Vitreous mediators in retinal hypoxic diseases

Roberto dell'Omo, Francesco Semeraro, Giulio Bamonte, Francesco Cifariello, Mario R Romano, Ciro Costagliola, Roberto dell'Omo, Francesco Semeraro, Giulio Bamonte, Francesco Cifariello, Mario R Romano, Ciro Costagliola

Abstract

The causes of retinal hypoxia are many and varied. Under hypoxic conditions, a variety of soluble factors are secreted into the vitreous cavity including growth factors, cytokines, and chemokines. Cytokines, which usually serve as signals between neighboring cells, are involved in essentially every important biological process, including cell proliferation, inflammation, immunity, migration, fibrosis, tissue repair, and angiogenesis. Cytokines and chemokines are multifunctional mediators that can direct the recruitment of leukocytes to sites of inflammation, promote the process, enhance immune responses, and promote stem cell survival, development, and homeostasis. The modern particle-based flow cytometric analysis is more direct, stable and sensitive than the colorimetric readout of the conventional ELISA but, similar to ELISA, is influenced by vitreous hemorrhage, disruption of the blood-retina barrier, and high serum levels of a specific protein. Finding patterns in the expression of inflammatory cytokines specific to a particular disease can substantially contribute to the understanding of its basic mechanism and to the development of a targeted therapy.

References

    1. Janáky M, Grósz A, Tóth E, Benedek K, Benedek G. Hypobaric hypoxia reduces the amplitude of oscillatory potentials in the human ERG. Documenta Ophthalmologica. 2007;114(1):45–51.
    1. Tinjust D, Kergoat H, Lovasik JV. Neuroretinal function during mild systemic hypoxia. Aviation Space and Environmental Medicine. 2002;73(12):1189–1194.
    1. Brown GC, Magargal LE. The ocular ischemic syndrome. Clinical, fluorescein angiographic and carotid angiographic features. International Ophthalmology. 1988;11(4):239–251.
    1. Purtscher O. Angiopathia retinae traumatica: lymphorrhagien desaugengrundes. Graefe's Archive for Clinical and Experimental Ophthalmology. 1912;82:347–371.
    1. Buckley SA, James B. Purtscher’s retinopathy. Postgraduate Medical Journal. 1996;72(749):409–412.
    1. Kitagawa K, Matsumoto M M, Tagaya M, et al. Ischemic tolerance’phenomenon found in the brain. Brain Research. 1990;528(1):21–24.
    1. Prass K, Scharff A, Ruscher K, et al. Hypoxia-induced stroke tolerance in the mouse is mediated by erythropoietin. Stroke. 2003;34(8):1981–1986.
    1. Vilcek J, Feldmann M. Historical review: cytokines as therapeutics and targets of therapeutics. Trends in Pharmacological Sciences. 2004;25(4):201–209.
    1. Struyf S, Proost P, van Damme J. Regulation of the immune response by the interaction of chemokines and proteases. Advances in Immunology. 2003;81:1–44.
    1. Aiello LP, Avery RL, Arrigg PG, et al. Vascular endothelial growth factor in ocular fluid of patients with diabetic retinopathy and other retinal disorders. The New England Journal of Medicine. 1994;331(22):1480–1487.
    1. Vignali DA. Multiplexed particle-based flow cytometric assays. Journal of Immunological Methods. 2000;243(1-2):243–255.
    1. Banerjee S, Savant V, Scott RAH, Curnow SJ, Wallace GR, Murray PL. Multiplex bead analysis of vitreous humor of patients with vitreoretinal disorders. Investigative Ophthalmology and Visual Science. 2007;48(5):2203–2207.
    1. Maier R, Weger M, Haller-Schober EM, et al. Multiplex bead analysis of vitreous and serum concentrations of inflammatory and proangiogenic factors in diabetic patients. Molecular Vision. 2008;14:637–643.
    1. Yoshimura T, Sonoda KH, Sugahara M, et al. Comprehensive analysis of inflammatory immune mediators in vitreoretinal diseases. PLoS ONE. 2009;4(12)e8158
    1. Szabo ME, Droy-Lefaix MT, Doly M, Carre C, Braquet P. Ischemia and reperfusion-induced histologic changes in the rat retina: demonstration of a free radical-mediated mechanism. Investigative Ophthalmology and Visual Science. 1991;32(5):1471–1478.
    1. Szabo ME, Droy-Lefaix MT, Doly M. Direct measurement of free radicals in ischemic/reperfused diabetic rat retina. Clinical Neuroscience. 1997;4(5):240–245.
    1. Kim SY, Kwak JS, Shin JP, Lee SH. The protection of the retina from ischemic injury by the free radical scavenger EGb 761 and zinc in the cat retina. Ophthalmologica. 1998;212(4):268–274.
    1. Hirose F, Kiryu J, Miyamoto K, et al. In vivo evaluation of retinal injury after transient ischemia in hypertensive rats. Hypertension. 2004;43(5):1098–1102.
    1. Kowluru RA, Kowluru V, Xiong Y, Ho YS. Overexpression of mitochondrial superoxide dismutase in mice protects the retina from diabetes-induced oxidative stress. Free Radical Biology and Medicine. 2006;41(8):1191–1196.
    1. Chen BH, Caballero S, Seo S, Grant MB, Lewin AS. Delivery of antioxidant enzyme genes to protect against ischemia/reperfusion-induced injury to retinal microvasculature. Investigative Ophthalmology and Visual Science. 2009;50(12):5587–5595.
    1. Dong A, Shen J, Krause M, et al. Superoxide dismutase 1 protects retinal cells from oxidative damage. Journal of Cellular Physiology. 2006;208(3):516–526.
    1. Nayak MS, Kita M, Marmor MF. Protection of rabbit retina from ischemic injury by superoxide dismutase and catalase. Investigative Ophthalmology and Visual Science. 1993;34(6):2018–2022.
    1. Esterbauer H, Schaur RJ, Zollner H. Chemistry and biochemistry of 4-hydroxynonenal, malonaldehyde and related aldehydes. Free Radical Biology and Medicine. 1991;11(1):81–128.
    1. Poli G, Schaur RJ. 4-Hydroxynonenal in the pathomechanisms of oxidative stress. IUBMB Life. 2000;50(4-5):315–321.
    1. Kaur C, Sivakumar V, Foulds WS. Early response of neurons and glial cells to hypoxia in the retina. Investigative Ophthalmology and Visual Science. 2006;47(3):1126–1141.
    1. Kashiwagi K, Iizuka Y, Mochizuki S, et al. Differences in nitric oxide production: a comparison of retinal ganglion cells and retinal glial cells cultured under hypoxic conditions. Molecular Brain Research. 2003;112(1-2):126–134.
