首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 140 毫秒
1.
糖尿病视网膜病变是糖尿病最常见、最主要的微血管并发症之一,其高发病率、高致盲率特征严重威胁着人类的健康和生存质量。长期的高糖状态导致视网膜缺血缺氧是糖尿病视网膜病变的主要发病原因。控制高血糖和改善组织缺氧无疑是防治糖尿病微血管病变的有效方法之一。而高压氧治疗是许多急慢性疾病的首选或辅助治疗方法。已有大量证据表明,高压氧治疗对视网膜静脉阻塞及黄斑囊样水肿、缺血性视神经病变、中心性浆液性脉络膜视网膜病变等眼病安全有效。本文就高压氧在糖尿病视网膜病变中的应用进行综述和讨论。  相似文献   

2.
糖尿病视网膜病变是糖尿病最常见、最主要的微血管并发症之一,具有高发病率,高致盲率的特点,严重影响了人类的生存质量。控制高血糖和改善组织缺氧无疑是防治糖尿病微血管病变的有效方法。如果对糖尿病视网膜病变及时进行治疗,能延缓其发展并能提高病人的生活质量。近年来,随着对糖尿病发病机制的深入研究,很多方法用于防治糖尿病视网膜病人都取得了一定的疗效。而高压氧治疗是许多急慢性疾病的首选治疗方法。已有基础和临床研究证实,高压氧治疗对糖尿病视网膜病变安全有效。因此,作为一种新疗法,高压氧疗法可能会为糖尿病视网膜病变的治疗带来更广泛的应用前景。  相似文献   

3.
汪梦竹  蔡畅  刘洋  宋洪元  沈炜 《生命科学》2023,(10):1380-1387
糖尿病视网膜病变(diabetic retinopathy, DR)是糖尿病患者最常见的微血管系统并发症之一,是视力丧失的主要原因。增殖性糖尿病视网膜病变(proliferative diabetic retinopathy, PDR)是DR的终末期表现,其主要的病理生理学特征是视网膜新生血管形成(retinal neovascularization, RNV)。但PDR现有治疗方式存在局限性。外泌体作为细胞间沟通交流的重要使者,其携带的非编码RNA和生物活性蛋白质等重要信号分子,通过影响血管内皮细胞的增殖和迁移,在RNV中发挥关键作用。巨噬细胞是一种多功能调节细胞,越来越多的研究表明巨噬细胞外泌体在调控新生血管形成中起重要作用。该文就巨噬细胞外泌体在增殖性糖尿病视网膜病变形成中的作用与机制研究进展进行综述。  相似文献   

4.
本研究通过体外实验与网络药理学相结合的方法分析芍药苷治疗糖尿病视网膜病变的相关作用机制。制备高糖环境下视网膜血管内皮细胞的体外模型,探究芍药苷对糖尿病视网膜病变的保护作用;从TCMSP、 UniProt、 PubChem、 PharmMapper、 SwisstargetPrediction、 GeneCards、 OMIM、 DrugBank、 TTD数据库获取芍药苷和糖尿病视网膜病变的作用靶点,使用韦恩图获取核心靶点;基于STRING数据库,采用Cytoscape构建蛋白质-蛋白质相互作用网络和“药物-靶点-疾病”网络;应用DAVID数据库对核心靶点进行GO功能富集分析和KEGG通路富集分析,并用AutoDock软件将芍药苷与核心靶点进行分子对接验证。体外实验结果显示,芍药苷能显著抑制视网膜血管内皮细胞的增殖,降低其迁移能力并减少管腔形成长度。网络药理学结果显示共获取芍药苷治疗糖尿病视网膜病变的核心靶点154个,其中度值较高的靶点包括VEGFA、 IL6等。GO功能共富集到705个GO条目,主要涉及细胞对药物的反应、血管内皮细胞迁移的调节、细胞增殖的调节等生物过程;KEGG通路富集...  相似文献   

