骨髓来源细胞在脉络膜新生血管发生发展中的作用及其治疗潜能
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
研究背景脉络膜新生血管(choroidal neovascularization, CNV)是近40种眼病的共有体征,视网膜下病理性新生血管的生长不但损伤正常脉络膜视网膜结构,而且常发生出血渗漏,往往造成严重视力损害。CNV的发生机制复杂,涉及多种细胞、因子和信号通路,确切机制尚未完全阐明。
     成年个体新生血管的形成包含两种机制:血管生成(angiogenesis)指原有的血管细胞发生增生、移行,形成新的血管;血管发生(vasculogenesis)指由干细胞分化而来的血管细胞构成新血管。干细胞在一般情况下处于“休眠”状态,在外因刺激或病理状态下会进入血液循环,移行至周边组织,表现出不同程度的更新和分化能力,参与组织修复和新生物形成。近期的研究表明,CNV的生成兼具这两种方式,既有原位组织细胞的增殖,又有骨髓来源细胞(bone marrow-derived cells, BMCs)的参与,但BMCs究竟以何种方式参与CNV生成、BMCs对CNV发展有何影响有待进一步阐明。此外,BMCs是一个异质性细胞群体,包含多种干/祖细胞,究竟是哪种干/祖细胞参与了CNV的生成、它们在CNV发生发展中发挥着怎样的作用、是否可作为CNV治疗的靶点等诸多问题有待研究。
     目的和内容探讨BMCs在CNV发生发展中的作用,在危险因素影响CNV发展中的介导作用,及作为干细胞疗法细胞载体的潜能。⑴建立C57-绿色荧光蛋白(green fluorescent protein, GFP)嵌合体小鼠,观察在嵌合体CNV发生发展过程中,BMCs在CNV聚集、分化和表达蛋白情况;⑵探讨尼古丁对参与CNV生成的BMCs的趋化、分化和蛋白表达能力的影响;⑶观察骨髓来源基质干细胞(mesenchymal stem cells, MSCs)向CNV的趋化特异性和时间窗,观察MSCs参与CNV生成情况,探讨MSCs是否具备作为药物载体的潜能;⑷构建表达人源性色素上皮衍生因子(pigment epithelium derived factor, PEDF)的腺病毒,观察腺病毒转导的MSCs对CNV的抑制作用,探讨利用MSCs靶向治疗CNV的新策略。
     方法⑴通过GFP转基因小鼠与野生型C57小鼠的骨髓移植建立嵌合体小鼠,利用532nm激光建立小鼠CNV模型,脉络膜铺片观察GFP-BMCs向CNV趋化、参与血管结构组成的情况,免疫荧光染色观察CNV中BMCs分化的细胞类型(包括血管内皮细胞、血管平滑肌细胞和巨噬细胞)及表达血管内皮生长因子(vascular endothelial growth factor, VEGF)和碱性成纤维细胞生长因子(basic fibroblast cell growth factor, bFGF)的情况;⑵激光光凝建立C57-GFP嵌合体小鼠CNV模型后,即日在其饮水中添加尼古丁(100μg/ml),对照组嵌合体小鼠建立CNV模型后普通饲养,4周后眼球切片HE染色观察CNV厚度与直径,脉络膜铺片观察CNV表面积、GFP-BMCs的趋化和参与构成血管情况,免疫荧光染色观察CNV中BMCs分化情况及各因子表达变化;⑶体外培养GFP转基因小鼠骨髓来源MSCs,野生型小鼠激光建模后0.5-1小时尾静脉注射4.0×106 GFP-BMCs或GFP-MSCs,激光后1、3、7天,心、肝、脾、肺、心脏切片荧光观察比较两种细胞在器官内的滞留情况;野生型小鼠激光后0.5-1小时、3天及6天,分别注射4.0×106 GFP-MSCs,激光后7天观察比较单次注射、双次注射和三次注射后GFP-MSCs在体内移行和向CNV趋化的情况;酶联免疫吸附测定(enzyme linked immunosorbent assay, ELISA)和免疫荧光染色检测激光后不同时间眼内干细胞趋化因子基质细胞衍生因子-1(stromal cell derived factor-1, SDF-1)的表达;脉络膜铺片检测参与构成新血管的GFP-MSCs,免疫荧光染色分析CNV中GFP-MSCs分化的细胞类型;⑷构建表达人源性PEDF、含报告基因GFP、E1 E3缺陷的5型腺病毒(adenovirus, Ad),转导体外培养的野生型小鼠骨髓来源MSCs,倒置荧光显微镜观察及流式细胞仪测定感染效率,ELISA检测体外人源性PEDF表达情况;激光后0.5-1小时小鼠尾静脉分别注射4.0×106 Ad-PEDF/MSCs、对照病毒Ad-GFP/MSCs、GFP-MSCs及0.4ml磷酸盐缓冲液(Phosphate buffered saline, PBS),分别于激光后1、3、5、7天利用免疫荧光染色和ELISA检测眼内人源性PEDF表达,激光后7天眼球切片组织学分析和脉络膜铺片分析比较各组CNV厚度、直径及表面积。体外建立视网膜色素上皮(retinal pigment epithelium, RPE)细胞与MSCs的共培养体系,分析MSCs分泌的PEDF对RPE细胞增生和移行的影响。