    1. Iadecola C. Bright and dark sides of nitric oxide in ischemic brain injury. Trends in Neurosciences. 1997;20(3):132–139.
    1. Toda N, Nakanishi-Toda M. Nitric oxide: ocular blood flow, glaucoma, and diabetic retinopathy. Progress in Retinal and Eye Research. 2007;26(3):205–238.
    1. Bolaños JP, Almeida A. Roles of nitric oxide in brain hypoxia ischemia. Biochimica et Biophysica Acta. 1999;1411(2-3):415–436.
    1. Fukumura D, Gohongi T, Kadambi A, et al. Predominant role of endothelial nitric oxide synthase in vascular endothelial growth factor-induced angiogenesis and vascular permeability. Proceedings of the National Academy of Sciences of the United States of America. 2001;98(5):2604–2609.
    1. Mishra OP, Zubrow AB, Ashraf QM. Nitric oxide-mediated activation of extracellular signal-regulated kinase (ERK) and c-Jun N-terminal kinase (JNK) during hypoxia in cerebral cortical nuclei of newborn piglets. Neuroscience. 2004;123(1):179–186.
    1. Zubrow AB, Delivoria-Papadopoulos M, Ashraf QM, Fritz KI, Mishra OP. Nitric oxide-mediated Ca2+/calmodulin-dependent protein kinase IV activity during hypoxia in neuronal nuclei from newborn piglets. Neuroscience Letters. 2002;335(1):5–8.
    1. Zubrow AB, Delivoria-Papadopoulos M, Ashraf QM, Ballesteros JR, Fritz KI, Mishra OP. Nitric oxide-mediated expression of Bax protein and DNA fragmentation during hypoxia in neuronal nuclei from newborn piglets. Brain Research. 2002;954(1):60–67.
    1. Nguyen T, Brunson D, Crespi CL, Penman BW, Wishnok JS, Tannenbaum SR. DNA damage and mutation in human cells exposed to nitric oxide in vitro. Proceedings of the National Academy of Sciences of the United States of America. 1992;89(7):3030–3034.
    1. Zhang RL, Chopp M, Chen H, Garcia JH. Temporal profile of ischemic tissue damage, neutrophil response, and vascular plugging following permanent and transient (2H) middle cerebral artery occlusion in the rat. Journal of the Neurological Sciences. 1994;125(1):3–10.
    1. Gross WL, Bak MI, Ingwall JS, et al. Nitric oxide inhibits creatinekinase and regulates rat heart contractile reserves. Proceedings of the National Academy of Sciences of the United States of America. 1996;93(11):5604–5609.
    1. Beckman JS, Beckman TW, Chen J, Marshall PA, Freeman BA. Apparent hydroxyl radical production by peroxynitrite: implications for endothelial injury from nitric oxide and superoxide. Proceedings of the National Academy of Sciences of the United States of America. 1990;87(4):1620–1624.
    1. Lipton SA, Choi YB, Pan ZH, et al. A redox-based mechanism for the neuroprotective and neurodestructive effects of nitric oxide and related nitroso-compounds. Nature. 1993;364(6438):626–632.
    1. Marumo T, Noll T, Schini-Kerth VB, et al. Significance of nitric oxide and peroxynitrite in permeability changes of the retinal microvascular endothelial cell monolayer induced by vascular endothelial growth factor. Journal of Vascular Research. 1999;36(6):510–515.
    1. Salgo MG, Bermúdez E, Squadrito GL, Pryor WA. Peroxynitrite causes DNA damage and oxidation of thiols in rat thymocytes. Archives of Biochemistry and Biophysics. 1995;322(2):500–505.
    1. Salvemini D, Wang Z, Stern M, Currie MG, Misko TP. Peroxynitrite decomposition catalysts: therapeutics for peroxynitrite-mediated pathology. Proceedings of the National Academy of Sciences of the United States of America. 1998;95(5):2695–2703.
    1. Anderson SM, Krinsky NI. Protective action of carotenoid pigments against photodynamic damage to liposomes. Photochemistry and Photobiology. 1973;18(5):403–408.
    1. Arstila AU, Smith MA, Trump BF. Microsomal lipid peroxidation: morphological characterization. Science. 1972;175(4021):530–533.
    1. Kondo T, Kinouchi H, Kawase M, Yoshimoto T. Differential response in the release of hydrogen peroxide between astroglial cells and endothelial cells following hypoxia/reoxygenation. Neuroscience Letters. 1996;215(2):103–106.
    1. Auge N, Pieraggi MT, Thiers JC, Negre-Salvayre A, Salvayre R. Proliferative and cytotoxic effects of mildly oxidized low-density lipoproteins on vascular smooth-muscle cells. Biochemical Journal. 1995;309(3):1015–1020.
    1. D’Amore PA, Sweet E. Effects of hyperoxia on microvascular cells in vitro. In Vitro Cellular and Developmental Biology. 1987;23(2):123–128.
    1. Halks-Miller M, Henderson M, Eng LF. α tocopherol decreases lipid peroxidation, neuronal necrosis, and reactive gliosis in reaggregate cultures of fetal rat brain. Journal of Neuropathology and Experimental Neurology. 1986;45(4):471–484.
    1. Carbajal JM, Schaeffer RC. H2O2 and genistein differentially modulate protein tyrosine phosphorylation, endothelial morphology, and monolayer barrier function. Biochemical and Biophysical Research Communications. 1998;249(2):461–466.
    1. Du Y, Miller CM, Kern TS. Hyperglycemia increases mitochondrial superoxide in retina and retinal cells. Free Radical Biology and Medicine. 2003;35(11):1491–1499.
    1. Kowluru RA. Diabetic retinopathy: mitochondrial dysfunction and retinal capillary cell death. Antioxidants and Redox Signaling. 2005;7(11-12):1581–1587.
    1. Nishikawa T, Araki E. Impact of mitochondrial ROS production in the pathogenesis of diabetes mellitus and its complications. Antioxidants and Redox Signaling. 2007;9(3):343–353.
    1. Kowluru RA. Effect of advanced glycation end products on accelerated apoptosis of retinal capillary cells under in vitro conditions. Life Sciences. 2005;76(9):1051–1060.
    1. Grammas P, Riden M. Retinal endothelial cells are more susceptible to oxidative stress and increased permeability than brain-derived endothelial cells. Microvascular Research. 2003;65(1):18–23.
    1. Du Y, Smith MA, Miller CM, Kern TS. Diabetes-induced nitrative stress in the retina, and correction by aminoguanidine. Journal of Neurochemistry. 2002;80(5):771–779.
