Notch信号途径对高糖条件下NF-κB和PARP介导的视网膜细胞凋亡的保护作用
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
本实验成功建立了糖尿病视网膜病变的小鼠模型,应用免疫组织化学法和Western Blot方法检测糖尿病视网膜病变小鼠视网膜中Notch1、Dll4、PARP、Akt、NF-κB和caspase-3表达量的变化,同时体外培养的人视网膜血管内皮细胞HRVEC,建立高糖培养的视网膜内皮细胞模型,应用免疫印记方法检测高糖培养的视网膜血管内皮细胞由Notch1、Dll4、PARP、Akt、NF-κB和caspase-3表达量的变化,发现糖尿病小鼠模型的视网膜和高糖培养的HRVECs中的PARP和caspase-3表达量比正常对照组增高显著,并与葡萄糖浓度正相关;而Notch1、Dll4及p-Akt表达量随葡萄糖浓度的降低而显著降低,说明高糖引起的细胞凋亡与PARP增加和Notch1、p-Akt下降有关。进一步通过免疫共沉淀及激光共聚焦的方法证明了PARP和NF-κB是相互作用的蛋白质,在高糖情况下,与PARP结合的NF-κB蛋白明显增多,说明PARP通过激活NF-κB诱导细胞凋亡。另外通过Western Blot检测高糖下Notch1拮抗PARP-1和NF-κB介导的细胞凋亡,发现Notch1能够抑制高糖高糖引起的细胞凋亡。另外本实验还体外构建了siNotch1表达载体,应用免疫共沉淀及免疫印记的方法检测siNotch1对D114的抑制作用,发现siNotch1能抑制D114的抗凋亡作用,即说明Notch信号对高糖导致的细胞凋亡有保护作用。另外,利用Akt特异抑制剂wortmannin研究Akt与Notch1的关系,发现wortmannin能抑制高糖情况下Notch1对细胞的保护作用。
     本实验的创新之处在于:
     1、证明了Notch1、p-Akt及D114蛋白表达量在高糖培养的HRVEC中比正常HRVEC显著降低。
     2、本实验发现并且证明了在高糖培养的视网膜血管内皮细胞中,Notch1/Akt信号途径能够抑制PARP-1和NF-κB介导的HRVECs凋亡
Diabetic retinopathy is one of serious microvascular complications caused by diabetes at later stage and is very common in clinic which leads to visual loss and blindness ultimately. As people's living standards and diet changes, the prevalence of diabetes increased and leads to the increase of the incidence of diabetic retinopathy significantly. Its incidence increased with age and longer duration of diabetes, the longer the duration of diabetes continued, the higher the changes in eye fundus occurs. The main symptoms of diabetic retinopathy are flash flu and vision loss and its fudus changes mainly contain capillary hemangioma, bleeding spots, hard exudates, cotton wool spots, retinal vascular disease, macular degeneration, vitreous and optic neuropathy and etc.
     So far, the pathogenesis mechanism of diabetic retinopathy mainly contains oxidative stress, advanced glycosylation end products formation, polyol pathway hyperthyroidism, protein kinase C activation, the effect of vascular endothelial growth factor (VEGF), basic fibroblast growth factor(bFGF), insulin-like grow factor-1(IGF-1), tumor mecrosis factor-α(TNF-α), transforming growth factor-β(TGF-β) and platelet-derived growth factor-B (PDGF-B). Some signal transduction pathway also involved in the pathological process of diabetic retinopathy. Recent studies indicated that PARP also participated in this process. The mechanism of diabetic retinopathy is still not completely clear because it is very intrcated. Studies indicated that Notch1/Dll4 played an important role in the process of retinal vascular development. The missing of either Notch1 or Dll4 will lead to serious deficiencies in retinal blood vessels which causes vascular diseases. So we speculate that Notchl also participated in the pathological process of diabetic retinopathy. This study successfully established early mouse model of diabetic retinopathy and high glucose in vitro cell model which were used to study the relationship between Notchl and the diabetic retinopathy. This study provided a new idea and theoretical foundation for further study the mechanism, and also provides a new target for treatment of the diseases.
     1. The expression of Notch1、D114、PARP、Akt、NF-κB and caspase-3 in the retina of diabetic mouse and in human retinal vascular endothelial cell exposed to high glucose.
     Bcl57/6 mice were intraperitoneal injected with streptozotocin to establish mouse model of early diabetic retinopathy which was always the in vivo animal model to study the pathogenesis of diabetic. And culturing retinal vascular endothelial cells exposed to high glucose in vitro was always the in vitro model to study the pathogenesis mechanism of diabetic. We still use these two models to detect the relationship among each protein and high glucose, and the expression changes. It has been proved that PARP expressed in retinal vascular endothelial cells in STZ-induced diabetic rats indicating that PARP participated in the pathogenesis mechanism of diabetic. High glucose also can activate NF-κB which can lead to the expression of proapoptosis gene and cause cell apoptosis. It has been proved that Akt also involved in, ROS generated in high glucose can dephosphorylated Akt and inhibit the activation of Akt, leading to human umbilical vascular endothelial cells(HUVECs) apoptosis. These protein and signal pathways were all related to the pathological process of diabetic retinopathy.
     Thus, we take advantage of animal and cell model and use immunohistochemical and western blot methods to detect the expression changes of Notch1, Dll4, PARP, Akt, NF-κB and caspase-3 in the retina of diabetic mouse and in human retinal vascular endothelial cell exposed to high glucose. We found that PARP and caspase-3 protein have low expression in normal mouse retina and HRVEC, and they have high expression in in the retina of diabetic mouse and in human retinal vascular endothelial cell exposed to high glucose. We also found that Notch1, p-Akt and Dll4 protein have high expression in normal mouse retina and HRVEC, and they have low expression in in the retina of diabetic mouse and in human retinal vascular endothelial cell exposed to high glucose. These results illustrated that the occurrence of diabetic retinopathy was related with the decreasing of Notch1, p-Akt and Dll4 protein and the increasing of PARP expression.
     2. Notch1 signal pathway inhibits retinal vascular endothelial cells apoptosis through regulating PARP and NF-κB interaction.
     Notch is an important transduction pathway highly conserved in animals which mainly effects blood vessels growth during embryonic development and regulates arteriovenous differentiation and cell proliferation and death. This signal pathway also plays an important role in the retinal vascular development. Notch signal can maintain the normal physiological environment of the retinal blood vessels. The missing of Notch can lead to retinal vascular and cell dysplasia, and retinal vascular homeostasis disorders indicating that Notch1 may involved in the pathogenesis mechanism of diabetic retinopathy. Our studies conformed that high glucose causes Notch1 decreasing also proved this view. In addition, it is also reported that high glucose can increase PARP, then PARP activate NF-κB which can then activate caspase-3 leading to cell apoptosis. Notchl may have relation with it.
     Thus, we take advantage of immunoprecipitation and confocal laser scanning microscope to detect PARP and NF-κB interation under high glucose. We found that PARP and NF-κB are interacted proteins, and PARP enters into nucleus after activation to combine with NF-κB which can activate apoptosis gene to induce cell apoptosis. In addition, we use Western Blot and immunoprecipitation to detect the relationship among PARP, NF-κB and Notch1, finding that Notchl can inhibit PARP and NF-κB mediated cell apoptosis under high glucose, which indicates Notch1 can protect the retina from injury under high glucose.
