SDF-1α、VEGF和bevacizumab在增殖性糖尿病视网膜病变继发新生血管性青光眼中的作用及机制研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
新生血管性青光眼(neovascular glaucoma, NVG)是一种导致视力破坏的严重青光眼,它是由新生血管破坏房水外流通道引起,通常继发于眼后段广泛的缺血性疾病,如增殖性糖尿病性视网膜病变(proliferative diabetic retinopathy, PDR)。血管内皮生长因子(vascular endothelial growth factor,VEGF)已被公认在眼内新生血管形成过程中起着重要作用。
     bevacizumab (Avastin)是抗VEGF的单克隆抗体,目前在眼科已越来越广泛地被应用。最新的临床研究表明玻璃体腔内注射bevacizumab(intravitreal injection of bevacizumab, IVB)在治疗眼内新生血管性疾病包括新生血管性青光眼方面取得了显著成果。虽然IVB可以显著抑制眼内新生血管的生成,但是临床观察发现部分患者即使使用了IVB,新生血管仍然在增多,病情仍会继续进展。
     基质细胞衍生因子- 1α(stromal cell derived factor -1α, SDF-1α),α组趋化因子家族的一个新成员,最早从骨髓的基质细胞中分离出来,其特异性受体CXCR4广泛地表达在许多组织和器官上,SDF-1/CXCR4在调节免疫和炎症反应、调控造血、恶性肿瘤细胞的浸润转移、血管生成等方面发挥着重要的作用。
     为了探讨SDF-1α、VEGF和bevacizumab在增殖性糖尿病视网膜病变继发新生血管性青光眼中的作用及机制,本研究首先在体外实验中,通过在体外血管生成检测和血管内皮细胞增殖检测中加入SDF-1α,观察SDF-1α在其中的作用;体内实验中,检测患者玻璃体中SDF-1α水平和患者玻璃体对血管内皮细胞增殖的作用。结果表明SDF-1α在新生血管形成过程中起到了促进作用,并参与了PDR继发NVG患者病理性新生血管形成过程。接着对PDR患者玻璃体中bevacizumab和VEGF的含量进行了检测和分析,以明确bevacizumab在人眼内的药代动力学情况并为其在临床的应用剂量和使用频率达到最佳化提供了理论依据。然后对不同剂量bevacizumab玻璃体腔注射治疗眼内缺血性疾病继发虹膜新生血管及新生血管性青光眼疗效进行了分析,为bevacizumab的玻璃体腔最佳给药剂量提供依据。进一步,在体外实验中,通过在毛细血管样结构形成和血管内皮细胞增殖检测中分别或同时加入VEGF、bevacizumab和SDF-1α,来观察bevacizumab和SDF-1α之间的关系,同时检测患者IVB前后玻璃体中SDF-1α水平和患者玻璃体对血管内皮细胞增殖的作用。结果显示bevacizumab对SDF-1α促进血管新生作用没有明显抑制,患者IVB后玻璃体中SDF-1α水平高,而且能明显促进血管内皮细胞增殖,表明SDF-1α在患者IVB后新生血管形成过程中起着重要作用。最后在体外实验毛细血管样结构形成和血管内皮细胞增殖检测中同时加入SDF-1α及其抗体,观察抗SDF-1α抗体的作用,结果表明抗SDF-1α抗体对SDF-1α诱导的新生血管形成有抑制作用,这些研究结果提示了抗SDF-1α抗体在治疗缺血性视网膜疾病及IVB治疗后病情进展和复发病例中的可行性。
Neovascular glaucoma (NVG) is a severe form of glaucoma with devastating visual outcome attributed to new blood vessels obstructing aqueous humor outflow, usually secondary to widespread posterior segment ischemia such as proliferative diabetic retinopathy (PDR), central retinal vein occlusion (CRVO) and ocular ischemic syndrome (OIS) et al. Angiogenesis is a physiologic process that involves endothelial cell proliferation, migration, and maturation, and sometimes plays an important role in ocular diseases such as PDR, CRVO, and NVG. Several angiogenic growth factors have been reported to contribute to pathologic angiogenesis. Of these, vascular endothelial growth factor (VEGF) has been shown to be a key molecule in promoting ocular neovascularization.
     Bevacizumab (Avastin) is a full-length humanized anti-VEGF monoclonal antibody that is approved for use as an antiangiogenetic agent for metastatic colorectal cancer. It has been increasingly used as an off-label drug in ophthalmology. Recent clinical studies have shown that intravitreal injections of bevacizumab (IVB) have had excellent results in the treatment of angiogenetic pathologies, including neovascular glaucoma.
     Although IVB may provide great benefits to halt neovascular activities in a variety of intraocular neovascular pathologies, advancement of neovascular activities may continue even after IVB. A recent study reported that IVB could not prevent re-bleeding in eyes undergoing IVB followed by pars plana vitrectomy to treat diabetic vitreous hemorrhage. We presume that there are other cytokines/chemokines that may have effects to ocular angiogenesis including stromal cell–derived factor–1α(SDF-1α).
     SDF-1α, a member of the CXC chemokine family, is a potent chemoattractant for mature mononuclear leukocytes, as well as hematopoietic stem and progenitor cells, and mediates its effects through the chemokine receptor CXCR4. SDF-1 has been reported to enhance adult angiogenesis induced by VEGF, by recruiting haemopoietic stem cells (HSCs). It has been shown to be both necessary and sufficient to promote proliferative retinopathy, and to be present at high concentrations in the vitreous of patients with PDR, retinal vein occlusion (ROV) and retinopathy of prematurity (ROP).