    1. Castilho Á, Aveleira CA, Leal EC, et al. Heme oxygenase-1 protects retinal endothelial cells against high glucose- and oxidative/nitrosative stress-induced toxicity. PLoS ONE. 2012;7(8)e42428
    1. Massey SC. Cell types using glutamate as a neurotransmitter in the vertebrate retina. Progress in Retinal and Eye Research. 1990;9:399–425.
    1. Benveniste H, Drejer J, Schousboe A, Diemer NH. Elevation of the extracellular concentrations of glutamate and aspartate in rat hippocampus during transient cerebral ischemia monitored by intracerebral microdialysis. Journal of Neurochemistry. 1984;43(5):1369–1374.
    1. Lu YM, Lu BF, Zhao FQ, Yan YL, Ho XP. Accumulation of glutamate is regulated by calcium and protein kinase C in rat hippocampal slices exposed to ischemic states. Hippocampus. 1993;3(2):221–227.
    1. Brandstätter JH, Koulen P, Wässle H. Diversity of glutamate receptors in the mammalian retina. Vision Research. 1998;38(10):1385–1397.
    1. Gründer T, Kohler K, Guenthe E. Alterations in NMDA receptor expression during retinal degeneration in the RCS rat. Visual Neuroscience. 2001;18(5):781–787.
    1. Sucher NJ, Lipton SA, Dreyer EB. Molecular basis of glutamate toxicity in retinal ganglion cells. Vision Research. 1997;37(24):3483–3493.
    1. MacDermott AB, Mayer ML, Westbrook GL. NMDA-receptor activation increases cytoplasmic calcium concentration in cultured spinal cord neurones. Nature. 1986;321(6069):519–522.
    1. Hollmann M, Hartley M, Heinemann S. Ca2+ permeability of KA-AMPA-gated glutamate receptor channels depends on subunit composition. Science. 1991;252(5007):851–853.
    1. Rörig B, Grantyn R. Rat retinal ganglion cells express Ca2+-permeable non-NMDA glutamate receptors during the period of histogenetic cell death. Neuroscience Letters. 1993;153(1):32–36.
    1. Siliprandi R, Canella R, Carmignoto G, et al. N-methyl-D-aspartate-induced neurotoxicity in the adult rat retina. Visual Neuroscience. 1992;8(6):567–573.
    1. Sucher NJ, Aizenman E, Lipton SA. N-methyl-D-aspartate antagonists prevent kainate neurotoxicity in rat retinal ganglion cells in vitro. Journal of Neuroscience. 1991;11(4):966–971.
    1. Sucher NJ, Wong LA, Lipton SA. Redox modulation of NMDA receptor-mediated Ca2+ flux in mammalian central neurons. NeuroReport. 1990;1(1):29–32.
    1. Nicotera P, Orrenius S. The role of calcium in apoptosis. Cell Calcium. 1998;23(2-3):173–180.
    1. Sattler R, Tymianski M. Molecular mechanisms of glutamate receptor-mediated excitotoxic neuronal cell death. Molecular Neurobiology. 2001;24(1–3):107–129.
    1. Dugan LL, Sensi SL, Canzoniero LMT, et al. Mitochondrial production of reactive oxygen species in cortical neurons following exposure to N-methyl-D-aspartate. Journal of Neuroscience. 1995;15(10):6377–6388.
    1. Katsura K, Kristian T, Siesjo BK. Energy metabolism, ion homeostasis, and cell damage in the brain. Biochemical Society Transactions. 1994;22(4):991–996.
    1. Garthwaite G, Garthwaite J. AMPA neurotoxicity in rat cerebellar and hippocampal slices: histological evidence for three mechanisms. European Journal of Neuroscience. 1991;3(8):715–728.
    1. Kiss JP, Vizi ES. Nitric oxide: a novel link between synaptic and nonsynaptic transmission. Trends in Neurosciences. 2001;24(4):211–215.
    1. De A, Krueger JM, Simasko SM. Glutamate induces the expression and release of tumor necrosis factor-α in cultured hypothalamic cells. Brain Research. 2005;1053(1-2):54–61.
    1. Noda M, Nakanishi H, Nabekura J, Akaike N. AMPA-kainate subtypes of glutamate receptor in rat cerebral microglia. Journal of Neuroscience. 2000;20(1):251–258.
    1. Matute C, Alberdi E, Domercq M, Pérez-Cerdá F, Pérez-Samartín A, Sánchez-Gómez MV. The link between excitotoxic oligodendroglial death and demyelinating diseases. Trends in Neurosciences. 2001;24:224–230.
    1. Hagan P, Poole S, Bristow AF, Tilders F, Silverstein FS. Intracerebral NMDA injection stimulates production of interleukin-1β in perinatal rat brain. Journal of Neurochemistry. 1996;67(5):2215–2218.
    1. Ratan RR, Murphy TH, Baraban JM. Oxidative stress induces apoptosis in embryonic cortical neurons. Journal of Neurochemistry. 1994;62(1):376–379.
    1. Meister A, Anderson ME. Glutathione. Annual Review of Biochemistry. 1983;52:711–760.
    1. Mizui T, Kinouchi H, Chan PH. Depletion of brain glutathione by buthionine sulfoximine enhances cerebral ischemic injury in rats. American Journal of Physiology. 1992;262(2):H313–H317.
    1. Bobyn PJ, Franklin JL, Wall CM, Thornhill JA, Juurlink BHJ, Paterson PG. The effects of dietary sulfur amino acid deficiency on rat brain glutathione concentration and neural damage in global hemispheric hypoxia—ischemia. Nutritional Neuroscience. 2002;5(6):407–416.
    1. Coyle JT, Puttfarcken P. Oxidative stress, glutamate, and neurodegenerative disorders. Science. 1993;262(5134):689–695.
    1. Harada T, Harada C, Nakamura K, et al. The potential role of glutamate transporters in the pathogenesis of normal tension glaucoma. Journal of Clinical Investigation. 2007;117(7):1763–1770.
    1. Yoshida S, Yoshida A, Ishibashi T. Induction of IL-8, MPC-1, and bFGF by TNF-α in retinal glial cells: implications for retinal neovascularization during post-ischemic inflammation. Graefe’s Archive for Clinical and Experimental Ophthalmology. 2004;242(5):409–413.
    1. Zheng L, Gong B, Hatala DA, Kern TS. Retinal ischemia and reperfusion causes capillary degeneration: similarities to diabetes. Investigative Ophthalmology and Visual Science. 2007;48(1):361–367.
    1. Joussen AM, Poulaki V, Qin W, et al. Retinal vascular endothelial growth factor induces intercellular adhesion molecule-1 and endothelial nitric oxide synthase expression and initiates early diabetic retinal leukocyte adhesion in vivo. American Journal of Pathology. 2002;160(2):501–509.