     结果⑴激光后大量GFP-BMCs聚集于激光斑处并整合入CNV中的血管结构内,由其组成的血管面积约占CNV表面积的16.22%。GFP-BMCs主要分布于CNV区域(包括CNV深层的脉络膜),少量散布于CNV表层的视网膜、角巩膜缘、睫状体、视盘周围及远离CNV的视网膜、脉络膜和巩膜。CD31或αSMA阳性的GFP+细胞仅出现于CNV区域,F4/80+/GFP+细胞不仅出现在CNV区域,还出现在CNV表层的视网膜、角巩膜缘和睫状体内。CNV内CD31/GFP、αSMA/GFP和F4/80/GFP双阳性细胞在全部GFP+细胞中的构成比随CNV进展而变化,CD31/GFP或F4/80/GFP双阳性细胞构成比的最高值出现于激光后第2周,而αSMA+/GFP+细胞构成比在激光后4周仍处上升趋势。CNV区域的一些GFP-BMCs可表达促血管形成因子VEGF和bFGF。⑵尼古丁处理组小鼠CNV长度和表面积增大,CNV内GFP-BMCs的面积和密度增加,GFP-BMCs来源的血管细胞面积增加,F4/80+/GFP+细胞构成比下降,CNV内VEGF和bFGF的表达及CNV下脉络膜内VCAM-1的表达上调。⑶在整个观察期间,肺、肝和脾内可见GFP+BMCs滞留,但未观测到GFP+MSCs。单次、双次或三次MSCs注射后趋化至CNV的MSCs的数量没有明显差异,双次和三次注射组小鼠的脾内发现大量GFP-MSCs。SDF-1免疫荧光染色显示,激光后初期,SDF-1在激光斑处RPE层表达,随着CNV生成,CNV内出现阳性染色;ELISA结果显示,SDF-1表达水平在最初24小时内迅速增加,在第2天时达顶峰,然后迅速下降。MSCs移植后外周血和骨髓的流式分析显示,MSCs仅于第1天内在骨髓中作短暂停留。脉络膜铺片显示,激光后第1天,GFP-MSCs散布于激光斑周围;第3天时,GFP-MSCs组成的“细胞环”围绕激光斑,显示出向CNV靠近的定向移行趋势;第7天,GFP-MSCs进入CNV内并加入血管组成中。眼球切片显示绝大多数GFP-MSCs位于激光斑处脉络膜与光感受器间的CNV内。在这些MSCs中发现了血管内皮细胞、血管平滑肌细胞、巨噬细胞、上皮细胞和成纤维细胞的标记物(CD31,αSMA, F4/80, keratin,和vimentin)的阳性表达。⑷Ad转导小鼠MSCs后24小时,倒置荧光显微镜检测到报告基因GFP的表达,流式分析显示(73.6±5.3)%的细胞转导成功,AdPEDF转导的MSCs在体外表达PEDF可持续至少8天。AdPEDF组小鼠的眼球切片中,人PEDF的阳性染色出现于MSCs内及其周围的细胞外基质中;ELISA结果显示,在整个观察期间,眼内人PEDF呈稳定表达,表达量是抑制CNV阈值的4倍多;其CNV的厚度、长度和表面积明显减小。与AdPEDF/MSCs共培养的RPE细胞较之其他共培养条件下的RPE细胞增生和移行更快。
     结论⑴BMCs分化为多种细胞类型参与CNV细胞构成,分泌促血管形成因子促进CNV发展,在CNV生成过程中发挥着重要作用。CNV微环境趋化BMCs,并支持和调控BMCs的分化方向。BMCs与CNV微环境可能存在相互作用。⑵尼古丁促进BMCs向CNV趋化和参与CNV生成,并影响CNV内BMCs的分化。尼古丁对BMCs的这种作用可能部分通过调节局部因子(如VEGF、VCAM-1)表达实现。⑶MSCs仅出现于CNV而不在其他器官停留(骨髓中一过性停留),提示其在CNV模型中具有比BMCs更为卓越的特异趋化能力;一次激光光凝引起的MSCs趋化具有有限的时间窗,其原因可能是激光后CNV区域趋化因子的表达规律。激光后MSCs逐步趋化至CNV内,并分化为CNV生成所需的多种细胞类型。MSCs具备作为细胞载体的潜能。⑷经基因修饰的MSCs到达CNV部位并在局部表达抗新生血管形成因子,从而抑制了CNV的生长,该效应可能部分通过在CNV发展中发挥重要作用的RPE细胞介导。MSCs有望作为抗新生血管药物的释放系统用于CNV相关疾病的治疗。