     To evaluate the role and mechanism of SDF-1αand VEGF and bevacizumab in NVG secondary to PDR, firstly we studied the role and correlation of SDF-1αand VEGF in NVG secondary to PDR. The role of SDF-1αand VEGF in the migration, proliferation and differentiation of human umbilical vascular endothelial cells (HUVECs) was evaluated by tube-like and capillary-like formation assay and 5′-bromo-2′-deoxyuridine (BrdU) incorporation in vitro. The vitreous samples from patients with NVG secondary to PDR were measured for concentrations of SDF-1αand VEGF by enzyme-linked immunosorbent assay. The correlation of the vitreous levels of SDF-1αand VEGF was analyzed. Secondly, the vitreous levels of bevacizumab and VEGF in patients with NVG secondary to PDR were measured by ELISA, to optimize reinjection intervals to avoid overdosing and underdosing. Bevacizumab enzyme-linked immunosorbent assay standard curve was obtained. Serial dilutions of bevacizumab were measured to calibrate the assay. The vitreous concentrations of VEGF and bevacizumab were assayed by ELISA.The correlation of the vitreous concentrations of VEGF and bevacizumab and the time after IVB and intraocular pharmacokinetics of bevacizumab were analyzed. Thirdly, the effects of different doses of IVB on ischemic retinal diseases were evaluated to optimize intravitreal injection dose. The concentration of VEGF in aqueous humor before IVB was measured by ELISA. The correlation of the concentration of VEGF in aqueous humor and intraocular pressure (IOP) before IVB were analyzed. The concentration of VEGF in aqueous humor and IOP was compared before and after the different dose of IVB. Finally, the role of SDF-1αafter IVB in patients with NVG secondary to PDR was evaluated. The role and correlation of SDF-1α, anti- SDF-1αantibody and bevacizumab in the migration, proliferation and differentiation of HUVECs was evaluated by capillary-like formation assay and BrdU incorporation in vitro. The effects of the vitreous samples on HUVECs proliferation were assayed by BrdU incorporation. Vitreous concentrations of VEGF and SDF-1αwere compared between the patients with NVG and without NVG (control group) and among bevacizumab 1mg group, bevacizumab 0.1mg group and no IVB group.
     In the current study, the results showed that HUVEC tube-like and capillary-like formation was significantly enhanced by SDF-1αand VEGF (P﹤0.05) . All of SDF 10ng/ml, SDF 100ng/ml and SDF 1000ng/ml group significantly promoted HUVECs proliferation (P<0.05); SDF 100ng/ml group was strongest and SDF 100ng/ml group was weakest in SDF 10ng/ml, SDF 100ng/ml and SDF 1000ng/ml group. The OD value of VEGF 100ng/ml + SDF 100ng/ml group was significantly higer than VEGF 100ng/ml group (P<0.05). In vitreous sample measurements, both VEGF and SDF-1αlevels in patients with NVG were significantly higher than those in patients without NVG (P<0.01). No significant correlation between VEGF and SDF-1αin vitreous was noted (r=0.41, P>0.05). The concentration of bevacizumab (the range 0-100ng/ml) exhibited a positive correlation with OD value by ELISA. The vitreous concentration of VEGF in patients with PDR showed a negative correlation with the bevacizumab concentration (R=0.96, P<0.001) and a positive correlation with the time after IVB (R=0.96, P<0.001). The vitreous concentration of bevacizumab showed a negative correlation with the time after IVB (R=0.98, P<0.001). The peak concentration (127.1ug/ml) of bevacizumab was observed on the first day after IVB. Estimated half-time (t1/2) of bevacizumab elimination from vitreous was 10.2 days. The average value of VEGF concentration in aqueous humor before IVB (control group 150.1pg/ml; iris neovascularization, INV group 1058.3pg/ml; open-angle neovascular glaucoma, O-NVG group 5856.2pg/ml; closed-angle neovascular glaucoma, C-NVG group 19153.2pg/ml) was obtained. The VEGF concentration of C-NVG group was significantly higher than control, INV and O-NVG group. Intraocular pressure (IOP) showed a positive correlation with the concentration of VEGF in aqueous humor. The average value of VEGF concentration in aqueous humor before and after different doses of IVB (1.0mg group before IVB, 14978.3pg/ml; 0.1mg group before IVB, 5718.2pg/ml; 1.0mg group after IVB, 28.9 pg/ml; 0.1mg group after IVB, 22.2pg/ml) was obtained. There was no difference between 1.0mg and 0.1mg group in the variation of before and after IVB (P=0.140). The VEGF concentration of 1.0mg and 0.1mg groups after IVB in aqueous humor was both lower than control group (150pg/ml). The average value of IOP before and after different doses of IVB (1.0mg group before IVB, 35mmHg; 0.1mg group before IVB, 34mmHg; 1.0mg group after IVB, 23mmHg; 0.1mg group after IVB, 24mmHg) was obtained. There was no difference between IOP of 1.0mg and 0.1mg groups before and after IVB (P=0.892). The enhancement of recombinant human VEGF (10 ng/ml) in HUVECs capillary-like formation was inhibited by treatment with bevacizumab (10 ug/ml). However, the enhancement of recombinant human SDF-1αwas not inhibited by treatment with bevacizumab (10ug/ml). SDF-1αpromoted HUVECs capillary-like formation with bevacizumab (P<0.05). However, the enhancement of recombinant human SDF-1αwas inhibited by treatment with anti- SDF-1αantibody in HUVECs capillary-like formation and proliferation (P<0.05). There was no difference between the treatments with VEGF 100ng/ml and bevacizumab 100ug/ml and the control group in HUVECs proliferation (P>0.05).The treatment with VEGF and bevacizumab and SDF-1αpromoted HUVECs proliferation (P<0.05). The promotion of the treatment with VEGF and bevacizumab and SDF-1αwas inhibited by anti- SDF-1αantibody (P<0.05). Both the vitreous samples before and after IVB in NVG significantly promoted HUVECs proliferation (P<0.01). The promotion of vitreous samples after IVB was weaker than those before IVB (P<0.05). Both VEGF and SDF-1αlevels in patients with NVG were significantly higher than those in patients without NVG (control group) (P<0.05). The vitreous level of VEGF significantly decreased after IVB (P<0.05). The vitreous level of SDF-1αin patients with NVG was higher than control group after IVB (P<0.05), although the vitreous level of SDF-1αdecreased.