    1. Wong D, Dorovini-Zis K. Upregulation of intercellular adhesion molecule-1 (ICAM-1) expression in primary cultures of human brain microvessel endothelial cells by cytokines and lipopolysaccharide. Journal of Neuroimmunology. 1992;39(1-2):11–22.
    1. Hess DC, Bhutwala T, Sheppard JC, Zhao W, Smith J. ICAM-1 expression on human brain microvascular endothelial cells. Neuroscience Letters. 1994;168(1-2):201–204.
    1. McHale JF, Harari OA, Marshall D, Haskard DO. TNF-α and IL-1 sequentially induce endothelial ICAM-1 and VCAM-1 expression in MRL/lpr lupus-prone mice. Journal of Immunology. 1999;163(7):3993–4000.
    1. Feuerstein G, Wang X, Barone FC. Cytokines in brain ischemia—the role of TNF-α . Cellular and Molecular Neurobiology. 1998;18(6):695–701.
    1. Wang J, Beekhuizen H, van Furth R. Surface molecules involved in the adherence of recombinant interferon-γ (rIFN-γ)-stimulated human monocytes to vascular endothelial cells. Clinical and Experimental Immunology. 1994;95(2):263–269.
    1. López-Figueroa MO, Day HE, Lee S, Rivier C, Akil H, Watson SJ. Temporal and anatomical distribution of nitric oxide synthase mRNA expression and nitric oxide production during central nervous system infl ammation. Brain Research. 2000;852(1):239–246.
    1. Kadhim H, Khalifa M, Deltenre P, Casimir G, Sébire G. Molecular mechanisms of cell death in periventricular leukomalacia. Neurology. 2006;67(2):293–299.
    1. Kern TS. Contributions of inflammatory processes to the development of the early stages of diabetic retinopathy. Experimental Diabetes Research. 2007;2007:14 pages.95103
    1. Iliaki E, Poulaki V, Mitsiades N, Mitsiades CS, Miller JW, Gragoudas ES. Role of α4 integrin (cd49d) in the pathogenesis of diabetic retinopathy. Investigative Ophthalmology and Visual Science. 2009;50(10):4890–4904.
    1. Adamis AP, Berman AJ. Immunological mechanisms in the pathogenesis of diabetic retinopathy. Seminars in Immunopathology. 2008;30(2):65–84.
    1. Tawfik A, Sanders T, Kahook K, Akeel S, Elmarakby A, Al-Shabrawey M. Suppression of retinal peroxisome proliferator-activated receptor γ in experimental diabetes and oxygen-induced retinopathy: role of NADPH oxidase. Investigative Ophthalmology and Visual Science. 2009;50(2):878–884.
    1. Tezel G, Wax MB. Increased production of tumor necrosis factor-α by glial cells exposed to simulated ischemia or elevated hydrostatic pressure induces apoptosis in cocultured retinal ganglion cells. Journal of Neuroscience. 2000;20(23):8693–8700.
    1. Carswell EA, Old LJ, Kassel RL, Green S, Fiore N, Williamson B. An endotoxin-induced serum factor that causes necrosis of tumors. Proceedings of the National Academy of Sciences of the United States of America. 1975;72(9):3666–3670.
    1. Fràter-Schröder M, Risau W, Hallmann R, Gautschi P, Böhlen P. Tumor necrosis factor type α, a potent inhibitor of endothelial cell growth in vitro, is angiogenic in vivo. Proceedings of the National Academy of Sciences of the United States of America. 1987;84(15):5277–5281.
    1. Rosenbaum JT, Howes ELJ, Rubin RM, Samples JR. Ocular inflammatory effects of intravitreally injected tumor necrosis factor. American Journal of Pathology. 1988;133(1):47–53.
    1. Yoshida S, Ono M, Shono T, et al. Involvement of interleukin-8, vascular endothelial growth factor, and basic fibroblast growth factor in tumor necrosis factor α-dependent angiogenesis. Molecular and Cellular Biology. 1997;17(7):4015–4023.
    1. Giraudo E, Primo L, Audero E, et al. Tumor necrosis factor-α regulates expression of vascular endothelial growth factor receptor-2 and of its co-receptor neuropilin-1 in human vascular endothelial cells. The Journal of Biological Chemistry. 1998;273(34):22128–22135.
    1. Fajardo LF, Kwan HH, Kowalski J, Prionas SD, Allison AC. Dual role of tumor necrosis factor-α in angiogenesis. American Journal of Pathology. 1992;140(3):539–544.
    1. Tartaglia LA, Ayres TM, Wong GHW, Goeddel DV. A novel domain within the 55 kd TNF receptor signals cell death. Cell. 1993;74(5):845–853.
    1. Song HY, Régnier CH, Kirschning CJ, Goeddel DV, Rothe M. Tumor necrosis factor (TNF)-mediated kinase cascades: bifurcation of nuclear factor-κB and c-jun N-terminal kinase (JNK/SAPK) pathways at TNF receptor-associated factor 2. Proceedings of the National Academy of Sciences of the United States of America. 1997;94(18):9792–9796.
    1. Ishida S, Usui T, Yamashiro K, et al. VEGF 164-mediated inflammation is required for pathological, but not physiological, ischemia-induced retinal neovascularization. Journal of Experimental Medicine. 2003;198(3):483–489.
    1. Lang RA, Bishop MJ. Macrophages are required for cell death and tissue remodeling in the developing mouse eye. Cell. 1993;74(3):453–462.
    1. Armstrong D, Ueda T, Ueda T, et al. Lipid hydroperoxide stimulates retinal neovascularization in rabbit retina through expression of tumor necrosis factor-α, vascular endothelial growth factor and platelet-derived growth factor. Angiogenesis. 1998;2(1):93–104.
    1. Camussi G, Albano E, Tetta C, Bussolino F. The molecular action of tumor necrosis factor-α . European Journal of Biochemistry. 1991;202(1):3–14.
    1. Majka S, McGuire PG, Das A. Regulation of matrix metalloproteinase expression by tumor necrosis factor in a murine model of retinal neovascularization. Investigative Ophthalmology and Visual Science. 2002;43(1):260–266.
    1. Derevjanik NL, Vinores SA, Xiao WH, et al. Quantitative assessment of the integrity of the blood-retinal barrier in mice. Investigative Ophthalmology and Visual Science. 2002;43(7):2462–2467.
    1. Wilkinson-Berka JL, Tan G, Jaworski K, Miller AG. Identification of a retinal aldosterone system and the protective effects of mineralocorticoid receptor antagonism on retinal vascular pathology. Circulation Research. 2009;104(1):124–133.
    1. Zeng HY, Green WR, Tso MOM. Microglial activation in human diabetic retinopathy. Archives of Ophthalmology. 2008;126(2):227–232.