Background choroidal neovascularization (CNV), which refers to the formation of new blood vessels that arise from the choriocapillaries through Bruch's membrane into the subretinal space causing damage of topic choriod and retina, exudation of fluid and hemorrhage, is now known to occur as a final common pathway in nearly 40 ophthalmic diseases leading to irreversible visual loss. The pathogenesis of CNV, which includes multiple cell-types, cytokines and signal transduction pathways, is quite complicated and still poorly understood.
     Postnatal Vessels are formed through two distinct processes. Angiogenesis involve the remodeling of established capillary networks and arterioles, while vasculogenesis involves the differentiation of stem/progenitor cells into mature vascular cells. Though being‘quiescence’generally, these stem/progenitor cells can be mobilized into circulation by physiological or pathologic stimulus to contribute to tissue repair and neoplasia formation at perienchyma. Recent studies have indicated that CNV occur through not only angiogenesis, but also vasculogenesis, that is, both bone marrow-derived cells (BMCs) and cells in situ participate in CNV development. However, little is known about the dynamic conduct of BMCs in the CNV microenvironment. BMCs, a heterogeneous cell population, is comprise of multiple stem/progenitor cell-types. Which types of them contribute to CNV and what role they play in the process? Whether they could be the new targets for CNV treatment? Lots of issues need to be studied.
     Objectives This study aims⑴to investigate the contribution of BMCs to CNV, and the dynamic conduct of BMCs in CNV microenvironment;⑵to investigate the effects of nicotine on BMCs’contribution to CNV and the underlying mechanism;⑶to investigate the specific recruitment of MSCs to CNV and their contribution to CNV formation, to confirm the potential of MSCs being cell vectors;⑷to explore a noval therapeutic strategy of applicating MSCs as delivery vehicles for CNV treatment.