     In conclusion, the present study demonstrates that both SDF-1αand VEGF may enhance angiogenesis by the promotion of migration and proliferation of HUVECs, and participate in promoting neovascularization in patients with NVG secondary to PDR. We obtain the standard curve of bevacizumab enzyme-linked immunosorbent assay. The vitreous half-life of 1.0 mg intravitreally injected bevacizumab in patients with PDR is 10.2 days. The vitreous concentration of bevacizumab in patients with PDR shows a negative correlation with the time after IVB. The vitreous concentration of VEGF in patients with PDR shows a negative correlation with the bevacizumab concentration and a positive correlation with the time after IVB. The intravitreally injected bevacizumab may be decreased to 0.1mg from 1.0 mg as preoperative adjunctive therapy with invariable therapeutic effect. SDF-1αmay participate in promoting neovascularization in patients with NVG secondary to PDR and sufficiently to facilitate the advancement of neovascular activities even after IVB.
引文
[1] Jones A, Harris AL. New developments in angiogenesis: a major mechanism for tumor growth and target for therapy. Cancer J Sci Am 1998;4:209–17.
    [2] Salcedo R, Wasserman K, Young HA, et al. Vascular endothelial growth factor and basic fibroblast growth factor induce expression of CXCR4 on human endothelial cells: in vivo neovascularization induced by stromal-derived factor-1alpha. Am J Pathol 1999; 154:1125–35.
    [3] Hanahan D, Folkman J. Patterns and emerging mechanisms of the angiogenic switch during tumorigenesis. Cell 1996;86:353–64.
    [4] Shima DT, Adamis AP, Ferrara N, et al. Hypoxic induction of endothelial cell growth factors in retinal cells: identification and characterization of vascular endothelial growth factor (VEGF) as the mitogen. Mol Med 1995;1:182–93.
    [5] Adamis AP, Miller JW, Bernal MT, et al. Increased vascular endothelial growth factor levels in the vitreous of eyes with proliferative diabetic retinopathy. Am J Ophthalmol 1994;118:445–50.
    [6] Wakabayashi T, Oshima Y, Sakaguchi H, et al. Intravitreal bevacizumab to treat iris neovascularization and neovascular glaucoma secondary to ischemic retinal diseases in 41 consecutive cases. Ophthalmology 2008;115:1571–1580.
    [7] Oshima Y, Shima C, Wakabayashi T, et al. Microincision vitrectomy surgery and intravitreal bevacizumab as a surgical adjunct to treat diabetic traction retinal detachment. Ophthalmology.2009 May;116(5):927-38.
    [8] Fang X, Sakaguchi H, Gomi F, et al. Efficacy and safety of one intravitreal injection of bevacizumab in diabetic macular oedema. Acta Ophthalmol. 2008 Nov;86(7):800-5.
    [9] Gomi F, Nishida K, Oshima Y, et al. Intravitreal bevacizumab for idiopathic choroidalneovascularization after previous injection with posterior subtenon triamcinolone.Am J Ophthalmol.2007 Mar;143(3):507-10.
    [10] Moschos MM, Brouzas D, Apostolopoulos M, et al. Intravitreal use of bevacizumab (Avastin) for choroidal neovascularization due to ARMD: A preliminary multifocal-ERG and OCT study: Ultifocal-ERG after use of bevacizumab in ARMD. Doc Ophthalmol. 2007;114:37–44.
    [11] Tewari A, Dhalla MS, Apte RS. Intravitreal bevacizumab for treatment of choroidal neovascularization in pathologic myopia. Retina. 2006;26:1093–4.
    [12] Haritoglou C, Kook D, Neubauer A, et al. Intravitreal bevacizumab (Avastin) therapy for persistent diffuse diabetic macular edema. Retina. 2006;26:999–1005.
    [13] Yoganathan P, Deramo VA, Lai JC, et al. Visual improvement following intravitreal bevacizumab (Avastin) in exudative age-related macular degeneration. Retina. 2006; 26:994–8.
    [14] Jonas JB, Harder B, Spandau UH, et al. Bevacizumab for occult subfoveal neovas- cularization in age-related macular degeneration. Eur J Ophthalmol. 2006;16:774–5.