     3. SiNotch1 and Wortmannin, a specific inhibitor of Akt, inhibit the protective effect of Notch1 on retina
     RNA interference is a defense mechanism that is highly conserved in evolution which is used to against the foreign gene or foreign virus. RNAi technology was intend to synthesize siRNA in vitro using Chemical and enzymatic methods through RNAi theory or construct plasmid and viral vector carrying siRNA, which then transfected into the cells to make the target gene silence. It was a common method that used in studying signal transduction. We also take advantage of this method to investigate the protective effect of Notchl on retina. Simultaneously, it is proved that ROS can dephosphorylate Akt under high glucose to inhibit activation of Akt, which can induce HUVECs apoptosis, the whole effect can be inhibited by wortmannin.
     siNotch1 expression vector was constructed in vitro and was transfected into each group to detect PARP and NF-κB express changes, finding that siNotchl can inhibit the effect of Notchl against PARP and NF-κB. At the same time, after the cells were treated with wortmannin, immunoprecipitation and Western Blot were used to detect the expression changes of PARP and NF-κB, finding that wortmannin can inhibit Notchl protective function on PARP and NF-κB. Those results indicate that Notchl inhibit cell apoptosis mediated by PARP and NF-κB under high glucose through Akt pathway.
     This research was mainly to explore the pathological mechanism of diabetic retinopathy and detect related factors using animal model in vivo and cell model in vitro. Our study illustrates that high glucose can activate PARP in HRVEC and then activate NF-κB which then activate caspase-3, and Notchl dephosphorylated Akt regulating PARP and NF-κB, further clarifies mechanism of diabetic retinopathy. It provides a good experimental basis and theoretical basis to further study the pathogenesis of diabetic retinopathy, and also provides a new direction and possibilities for the treatment of diabetic retinopathy.
引文
[1]Artavanis-Tsakonas S, Rand MD, Lake RJ. Notch signaling cell fate control and signal integration in development. Science,1999,284:770-764.
    [2]Kopan and M.X. Ilagan, The canonical Notch signaling pathway:Unfolding the activation mechanism, Cell.2009,137,216-233.
    [3]Mumm JS, and Kopan R. Notch signaling:from the outside in. Dev Biol.2000,228, 156-65.
    [4]Milner LA and Bigas A. Notch as a mediator of cell fate determination in hematopoiesis: Evidence and speculation. Blood,1999,93,2431-48.
    [5]Rebay I, Fleming RJ, Fehon RG, et, al. Specific EGF repeats of Notch mediate interactions with Delta and Serrate:implications for Notch as a multifunctional receptor. Cell,1991,67,687-99.
    [6]Fleming RJ. Stuctural conservation of Notch receptors and ligands. Seminars in Cell and Developmental Biology,1998,9,559-607.
    [7]Allman D, Punt JA, Izon DJ. An invitation to T and more:notch signaling in lymphopoiesis.2002,109 Suppl, S1-11.
    [8]Greenwald 1, Notch signaling during vascular development. Proc Natl Acad Sci USA, 1998,98,1752-62.
    [9]Kimble J, Henderson S, Crittenden S. Notch/LIN-12 signaling:transduction by regulared protein slicing. Trends Biochem Sci,1998,23,353-7.
    [10]Limber T, Kidd S, Alcamo E. Antineurogenic phenotypes induced by truncated Notch proteins indicate a role in signal transduction and may point to a novel function for Notch in nuclei. Genes Dev,1993,7,1949-65.
    [11]Kimble J and Simpson P. The LIN-12/NOTCH signaling pathway and its regulation. Annual Review of Cell and Developmental Biology.1997,13,333-361.
    [12]Mizutani T, Taniguchi Y, Aoki T, et al. Conservation of the biochemical mechanisms of signal transduction among mammalian Notch family members. Proc Natl Acad Sci USA.2001,98,9026-31.
    [13]Shutter JR, Scully S, Fan W, et al. D114, a novel Notch ligand expressed in arterial endothelium. Genes Dev,2000,14,1313-8.
    [14]Fleming RJ, Purcell K, Artavanistsakonas S. The Notch Receptor and its ligand. Trends in Cell Biology.1997b,7,437-441.
    [15]Le Bergne, Bardin A, Schweisgurh F. The roles of receptor and ligand endocytosis in regulating Notch signaling[J]. Development,2005,132(8):1751-1762.
    [16]Beckstead BL, Santosa DM, Giachelli CM. Mimicking cell-cell interactions at the biomaterial-cell interface for control of stem cell differentiation[J]. J Biomed Mater Res A,2006,79(1):94-103.
    [17]Breunig JJ, Silbereis J, Vaccarino FM, et al. Notch regulates cell fate and dendrite morphology of newborn neurons in the postnatal dentate gyrus[J]. Proc Natl Acad Sci USA,2007,104(51):20558-20563.
    [18]Kopan R and Goate A. A commom enzyme connects notch signaling and Alzheimer's disease. Genes Dev.2000,14,2799-806.
    [19]Struhl G and Greenwald I. Presenilin-mediated transmembrane cleavage is required for Notch signal transduction in Drosophila. Proc Natl Acad Sci USA,2001,98,229-34.
    [20]Aster, J.C, Robertson, E.S., Hasserjian, R.P., et al. Oncogenic forms of Notch 1 lacking either the primary binding site for RBP-J kappa or nuclear localization sequences retain the ability to associate with RBP-Jkappa and activate transcription. J Biol Chem,1997, 272,11336-43.
    [21]Fortini, M.E. and Artavanis-Tsakonas, S. The suppressor of hairless protein participates in notch receptor signaling. Cell.1994,79,273-82.
    [22]Hsieh, J.J., Henkel, T., Salmon, P., et al. Truncated mammalian Notchl activates CBF1/RBPJk-repressed genes by a mechanism resembling that of Epstein-Barr virus EBNA2. Mol Cell Biol.1996,16,952-9.
    [23]Matsuno, K., Go, M.J., Sun, X., et al. Suppressor of Hairless-independent events in Notch signaling imply novel pathway elements. Development.1997,124,4265-73.
    [24]Tamura, K., Taniguchi, Y., Minoguchi, S., et al. Physical interaction between a novel domain of the receptor Notch and the transcription factor RJ3P-J kappa/Su(H). Curr Biol.1995,5,1416-23.
    [25]Kato, H., Taniguchi, Y., Kurooka, H., et al. Involvement of RBP-J in biological functions of mouse Notch1 and its derivatives. Development,1997,124,4133-41.
    [26]Zhou, S., Fujimuro, M., Hsieh, J.J., et al. SKIP, a CBF1-associated protein, interacts with the ankyrin repeat domain of NotchIC To facilitate NotchIC function. Mol Cell Biol.2000b,20,2400-10.
    [27]Bray, S. and Furriols, M. Notch pathway:making sense of suppressor of hairless. Curr Biol,2001,11, R217-21.
    [28]Hsieh, J.J. and Hayward, S.D. Masking of the CBFI/RBPJ kappa transcriptional repression domain by Epstein-Barr virus EBNA2. Science,1995,268,560-3.
    [29]Piccoli, D.A. and Spinner, N.B. Alagille syndrome and the Jaggedl gene. Semin Liver Dis,2001,21,525-34.
    [30]Hsieh, J.J., Zhou, S., Chen, L., et al. CIR, a corepressor linking the DNA binding factor CBF1 to the histone deacetylase complex. Proc Natl Acad Sci USA,1999,96,23-8.