    1. Joussen AM, Poulaki V, Mitsiades N, et al. Nonsteroidal anti-inflammatory drugs prevent early diabetic retinopathy via TNF-α suppression. The FASEB Journal. 2002;16(3):438–440.
    1. Demircan N, Safran BG, Soylu M, Ozcan AA, Sizmaz S. Determination of vitreous interleukin-1 (IL-1) and tumour necrosis factor (TNF) levels in proliferative diabetic retinopathy. Eye. 2006;20(12):1366–1369.
    1. Funatsu H, Yamashita H, Noma H, et al. Aqueous humor levels of cytokines are related to vitreous levels and progression of diabetic retinopathy in diabetic patients. Graefe’s Archive for Clinical and Experimental Ophthalmology. 2005;243(1):3–8.
    1. Malik RA, Li C, Aziz W, Olson JA, et al. Elevated plasma CD105 and vitreous VEGF levels in diabetic retinopathy. Journal of Cellular and Molecular Medicine. 2005;9(3):692–697.
    1. Murugeswari P, Shukla D, Rajendran A, Kim R, Namperumalsamy P, Muthukkaruppan V. Proinflammatory cytokines and angiogenic and anti-angiogenic factors in vitreous of patients with proliferative diabetic retinopathy and eales’ disease. Retina. 2008;28(6):817–824.
    1. Hiasa KI, Ishibashi M, Ohtani K, et al. Gene transfer of stromal cell-derived factor-1α enhances ischemic vasculogenesis and angiogenesis via vascular endothelial growth factor/endothelial nitric oxide synthase-related pathway: next-generation chemokine therapy for therapeutic neovascularization. Circulation. 2004;109(20):2454–2461.
    1. Arjamaa O, Pöllönen M, Kinnunen K, Ryhänen T, Kaarniranta K. Increased IL-6 levels are not related to NF-κB or HIF-1α transcription factors activity in the vitreous of proliferative diabetic retinopathy. Journal of Diabetes and Its Complications. 2011;25(6):393–397.
    1. Goodwin GH, Sanders C, Johns EW. A new group of chromatin associated proteins with a high content of acidic and basic amino acids. European Journal of Biochemistry. 1973;38(1):14–19.
    1. Lotze MT, Tracey KJ. High-mobility group box 1 protein (HMGB1): nuclear weapon in the immune arsenal. Nature Reviews Immunology. 2005;5(4):331–342.
    1. Arimura N, Ki-I Y, Hashiguchi T, et al. Intraocular expression and release of high-mobility group box 1 protein in retinal detachment. Laboratory Investigation. 2009;89(3):278–289.
    1. Watanabe T, Keino H, Sato Y, Kudo A, Kawakami H, Okada AA. High mobility group box protein-1 in experimental autoimmune uveoretinitis. Investigative Ophthalmology and Visual Science. 2009;50(5):2283–2290.
    1. Hori O, Brett J, Slattery T, et al. The receptor for advanced glycation end products (RAGE) is a cellular binding site for amphoterin. Mediation of neurite outgrowth and co-expression of RAGE and amphoterin in the developing nervous system. The Journal of Biological Chemistry. 1995;270(43):25752–25761.
    1. Jakuš V, Rietbrock N. Advanced glycation end-products and the progress of diabetic vascular complications. Physiological Research. 2004;53(2):131–142.
    1. Yang S, Hirooka K, Liu Y, et al. Deleterious role of anti-high mobility group box 1 monoclonal antibody in retinal ischemia-reperfusion injury. Current Eye Research. 2011;36(11):1037–1046.
    1. Klune JR, Dhupar R, Cardinal J, Billiar TR, Tsung A. HMGB1: endogenous danger signaling. Molecular Medicine. 2008;14(7-8):476–484.
    1. Hreggvidsdottir HS, Östberg T, Wähämaa H, et al. The alarmin HMGB1 acts in synergy with endogenous and exogenous danger signals to promote inflammation. Journal of Leukocyte Biology. 2009;86(3):655–662.
    1. Mitola S, Belleri M, Urbinati C, et al. Cutting edge: extracellular high mobility group box-1 protein is a proangiogenic cytokine. Journal of Immunology. 2006;176(1):12–15.
    1. Biscetti F, Straface G, de Cristofaro R, et al. High-mobility group box-1 protein promotes angiogenesis after peripheral ischemia in diabetic mice through a VEGF-dependent mechanism. Diabetes. 2010;59(6):1496–1505.
    1. Lee JJ, Hsiao CC, Yang I, et al. High-mobility group box 1 protein is implicated in advanced glycation end products-induced vascular endothelial growth factor a production in the rat retinal ganglion cell line RGC-5. Molecular Vision. 2012;18:838–850.
    1. Murphy PM, Baggiolini M, Charo IF, et al. International union of pharmacology. XXII. Nomenclature for chemokine receptors. Pharmacological Reviews. 2000;52(1):145–176.
    1. Abu El-Asrar AM, Struyf S, Kangave D, Geboes K, van Damme J. Chemokines in proliferative diabetic retinopathy and proliferative vitreoretinopathy. European Cytokine Network. 2006;17(3):155–165.
    1. Hong KH, Ryu J, Han KH. Monocyte chemoattractant protein-1-induced angiogenesis is mediated by vascular endothelial growth factor-A. Blood. 2005;105(4):1405–1407.
    1. Silverman MD, Zamora DO, Pan Y, et al. Constitutive and inflammatory mediator-regulated fractalkine expression in human ocular tissues and cultured cells. Investigative Ophthalmology and Visual Science. 2003;44(4):1608–1615.
    1. You JJ, Yang CH, Huang JS, Chen MS, Yang CM. Fractalkine, a CX3C chemokine, as a mediator of ocular angiogenesis. Investigative Ophthalmology and Visual Science. 2007;48(11):5290–5298.
    1. Wakabayashi Y, Usui Y, Okunuki Y, et al. Increased levels of monokine induced by interferon-γ (Mig) in the vitreous of patients with diabetic retinopathy. Diabetic Medicine. 2008;25(7):875–877.
    1. Joussen AM, Poulaki V, Le ML, et al. A central role for inflammation in the pathogenesis of diabetic retinopathy. The FASEB Journal. 2004;18(12):1450–1452.
    1. Yamaguchi JI, Kusano KF, Masuo O, et al. Stromal cell-derived factor-1 effects on ex vivo expanded endothelial progenitor cell recruitment for ischemic neovascularization. Circulation. 2003;107(9):1322–1328.
    1. de Falco E, Porcelli D, Torella AR, et al. SDF-1 involvement in endothelial phenotype and ischemia-induced recruitment of bone marrow progenitor cells. Blood. 2004;104(12):3472–3482.
