HIF1α pathway activation. The effects of ICA on pathological neovascularization, microglial activation and BRB damage disappeared after HK2 overexpression in vivo. Similarly, the endothelial cell function was revised after HK2 overexpression. HK2 overexpression reversed ICA-induced AKT/mTOR/HIF1α pathway inhibition in vivo and in vitro. Therefore, ICA prevented pathological angiogenesis in OIR in an HK2-dependent manner, implicating ICA as a potential therapeutic agent for ROP.
    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