    [15] Rosenfeld PJ, Fung AE, Puliafito CA. Optical coherence tomography findings after an intravitreal injection of bevacizumab (Avastin?) for macular edema from central retinal vein occlusion. Ophthalmic Surg Lasers Imaging 2005;36:336–339.
    [16] Michels S, Rosenfeld PJ, Puliafito CA, et al. Systemic bevacizumab (Avastin) therapy for neovascular age-related macular degeneration: twelve-week results of an uncontrolled open-label clinical study. Ophthalmology 2005;112:1035–1047.
    [17] Davidorf FH, Mouser JG, Derick RJ. Rapid improvement of rubeosis iridis from a single bevacizumab (Avastin) injection. Retina 2006;26:354–356.
    [18] Oshima Y, Sakaguchi H, Gomi F, et al. Regression of iris neovascularization afterintravitreal injection of bevacizumab in patients with proliferative diabetic retinopathy. Am J Ophthalmol 2006;142: 155–158.
    [19] Nagasawa T, Hirota S, Tachibana K, et al. 1996. Defects of B-cell lymphopoiesis and bone marrow myelopoiesis in mice lacking the CXC chemokine PBSF/SDF-1. Nature 382: 635–638.
    [20] Ma Q, Jones D, Borghesani PR, et al. 1998. Impaired B-lymphopoiesis, myelopoiesis, and derailed cerebellar neuron migration in CXCR4 and SDF-1–deficient mice. Proc. Natl. Acad. Sci. USA 95: 9448–9453.
    [21] Tachibana K, Hirota S, Iizasa H, et al. 1998. The chemokine receptor CXCR4 is essential for vascularization of the gastrointestinal tract. Nature 393: 591–594.
    [22] Zou YR, Kottmann AH, Kuroda M, et al. 1998. Function of the chemokine receptor CXCR4 in haematopoiesis and cerebellar development. Nature 393: 595–599.
    [23] Tashiro K, Tada H, Heilker R, et al. Signal sequence trap: a cloning strategy for secreted proteins and type I membrane proteins. Science 1993; 261(5121): 600-3.
    [24] Nagasawa T, Kikutani H, Kishimoto T. Molecular cloning and structure of a pre-B-cell growth-stimulating factor. Proc Natl Acad Sci USA 1994; 91(6): 2305-9.
    [25] Shirozu M, Nakano T, Inazawa J, et al. Structure and chromosomal localization of the human stromal cell-derived factor 1 (SDF1) gene. Genomics 1995; 28(3): 495-500.
    [26] Gleichmann M,Gillen C, Czardybon M, et al. Cloning and characterization of SDF-1 gamma, a novel SDF-1 chemokine transcript with developmentally regulated expression in the nervous system[J]. Eur J Neuro Sci , 2000, 12 (6): 1857-1866.
    [27] Tashiro K, Tada H, Heilker R, et al. Signal sequence trap: a cloning strategy for secreted proteins and type I membrane proteins. Science 1993; 261(5121): 600-3.
    [28] Nagasawa T, Kikutani H, Kishimoto T. Molecular cloning and structure of a pre-B-cellgrowth-stimulating factor. Proc Natl Acad Sci USA 1994; 91(6): 2305-9.
    [29] Shirozu M, Nakano T, Inazawa J, et al. Structure and chromosomal localization of the human stromal cell-derived factor 1 (SDF1) gene. Genomics 1995; 28(3): 495-500.
    [30] Nagasawa T, Hirota S, Tachibana K, et al. Defects of B-cell lymphopoiesis and bonemarrow myelopoiesis in mice lacking the CXC chemokine PBSF/SDF-1. Nature 1996; 382(6592): 635-8.
    [31] Bourcier T, Berbar T, Paquet S, et al. Characterization and functionality of CXCR4 chemokine receptor and SDF-1 in human corneal fibroblasts. MolVis2003:9(1):96-102.
    [32] Crane IJ, Wallace CA, McKillop-Smith S, et al. CXCR4 receptor expression on human retinal pigment epithelial cells fromthe blood-retina barrier leads to chemokinesecretion andmigration in response to stromal cell-derived factor1 alpha. J Immuno l2000: 165 (8):4372-4378.
    [33] Shirozu M, Nakano T, Inazawa J, et al. Structure and chromosomal localization of the human stromal cell-derived factor 1 (SDF1) gene. Genomics 1995; 28(3): 495-500.
    [34] Bleul CC, Fuhlbrigge RC, Casasnovas JM, et al. A highly efficacious lymphocyte chemoattractant, stromal cellderived factor 1 (SDF-1). J Exp Med 1996; 184(3): 1101-9.
    [35] Loetscher M, Geiser T, O’Reilly T, et al. Cloning of a human seven-transmembrane domain receptor, LESTR, that is highly expressed in leukocytes [J]. J Biol Chem, 1994, 269(1):232-237.
    [36] Berson JF, Long D, Doranz BJ, et al. A seven-transmembrane domain receptor involved in fusion and entry of T-cell-tropic human immunodeficiency virus type 1 strains. J Virol 1996; 70(9): 6288-95.
    [37] Horuk R. Chemokine receptors. Cytokine Growth Factor Rev 2001;12(4): 313-335.
    [38] Bleul CC, Wu L, Hoxie JA, et al. The HIV coreceptors CXCR4 and CCR5 aredifferentially expressed and regulated on human T lymphocytes. Proc Natl Acad Sci USA 1997; 94(5): 1925-1930.