    1. Walter DH, Haendeler J, Reinhold J, et al. Impaired CXCR4 signaling contributes to the reduced neovascularization capacity of endothelial progenitor cells from patients with coronary artery disease. Circulation Research. 2005;97(11):1142–1151.
    1. Reddy K, Zhou Z, Jia SF, et al. Stromal cell-derived factor-1 stimulates vasculogenesis and enhances Ewing’s sarcoma tumor growth in the absence of vascular endothelial growth factor. International Journal of Cancer. 2008;123(4):831–837.
    1. Stellos K, Langer H, Daub K, et al. Platelet-derived stromal cell-derived factor-1 regulates adhesion and promotes differentiation of human CD34+ cells to endothelial progenitor cells. Circulation. 2008;117(2):206–215.
    1. Butler JM, Guthrie SM, Koc M, et al. SDF-1 is both necessary and sufficient to promote proliferative retinopathy. Journal of Clinical Investigation. 2005;115(1):86–93.
    1. Blom IE, Goldschmeding R, Leask A. Gene regulation of connective tissue growth factor: new targets for antifibrotic therapy? Matrix Biology. 2002;21(6):473–482.
    1. Abu El-Asrar AM, Struyf S, Verbeke H, van Damme J, Geboes K. Circulating bone-marrow-derived endothelial precursor cells contribute to neovascularization in diabetic epiretinal membranes. Acta Ophthalmologica. 2011;89(3):222–228.
    1. Déry MAC, Michaud MD, Richard DE. Hypoxia-inducible factor 1: regulation by hypoxic and non-hypoxic activators. International Journal of Biochemistry and Cell Biology. 2005;37(3):535–540.
    1. Mazure NM, Brahimi-Horn MC, Berta MA, et al. HIF-1: master and commander of the hypoxic world: a pharmacological approach to its regulation by siRNAs. Biochemical Pharmacology. 2004;68(6):971–980.
    1. Pugh CW, Ratcliffe PJ. Regulation of angiogenesis by hypoxia: role of the HIF system. Nature Medicine. 2003;9(6):677–684.
    1. Lukiw WJ, Ottlecz A, Lambrou G, et al. Coordinate activation of HIF-1 and NF-κB DNA binding and COX-2 and VEGF expression in retinal cells by hypoxia. Investigative Ophthalmology and Visual Science. 2003;44(10):4163–4170.
    1. Gao W, Ferguson G, Connell P, et al. High glucose concentrations alter hypoxia-induced control of vascular smooth muscle cell growth via a HIF-1α-dependent pathway. Journal of Molecular and Cellular Cardiology. 2007;42(3):609–619.
    1. Treins C, Giorgetti-Peraldi S, Murdaca J, Monthouël-Kartmann MN, van Obberghen E. Regulation of hypoxia-inducible factor (HIF)-1 activity and expression of HIF hydroxylases in response to insulin-like growth factor I. Molecular Endocrinology. 2005;19(5):1304–1317.
    1. Poulaki V, Qin W, Joussen AM, et al. Acute intensive insulin therapy exacerbates diabetic blood-retinal barrier breakdown via hypoxia-inducible factor-1α and VEGF. Journal of Clinical Investigation. 2002;109(6):805–815.
    1. El-Asrar AMA, Missotten L, Geboes K. Expression of hypoxia-inducible factor-1α and the protein products of its target genes in diabetic fibrovascular epiretinal membranes. British Journal of Ophthalmology. 2007;91(6):822–826.
    1. Lim JI, Spee C, Hinton DR. A comparison of hypoxia-inducible factor-α in surgically excised neovascular membranes of patients with diabetes compared with idiopathic epiretinal membranes in nondiabetic patients. Retina. 2010;30(9):1472–1478.
    1. Frede S, Stockmann C, Freitag P, Fandrey J. Bacterial lipopolysaccharide induces HIF-1 activation in human monocytes via p44/42 MAPK and NF-κB. Biochemical Journal. 2006;396(3):517–527.
    1. Bonello S, Zähringer C, BelAiba RS, et al. Reactive oxygen species activate the HIF-1α promoter via a functional NFκB site. Arteriosclerosis, Thrombosis, and Vascular Biology. 2007;27(4):755–761.
    1. Kinnunen K, Puustjärvi T, Teräsvirta M, et al. Differences in retinal neovascular tissue and vitreous humour in patients with type 1 and type 2 diabetes. British Journal of Ophthalmology. 2009;93(8):1109–1115.
    1. Arjamaa O, Nikinmaa M, Salminen A, Kaarniranta K. Regulatory role of HIF-1α in the pathogenesis of age-related macular degeneration (AMD) Ageing Research Reviews. 2009;8(4):349–358.
    1. Salminen A, Kaarniranta K. Insulin/IGF-1 paradox of aging: regulation via AKT/IKK/NF-κB signaling. Cellular Signalling. 2010;22(4):573–577.
    1. Ferrara N, Gerber HP, LeCouter J. The biology of VEGF and its receptors. Nature Medicine. 2003;9(6):669–676.
    1. Takahashi H, Shibuya M. The vascular endothelial growth factor (VEGF)/VEGF receptor system and its role under physiological and pathological conditions. Clinical Science. 2005;109(3):227–241.
    1. Shibuya M, Ito N, Claesson-Welsh L. Structure and function of vascular endothelial growth factor receptor-1 and -2. Current Topics in Microbiology and Immunology. 1999;237:60–83.
    1. Campochiaro PA. Ocular neovascularisation and excessive vascular permeability. Expert Opinion on Biological Therapy. 2004;4(9):1395–1402.
    1. Hiratsuka S, Kataoka Y, Nakao K, et al. Vascular endothelial growth factor A (VEGF-A) is involved in guidance of VEGF receptor-positive cells to the anterior portion of early embryos. Molecular and Cellular Biology. 2005;25(1):355–363.
    1. Sakurai Y, Ohgimoto K, Kataoka Y, Yoshida N, Shibuya M. Essential role of Flk-1 (VEGF receptor 2) tyrosine residue 1173 in vasculogenesis in mice. Proceedings of the National Academy of Sciences of the United States of America. 2005;102(4):1076–1081.
    1. Asahara T, Bauters C, Pastore C, et al. Local delivery of vascular endothelial growth factor accelerates reendothelialization and attenuates intimal hyperplasia in balloon-injured rat carotid artery. Circulation. 1995;91(11):2793–2801.
    1. Gennaro G, Ménard C, Michaud SE, Rivard A. Age-dependent impairment of reendothelialization after arterial injury: role of vascular endothelial growth factor. Circulation. 2003;107(2):230–233.