    [31]Zhou, S., Fujimuro, M., Hsieh, J.J., et al. A role for SKIP in EBNA2 activation of CBF1-repressed promoters. J Virol,2000a,74,1939-47.
    [32]Jarriault, S., Brou, C, Logeat, F., et al. Signalling downstream of activated mammalian Notch. Nature.1995,311,355-8.
    [33]Kopan, R., Nye, J.S. and Weintraub, H. The intracellular domain of mouse Notch:a constitutively activated repressor of myogenesis directed at the basic helix-loop-helix region of MyoD. Development,1994,120,2385-96.
    [34]Tomlinson, A. and Struhl, G. Decoding vectorial information from a gradient: sequential roles of the receptors Frizzled and Notch in establishing planar polarity in the Drosophila eye. Development,1999,126,5725-38
    [35]Struhl, G. and Greenwald, I. Presenilin is required for activity and nuclear access of Notch in Drosophila. Nature,1999,398,522-5.
    [36]Petcherski, A.G. and Kimble, J. LAG-3 is a putative transcriptional activator in the C. elegans Notch pathway. Nature,2000,405,364-8.
    [37]Bailey, A.M. and Posakony, J.W. Suppressor of hairless directly activates transcription of enhancer of split complex genes in response to Notch receptor activity. Genes Dev, 1995,9,2609-22.
    [38]Chen, H., Thiagalingam, A., Chopra, H., et al. Conservation of the Drosophila lateral inhibition pathway in human lung cancer:a hairy-related protein (HES-1) directly represses achaete-scute homolog-1 expression. Proc Natl Acad Sci U SA,1997,94, 5355-60.
    [39]Ishibashi, M., Ang, S.L., Shiota, K., et al. Targeted disruption of mammalian hairy and Enhancer of split homolog-1 (HES-1) leads to up-regulation of neural helix-loop-helix factors, premature neurogenesis, and severe neural tube defects. Genes Dev,1995,9, 3136-48.
    [40]Ohtsuka, T., Ishibashi, M., Gradwohl, G., et al. Hesl and Hes5 as Notch effectors in mammalian neuronal differentiation. EMBO Journal,1999,18,2196-2207.
    [41]Iso, T., Kedes, L. and Hamamori, Y. HES and HERP families:multiple effectors of the Notch signaling pathway. J Cell Physiol,2003,194,237-55.
    [42]Iso, T., Sartorelli, V., Poizat, C., et al. HERP, a novel heterodimer partner of HES/E(spl) in Notch signaling. Mol Cell Biol,2001,21,6080-9.
    [43]Fisher, A.L., Ohsako, S. and Caudy, M. The WRPW motif of the hairy-related basic helix-loop-helix repressor proteins acts as a 4-amino-acid transcription repression and protein-protein interaction domain. Mol Cell Biol,1996,16,2670-7.
    [44]Grbavec, D. and Stifani, S. Molecular Interaction Between Tlel and the Carboxyl-Terminal Domain of Hes-1 Containing the Wrpw Motif. Biochemical & Biophysical Research Communications,1996,223,701-705.
    [45]Paroush, Z., Finley, R.L., Jr., Kidd, T., et al. Groucho is required for Drosophila neurogenesis, segmentation, and sex determination and interacts directly with hairy-related bHLH proteins. Cell,1994,79,805-15.
    [46]Akazawa, C, Sasai, Y., Nakanishi, S. and Kageyama, R. Molecular characterization of a rat negative regulator with a basic helix-loop-helix structure predominantly expressed in the developing nervous system. J Biol Chem,1992,267,21879-85.
    [47]Bae, S., Bessho, Y., Hojo, M. and Kageyama, R. The bHLH gene Hes6, an inhibitor of Hesl, promotes neuronal differentiation. Development,2000,127,2933-43.
    [48]Maier, M.M. and Gessler, M. Comparative analysis of the human and mouse Heyl promoter:Hey genes are new Notch target genes. Biochem Biophys Res Commun, 2000,275,652-60.
    [49]Izon, D.J., Aster, J.C, He, Y., et al. Deltexl redirects lymphoid progenitors to the B cell lineage by antagonizing Notchl. Immunity,2002,16,231-43.
    [50]Hamada, Y., Kadokawa, Y., Okabe, M. et al. Mutation in ankyrin repeats of the mouse Notch2 gene induces early embryonic lethality. Development,1999,126,3415-24.
    [51]Dunwoodie, S.L., Clements, M., Sparrow, D.B., et al. Axial skeletal defects caused by mutation in the spondylocostal dysplasia/pudgy gene D113 are associated with isruption of the segmentation clock within the presomitic mesoderm. Development, 2002,129,1795-806.
    [52]de la Pompa, J.L., Wakeham, A., Correia, K. M., et al. Conservation of the Notch signalling pathway in mammalian neurogenesis. Development,1997,124,1139-48.
    [53]Zecchini, V., Brennan, K. and Martinez-Arias, A. An activity of Notch regulates INK signalling and affects dorsal closure in Drosophila. Curr Biol,1999,9,460-9.
    [54]Ordentlich, P., Lin, A., Shen, CP., et al. Notch inhibition of E47 supports the existence of a novel signaling pathway. Mol Cell Biol,1998,18,2230-9.
    [55]Miyoshi G, Bessho Y, Yamada S, et al. Identification of a novel basic helix-loop-helix gene, Heslike and its role in GABAergic neurogenesis. The Journal of Neuroscience. 2004,24(14):3672-3682.
    [56]Fortini, M.E., Rebay, I., Caron, L.A. et al. An activated Notch receptor blocks cell-fate commitment in the developing Drosophila eye. Nature,1993,365,555-7.
    [57]Bruckner, K., Perez, L., Clausen, H.et al. Glycosyltransferase activity of Fringe modulates Notch-Delta interactions. Nature,2000,406,411-5.
    [58]Fleming, R.J., Gu, Y. and Hukriede, N.A. Serrate-mediated activation of Notch is specifically blocked by the product of the gene fringe in the dorsal compartment of the Drosophila wing imaginal disc. Development,1997a,124,2973-81
    [59]Correia T, Papayannopoulos V, Panin V, et al. Molecular genetic analysis of the glycosyltransferase Fringe in Drosophila. Proc Natl Acad Sci USA,2003,100 (11): 6404~6409
    [60]Rulifson, E.J., Micchelli, C.A., Axelrod, J.D., et al. wingless refines its own expression domain on the Drosophila wing margin. Nature,1996,384,72-4.
    [61]Wesley, C.S. Notch and wingless regulate expression of cuticle patterning genes. Mol Cell Biol,1999,19,5743-58.
    [62]Aravind, L. The WWE domain:a common interaction module in protein ubiquitination and ADP ribosylation. Trends Biochem Sci,2001,26,273-5.
    [63]Matsuno, K., Eastman, D., Mitsiades, T., et al. Human deltex is a conserved regulator of Notch signalling. Nat Genet,1998,19,74-8.
    [64]Matsuno, K., Diederich, R.J., Go, M.J., Blaumueller, C M. and Artavanis-Tsakonas, S. Deltex acts as a positive regulator of Notch signaling through interactions with the Notch ankyrin repeats. Development,1995,121,2633-44.
    [65]Spana, E.P. and Doe, C Q. Numb antagonizes Notch signaling to specify sibling neuron cell fates. Neuron,1996,17,21-6.