    [39] Gupta SK, Lysko PG, Pillarisetti K, et al. Chemokine receptors in human endothelial cells. Functional expression of CXCR4 and its transcriptional regulation by inflammatory cytokines. J Biol Chem 1998; 273(7): 4282-4287.
    [40] Hesselgesser J, Halks-Miller M, DelVecchio V, et al. CD4-independent association between HIV-1 gp120 and CXCR4: functional chemokine receptors are expressed in human neurons. Curr Biol 1997; 7(2): 112-121.
    [41] Bajetto A, Bonavia R, Barbero S, et al. Glial and neuronal cells express functional chemokine receptor CXCR4 and its natural ligand stromal cell-derived factor 1. J Neurochem 1999; 73(6): 2348-2357.
    [42] Ohtani Y, Minami M, Kawaguchi N, et al. Expression of stromal cell-derived factor-1 and CXCR4 chemokine receptor mRNAs in cultured rat glial and neuronal cells. Neurosci Lett 1998; 249(2-3): 163-166.
    [43] Nagasawa T, Nakajima T, Tachibana K, et al. Molecular cloning and characterization of a murine pre-B-cell growth-stimulating factor/stromal cell-derived factor 1 receptor, a murine homolog of the human immunodeficiency virus 1 entry coreceptor fusin. Proc Natl Acad Sci U S A. 1996 Dec 10;93(25):14726-9.
    [44] Kollet O, Petit I, Kahn J, et al. Human CD34+ CXCR4+, which can be functionally expressed and provide NOD/SCID repopulation [J]. Blood, 2002,100(8): 2778-2786.
    [45] Ma Q, Jones D, Borghesani PR, et al. Impaired B-lymphopoiesis, myelopoiesis, and derailed cerebellar neuron migration in CXCR4 and SDF-1-deficient mice. Proc Natl Acad Sci U S A. 1998 Aug 4;95(16):9448-53.
    [46] M?hle R, Bautz F, Rafii S, et al. The chemokine receptor CXCR-4 is expressed on CD34+hematopoietic progenitors and leukemic cells and mediates transendothelial migration induced by stromal cell-derived factor-1. Blood. 1998 Jun 15;91(12):4523-4530.
    [47] Zhang XF, Wang JF, Matczak E, et al. Janus kinase 2 is involved in stromal cell-derived factor-1alpha-induced tyrosine phosphorylation of focal adhesion proteins and migration of hematopoietic progenitor cells. Blood. 2001 Jun 1;97(11):3342-3348.
    [48] Hatton K,Heissig B,Tashiro K,et al. Plasma elevation of stromal cell-derived factor-1 induces mobilization of mature and immature hematopoietic progenitor cells[J]. Blood, 2001, 97(11):3354-3360.
    [49] Peled A, Kollet O, Ponomaryov T, et al. The chemokine SDF-1 activates the integrins LFA-1, VLA-4, and VLA-5 on immature human CD34(+) cells: role in transendothelial/ stromal migration and engraftment of NOD/SCID mice. Blood. 2000 Jun 1;95 (11): 3289-96.
    [50] Lee Y, Gotoh A, Kwon HJ, et al. Enhancement of intracellular signaling associated with hematopoietic progenitor cell survival in response to SDF-1/CXCL12 in synerge with other cytokines[J]. Blood, 2001, 99(12):4307-4311.
    [51] Kantele JM,Kurk S,Jutila MA,et al. Efffects of continuous exposure of stromal cell-derived factor-lαon T cells [J]. J Immunol, 2000, 164(10):5010-5014.
    [52] Nanki T,Lipsky PE. Cutting Edge: Stromal cell-derived factor-1 is a co-stimulator for CD 4+ T cell activation [J].J Immunol,2000, 164(10):5010-5014.
    [53] NankiT, Hayashida K, El-GabalawyHS, et al. Stromal cell-derived factor-1 CXC chemokine receptor4 interactionsplay a central role inCD4+ T cell accumulation in rheumatoid arthritis synovium. J Immunol2000: 165 (11):6590-6598.
    [54] TakanoY, Shinokado K, Hata Y, et al. HIV envelope protein gp120-triggered CD4+ T-cell adhesion to vascular endothelium is regulated via CD4 and CXCR4 receptors. BiochinBiophys Acta2007;1772(5):549-555.
    [55] Mueller A,Homey B,Soto H, et al.Involvement of chemokine receptors in breast cancer metastasis[J].Nature,2001,410(6824):50-56.
    [56] Sanz Rodriguez F,Hidalgo A,Teixido J.Chemokine stromal cell-derived factor-l alpha modulates VLA-4 integrin-mediated multiple myeloma cell adhesion to CS-1/fibronection and VCAM-1[J]. Blood, 2001, 97(2):346-351.
    [57] Nishimura H, Asahara T. Bone marrow-derived endothelial progenitor cells for neovascular formation. EXS, 2005, (94):147-154.
    [58] Urbich C, Dimmeler S. Endothelial progenitor cells: characterization and role in vascular biology. Circ Res, 2004, 95:343-353.
    [59] Asahara T, Kawamoto A. Endothelial progenitor cells for postnatal vasculogenesis. Am J Physiol Cell Physiol, 2004, 287:C572-579.