    1. Safran M, Kaelin WG., Jr. HIF hydroxylation and the mammalian oxygen-sensing pathway. Journal of Clinical Investigation. 2003;111(6):779–783.
    1. Ikeda E, Achen MG, Breier G, Risau W. Hypoxia-induced transcriptional activation and increased mRNA stability of vascular endothelial growth factor in C6 glioma cells. The Journal of Biological Chemistry. 1995;270(34):19761–19766.
    1. Levy AP, Levy NS, Wegner S, Goldberg MA. Transcriptional regulation of the rat vascular endothelial growth factor gene by hypoxia. The Journal of Biological Chemistry. 1995;270(22):13333–13340.
    1. Stein I, Neeman M, Shweiki D, Itin A, Keshet E. Stabilization of vascular endothelial growth factor mRNA by hypoxia and hypoglycemia and coregulation with other ischemia-induced genes. Molecular and Cellular Biology. 1995;15(10):5363–5368.
    1. Melder RJ, Koenig GC, Witwer BP, Safabakhsh N, Munn LL, Jain RK. During angiogenesis, vascular endothelial growth factor and basic fibroblast growth factor regulate natural killer cell adhesion to tumor endothelium. Nature Medicine. 1996;2(9):992–997.
    1. Lu PCS, Ye H, Maeda M, Azar DT. Immunolocalization and gene expression of matrilysin during corneal wound healing. Investigative Ophthalmology and Visual Science. 1999;40(1):20–27.
    1. Min JK, Kim YM, Kim SW, et al. TNF-related activation-induced cytokine enhances leukocyte adhesiveness: induction of ICAM-1 and VCAM-1 via TNF receptor-associated factor and protein kinase C-dependent NF-κB activation in endothelial cells. Journal of Immunology. 2005;175(1):531–540.
    1. Clermont AC, Aiello LP, Mori F, Aiello LM, Bursell SE. Vascular endothelial growth factor and severity of nonproliferative diabetic retinopathy mediate retinal hemodynamics in vivo: a potential role for vascular endothelial growth factor in the progression of nonproliferative diabetic retinopathy. American Journal of Ophthalmology. 1997;124(4):433–446.
    1. Adamis AP, Miller JW, Bernal MT, et al. Increased vascular endothelial growth factor levels in the vitreous of eyes with proliferative diabetic retinopathy. American Journal of Ophthalmology. 1994;118(4):445–450.
    1. Matsunaga N, Chikaraishi Y, Izuta H, et al. Role of soluble vascular endothelial growth factor receptor-1 in the vitreous in proliferative diabetic retinopathy. Ophthalmology. 2008;115(11):1916–1922.
    1. Ozaki H, Seo MS, Ozaki K, et al. Blockade of vascular endothelial cell growth factor receptor signaling is sufficient to completely prevent retinal neovascularization. American Journal of Pathology. 2000;156(2):697–707.
    1. Kaur C, Sivakumar V, Foulds WS. Early response of neurons and glial cells to hypoxia in the retina. Investigative Ophthalmology and Visual Science. 2006;47(3):1125–1141.
    1. Kaur C, Sivakumar V, Yong Z, Lu J, Foulds WS, Ling EA. Blood-retinal barrier disruption and ultrastructural changes in the hypoxic retina in adult rats: the beneficial effect of melatonin administration. Journal of Pathology. 2007;212(4):429–439.
    1. Penn JS, Madan A, Caldwell RB, Bartoli M, Caldwell RW, Hartnett ME. Vascular endothelial growth factor in eye disease. Progress in Retinal and Eye Research. 2008;27(4):331–371.
    1. Kondo S, Kubota S, Shimo T, et al. Connective tissue growth factor increased by hypoxia may initiate angiogenesis in collaboration with matrix metalloproteinases. Carcinogenesis. 2002;23(5):769–776.
    1. Pi L, Xia H, Liu J, Shenoy AK, Hauswirth WW, Scott EW. Role of connective tissue growth factor in the retinal vasculature during development and ischemia. Investigative Ophthalmology and Visual Science. 2011;52(12):8701–8710.
    1. Moussad EE, Brigstock DR. Connective tissue growth factor. What’s in a name? Molecular Genetics and Metabolism. 2000;71(1-2):276–292.
    1. Kita T, Hata Y, Miura M, Kawahara S, Nakao S, Ishibashi T. Functional characteristics of connective tissue growth factor on vitreoretinal cells. Diabetes. 2007;56(5):1421–1428.
    1. Kuiper EJ, van Nieuwenhoven FA, de Smet MD, et al. The angio-fibrotic switch of VEGF and CTGF in proliferative diabetic retinopathy. PLoS ONE. 2008;3(7)e2675
    1. Fazel SS, Chen L, Angoulvant D, et al. Activation of c-kit is necessary for mobilization of reparative bone marrow progenitor cells in response to cardiac injury. The FASEB Journal. 2008;22(3):930–940.
    1. Huang PH, Chen YH, Wang CH, et al. Matrix metalloproteinase-9 is essential for ischemia-induced neovascularization by modulating bone marrow-derived endothelial progenitor cells. Arteriosclerosis, Thrombosis, and Vascular Biology. 2009;29(8):1179–1184.
    1. Li TS, Hamano K, Nishida M, et al. CD117+ stem cells play a key role in therapeutic angiogenesis induced by bone marrow cell implantation. American Journal of Physiology. 2003;285(3):H931–H937.
    1. Miyamoto Y, Suyama T, Yashita T, Akimaru H, Kurata H. Bone marrow subpopulations contain distinct types of endothelial progenitor cells and angiogenic cytokine-producing cells. Journal of Molecular and Cellular Cardiology. 2007;43(5):627–635.
    1. Dentelli P, Rosso A, Balsamo A, et al. C-KIT, by interacting with the membrane-bound ligand, recruits endothelial progenitor cells to inflamed endothelium. Blood. 2007;109(10):4264–4271.
    1. Matsui J, Wakabayashi T, Asada M, Yoshimatsu K, Okada M. Stem cell factor/c-kit signaling promotes the survival, migration, and capillary tube formation of human umbilical vein endothelial cells. The Journal of Biological Chemistry. 2004;279(18):18600–18607.
    1. van Huyen JPD, Smadja DM, Bruneval P, et al. Bone marrow-derived mononuclear cell therapy induces distal angiogenesis after local injection in critical leg ischemia. Modern Pathology. 2008;21(7):837–846.
    1. Kelly DJ, Zhang Y, Gow RM, Itescu S, Gilbert RE. Cells expressing the stem cell factor receptor, c-kit, contribute to neoangiogenesis in diabetes. Diabetes and Vascular Disease Research. 2005;2(2):76–80.