    [66]McGill, M.A. and McGlade, C J. Mammalian numb proteins promote Notchl receptor ubiquitination and degradation of the Notchl intracellular domain. J Biol Chem, 2003,278,23196-203.
    [67]Jafar-Nejad, H., Norga, K. and Bellen, H. Numb:"Adapting" notch for endocytosis. Dev Cell,2002,3,155-6.
    [68]Lai E C. Protein degradation:four E3s for the Notch pathway. Curr Biol,2002,12 (2): 74-78.
    [69]Gupta2Rossi N, Le Bail O, Gonen H, et al. Functional interaction between SEL210, an F2box protein, and the nuclear form of activated Notchl receptor. J Biol Chem,2001, 276 (37):34371~34378.
    [70]Lamar E, Deblandre G, Wettstein D, et al. Nrarp is a novel intracellular component of the Notch signaling pathway. Genes Dev,2001,15 (15):1885~1899.
    [71]Lai EC, Deblandre GA, Kintner C, et al. Drosophila neuralized is a ubiquitin ligase that promotes the internalization and degradation of delta. Dev Cell,2001,1 (6):783~794.
    [72]Itoh M, Kim CH, Palardy G, et al. Mind bomb is a ubiquitin ligase that is essential for efficient activation of Notch signaling by Delta. Dev Cell,2003,4(1):67~82
    [73]Parks A L, Klueg KM, Stout J R, et al. Ligand endocytosis drives receptor dissociation and activation in the Notch pathway. Development,2000,127 (7):1373~1385
    [74]Pourquie O. The segmentation clock:converting embryonic time into spatial pattern. Science,2003,301 (5631):328~330
    [75]Limbourg, F. P., Takeshita, K., Radtke, F., et al. Essential role of endothelial Notchl in angiogenesis. Circulation.2005,111,1826-1832.
    [76]Carlson TR, Yan Y, Wu, X, et al. Endothelial expression of constitutively active Notch4 elicits reversible arteriovenous malformations in adult mice. Proc. Natl. Acad. Sci. USA 2005,102,9884-9889.
    [77]Xue, Y., Gao, X., Lindsell, C. E., et al. Embryonic lethality and vascular defects in mice lacking the Notch ligand Jaggedl. Hum. Mol. Genet.1999,8,723-730.
    [78]Krebs, LT., Shutter, J. R., Tanigaki, K., et al. Haploinsufficient lethality and formation of arteriovenous malformations in Notch pathway mutants. Genes Dev.2004,18, 2469-2473.
    [79]Barsi JC., Rajendra R., Wu, JI. et, al. Mind bombl is a ubiquitin ligase essential for mouse embryonic development and Notch signaling. Mech. Dev.2005,122, 1106-1117.
    [80]Li, T., Ma, G., Cai, H., et al. Nicastrin is required for assembly of presenilin/gamma-secretase complexes to mediate Notch signaling and for processing and trafficking of beta-amyloid precursor protein in mammals. J. Neurosci.2003,23,3272-3277.
    [81]Herreman, A., Hartmann, D., Annaert, W., et al. Presenilin 2 deficiency causes a mild pulmonary phenotype and no changes in amyloid precursor protein processing but enhances the embryonic lethal phenotype of presenilin 1 deficiency. Proc. Natl. Acad. Sci.USA.1999,96,11872-11877.
    [82]Kokubo, H., Miyagawa-Tomita, S., Nakazawa, M., et al. Mouse hesrl and hesr2 genes are redundantly required to mediate Notch signaling in the developing cardiovascular system. Dev. Biol.2005,278,301-309.
    [83]Jones, E. A., le Noble, F. and Eichmann, A. What determines blood vessel structure? Genetic prespecification vs. hemodynamics. Physiology.2006,21,6388-395.
    [84]Vogeli, K. M., Jin, S. W., Martin, G. R. et al. A common progenitor for haematopoietic and endothelial lineages in the zebrafish gastrula. Nature.2006,443,337-339.
    [85]Shibuya, M. and Claesson-Welsh, L. Signal transduction by VEGF receptors in regulation of angiogenesis and lymphangiogenesis. Exp. Cell Res.2006,312,549-560.
    [86]Lawson, N. D., Vogel, A. M. and Weinstein, B. M. sonic hedgehog and vascular endothelial growth factor act upstream of the Notch pathway during arterial endothelial differentiation. Dev. Cell.2002,3,127-136.
    [87]Lawson, N. D., Scheer, N., Pham, V. N., et al. Notch signaling is required for arterial-venous differentiation during embryonic vascular development. Development. 2001,128,3675-3683.
    [88]Zhong, T. P., Childs, S., Leu, J. P. and Fishman, M. C. Gridlock signaling pathway fashions the first embryonic artery. Nature.2001,414,216-220.
    [89]Liu, Z. J., Shirakawa, T., Li, Y., et al. Regulation of Notchl and D114 by vascular endothelial growth factor in arterial endothelial cells:implications for modulating arteriogenesis and angiogenesis. Mol. Cell. Biol.2003,23,14-25.
    [90]Carmeliet P, Ferreira V, Breier G, et al. Abnormal blood vessel development and lethality in embryos lacking a single VEGF allele. Nature.1996,380,435-439.
    [91]Stalmans,1., Ng, Y. S., Rohan, R., et al. Arteriolar and venular patterning in retinas of mice selectively expressing VEGF isoforms. J. Clin Invest.2002,109,327-336.
    [92]Visconti, R. P., Richardson, C. D. and Sato, T. N. Orchestration of angiogenesis and arteriovenous contribution by angiopoietins and vascular endothelial growth factor (VEGF). Proc. Natl. Acad. Sci. USA.2002,99,8219-8224.
    [93]Mukouyama, Y. S., Gerber, H. P., Ferrara, N., et al. Peripheral nerve-derived VEGF promotes arterial differentiation via neuropilin 1-mediated positive feedback. Development.2005,132,941-952.
    [94]Duarte, A., Hirashima, M., Benedito, R., et al. Dosage-sensitive requirement for mouse D114 in artery development. Genes Dev.2004,18,2474-2478.
    [95]Grego-Bessa, J., Luna-Zurita, L., Del Monte, G. et al. Notch signaling is essential for ventricular chamber development. Dev. Cell.2007,12,415-429.
    [96]Kaestner, K. H., Knochel, W. and Martinez, D. E. Unified nomenclature for the winged helix/forkhead transcription factors. Genes Dev.2000,14,142-146.
    [97]Kume, T., Jiang, H., Topczewska, J. M. et al. The murine winged helix transcription factors, Foxcl and Foxc2, are both required for cardiovascular development and somitogenesis. Genes Dev.2001,15,2470-2482.
    [98]Seo, S., Fujita, H., Nakano, A., et al. The forkhead transcription factors, Foxcl and Foxc2, are required for arterial specification and lymphatic sprouting during vascular development. Dev. Biol.2006,294,458-470.
    [99]Kurpinski K, Lam H, Chu J, et al. Transforming Growth Factor-(3 and Notch Signaling Mediate Stem Cell Differentiation into Smooth Muscle Cells. Stem Cells.2010,4(28), 734-742.
    [100]vander Loop, F. T., Gabbiani, G., Kohnen, G.,et al. Differentiation of smooth muscle cells in human blood vessels as defined by smoothelin, a novel marker for the contractile phenotype. Arterioscler. Thromb. Vasc. Biol.1997,17,665-671.