    [60] Ruiz de Almodovar C, Luttun A, Carmeliet P. An SDF-1 trap for myeloid cells stimulates angiogenesis.Cell,2006; 124(1):18-21.
    [61] Carr AN, Howard BW, Yang HT, et al. Efficacy of systemic administration of SDF-1 in a model of vascular insufficiency: support for an endothelium-dependent mechanism. Cardiovasc Res,2006;69(4):925-935.
    [62] Tomita M, Yamada H, Adachi Y, et al. Choroidal neovascularization is provided by bone marrow cells [J]. Stem Cells2004;22(1):21-26.
    [63] De Falco E, Porcelli D, Torella AR, et al. SDF-1 involvement in endothelial phenotype and ischemia-induced recruitment of bone marrow progenitor cells[J ].Blood. 2004, 104(12): 3472-3482.
    [64] Togel F, Isaac J, Hu Z, et al. Renal SDF-1 signalsmobilization and homing of CXCR4- positive cells to the kidney after ischemic injury [J]. Kidney Int. 2005, 67(5): 1772-84.
    [65] Yamaguchi J, Kusano KF, Masuo O, et al. Stromal cell-derived factor-1 effects on ex vivo expanded endothelial progenitor cells recruitment for ischemic neovascularization [ J ]. Circulation. 2003, 107(9): 1322-1328.
    [66] Salvucci O, Yao L, Villalba S, et al. Regulation of endothelial cell branching morphogenesis by endogenous chemokine stromal-derived factor-1. Blood. 2002 Apr 15;99(8):2703-2711.
    [67] Netelenbos T, Zuijderduijn S, Van Den Born J, et al. Proteoglycans guide SDF-1-induced migration of hematopoietic progenitor cells. J Leukoc Biol. 2002 Aug;72(2):353-362.
    [68] Kelly DJ, Zhang Y, Gow RM, et al. Cells expressing the stem cell factor receptor, c-kit, contribute to neoangiogenesis in diabetes. Diab Vasc Dis Res, 2005, 2:76-80.
    [69] Nagasawa T, Hirota S, Tachibana K, et al. Defects of B-cell lymphopoiesis and bone- marrow myelopoiesis in mice lacking the CXC chemokine PBSF/SDF-1. Nature. 1996 Aug 15;382(6592):635-8.
    [70] Akimoto M, Hashimoto H, Maeda A, et al. Roles of angiogenic factors and endothelin-1 in gastric ulcer healing. Clin Sci (Lond). 2002 Aug;103 Suppl 48:450-454.
    [71] Kanda S, Mochizuki Y, Kanetake H. Stromal cell-derived factor-1alpha induces tube-like structure formation of endothelial cells through phosphoinositide 3-kinase. J Biol Chem, 2003 Jan 3;278(1):257-262.
    [72] Hitchon C, Wong K, Ma G. Hypoxia-induced production of stromal cell-derived factor 1(CXCL12) and vascular endothelial growth factor by synovial fibroblasts. Arthritis Rheum, 2002, 46: 2587-2597.
    [73] Salcedo R, Wasserman K, Young HA, et al. Vascular endothelial growth factor and basic fibroblast growth factor induce expression of CXCR4 on human endothelial cells: in vivo neovascularization induced by stromal-derived factor-1alpha. Am J Pathol, 1999, 154:1125-1135.
    [74] Kijowski J, Baj-Krzyworzeka M, Majka M, et al. The SDF-1-CXCR4 axis stimulates VEGF secretion and activates integrins but does not affect proliferation and survival in lymphohematopoietic cells. Stem Cells, 2001,19:453-466.
    [75] Brooks HL, Caballero S, Newell CK, et al. Vitreous levels of vascular endothelial growth factor and stromal-derived factor-1 in patients with diabetic retinopathy and cystoid macular edema before and after intraocular injection of triamcinolone. Arch Ophthalmol, 2004,112:1801-1807.
    [76] Sengupta N, Caballero S, Mames RN, et al. Preventing stem cell incorporation into choroidal neovascularization by targeting homing and attachment factors. Invest OphthalmolVis Sci2005;46(1):343-348.
    [77] Grant MB, May WS, Caballero S, et al. Adult hematopoietic stem cells provide functional hemangiohlast activity during re-tinal neovascularization. NatMed2002;8:607-612.
    [78] Li Y, Reca RG, Atmaca-Sonmez P, et al. Retinal pigment epithelium damage enhances expression of chemoattractants andmigration of bone marrow-derived stem cells. Invest Ophthalmol Vis Sci 2006;47(4):1646-1652.
    [79] Butler JM, Guthrie SM, KocM, et al. SDF-1 is both necessary and sufficient to promote proliferative retinopathy. J Clin Invest 2005; 115(1):86-93.
    [80] Lima e Silva R, Shen J, Hackett SF, et al. The SDF-1/CXCR4 ligand/receptorpair is an important contributor to several types of ocular neovascularization. FASEB J. 2007 Oct; 21(12):3219-3230.
    [81] Otani A, Kinder K, Ewalt K, et al. Bone marrow-derived stem cells target retinal astrocytes and can promote or inhibit retinal angiogenesis. Nat Med, 2002, 8:1004-1010.
    [82] Romano MR, Gibran SK, Marticorena J, et al. Can an intraoperative bevacizumabinjection prevent recurrent postvitrectomy diabetic vitreous hemorrhage?Eur J Ophthalmol. 2009 Jul-Aug;19(4):618-21.