     Methods⑴Green fluorescent protein (GFP) chimeric mice were developed by transplanting unpurified bone marrow cells from GFP +/+ transgenic mice to wild-type adult C57 mice. The qualified chimeric mice underwent laser rupture of Bruch membrane to induce CNV. Choroidal flatmount was performed to detect BMCs recruitment to and participation in CNV lesions, and immunofluorescent staining was performed to detect the phenotype of GFP+ cells (including vascular endothelial cells (VEC), vascular smooth muscle cells (VSMC) and macrophages) and expression of vascular endothelia growth factor (VEGF) and basic fibroblast cell growth factor (bFGF) in CNV;⑵GFP chimeric mice were developed. CNV was induced by lasering, and nicotine was administered orally in drinking water of mice in nicotine group on that very day. The chimeric mice in control group were normal raised. Four weeks later, histopathologic study and choroidal flatmount were performed to measure the CNV severity and BMCs recruitment. The differentiation of BMCs in CNV and local expressions of VEGF, bFGF and vascular cell adhesion molecule-1 (VCAM-1) were detected by immunofluorescent staining;⑶MSCs derived from GFP transgenic C57 mice were enriched and cultured. 4.0×106 GFP-MSCs or unpurified bone marrow cells were injected into wild type C57 mice through tail veins 0.5-1 hour after laser photocoagulation induction of CNV. To compare the migration of these two cell-types, GFP+ cells were detected in peripheral blood, bone marrow, heart, liver, spleen and lung. To study the time window of MSCs recruitment, mice were randomly divided into three groups. Mice in one group received single injection of 4.0×106 GFP-MSCs 0.5-1 hours after laser photocoagulation. The second group received twice injection at 0.5-1 hours and 3 days after lasering, and mice in another group received three times injections at 0.5-1 hours, 3 days and 6 days after lasering, respectively. Migration of GFP-MSCs in vivo was observed and compared. Expression of SDF-1, an important chemotactic factor for stem cells, in eyes after lasering was detected by using ELISA and immunofluorescent staining. Seven days after laser, choroidal flatmount was performed to investigate the participate of MSCs in CNV, and the differentiation of MSCs in CNV were analyzed by cell-markers immunofluorescent staining;⑷Adenoviral vectors (Ad) expressing human pigment epithelial-derived factor (PEDF) were generated with a reporter gene (GFP). Control vectors that do not express PEDF were constructed and produced in parallel (AdNull). MSCs were transduced. Transduction efficiency was detected by inverted microscopy and flow cytometry. Concentrations of human PEDF released by the transduced MSCs were measured by ELISA. AdPEDF/ MSCs, AdNull/MSCs, GFP-MSCs (4.0×106 cells/mouse) or 0.4ml PBS were injected into C57 mice tail veins 0.5-1 hour after laser photocoagulation. Human PEDF expression in mice eyes was examined by ELISA and immunofluorescent staining. One week after laser, CNV lesion severity was measured by quantitative analysis of both histopathology and choroidal flatmount. To test the impact of PEDF secreted by transduced MSCs on the RPE proliferation and migration, coculture system of AdPEDF/MSCs and RPE cells was established.
     Results⑴Large number of GFP-BMCs appeared in the CNV lesions and integrated into CNV, constituting 16.22% of the total vascular area. Most GFP-BMCs appeared in CNV lesions (including choroid beneath CNV lesion), and a few GFP-BMCs were in neurosensory retina over CNV, corneoscleral limbus, ciliary body, optic disc and sclera, retina and choroid distant from CNV. GFP+ cells, which were immunoreactive forαSMA or CD31, appeared in CNV lesions only. However, F4/80+/GFP+ cells can be also detected in neurosensory retina over CNV, corneoscleral limbus and ciliary body. The constituent ratio of those three cell-types in total GFP-BMCs in CNV altered as CNV developed. The maximal ratios of CD31-labeled cells and F4/80-labeled cells presented at 2 week, while the ratio ofαSMA-labeled cells upgraded continuously. Immunofluorescent staining showed some BMCs in CNV were VEGF or bFGF positive.⑵Nicotine administration resulted in larger diameter and surface area of CNV. Nicotine-exposed mice demonstrated increased area and density of GFP+BMCs, increased GFP+ vascular cells area, and decreased ratio of BMCs expressing F4/80 in CNV. Furthermore, the expression of VEGF and bFGF within CNV and VCAM-1 in choroid beneath CNV was up-regulated in nicotine-exposed mice.⑶GFP-labeled BMCs , rather than MSCs, were found in lungs, livers and spleens at all time point. The quantity of MSCs recruited to CNV after singer, twice, or thrice MSCs-injections was compared, and there was no statistic difference between each two groups, while lots of GFP-MSCs were found in spleens of mice in the latter two groups. SDF-1 expression in eyes of mice models was examined. Initially, SDF-1 was expressed in RPE layer. As CNV formed, positive staining presented in CNV. The level of SDF-1 expression increased rapidly in the first 24h, reached peak at the second day, and then decreased sharply. Flow cytometry analysis showed MSCs underwent a transient retention in bone marrow. Choroidal flatmount indicated that 1 day after laser, GFP-MSCs were found dispersed around the laser sites. Subsequently, a directional movement of GFP-MSCs coming closer toward the CNV lesions presented. On day 7, GFP-MSCs were found in CNV and appeared to participate in vascular structure formation. Most GFP-MSCs were located in CNV between choroid and photoreceptors. In differentiation analysis, positive expression was found for cell-markers of VEC, VSMC, macrophage, fibroblast and epithelial cell in MSCs examined.⑷Reporter GFP expression in MSCs was found 24 hours after transduction by using inverted fluorescent microscope. Flow cytometry analysis further showed (73.6±5.3)% of the total cell population was GFP positive. Human PEDF production by AdPEDF/MSCs persisted for at least 8 days in vitro. After transplantation of AdPEDF/MSCs, human PEDF positive staining was found in MSCs and the nearby extracellular matrix in CNV. ELISA analysis showed that PEDF expression persisted for at least one week, the average production of human PEDF in the eyes was steady and the level was 4 folds higher than the quantity required to elicit anti-angiogenic effects for CNV. Histopathology and flatmount analysis indicated that there was significant reduction in CNV thickness, diameter and surface area in the AdPEDF group. The proliferation and migration of RPE cells which cocultured with AdPEDF/MSCs were enhanced.