    [101]Moessler, H., Mericskay, M., Li, Z., et al. The SM 22 promoter directs tissue-specific expression in arterial but not in venous or visceral smooth muscle cells in transgenic mice. Development.1996,122,2415-2425.
    [102]Gerhardt, H., Ruhrberg, C., Abramsson, A., et al. Neuropilin-1 is required for endothelial tip cell guidance in the developing central nervous system. Dev. Dyn.2004, 231,503-509.
    [103]Sainson, R. C., Aoto, J., Nakatsu, M. N., et al. Cell-autonomous notch signaling regulates endothelial cell branching and proliferation during vascular tubulogenesis. FASEB J.2005,19,1027-1029.
    [104]Hellstrom, M., Phng, L. K., Hofmann, J. J., et al. D114 signalling through Notchl regulates formation of tip cells during angiogenesis. Nature,2007,445,776-780.
    [105]Lobov, I. B., Renard, R. A., Papadopoulos, N., et al. Delta-like ligand 4 (Dll4) is induced by VEGF as a negative regulator of angiogenic sprouting. Proc. Natl. Acad. Sci. USA.2007,104,3219-3224.
    [106]Leslie, J. D., Ariza-McNaughton, L., Bermange, A. L., et al. Endothelial signalling by the Notch ligand Delta-like 4 restricts angiogenesis. Development.2007,134, 839-844.
    [107]Scehnet, J. S., Jiang, W., Kumar, S. R., et al. Inhibition of D114 mediated signaling induces proliferation of immature vessels and results in poor tissue perfusion. Blood 2007,109,4753-4760.
    [108]Dorrell, M. I. and Friedlander, M. Mechanisms of endothelial cell guidance and vascular patterning in the developing mouse retina. Prog. Retin. Eye Res.2006,25, 277-295.
    [109]Siekmann, A. F. and Lawson, N. D. Notch signalling limits angiogenic cell behaviour in developing zebrafish arteries. Nature.2007,445,781-784.
    [110]Proweller, A., Pear, W. S. and Parmacek, M. S. Notch signaling represses myocardin-induced smooth muscle cell differentiation. J. Biol. Chem.2005,280, 8994-9004.
    [111]High, F. A., Zhang, M., Proweller, A., et al. An essential role for Notch in neural crest during cardiovascular development and smooth muscle differentiation. J. Clin. Invest. 2007,117,353-363.
    [112]Doi, H., Iso, T., Sato, H., et al. Jagged1-selective notch signaling induces smooth muscle differentiation via a RBP-Jkappa-dependent pathway. J. Biol. Chem.2006,281, 28555-28564.
    [113]Noseda, M., Fu, Y., Niessen, K.,et al. Smooth Muscle alpha-actin is a direct target of Notch/CSL. Circ. Res.2006,98,1468-1470.
    [114]Sakata, Y., Xiang, F., Chen, Z., et al. Transcription factor CHF1/Hey2 regulates neointimal formation in vivo and vascular smooth muscle proliferation and migration in vitro. Arterioscler. Thromb. Vasc. Biol.2004,24,2069-2074.
    [115]Wang, W., Prince, C. Z., Hu, X. and Pollman, M. J. HRT1 modulates vascular smooth muscle cell proliferation and apoptosis. Biochem. Biophys. Res. Commun.2003,308, 596-601.
    [116]Havrda, M. C., Johnson, M. J., O'Neill, C. F. et al. A novel mechanism of transcriptional repression of p27kipl through Notch/HRT2 signaling in vascular smooth muscle cells. Thromb. Haemost.2006,96,361-370.
    [117]Morrow, D., Sweeney, C., Birney, Y. A., et al. Cyclic strain inhibits Notch receptor signaling in vascular smooth muscle cells in vitro. Circ. Res.2005b,96,567-575.
    [118]Nicolas M, Wolfer A, Raj K, et al. Notchl functions as a tumor suppressor in mouse skin. Nat Genet.2003,33:416-21.
    [119]Rangarajan A, Talora C, Okuyama R, et al. Notch signaling is a direct determinant of keratinocyte growth arrest and entry into differentiation. EMBO J.2001,20:3427-36.
    [120]Lowell S, Jones P, Le Roux I, et al. Stimulation of human epidermal differentiation by δ-notch signalling at the boundaries of stem-cell clusters. Curr Biol.2000,10:491-500.
    [121]Massi D, Tarantini F, Franchi A, et al. Evidence for differential expression of Notch receptors and their ligands in melanocytic nevi and cutaneous malignant melanoma. Modern Pathology.2006,19:246-259.
    [122]Pinnix CC, Lee JT, Liu ZJ, et al. Active Notchl confers a transformed phenotype to primary human melanocytes. Cancer Res.2009,69:5312-5320.
    [123]Kang DE, Soriano S, Xia X, et al. Presenilin couples the paired phosphorylation of beta-catenin independent of axin:implications for beta-catenin activation in tumorigenesis. Cell.2002,110:751-762.
    [124]Li G, Satyamoorthy K, Herlyn M:N-cadherin-mediated intercellular interactions promote survival and migration of melanoma cells. Cancer Res.2001,61:3819-3825.
    [125]Hainaud, P., Contreres, J. O., Villemain, A.,et al. The role of the vascular endothelial growth factor-deltalike 4 ligand/Notch4-ephrin B2 cascade in tumor vessel remodeling and endothelial cell functions. Cancer Res.2006,66,8501-8510.
    [126]Noguera-Troise, I., Daly, C., Papadopoulos, N. J., et al. Blockade of D114 inhibits tumour growth by promoting non-productive angiogenesis. Nature.2006,444, 1032-1037.
    [127]Jain, R. K., Duda, D. G., Clark, J. W, et al. Lessons from phase III clinical trials on anti-VEGF therapy for cancer. Nat. Clin. Pract. Oncol.2006,3,24-40.
    [128]Marquardt, T., and Gruss, P. Generating neuronal diversity in the retina:one for nearly all. Trends Neurosci.2002,25:32-38.
    [129]Fruttiger, M. Development of the retinal vasculature. Angiogenesis.2007,10:77-88.
    [130]Chow, R.L., and Lang, R.A. Early eye development in vertebrates. Annu. Rev. Cell Dev. Biol.2001,17:255-296.
    [131]Poggi, L., Zolessi, F.R., and Harris, W.A. Time-lapse analysis of retinal differentiation. Curr. Opin. Cell Biol.2005,17:676-681.
    [132]Fu, X., Sun, H., Klein, W.H., and Mu, X. Beta-catenin is essential for lamination but not neurogenesis in mouse retinal development. Dev. Biol.2006,299:424-437.
    [133]Campochiaro, P.A. Retinal and choroidal neovascularization. J. Cell Physiol.2000, 184:301-310.
    [134]Carmeliet, P. Mechanisms of angiogenesis and arteriogenesis. Nat. Med.2000,6: 389-395.
    [135]Gao, F., Zhang, Q., Zheng, M.H., et al. Transcription factor RBP-J-mediated signaling represses the differentiation of neural stem cells into intermediate neural progenitors. Mol. Cell. Neurosci.2009,40:442-450.
    [136]Louvi, A., and Artavanis-Tsakonas, S. Notch signalling in vertebrate neural development. Nat. Rev. Neurosci.2006,7:93-102.