    [83] Sivack-Callcott JA, O’Day DM, Gass DM, et al. Evidence-based recommendations for the diagnosis and treatment of neovascular glaucoma. Ophthalmology 2001;108:1767–76.
    [84] Akivama Y, Yamasaki T, Kagawa T, et al. Empty sella syndrome. Nippon Rinsho, 1993; 51(10):2731-2736.
    [85] Aiello LP, Brucker AJ, Chang S, et al. Evolving guidelines for intravitreous injections. Retina 2004;24(suppl):S3–19.
    [86] Schor AM, Schor SL. Tumour angiogenesis.J Pathol. 1983 Nov;141(3):385-413.
    [87] Asahara T, Murohara T, Sullivan A, et al. Isolation of putative progenitor endothelial cells for angiogenesis[J]. Science , 1997 , 275(14) :964 -967.
    [88] Lin Y, Weisdorf DJ, Solovey A, et al. Origins of circulating endothelial cells and endothelial outgrowth from blood [J]. J Clin Invest2000;105(1):71-77.
    [89] Askari AT, Unzek S, Popovic ZB, et al. Effect of stromal cell-derived factor-1 on stem cell homing and tissue regeneration in ischemic cardiomyopathy[J ].Lancet. 2003, 362(9385): 697-703.
    [90] Schober A, Knarren S, Lietz M, et al. Crucial role of stromal cell-derived factor-1alpha in neointima formation after vascular injury in apolipoprotein E-deficientmice[J ].Circulation. 2003, 108(20):2491-2497.
    [91] Yamaguchi J, Kusano KF, Masuo O, et al. Stromal cell-derived factor-1 effects on ex vivo expanded endothelial progenitor cells recruitment for ischemic neovascularization [ J ]. Circulation. 2003, 107(9): 1322-1328.
    [92] Lataillade JJ, Clay D, Dupuy C, et al. Chemokine SDF-1 enhances circulating CD34(+) cell proliferation in synergy with cytokines: possible role in progenitor survival. Blood.2000 Feb 1;95(3):756-68.
    [93] TogelF, Isaac J, Hu Z, et al. Renal SDF-1 signals mobilization and homing of CXCR4- positive cells to the kidney after ischemic injury [J]. Kidney Int. 2005, 67(5): 1772-84.
    [94] De Falco E, Porcelli D, Torella AR, et al. SDF-1 involvement in endothelial phenotype and ischemia-induced recruitment of bone marrow progenitor cells [J]. Blood. 2004, 104(12): 3472-3482.
    [95]王承艳,苗振川,丰美福.基质细胞衍生因子SDF及其受体CXCR4在造血干/祖细胞动员及归巢过程中的作用[J ].中国实验血液学杂志, 2004, 12 (1) : 115-119.
    [96] Hiasa K, IshibashiM, Ohtani K, et al. Gene transfer of stromal cell-derived factor-1alpha enhances ischemic vasculogenesis and angiogenesis via vascular endothelial growth factor/endothelial nitric oxide synthase-related pathway: next-generation chemokine therapy for therapeutic neovascularization [J]. Circulation. 2004, 109(20):2454-2461.
    [97] Michels S, Rosenfeld PJ, Puliafito CA, et al. Systemic bevacizumab (Avastin) therapy for neovascular age-related macular degeneration twelve-week results of an uncontrolled open-label clinical study. Ophthalmology. 2005 Jun;112(6):1035-47.
    [98] Rosenfeld PJ, Moshfeghi AA,Puliafito CA,et al. Optical coherence tomography findings after an intravitreal injection of bevacizumab (Avastin) for neovascular age-related macular degeneration. Ophthalmic Surg Lasers Imaging. 2005 Jul-Aug;36(4):331-335.
    [99] Krohne TU, Eter N, Holz FG, et al. Intraocular pharmacokinetics of bevacizumab after a single intravitreal injection in humans. Am J Ophthalmol. 2008 Oct;146(4):508-12.
    [100]Bakri SJ, Snyder MR, Reid JM, et al. Pharmacokinetics of intravitreal bevacizumab (Avastin). Ophthalmology. 2007;114:855–859.
    [101]Wang Y, Fei D, Vanderlaan M, et al. Biological activity of bevacizumab, a humanized anti-VEGF antibody in vitro. Angiogenesis. 2004;7:335– 45.
    [102]Fernández-Vigo J, Castro J, Macarro A. Diabetic iris neovascularization. Natural history and treatment. Acta Ophthalmol Scand. 1997 Feb;75(1):89-93.
    [103]Bartz-Schmidt KU, Thumann G, Psichias A, et al. Pars plana vitrectomy, endolaser coagulation of the retina and the ciliary body combined with silicone oil endotamponade in the treatment of uncontrolled neovascular glaucoma. Graefes Arch Clin Exp Ophthalmol. 1999 Dec;237(12):969-75.
    [104]Aiello LP, Avery RL, Arrigg PG, et al. Vascular endothelial growth factor in ocular fluid of patients with diabetic retinopathy and other retinal disorders. N Engl J Med. 1994 Dec 1;331(22):1480-1487.
    [105]Tripathi RC, Li J, Tripathi BJ, et al. Increased level of vascular endothelial growth factor in aqueous humor of patients with neovascular glaucoma. Ophthalmology. 1998 Feb; 105(2):232-7.