     Conclusion⑴BMCs play an important role in CNV by differentiating into multiple cell-types to compose CNV and secreting angiogenic factors. CNV microenvironment recruits BMCs, supports and regulates the differentiation of BMCs. There may be interaction between BMCs and the CNV microenvironment.⑵Nicotine promotes recruitment and incorporation of BMCs into CNV, and affects differentiation of BMCs in CNV. These effects may be partly due to regulation of related factors (e.g. VEGF or VCAM-1) expression by nicotin.⑶Comparing with unpurified BMCs, MSCs could be specifically recruited into CNV lesions, without stagnation in other organs, indicating their predominant potential for specific recruitment to CNV. Single lasering induction results in a finite time window for MSCs recruitment, it may due to the expression pattern of chemotatic factor, such as SDF-1, in CNV. After laser photocoagulation, MSCs were specifically recruited into CNV lesions gradually, and differentiated into multiple cell types required for CNV formation.⑷Genetic engineered MSCs localized and produced anti-angiogenic factor in CNV lesion. Thus, the growth of CNV was inhibited in vivo. The effect may be mediated, partly at least, by RPE cells, which function as an important regulator for CNV development. MSCs could serve as delivery vehicles of anti-angiogenic agents for the treatment of a range of CNV-associated diseases.
引文
1. Tong JP, Yao YF. Contribution of VEGF and PEDF to choroidal angiogenesis: a need for balanced expressions. Clin Biochem. 2006;39:267-276.
    2. Grossniklaus HE, Green WR. Choroidal neovascularization. Am J Ophthalmol. 2004;137:496-503.
    3. Noel A, Jost M, Lambert V, Lecomte J, Rakic JM. Anti-angiogenic therapy of exudative age-related macular degeneration: current progress and emerging concepts. Trends Mol Med. 2007;13:345-352.
    4. Eichmann A, Pardanaud L, Yuan L, Moyon D. Vasculogenesis and the search for the hemangioblast. J Hematother Stem Cell Res. 2002;11:207-214.
    5. Aghi M, Chiocca EA. Contribution of bone marrow-derived cells to blood vessels in ischemic tissues and tumors. Mol Ther. 2005;12:994-1005.
    6. Espinosa-Heidmann DG, Caicedo A, Hernandez EP, Csaky KG, Cousins SW. Bone marrow-derived progenitor cells contribute to experimental choroidal neovascularization. Invest Ophthalmol Vis Sci. 2003;44:4914-4919.
    7. Tomita M, Yamada H, Adachi Y, et al. Choroidal neovascularization is provided by bone marrow cells. Stem Cells. 2004;22:21-26.
    8. Sata M. Circulating progenitors contribute to angiogenesis, vascular repair, and lesion formation. Rinsho Byori. 2005;53:70-76.
    9. Sheridan CM, Rice D, Hiscott PS, Wong D, Kent DL. The presence of AC133-positive cells suggests a possible role of endothelial progenitor cells in the formation of choroidal neovascularization. Invest Ophthalmol Vis Sci. 2006;47:1642-1645.
    10. Hou HY, Wang YS, Xu JF, Wang YC, Liu JP. The dynamic conduct of bone marrow-derived cells in the choroidal neovascularization microenvironment. Curr Eye Res. 2006;31:1051-1061.