    1. Bosch-Marce M, Okuyama H, Wesley JB, et al. Effects of aging and hypoxia-inducible factor-1 activity on angiogenic cell mobilization and recovery of perfusion after limb ischemia. Circulation Research. 2007;101(12):1310–1318.
    1. El-Asrar AMAbu, Struyf S, Opdenakker G, van Damme J, Geboes K. Expression of stem cell factor/c-kit signaling pathway components in diabetic fibrovascular epiretinal membranes. Molecular Vision. 2010;16:1098–1107.
    1. Poulaki V, Joussen AM, Mitsiades N, Mitsiades CS, Iliaki EF, Adamis AP. Insulin-like growth factor-I plays a pathogenetic role in diabetic retinopathy. American Journal of Pathology. 2004;165(2):457–469.
    1. Uthra S, Raman R, Mukesh BN, et al. Diabetic retinopathy and IGF-1 gene polymorphic cytosine-adenine repeats in a Southern Indian cohort. Ophthalmic Research. 2007;39(5):294–299.
    1. Bikfalvi A, Han ZC. Angiogenic factors are hematopoietic growth factors and vice versa. Leukemia. 1994;8(3):523–529.
    1. Krantz SB. Erythropoietin. Blood. 1991;77(3):419–434.
    1. Watanabe D, Suzuma K, Matsui S, et al. Erythropoietin as a retinal angiogenic factor in proliferative diabetic retinopathy. The New England Journal of Medicine. 2005;353(8):782–792.
    1. Takagi H, Watanabe D, Suzuma K, et al. Novel role of erythropoietin in proliferative diabetic retinopathy. Diabetes Research and Clinical Practice. 2007;77(3, supplement):S62–S64.
    1. Becerra SP, Amaral J. Erythropoietin—an endogenous retinal survival factor. The New England Journal of Medicine. 2002;347(24):1968–1970.
    1. Wilkinson-Berka JL, Fletcher EL. Angiotensin and bradykinin: targets for the treatment of vascular and neuro-glial pathology in diabetic retinopathy. Current Pharmaceutical Design. 2004;10(27):3313–3330.
    1. Kim JH, Kim JH, Yu YS, Cho CS, Kim KW. Blockade of angiotensin II attenuates VEGF-mediated blood-retinal barrier breakdown in diabetic retinopathy. Journal of Cerebral Blood Flow and Metabolism. 2009;29(3):621–628.
    1. Yamagishi SI, Nakamura K, Imaizumi T. Advanced glycation end products (AGEs) and diabetic vascular complications. Current Diabetes Reviews. 2005;1(1):93–106.
    1. Hughes JM, Kuiper EJ, Klaassen I, et al. Advanced glycation end products cause increased CCN family and extracellular matrix gene expression in the diabetic rodent retina. Diabetologia. 2007;50(5):1089–1098.
    1. Yamagishi SI, Matsui T, Nakamura K, et al. Olmesartan blocks advanced glycation end products (AGEs)-induced angiogenesis in vitro by suppressing receptor for AGEs (RAGE) expression. Microvascular Research. 2008;75(1):130–134.
    1. Chmielewska K, Robaszkiewicz J, Kosatka M. Role of the retinal pigment epithelium (RPE) in the pathogenesis and treatment of diabetic macular edema (DME) Klinika Oczna. 2008;110(7-9):318–320.
    1. Horiuchi K, Amizuka N, Takeshita S, et al. Identification and characterization of a novel protein, periostin, with restricted expression to periosteum and periodontal ligament and increased expression by transforming growth factor β . Journal of Bone and Mineral Research. 1999;14(7):1239–1249.
    1. Yoshida S, Ishikawa K, Asato R, et al. Increased expression of periostin in vitreous and fibrovascular membranes obtained from patients with proliferative diabetic retinopathy. Investigative Ophthalmology and Visual Science. 2011;52(8):5670–5678.
    1. Ishikawa K, Yoshida S, Kadota K, et al. Gene expression profile of hyperoxic and hypoxic retinas in a mouse model of oxygen-induced retinopathy. Investigative Ophthalmology and Visual Science. 2010;51(8):4307–4319.
    1. Yoshida S, Yoshida A, Ishibashi T, Elner SG, Elner VM. Role of MCP-1 and MIP-1α in retinal neovascularization during postischemic inflammation in a mouse model of retinal neovascularization. Journal of Leukocyte Biology. 2003;73(1):137–144.
    1. Tatemoto K, Hosoya M, Habata Y, et al. Isolation and characterization of a novel endogenous peptide ligand for the human APJ receptor. Biochemical and Biophysical Research Communications. 1998;251(2):471–476.
    1. Masri B, Knibiehler B, Audigier Y. Apelin signalling: a promising pathway from cloning to pharmacology. Cellular Signalling. 2005;17(4):415–426.
    1. Devic E, Rizzoti K, Bodin S, Knibiehler B, Audigier Y. Amino acid sequence and embryonic expression of msr/apj, the mouse homolog of Xenopus X-msr and human APJ. Mechanisms of Development. 1999;84(1-2):199–203.
    1. Kleinz MJ, Davenport AP. Immunocytochemical localization of the endogenous vasoactive peptide apelin to human vascular and endocardial endothelial cells. Regulatory Peptides. 2004;118(3):119–125.
    1. Cox CM, D’Agostino SL, Miller MK, Heimark RL, Krieg PA. Apelin, the ligand for the endothelial G-protein-coupled receptor, APJ, is a potent angiogenic factor required for normal vascular development of the frog embryo. Developmental Biology. 2006;296(1):177–189.
    1. Qian J, Lu Q, Tao Y, Jiang YR. Vitreous and plasma concentrations of apelin and vascular endothelial growth factor after intravitreal bevacizumab in eyes with proliferative diabetic retinopathy. Retina. 2011;31(1):161–168.
    1. Ferrara N, Mass RD, Campa C, Kim R. Targeting VEGF-A to treat cancer and age-related macular degeneration. Annual Review of Medicine. 2007;58:491–504.
    1. Frystyk J, Tarnow L, Krarup Hansen T, Parving HH, Flyvbjerg A. Increased serum adiponectin levels in type 1 diabetic patients with microvascular complications. Diabetologia. 2005;48(9):1911–1918.
    1. Mohan N, Monickaraj F, Balasubramanyam M, Rema M, Mohan V. Imbalanced levels of angiogenic and angiostatic factors in vitreous, plasma and postmortem retinal tissue of patients with proliferative diabetic retinopathy. Journal of Diabetes and Its Complications. 2012;26(5):435–441.
    1. Mao D, Peng H, Li Q, et al. Aqueous humor and plasma adiponectin levels in proliferative diabetic retinopathy patients. Current Eye Research. 2012;37(9):803–808.

Source: PubMed

3
订阅