5.
糖尿病微血管病变严重影响了患者生活质量,是患者致死致残主要原因。微血管病变主要表现在视网膜、肾、神经、心肌组织。微血管病变的机制尚未完全清楚,近年越来越多研究发现血管内皮祖细胞(endothelial progenitor cells,EPCs)是该病发病重要原因。EPCs有分化为成熟的内皮细胞并且参与新血管形成和新生的能力。正常情况下内皮损失和EPCs对内皮的修复作用处于动态平衡状态,一旦EPCs受损,内皮损害和修复之间的平衡被打破,内皮层的完整性遭到破坏,必然参与糖尿病血管病变的发生发展。国内外大量研究证明糖尿病合并大血管病变EPCs数目功能改变,而糖尿病合并微血管病变EPCs的怎样变化?本文就EPCs与糖尿病微血管病变的关系进行系统综述。  相似文献   

6.
糖尿病的发病率逐年上升,其并发症的严重性日趋明显,特别是糖尿病视网膜病变导致视力下降和丧失已经引起了广泛关注,所以研究糖尿病视网膜病变的发病机制及其防治是必要的。糖尿病视网膜病变是一种多种机制共同作用的复杂性疾病,而细胞凋亡在糖尿病视网膜病变的发生和发展中起着重要的作用,所以研究细胞凋亡对糖尿病视网膜病变的治疗有着重要意义。由于细胞凋亡研究的深入,人们将注意力集中于糖尿病视网膜细胞凋亡能否得到抑制和逆转的问题上。研究发现,糖尿病视网膜病变细胞凋亡可能与视网膜新生血管形成、VEGF水平增高等因素有关。当前对葛根素的研究表明,葛根素能有效抑制视网膜新生血管形成,并且对于缺血、缺氧等因素引起的损害有很强的改善作用,葛根素还可以降低糖尿病糖基化终产物水平,甚至对视网膜超微结构的损害具有一定的保护作用,所以葛根素可能是治疗糖尿病性视网膜病变的新策略。本文就近期糖尿病视网膜病变中细胞凋亡的有关研究和葛根素的抗细胞凋亡作用做一综述,提示在糖尿病视网膜病变中葛根素的不可忽视的作用。  相似文献   

7.

视网膜血管疾病如早产儿视网膜病变、糖尿病视网膜病变和视网膜静脉阻塞等以异常增生的视网膜新生血管为主要病理表现。骨髓间充质干细胞来源外泌体通过旁分泌作用传递生物活性分子介导细胞间的物质与信息交换。其中,miRNA等内容物在传递信息中起关键作用,可调控缺血缺氧环境下内皮细胞的增殖、管腔形成和新生血管的形成。并且能够通过血视网膜屏障而不引起免疫、炎症反应,在眼科疾病治疗中极具潜力。本文总结骨髓间充质干细胞衍生外泌体中miRNA在视网膜新生血管形成中的作用和可能的作用机制,以期为外泌体在眼科疾病诊治中的应用拓宽新思路。  相似文献   


8.
封凡  许峰晟  杜飞  杨康鹃 《生命科学》2020,32(5):461-470
2型糖尿病(type 2 diabetes mellitus, T2DM)是一种复杂的以慢性高血糖为特征的代谢性疾病。近年来,越来越多的研究发现T2DM患者体内的高糖环境可以改变DNA甲基化程度,影响相关基因功能,从而导致糖代谢、脂肪代谢和能量代谢紊乱或炎症反应,进而造成血管病变。T2DM常见的血管并发症有大血管病变引起的心脑血管疾病,以及微血管病变引起的糖尿病肾病、糖尿病视网膜病变、糖尿病周围神经病变等。DNA甲基化标记在T2DM血管并发症的早期诊断与治疗中可能有重要意义,已成为相关领域的研究热点。现主要对近期DNA甲基化在T2DM血管并发症方面的研究及其研究策略加以综述,以期深入了解T2DM血管并发症发生发展的表观遗传机制和可能的防治途径。  相似文献   