    11. Espinosa-Heidmann DG, Reinoso MA, Pina Y, Csaky KG, Caicedo A, Cousins SW. Quantitative enumeration of vascular smooth muscle cells and endothelial cells derived from bone marrow precursors in experimental choroidal neovascularization. Exp Eye Res. 2005;80:369-378.
    12. Sengupta N, Caballero S, Mames RN, Butler JM, Scott EW, Grant MB. The role of adult bone marrow-derived stem cells in choroidal neovascularization. Invest Ophthalmol Vis Sci. 2003;44:4908-4913.
    13. Sasahara M, Otani A, Yodoi Y, Gotoh N, Kameda T, Yoshimura N. Circulating hematopoietic stem cells in patients with choroidal neovascularization secondary to pathologic myopia. Eye. 2009;23:718-726.
    14. Sasahara M, Otani A, Yodoi Y, Yoshimura N. Circulating hematopoietic stem cells in patients with idiopathic choroidal neovascularization. Invest Ophthalmol Vis Sci.2009;50:1575-1579.
    15. Yodoi Y, Sasahara M, Kameda T, Yoshimura N, Otani A. Circulating hematopoietic stem cells in patients with neovascular age-related macular degeneration. Invest Ophthalmol Vis Sci. 2007;48:5464-5472.
    16. Chan-Ling T, Baxter L, Afzal A, et al. Hematopoietic stem cells provide repair functions after laser-induced Bruch's membrane rupture model of choroidal neovascularization. Am J Pathol. 2006;168:1031-1044.
    17. Sengupta N, Caballero S, Mames RN, Timmers AM, Saban D, Grant MB. Preventing stem cell incorporation into choroidal neovascularization by targeting homing and attachment factors. Invest Ophthalmol Vis Sci. 2005;46:343-348.
    18. Lee ES, Rewolinski D. Evaluation of CXCR4 inhibition in the prevention and intervention model of laser-induced choroidal neovascularization. Invest Ophthalmol Vis Sci. 2009.
    19. Ozawa K, Sato K, Oh I, et al. Cell and gene therapy using mesenchymal stem cells (MSCs). J Autoimmun. 2008;30:121-127.
    20. Gao F, He T, Wang H, et al. A promising strategy for the treatment of ischemic heart disease: Mesenchymal stem cell-mediated vascular endothelial growth factor gene transfer in rats. Can J Cardiol. 2007;23:891-898.
    21. Liu XH, Bai CG, Xu ZY, et al. Therapeutic potential of angiogenin modified mesenchymal stem cells: angiogenin improves mesenchymal stem cells survival under hypoxia and enhances vasculogenesis in myocardial infarction. Microvasc Res. 2008;76:23-30.
    22. Hall B, Dembinski J, Sasser AK, Studeny M, Andreeff M, Marini F. Mesenchymal stem cells in cancer: tumor-associated fibroblasts and cell-based delivery vehicles. Int J Hematol. 2007;86:8-16.
    23. Hall B, Andreeff M, Marini F. The participation of mesenchymal stem cells in tumor stroma formation and their application as targeted-gene delivery vehicles. Handb Exp Pharmacol. 2007:263-283.
    24. Mohr A, Lyons M, Deedigan L, et al. Mesenchymal stem cells expressing TRAIL lead to tumour growth inhibition in an experimental lung cancer model. J Cell Mol Med. 2008;12:2628-2643.
    25. Campochiaro PA, Soloway P, Ryan SJ, Miller JW. The pathogenesis of choroidal neovascularization in patients with age-related macular degeneration. Mol Vis. 1999;5:34.
    26. Yoshida T. Molecular mechanism of choroidal neovascularization in age-related macular degeneration. Nippon Ganka Gakkai Zasshi. 2007;111:881-891.
    27. Baffi JZ BG, Wolfe JD. Adult hematopoetic stem cells provide functional hemangioblast activity during choroidal neovascularization in mice. Invest Ophthalmol Vis Sci. 2003;44:1825.
    28. Nishimura H, Asahara T. Bone marrow-derived endothelial progenitor cells for neovascular formation. EXS. 2005:147-154.
    29. Choi K, Kennedy M, Kazarov A, Papadimitriou JC, Keller G. A common precursor for hematopoietic and endothelial cells. Development. 1998;125:725-732.