    [137]Han, H., Tanigaki, K., Yamamoto, N., et al. Inducible gene knockout of transcription factor recombination signal binding protein-J reveals its essential role in T versus B lineage decision. Int. Immunol.2002,14:637-645.
    [138]Radtke, F., Fasnacht, N., and Macdonald, H.R. Notch signaling in the immune system. Immunity.2010,32:14-27.
    [139]Grandbarbe, L., Bouissac, J., Rand, M., et al. Delta-Notch signaling controls the generation of neurons/glia from neural stem cells in a stepwise process. Development. 2003,130:1391-1402.
    [140]Dou, G.R., Wang, Y.C., Hu, X.B., et al. RBP-J, the transcription factor downstream of Notch receptors, is essential for the maintenance of vascular homeostasis in adult mice. FASEB J.2008,22:1606-1617.
    [141]Dorsky, R.I., Rapaport, D.H., and Harris, W.A. Notch inhibits cell differentiation in the Xenopus retina. Neuron.1995,14:487-496.
    [142]Rocha, S.F., Lopes, S.S., Gossler, A., and Henrique, D. Dll1 and D114 function sequentially in the retina and pV2 domain of the spinal cord to regulate neurogenesis and create cell diversity. Dev. Biol.2009,328:54-65.
    [143]Yaron, O., Farhy, C., Marquardt, T., et al. Notch1 functions to suppress cone-photoreceptor fate specification in the developing mouse retina. Development. 2006,133:1367-1378.
    [144]Riesenberg, A.N., Liu, Z., Kopan, R., and Brown, N.L. Rbpj cell autonomous regulation of retinal ganglion cell and cone photoreceptor fates in the mouse retina. J. Neurosci.2009,29:12865-12877.
    [145]Jadhav, A.P., Cho, S.H., and Cepko, C.L. Notch activity permits retinal cells to progress through multiple progenitor states and acquire a stem cell property. Proc. Natl. Acad. Sci. USA.2006a,103:18998-19003.
    [146]Zheng, M.H., Shi, M., Pei, Z., et al. The transcription factor RBP-J is essential for retinal cell differentiation and lamination. Mol. Brain.2009,2:38.
    [147]Takatsuka, K., Hatakeyama, J., Bessho, Y., and Kageyama, R. Roles of the bHLH gene Hesl in retinal morphogenesis. Brain Res.2004,1004:148-155.
    [138]Hojo, M., Ohtsuka, T., Hashimoto, N.,et al. Glial cell fate specification modulated by the bHLH gene Hes5 in mouse retina. Development.2000,127:2515-2522.
    [149]Jadhav AP, cho sH, cepko cL. Notch activity permits retinal cells to progress through multiple progenitor states and acquire a stem cell property. [J]. Proc Natl Acad Sci USA,2006,103:18998-19003
    [150]Ll Y, Baker NE. Proneural enhancement by Notch overcomes Suppressor-of-hairless repressor function in developing Drosophila eye. [J]. Curr Biol.2001,11:330-338.
    [151]Hurlbut GD, Kankel MW, Lake RJ, et al. Crossing Paths with Notch in the hyper-network[J]. Curr Opin cell Biol,2007,19:166-175..
    [152]Schneider ML, Turner DL, Vetter ML. Notch signaling can inhibit Xath5 function in the neural plate and developing retina.[J]. Mo cell Neurosci,2001,18:458-472.
    [153]Ohnuma S, Hopper S, Wang KC, et al. Co-ordinating retinal histogenesis:early cell cycle exit enhances early cell fate determination in the Xenopus retina.[J]. Development,2002,129:2435-2446
    [154]Brown NL, Paiel S, Brzezinski J, et al. Math5 is required for retinal ganglion cell and optic nerve formation. [J] Development,2001,128:2497-2508.
    [155]Nelson BR, Cumuscu B, Hartman BH, el al. Notch activity is downregulated just prior to retina ganglion cell differentiation. [J]. Dev Neorosci.2006,28:128-141.
    [156]Nam Y, Aster JC, Blacklow SC. Notch signaling as a therapeutic target.[J]. Curr Opin Chem Biol.2002,6:501-509.
    [157]Bernardos RL, Lentz SI, Wolfe MS, et al. Notch-Delta signaling is required for spatial patterning and Muller glia differentiation in the zebrafish retina[J]. Dev Biol,2005, 278:381-395.
    [158]Marquardt T, Ashery-Padan R, Andrejewski N, et al. Pax6 is required for the multipotent state of retinal progenitor cells [J]cell.2001,105:43-55.
    [159]Silva AO, Ercole CE, Mcloon SC. Regulation of ganglion cell production by Notch signaling during retinal development [J]. J Neurobiol.2003,54:511-524.
    [160]Duncan, A. W. et al. Integration of Notch and Wnt signaling in hematopoietic stem cell maintenance. Nature Immunol.2005,6,314-322.
    [161]Claxton, S.& Fruttiger, M. Periodic Delta-like 4 expression in developing retinal arteries. Gene Expr. Patterns.2004,5,123-127.
    [162]Benedito, R., Roca, C., Sorensen, I., et al. The notch ligands D114 and Jaggedl have opposing effects on angiogenesis. Cell.2009,137:1124-1135.
    [163]Joutel, A., and Tournier-Lasserve, E. Notch signalling pathway and human diseases. Semin. Cell Dev. Biol.1998,9:619-625.
    [164]Delaney, C., Heimfeld, S., Brashem-Stein, C., et al. Notch-mediated expansion of human cord blood progenitor cells capable of rapid myeloid reconstitution. Nat. Med. 2010,16:232-236.
    [165]Sakada, F., Ikeda, H., Mandai, M., et al. Toward the generation of rod and cone hotoreceptors from mouse, monkey and human embryonic stem cells. Nat. Biotechnol. 2008,26:215-224.
    [166]Carolyn Chen. Review of therapeutic advances in diabetic retinopathy. Therapeutic Advances in Endocrinology and Metabolism.2011,2 (1):39-53.
    [167]Klein R, Klein BE, Moss SE, et al. Glycosylated hemoglobin predicts the incidence and progression of diabetic retinopathy. JAMA.1988,260:2864-2871.
    [168]Antonelli-Orlidge A, Smith SR, D'Amore PA:Influence of pericytes on capillary endothelial cell growth. Am Rev Respir Dis.1989,140:1129-1131.
    [169]Ciulla TA, Harris A, Latkany P, et al. Ocular perfusion abnormalities in diabetes. Acta Ophthalmol Scand,2002,80:468-477.
    [170]Speiser P, Gittelsohn AM, Patz A:Studies on diabetic retinopathy, III:influence of diabetes on intramural pericytes. Arch Ophthalmol,1968,80:332-337.
    [171]Miyamoto K, Ogura Y:Pathogenetic potential of leukocytes in diabetic retinopathy. Semin Ophthalmol,1999,14:233-239
    [172]Miyamoto K, Khosrof S, Bursell SE, et al. Prevention of leukostasis and vascular leakage in streptozotocin-induced diabetic retinopathy via intercellular adhesion molecule-1 inhibition. Proc Natl Acad Sci U S A,1999,96:10836-10841.
    [173]Miller JW, Adamis AP, Aiello LP:Vascular endothelial growth factor in ocular neovascularization and proliferative diabetic retinopathy. Diabetes Metab Rev.1997, 13:37-50.