9.
糖尿病视网膜病变引起的视力丧失目前已经引起了广泛的关注,但是由于糖尿病是一种多种机制作用的复杂疾病,对它的研究尚不清楚.本文就近期糖尿病视网膜病变中各种发病机制所致的视网膜血管和神经节细胞凋亡的有关研究做一综述.提示在糖尿病视网膜病变中各种机制均可引起视网膜血管和神经节细胞的凋亡,细胞凋亡在糖尿病视网膜病变的发生和发展中起着重要的作用.  相似文献   

10.
延长糖尿病模型大鼠生存期对糖尿病视网膜病变的影响   总被引:1,自引:1,他引:0  
目的延长糖尿病模型大鼠生存期,动态观察糖尿病视网膜病变(DR)的形成和发展过程。方法雄性SD大鼠70只,随机分成对照组(20只)和模型组(50只),采用链脲佐菌素(STZ)60 mg/(kg.bw)体重腹腔1次注射造模,分别于69、、12月时处死取眼球,采用视网膜微血管消化铺片技术观察糖尿病视网膜病变的微血管形态学改变。结果糖尿病大鼠DR样病变随着病程的延长病变呈多样性改变,以12月DR出现的小动脉硬化尤为严重。结论糖尿病大鼠生存期的延长对糖尿病视网膜病变的研究有着积极的意义。  相似文献   

11.
Diabetic retinopathy is a sight-threatening complication of diabetes, affecting 65% of patients after 10 years of the disease. Diabetic metabolic insult leads to chronic low-grade inflammation, retinal endothelial cell loss and inadequate vascular repair. This is partly due to bone marrow (BM) pathology leading to increased activity of BM-derived pro-inflammatory monocytes and impaired function of BM-derived reparative circulating angiogenic cells (CACs). We propose that diabetes has a significant long-term effect on the nature and proportion of BM-derived cells that circulate in the blood, localize to the retina and home back to their BM niche. Using a streptozotocin mouse model of diabetic retinopathy with GFP BM-transplantation, we have demonstrated that BM-derived circulating pro-inflammatory monocytes are increased in diabetes while reparative CACs are trapped in the BM and spleen, with impaired release into circulation. Diabetes also alters activation of splenocytes and BM-derived dendritic cells in response to LPS stimulation. A majority of the BM-derived GFP cells that migrate to the retina express microglial markers, while others express endothelial, pericyte and Müller cell markers. Diabetes significantly increases infiltration of BM-derived microglia in an activated state, while reducing infiltration of BM-derived endothelial progenitor cells in the retina. Further, control CACs injected into the vitreous are very efficient at migrating back to their BM niche, whereas diabetic CACs have lost this ability, indicating that the in vivo homing efficiency of diabetic CACs is dramatically decreased. Moreover, diabetes causes a significant reduction in expression of specific integrins regulating CAC migration. Collectively, these findings indicate that BM pathology in diabetes could play a role in both increased pro-inflammatory state and inadequate vascular repair contributing to diabetic retinopathy.  相似文献   

12.
Altered insulin signaling in retinal tissue in diabetic states   总被引:3,自引:0,他引:3  
Both type 1 and type 2 diabetes can lead to altered retinal microvascular function and diabetic retinopathy. Insulin signaling may also play a role in this process, and mice lacking insulin receptors in endothelial cells are protected from retinal neovascularization. To define the role of diabetes in retinal function, we compared insulin signaling in the retinal vasculature of mouse models of type 1 (streptozotocin) and type 2 diabetes (ob/ob). In streptozotocin mice, in both retina and liver, insulin receptor (IR) and insulin receptor substrate (IRS)-2 protein and tyrosine phosphorylation were increased by insulin, while IRS-1 protein and its phosphorylation were maintained. By contrast, in ob/ob mice, there was marked down-regulation of IR, IRS-1, and IRS-2 protein and phosphorylation in liver; these were maintained or increased in retina. In both mice, Phosphatidylinositol 3,4,5-trisphosphate generation by acute insulin stimulation was enhanced in retinal endothelial cells. On the other hand, protein levels and phosphorylation of PDK1 and Akt were decreased in retina of both mice. Interestingly, phosphorylation of p38 mitogen-activated protein kinase and ERK1 were responsive to insulin in retina of both mice but were unresponsive in liver. HIF-1alpha and vascular endothelial growth factor were increased and endothelial nitric-oxide synthase was decreased in retina. These observations indicate that, in both insulin-resistant and insulin-deficient diabetic states, there are alterations in insulin signaling, such as impaired PDK/Akt responses and enhanced mitogen-activated protein kinases responses that could contribute to the retinopathy. Furthermore, insulin signaling in retinal endothelial cells is differentially altered in diabetes and is also differentially regulated from insulin signaling in classical target tissues such as liver.  相似文献   