    [106]Sivack-Callcott JA, O’Day DM, Gass DM, et al. Evidence-based recommendations for the diagnosis and treatment of neovascular glaucoma. Ophthalmology 2001;108:1767–76.
    [107]Aiello LP, Avery RL, Arrigg PG, et al. Vascular endothelial growth factor in ocular fluid of patients with diabetic retinopathy and other retinal disorders. N Engl J Med 1994;331: 1480–7.
    [108]Tripathi RC, Li J, Tripathi BJ, et al. Increased level of vascular endothelial growth factor in aqueous humor of patients with neovascular glaucoma. Ophthalmology 1998; 105: 232–7.
    [109]Wand M, Dueker DK, Aiello LM, et al. Effects of panretinal photocoagulation on rubeosis iridis, angle neovascularization, and neovascular glaucoma. Am J Ophthalmol 1978;86:332–9.
    [110]Murphy RP, Egbert PR. Regression of iris neovascularization following panretinalphotocoagulation. Arch Ophthalmol. 1979;97:700–2.
    [111]Tasman W, Magargal LE, Augsburger JJ. Effects of argon laser photocoagulation on rubeosis iridis and angle neovascularization. Ophthalmology 1980;87:400–2.
    [112]Laatikainen L. A prospective follow-up study of panretinal photocoagulation in preventing neovascular glaucoma following ischaemic central retinal vein occlusion. Graefes Arch Clin Exp Ophthalmol 1983;220:236–9.
    [113]Pavan PR, Folk JC, Weingeist TA, et al. Diabetic rubeosis and panretinal photocoagulation. Arch Ophthalmol 1983;101:882–4.
    [114]Tripathi RC, Li J, Tripathi BJ, et al. Increased level of vascular endothelial growth factor in aqueous humor of patients with neovascular glaucoma. Ophthalmology 1998; 105: 232–7.
    [115]Wakabayashi T, Oshima Y, Sakaguchi H, et al. Intravitreal bevacizumab to treat iris neovascularization and neovascular glaucoma secondary to ischemic retinal diseases in 41 consecutive cases. Ophthalmology 2008;115:1571–1580.
    [116]Davidorf FH, Mouser JG, Derick RJ. Rapid improvement of rubeosis iridis from a single bevacizumab (Avastin) injection. Retina. 2006 Mar;26(3):354-356.
    [117]Bakri SJ, Snyder MR, Reid JM, et al. Pharmacokinetics of intravitreal bevacizumab (Avastin). Ophthalmology. 2007 May; 114(5):855-9.
    [118]Shima C, Sakaguchi H, Gomi F, et al. Complications in patients after intravitreal injection of bevacizumab. Acta Ophthalmol. 2008 Jun;86(4):372-376.
    [119]Nagasawa T, Hirota S, Tachibana K, et al. 1996. Defects of B-cell lymphopoiesis and bone marrow myelopoiesis in mice lacking the CXC chemokine PBSF/SDF-1. Nature 382: 635–638.
    [120]Ma Q, Jones D, Borghesani PR, et al. 1998. Impaired B-lymphopoiesis, myelopoiesis,and derailed cerebellar neuron migration in CXCR4 and SDF-1–deficient mice. Proc. Natl. Acad. Sci. USA 95: 9448–9453.
    [121]Tachibana K, Hirota S, Iizasa H, et al. 1998. The chemokine receptor CXCR4 is essential for vascularization of the gastrointestinal tract. Nature 393: 591–594.
    [122]Zou YR, Kottmann AH, Kuroda M, et al. 1998. Function of the chemokine receptor CXCR4 in haematopoiesis and cerebellar development. Nature 393: 595–599.
    [123]Ruiz de Almodovar C, Luttun A, Carmeliet P. An SDF-1 trap for myeloid cells stimulates angiogenesis. Cell. 2006;124:18–21.
    [124]Grunewald M, Avraham I, Dor Y, et al. VEGF-induced adult neovascularization: Recruitment, retention, and role of accessory cells. Cell.2006;124:175–189.
    [125]Ki-I Y, Arimura N, Noda Y, et al. Stromal-derived factor-1 and inflammatory cytokines in retinal vein occlusion. Curr Eye Res. 2007 Dec;32(12):1065-72.
    [126]Sonmez K, Drenser KA, Capone A Jr, et al. Vitreous levels of stromal cell-derived factor 1 and vascular endothelial growth factor in patients with retinopathy of prematurity. Ophthalmology. 2008 Jun;115(6):1065-1070.
    [127]Shima DT, Adamis AP, Ferrara N, et al. Hypoxic induction of endothelial cell growth factors in retinal cells: identification and characterization of vascular endothelial growth factor (VEGF) as the mitogen. Mol Med. 1995;1:182–93.
    [128]Adamis AP, Miller JW, Bernal MT, et al. Increased vascular endothelial growth factor levels in the vitreous of eyes with proliferative diabetic retinopathy. Am J Ophthalmol. 1994;118:445–50.
    [129]Yazdani S, Hendi K, Pakravan M. Intravitreal bevacizumab (Avastin) injection for neovascular glaucoma. J Glaucoma 2007;16: 437–439.

© 2004-2018 中国地质图书馆版权所有 京ICP备05064691号 京公网安备11010802017129号

地址:北京市海淀区学院路29号 邮编:100083

电话:办公室:(+86 10)66554848;文献借阅、咨询服务、科技查新:66554700