    [174]Barber A.J., Gardner T.W., Steven F. Abcouwer.The Significance of Vascular and Neural Apoptosis to the Pathology of Diabetic Retinopathy. IOVS.2011,52(2): 1156-1163.
    [175]Zeng, X.X., Ng, Y.K., Ling, E.A., Neuronal and microglial response in the retina of streptozotocin-induced diabetic rats. Vis. Neurosci.2000,17,463-471.
    [176]Mohr, S., Xi, X., Tang, J., Kern, T.S., Caspase activation in retinas of diabetic and galactosemic mice and diabetic patients. Diabetes.2002,51,1172-1179.
    [177]Pan HZ, Zhang H, Chang D, et al. The change of oxidative stress products in diabetes mellitus and diabetic retinopathy[J]. Br J Ophthalmol,2008,92(4):548-551.
    [178]Sergio Li Calzi, Matthew B. Neu, Lynn C. Shaw, et al. Endothelial progenitor dysfunction in the pathogenesis of diabetic retinopathy:treatment concept to correct diabetes-associated deficits. EPMA Journal.2010,1:88-100.
    [179]Anuradha CD, Kanno S, H irano S. Oxidative damage t o mitochondria is a preliminary step to caspase-3 activation in fluoride-induced apoptosis in H L-60 cells[J]. Free Radic Biol Med,2001,31(3):367-373.
    [180]Sofiya Milman MD and Jill P. Crandall MD. Mechanisms of Vascular Complications in Prediabetes. Medical Clinics of North America.2011,2(95):309-325.
    [181]Garcia Soriano F,Virag L, Jagtap P, et al. Diabetic endothelial dysfunction:the role of poly (ADP-ribose) polymerase activation. Nat Med,2001,7:108-113.
    [182]Pacher P,Liaudet L,Garcia Soriano F, et al. The role of poly (ADP-ribose) polymerase activation in the development of myocardial and endothelial dysfunction in diabetes. Diabetes.2002,51:514-521.
    [183]Yamagishi, Sho-ichi, Matsui, et al. Advanced Glycation End Products (AGEs), Oxidative Stress and Diabetic Retinopathy. Current Pharmaceutical Biotechnology. 2011,12(7):362-368.
    [184]Stit t AW. The role of advanced glycation in the pathogenesis of diabetic retinopathy [J]. Exp Mol Pathol,2003,75(1):95-108.
    [185]Chiarelli F, S pagnoli A, Basciani F, et al. Vascular endothelial growth factor (VEGF) in children, adolescents and young adults with type 1 diabetes mellitus:relation to glycaemic control and microvascular complications[J]. Diabet Med,2002,17(9):650-656.
    [186]Takashi Dan, Charles van Ypersele de Strihou and Toshio Miyata. Advanced Glycation End Products Inhibitor. Biomedical and Life Sciences. Oxidative Stress in Applied Basic Research and Clinical Practice,2011, Part 3,389-406, DOI:10. 1007/97 8-1-60761-857-7 20.
    [187]Wang Y, Vom Hagen F, Pfister F, et al. Receptor for advanced glycation end product expression in experiment al diabetic retinopathy [J]. AnnN YAca Sci,2008,1126: 42-45.
    [188]Kowluru RA. Effect of advanced glycation end products on accelerated apoptosis of retinal capillary cells under in vitro conditions[J]. Life Sci,2005,76(9):1051-1060.
    [189]Hammes HP, Martin S, Federlin K, et al. Aminoguanidine treatment inhibits the development of experimental diabetic retinopathy. Proc Natl Acad Sci USA,1991 88:11555-11558.
    [190]Gabbay KH:Hyperglycemia, polyol metabolism, and complications of diabetes mellitus. Annu Rev Med.1975,26:521-535.
    [191]Naruse K, Nakamura J, Hamada Y, et al. Aldose reductase inhibition prevents glucose-induced apoptosis in cultured bovine retinal microvascular pericytes[J]. Exp Eye Res,2000,71(3):309-315.
    [192]Sato S, Secchi EF, Lizak MJ, et al. Polyol formation and NADPH-dependent reductases in dog retinal capillary pericytes and endothelial cells. Invest Ophthalmol Vis Sci,1999,40:697-704.
    [193]Del Monte MA, Rabbani R, Diaz TC, et al. Sorbitol, myo-inositol, and rod outer segment phagocytosis in cultured hRPE cells exposed to glucose:in vitro model of myo-inositol depletion hypothesis of diabetic complications. Diabetes,1991, 40:1335-1345.
    [194]Miwa K, Nakamura J, Hamada Y, et al. T he role of polyol pathway in glucose-induced apoptosis of cultured retinal pericytes[J]. Diabetes Res Clin Pract, 2003,60(1):1-9.
    [195]Lassegue B, Clempus RE. Vascular NAD(P) H oxidases:specific features, expression, and regulation [J]. Am J Physiol Regul Integr Comp Physiol,2003,285(2):277-297.
    [196]Sheet z MJ, King GL. Molecular understanding of hyperglycemia's adverse effects for diabetic complications [J]. J AMA,2002,288 (20):2579-2588.
    [197]Alghadyan, A.A. Diabetic retinopathy-An update. Saudi Journal of Ophthalmology. 2011,doi:10.1016/j.sjopt.2011.01.009.
    [198]I. Lopez Galvez, M. Protein Kinase C Inhibitors in the Treatment of Diabetic Retinopathy. Review. Current Pharmaceutical Biotechnology.2011,12(6):386-391.
    [199]Nishikawa T, Edelstein D, Brownlee M:The missing link:a single unifying mechanism for diabetic complications.2000, Kidney Int 58 (Suppl.77):S26-S30.
    [200]Nagpala P, Malik AB, Vuong PT, Lum H:Protein kinase C b1 overexpression augments phorbol ester-induced increase in endothelial permeability. J Cell Physiol. 1996,166:249-255,
    [201]Takahara N, Clermont A, Kagokawa H, et al. Transgenic mice overexpressing protein kinase C β isoform mimicked the early changes of diabetes (Abstract). Diabetes.1999, 48:A138.
    [202]Yokota T, Ma RC, Park JY, et al. Role of protein kinase C on the expression of platelet-derived growth factor and endothelin-1 in the retina of diabetic rats and cultured retinal capillary pericytes[J]. Diabetes,2003,52(3):838-845.
    [203]Ishii H, Jirousek MR, Koya D, et al. Amelioration of vascular dysfunctions in diabetic rats by an oral PKC-(3 inhibitor. Science,1996,272:728-731.
    [204]Danis RP, Bingaman DP, Jirousek MR, Yang Y:Inhibition of intraocular neovascularization caused by retinal ischemia in pigs by PKC (3 inhibition with LY333531. Invest Ophthalmol Vis Sci,1998,39:171-179
    [205]Geraldes P, Hiraoka-Yamamot o J, Matsumoto M, et al. Activation of PKC-delta and SH P-1 by hyperglycemia causes vascular cell apoptosis and diabetic retinopathy[J]. Nat Med,2009,15(11):1298-1306.
    [206]Mark Slevin, Therapeutic Angiogenesis for Vascular Diseases:Molecular Mechanisms and Targeted Clinical Approaches for the Treatment of Angiogenic Disease. Springer. 2011, DOI 10.1007/978-90-481-9495-7.
    [207]Simo R, Carrasco E, Garcia-Ramirez M, et al. Angiogenic and antiangiogenic factors in proliferative diabetic retinopathy[J]. Curr Diabetes Rev,2006,2(1):71-98.