13.
BackgroundThe accumulation of advanced glycated end products (AGEs) in retinal blood vessels is one of the major etiological factors contributing to diabetic retinopathy. Aminoguanidine (AG) is one of the most extensively used inhibitors of AGEs formation. The aim of this study was to investigate whether AG could protect the development of diabetic retinopathy through inhibition of AGEs.MethodsRat diabetes was induced by intraperitoneal injection with streptozotocin (STZ). AG was given to rats in drinking water. Retina was extracted 3 and 6 months following STZ and AG administration. Immunochemistry and transmission electron microscope were used to detect the expression of AGEs and retina morphology.ResultsExtensive staining of AGEs was detected in retinal blood vessels of 3- and 6-month diabetic rats, while no significant staining was found in the control non-diabetic retina or AG treated groups. Pericyte loss, endothelial cell proliferation, increased ratio of endothelial cells/pericytes, acellular capillaries and capillary occlusion were observed in the retina of 6-month diabetic rats. The increased electron density of retinal capillary basement membrane, mitochondrial swelling in pericytes and endothelial cells were also found in 6-month diabetic rats. The 3-month diabetic rats and the AG-treated rats did not have similar morphological changes compared to control group. The AGEs staining in AG-treated rats was still weakly positive.ConclusionsAGEs plays pivotal roles in diabetic retinopathy. AGE deposition occurs prior to retinal microvasculature changes. AG could prevent the onset and development of diabetic retinopathy through inhibition of AGEs.  相似文献   

14.
To investigate the effect of protein kinase C (PKC)-ζ inhibition on vascular leakage in diabetic retinopathy, streptozotocin-induced diabetic mice were intravitreously injected with siPKC-ζ. According to the fluorescein angiography of the retinal vessels, suppression of PKC-ζ effectively attenuated vascular leakage in diabetic retina. Further evaluation on the retina with western blot analysis and immunohistochemistry revealed accompanying restoration of tight junction proteins on retinal vessels. As two major contributors to vascular leakage in diabetic retinopathy, vascular endothelial growth factor (VEGF) and advanced glycation end products (AGEs) were investigated on the tight junction protein expression in endothelial cells. Inhibition of PKC-ζ attenuated VEGF-induced decrease of tight junction proteins and accompanying hyperpermeability in human retinal microvascular endothelial cells (HRMECs). PKC-ζ inhibition also attenuated AGE-induced decrease of tight junction proteins in HRMECs. Our findings suggest that inhibition of PKC-ζ could be an alternative treatment option for compromised blood-retinal barrier in diabetic retinopathy.  相似文献   