    30. Naylor AR, Bolia A, Abbott RJ, et al. Randomized study of carotid angioplasty and stenting versus carotid endarterectomy: a stopped trial. J Vasc Surg. 1998;28:326-334.
    31. Hristov M, Weber C. Endothelial progenitor cells: characterization, pathophysiology, and possible clinical relevance. J Cell Mol Med. 2004;8:498-508.
    32. Takahashi H, Yanagi Y, Tamaki Y, Muranaka K, Usui T, Sata M. Contribution of bone-marrow-derived cells to choroidal neovascularization. Biochem Biophys Res Commun. 2004;320:372-375.
    33. Caicedo A, Espinosa-Heidmann DG, Pina Y, Hernandez EP, Cousins SW. Blood-derived macrophages infiltrate the retina and activate Muller glial cells under experimental choroidal neovascularization. Exp Eye Res. 2005;81:38-47.
    34. Espinosa DG. CA, Hernandez EP. Adult bone marrow derived progenitor cells contribute to choroidal neovascularization and modulate the severity. Invest Ophthalmol Vis Sci. 2003;44:3936.
    35. Csaky KG, Baffi JZ, Byrnes GA, et al. Recruitment of marrow-derived endothelial cells to experimental choroidal neovascularization by local expression of vascular endothelial growth factor. Exp Eye Res. 2004;78:1107-1116.
    36. Reinoso MA CA, Espinosa-Heidmann DG. Blood-derived macrophages and bone marrow derived progenitor cells contribute to intrachoroidal vascular changes in experimental choroidal neovascularization. Invest Ophthalmol Vis Sci. 2004;45:3386.
    37. Espinosa DG. CA, Hernandez EP. Aged bone marrow transfers age-related pathology into young recipients in experimental choroidal neovascularization. Invest Ophthalmol Vis Sci. 2004;45.
    38. Espinosa DG PS, Hernandez EP. Vascular precursor cells from aged mice transfer age-related pathology into young recipients. Invest Ophthalmol Vis Sci. 2005;46:1216.
    39. Tsutsumi C SK, Ishibashi T. The relative contribution of infiltrating bone marrow-derived cells in developing choroidal neovascularization. Invest Ophthalmol Vis Sci. 2003;44:3944.
    40. Gill M, Dias S, Hattori K, et al. Vascular trauma induces rapid but transient mobilization of VEGFR2(+)AC133(+) endothelial precursor cells. Circ Res. 2001;88:167-174.
    41. Veikkola T, Alitalo K. VEGFs, receptors and angiogenesis. Semin Cancer Biol. 1999;9:211-220.
    42. Asahara T, Takahashi T, Masuda H, et al. VEGF contributes to postnatal neovascularization by mobilizing bone marrow-derived endothelial progenitor cells. EMBO J. 1999;18:3964-3972.
    43. Shalaby F, Rossant J, Yamaguchi TP, et al. Failure of blood-island formation and vasculogenesis in Flk-1-deficient mice. Nature. 1995;376:62-66.
    44. Ceradini DJ, Kulkarni AR, Callaghan MJ, et al. Progenitor cell trafficking is regulated by hypoxic gradients through HIF-1 induction of SDF-1. Nat Med. 2004;10:858-864.
    45. Takahashi T, Kalka C, Masuda H, et al. Ischemia- and cytokine-induced mobilization of bone marrow-derived endothelial progenitor cells for neovascularization. Nat Med. 1999;5:434-438.
    46. Lai CC, Wu WC, Chen SL, et al. Suppression of choroidal neovascularization by adeno-associated virus vector expressing angiostatin. Invest Ophthalmol Vis Sci. 2001;42:2401-2407.
    47. Iwaguro H, Yamaguchi J, Kalka C, et al. Endothelial progenitor cell vascular endothelial growth factor gene transfer for vascular regeneration. Circulation. 2002;105:732-738.
    48. Guymer RH, Chong EW. Modifiable risk factors for age-related macular degeneration. Med J Aust. 2006;184:455-458.
    49. Suner IJ, Espinosa-Heidmann DG, Marin-Castano ME, Hernandez EP, Pereira-Simon S, Cousins SW. Nicotine increases size and severity of experimental choroidal neovascularization. Invest Ophthalmol Vis Sci. 2004;45:311-317.