    [208]Wu Y, Zhang Q, Ann DK, et al. Increased vascular endothelial growth factor may account for elevated level of plasminogen activator inhibitor-1 via activating ERK1/2 in keloid fibroblasts [J]. Am J Physiol Cell Physiol,2004,286 (4):905-912.
    [209]Mark R Cooson, Paul G. Ince, Philip A Usher, Pamela J Shaw. Poly(ADP-ribose) polymerase in found in both the nucleus and cytoplasm of human CNS neurons [J]. Brain Research,1999,834:182-185.
    [210]Merci P, eigneur M, Bilhou-Nabera C, et al. Nuclear transcription factor kappa B (NF-kappa B)[J]. Rev Med Interne.1998, (19),12:945.
    [211]Van Waes C. Nuclear factor-kappaB in development, prevention, and therapy of cancer[J]. Clin Cancer Res,2007,13(4):1076-1082.
    [212]Baldwin AS Jr. Series introduction:the transcription factor NF-kappaB and human disease[J]. J Clin Invest,2001,107(1):3-6.
    [213]Kaur H, Chen S, Xin X, el al. Diabetes induced extracellular matrix protein expression is mediated by transcription coactivator P300. Diabetes.2006,55(11):3104-3411.
    [214]Zheng L, Szabo C, KemTS. Poly(ADP-ribose)polymerase is involved in the development of diabetic retinopathy via regulation of nuclear factor kappa B. Diabetes. 2004,53:2960-2967.
    [215]Du X. Matsumurat T, Michael B, et al. Inhibition of GAPDH activity by poly(ADP-ribose)polymerase activates three major pathways of hyperglycemic damage in endothelial cells. J Clin Invest.2003,112:1049-1057.
    [216]Xu Y, Hutmg S, Lju ZG, et al. Poly(ADP-ribose)polymerase-1 signaling to mitochondria in necrotic cell death requires RIP1/TRAF2 mediated JNK1 activation. J Biol chem,2006,281:8788-8795.
    [217]Romeo G, Liu WH, Asnaghi V, et al. Activation of nuclear factor-κappa B induced by diabetes and high glucose regulates a proapoptotic program in retinal pericytes [J]. Diabetes,2002,51(7):2241-2248.
    [218]Remeo G, Liu WH, Asnaghi V, et al. Activated of nuclear factor-B induced by diabetes and high glucose regulates a proapoptotic program in retinal pericytes[J]. Diabetes,2002,51(7):2241-2248.
    [219]Kim J, Son JW, Lee JA, et al. Methylglyoxal induces apoptosis mediated by reactive oxygen species in bovine retinal pericytes [J]. J Korean Med Sci,2004,19 (1):95-100.
    [220]Add M, Nala T, MorisMta R, et al. Endothelial apoptosis induced by oxidative stress throngh activation of NF-kappa B:antiapoptotic effect of antioxidant agents on endothelial cells. Hypertension,2001,3s(1):48-55.
    [221]Ho FM, Lin WW, Chon BC, el al. High glucose-induced apoptosis in human vascular endothelial cells is mediated through NF-kappa B and c-Jun NH2-terminal kinase pathway and prevented by PI3K/Akt/eNos pathway. Cell Signal.2006,18(3): 391-399.
    [222]Hassa PO, Buerki C, Lombardi C, et al. Transcriptional coactivation of nuclear factor-kappa B dependent gene expression by p300 is regulated by poly(ADP)-ribose polymerase-1. J Biol Chem.2003,278:45145-45153.
    [223]Rajala MS, Rajala RV, Astey RA, et al. Corneal Cell Survival in Adenovirus type 19 infection Requires Phosphoinositide 3-Kinase/Akt Activation. J Virol.2005,79 (19): 12332-12341.
    [224]Varma S, Lal BK, Zheng R, et al. Hyperglycemia alters P13K and Akt signaling and leads to endothelial cell proliferative dysfunction. Am J Physiol Heart Circ Physiol, 2005,289(4):H1744-H1751.
    [225]Gusfin JA, Ozes ON, Akea H. cell type-specific expression of the kappaB kinases determines the significance of phosphalidylinositol 3-kinase/Akt signaling to NF-kappa B activation. J Biol Chem.2004,279(3):1615-1620.
    [226]Meng F, DMello SR. NF-kappa B stimulates Akt phosphorylation and gene expression by distinct signaling mechanisms. Bioehim Biophys Acta,2003,1630(1):35-40.
    [227]Xin X, Khan ZA, Chen S, et al. Glucose-induced Aktl activation mediates fibronectin synthesis in endothelial cells. Diabetologia,2005,48(11):2428-2436.
    [228]Hennig B, Lei W, Arzuaga X, et al. Linoleic acid induces proinflammatoty events in vascular endothelial cells via activation of PI3K/Akt and ERK1/2 signaling. J Nutr Biochem.2006,17(11):766-772.
    [229]Sainson RC, Harris AL. Regulation of angiogenesis by homotypic and heterotypic notch signalling in endothelial cells and pericytes:from basic research to potential therapies. Angiogenesis,2008,11:41-51.
    [230]Thurston E, Noguera-Troise I, Yancopoulos GD. The Delta paradox:DLL4 blockade leads to more tumour vessels but less tumour growth. Nat. Rev. Cancer.2007,7: 327-331.
    [231]Siekmann AF, Lawson ND. Notch signalling limits angiogenic cell behaviour in developing zebrafish arteries. Nature.2007,445:781-784.
    [232]Guan, E., Wang, J., Laborda, J., et al. T cell leukemia-associated human Notch/translocation-associated Notch homologue has I kappa B-like activity and physically interacts with nuclear factor-kappa B proteins in T cells. J. Exp. Med. 1996,183,2025-2032.
    [233]Kaisho, T., Takeda, K., Tsujimura, T., et al. IkappaB kinase alpha is essential for mature B cell development and function. J. Exp. Med.2001,193,417-426.
    [234]Cheng P, Zlobin A, Volgina V, Gottipati S, Osborne B, et al. Notch-1 regulates NF-kappaB activity in hemopoietic progenitor cells. J Immunol.2001, 167:4458-4467.
    [235]Oakley F, Mann J, Ruddell RG, Pickford J, Weinmaster G, Mann DA. Basal expression of IkappaBalpha is controlled by the mammalian transcriptional repressor RBP-J (CBF1) and its activator Notch1. J Biol Chem.2003,278:24359-24370.
    [236]YAO J, DUAN L, FAN M, el al. Notch1 induces cell cycle arrest and apoptosis in human cervical cancer cells:involvement of nuclear factor kappa B inhibition[J]. Int J Gynecol Cancer.2007,17:502-510.
    [237]Eager, T.N., Tang Q., Wolfe M., et al. Notchl signaling regulates peripheral T cell activation. Immunity.2004,20,407-15.
    [238]Sade H., Krishna S and Sarin A. The anti-apoptosis effect of Notchl requires p561ck-dependent, Akt/PKB-mediated signaling in T cells. Journal of Biological Chemistry.2004,279,2973-44.
    [239]Ciofany M, and Zuniga-Pflucker JC. Notch promotes survival of pre-T cells at the beta-selection checkout by regulating cellular metabolism. Nature Immunology.2005, 6,881-8.
    [240]Brantl S. Antisense RNA regulation and RNA interference. Biochim Biophys Acta. 2002,1575(1-3):15-25.