15.
Diabetic retinopathy is one of the main microvascular complications of diabetes and remains one of the leading causes of blindness worldwide. Recent studies have revealed an important role of inflammatory and proangiogenic high mobility group 1 (HMGB-1) cytokine in diabetic retinopathy. To elucidate cellular mechanisms of HMGB-1 activity in the retina, we performed this study. The histological features of diabetic retinopathy include loss of blood-vessel pericytes and endothelial cells, as well as abnormal new blood vessel growth. To establish the role of HMGB-1 in vulnerability of endothelial cells and pericytes, cultures of these cells, or co-cultures with glial cells, were treated with HMGB-1 and assessed for survival after 24 hours. The expression levels of the cytokines, chemokines, and cell adhesion molecules in glial and endothelial cells were tested by quantitative RT-PCR to evaluate changes in these cells after HMGB-1 treatment. Animal models of neovascularization were also used to study the role of HMGB-1 in the retina. We report that pericyte death is mediated by HMGB-1-induced cytotoxic activity of glial cells, while HMGB-1 can directly mediate death of endothelial cells. We also found that HMGB-1 affects endothelial cell activity. However, we did not observe a difference in the levels of neovascularization between HMGB-1-treated eyes compared to the control eyes, nor in the levels of proangiogenic cytokine VEGF-A expression between glial cells treated with HMGB-1 and control cells. Our data also indicate that HMGB-1 is not involved in retinal neovascularization in the oxygen-induced retinopathy model. Thus, our data suggest that retinal pericyte and endothelial injury and death in diabetic retinopathy may be due to HMGB-1-induced cytotoxic activity of glial cells as well as the direct effect of HMGB-1 on endothelial cells. At the same time, our findings indicate that HMGB-1 plays an insignificant role in retinal and choroidal neovascularization.  相似文献   

16.
In the pathogenesis of diabetic retinopathy, H-Ras (a small molecular weight G-protein) and matrix metalloproteinase-9 (MMP9) act as pro-apoptotic, accelerating the apoptosis of retinal capillary cells, a phenomenon that predicts its development and the activation of MMP9 is under the control of H-Ras. The goal of this study is to elucidate the cellular mechanism by which H-Ras activates MMP9 culminating in the development of diabetic retinopathy. Using isolated retinal endothelial cells, the effect of regulation of H-Ras downstream signaling cascade, Raf-1, MEK, and ERK, was investigated on glucose-induced activation of MMP9. In vitro results were confirmed in the retina obtained from diabetic mice manipulated for MMP9 gene, and also in the retinal microvasculature obtained from human donors with diabetic retinopathy. Regulation of Raf-1/MEK/ERK by their specific siRNAs and pharmacologic inhibitors prevented glucose-induced activation of MMP9 in retinal endothelial cells. In MMP9-KO mice, diabetes had no effect on retinal MMP9 activation, and H-Ras/Raf-1/MEK signaling cascade remained normal. Similarly, donors with diabetic retinopathy had increased MMP9 activity in their retinal microvessels, the site of histopathology associated with diabetic retinopathy, and this was accompanied by activated H-Ras signaling pathway (Raf-1/ERK). Collectively, these results suggest that Ras/Raf-1/MEK/ERK cascade has an important role in the activation of retinal MMP9 resulting in the apoptosis of its capillary cells. Understanding the upstream mechanism responsible for the activation of MMP9 should help identify novel molecular targets for future pharmacological interventions to inhibit the development/progression of diabetic retinopathy.  相似文献   

17.
The progressive alterations to the retinal microvasculature in diabetic retinopathy are known to cause vision loss. Chemokines are characterized by their ability to induce cell invasion, adhesion and migration. In this study, we used double siRNA transfection to transiently and selectively decrease the level of the endogenous CXCR4 in human retinal microvascular endothelial cells (HRMECs). The functional consequences of silencing CXCR4 expression in HRMECs were investigated using an endothelial cell migration assay and tubule formation in Matrigel. When CXCR4 expression was decreased with siRNA, HRMECs were less invasive and also resulted in markedly diminished vascular networks on Matrigel as compared to the controls. Additionally, hypoxia and VEGF, the factors affecting microvascular, regulate the expression level of CXCR4 in HRMECs, respectively, which have synergistic, additive effect in the HRMECs. As such, CXCR4 antagonists may become a therapeutic target for the treatment of retinal angiopathies.  相似文献   

18.
Many molecular and cellular abnormalities detected in the diabetic retina support a role for IL-1β-driven neuroinflammation in the pathogenesis of diabetic retinopathy. IL-1β is well known for its role in the induction and, through autostimulation, amplification of neuroinflammation. Upregulation of IL-1β has been consistently detected in the diabetic retina; however, the mechanisms and cellular source of IL-1β overexpression are poorly understood. The aim of this study was to investigate the effect of high glucose and IL-1β itself on IL-1β expression in microglial, macroglial (astrocytes and Müller cells) and retinal vascular endothelial cells; and to study the effect of diabetes on the expression of IL-1β in isolated retinal vessels and on the temporal pattern of IL-1β upregulation and glial reactivity in the retina of streptozotocin-diabetic rats. IL-1β was quantified by RealTime RT-PCR and ELISA, glial fibrillar acidic protein, α2-macroglobulin, and ceruloplasmin by immunoblotting. We found that high glucose induced a 3-fold increase of IL-1β expression in retinal endothelial cells but not in macroglia and microglia. IL-1β induced its own synthesis in endothelial and macroglial cells but not in microglia. In retinal endothelial cells, the high glucose-induced IL-1β overexpression was prevented by calphostin C, a protein kinase C inhibitor. The retinal vessels of diabetic rats showed increased IL-1β expression as compared to non-diabetic rats. Retinal expression of IL-1β increased early after the induction of diabetes, continued to increase with progression of the disease, and was temporally associated with upregulation of markers of glial activation. These findings point to hyperglycemia as the trigger and to the endothelium as the origin of the initial retinal upregulation of IL-1β in diabetes; and to IL-1β itself, via autostimulation in endothelial and macroglial cells, as the mechanism of sustained IL-1β overexpression. Interrupting the vicious circle triggered by IL-1β autostimulation could limit the progression of diabetic retinopathy.  相似文献   

19.
《Free radical research》2013,47(9):1083-1092
Abstract

The aim of this study was to determine the reasons why the intravitreal level of extracellular-superoxide dismutase (EC-SOD) increases in proliferative diabetic retinopathy patients by the investigation of two possibilities: first, change of EC-SOD expression in the retina; and secondly, leakage of EC-SOD through the endothelial monolayer by the treatment with endoplasmic reticulum (ER) stress inducers because ER stress is known to be involved in the vascular impairment in diabetic retinopathy. Intravitreous injection of tunicamycin in mice increased the permeability of tracer dye across retinal blood vessels while the retinal EC-SOD mRNA level was not changed. The leakage of EC-SOD through the retinal endothelial cell layer was elevated by the treatment with thapsigargin or tunicamycin. The expression of claudin-5 was significantly decreased by the treatment with the ER stress inducers. These phenomena were significantly suppressed by the pre-treatment of endothelial cells with a chemical chaperone 4-phenylbutyric acid. Our observations suggest that ER stress leads to the down-regulation of claudin-5 among tight junction proteins and may induce the elevation of endothelial permeability and leakage of EC-SOD into the vitreous body.  相似文献   

20.
Angiotensin and diabetic retinopathy   总被引:2,自引:0,他引:2  
Diabetic retinopathy develops in patients with both type 1 and type 2 diabetes and is the major cause of vision loss and blindness in the working population. In diabetes, damage to the retina occurs in the vasculature, neurons and glia resulting in pathological angiogenesis, vascular leakage and a loss in retinal function. The renin-angiotensin system is a causative factor in diabetic microvascular complications inducing a variety of tissue responses including vasoconstriction, inflammation, oxidative stress, cell hypertrophy and proliferation, angiogenesis and fibrosis. All components of the renin-angiotensin system including the angiotensin type 1 and angiotensin type 2 receptors have been identified in the retina of humans and rodents. There is evidence from both clinical and experimental models of diabetic retinopathy and hypoxic-induced retinal angiogenesis that the renin-angiotensin system is up-regulated. In these situations, retinal dysfunction has been linked to angiotensin-mediated induction of growth factors including vascular endothelial growth factor, platelet-derived growth factor and connective tissue growth factor. Evidence to date indicates that blockade of the renin-angiotensin system can confer retinoprotection in experimental models of diabetic retinopathy and ischemic retinopathy. This review examines the role of the renin-angiotensin system in diabetic retinopathy and the potential of its blockade as a treatment strategy for this vision-threatening disease.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号