    50. Khan JC, Thurlby DA, Shahid H, et al. Smoking and age related macular degeneration: the number of pack years of cigarette smoking is a major determinant of risk for both geographic atrophy and choroidal neovascularisation. Br J Ophthalmol. 2006;90:75-80.
    51. Espinosa-Heidmann DG, Suner IJ, Catanuto P, Hernandez EP, Marin-Castano ME, Cousins SW. Cigarette smoke-related oxidants and the development of sub-RPE deposits in an experimental animal model of dry AMD. Invest Ophthalmol Vis Sci. 2006;47:729-737.
    52. Scott WK, Schmidt S, Hauser MA, et al. Independent effects of complement factor H Y402H polymorphism and cigarette smoking on risk of age-related macular degeneration. Ophthalmology. 2007;114:1151-1156.
    53. Evans JR, Fletcher AE, Wormald RP. 28,000 Cases of age related macular degeneration causing visual loss in people aged 75 years and above in the United Kingdom may be attributable to smoking. Br J Ophthalmol. 2005;89:550-553.
    54. Evans JR. Risk factors for age-related macular degeneration. Prog Retin Eye Res. 2001;20:227-253.
    55. la Cour M, Kiilgaard JF, Nissen MH. Age-related macular degeneration: epidemiology and optimal treatment. Drugs Aging. 2002;19:101-133.
    56. Krishnaiah S, Das T, Nirmalan PK, et al. Risk factors for age-related macular degeneration: findings from the Andhra Pradesh eye disease study in South India. Invest Ophthalmol Vis Sci. 2005;46:4442-4449.
    57. Thornton J, Edwards R, Mitchell P, Harrison RA, Buchan I, Kelly SP. Smoking and age-related macular degeneration: a review of association. Eye (Lond). 2005;19:935-944.
    58. Penfold PL, Madigan MC, Gillies MC, Provis JM. Immunological and aetiological aspects of macular degeneration. Prog Retin Eye Res. 2001;20:385-414.
    59. Mitchell P, Chapman S, Smith W. Smoking is a major cause of blindness. Med J Aust. 1999;171:173-174.
    60. Schaumberg DA, Hankinson SE, Guo Q, Rimm E, Hunter DJ. A prospective study of 2 major age-related macular degeneration susceptibility alleles and interactions with modifiable risk factors. Arch Ophthalmol. 2007;125:55-62.
    61. Schmidt S, Hauser MA, Scott WK, et al. Cigarette smoking strongly modifies the association of LOC387715 and age-related macular degeneration. Am J Hum Genet. 2006;78:852-864.
    62. DeAngelis MM, Ji F, Kim IK, et al. Cigarette smoking, CFH, APOE, ELOVL4, and risk of neovascular age-related macular degeneration. Arch Ophthalmol. 2007;125:49-54.
    63. Schmidt S, Haines JL, Postel EA, et al. Joint effects of smoking history and APOE genotypes in age-related macular degeneration. Mol Vis. 2005;11:941-949.
    64. Dandekar SS, Jenkins SA, Peto T, Bird AC, Webster AR. Does smoking influence the type of age related macular degeneration causing visual impairment? Br J Ophthalmol. 2006;90:724-727.
    65. Wimpissinger B, Resch H, Berisha F, Weigert G, Polak K, Schmetterer L. Effects of isometric exercise on subfoveal choroidal blood flow in smokers and nonsmokers. Invest Ophthalmol Vis Sci. 2003;44:4859-4863.
    66. Wimpissinger B, Resch H, Berisha F, Weigert G, Schmetterer L, Polak K. Response of choroidal blood flow to carbogen breathing in smokers and non-smokers. Br J Ophthalmol. 2004;88:776-781.
    67. Gundogan FC, Erdurman C, Durukan AH, Sobaci G, Bayraktar MZ. Acute effects of cigarette smoking on multifocal electroretinogram. Clin Experiment Ophthalmol. 2007;35:32-37.
    68. Gottsch JD, Bynoe LA, Harlan JB, Rencs EV, Green WR. Light-induced deposits in Bruch's membrane of protoporphyric mice. Arch Ophthalmol. 1993;111:126-129.
    69. Solberg Y, Rosner M, Belkin M. The association between cigarette smoking and ocular diseases. Surv Ophthalmol. 1998;42:535-547.
    70. Strunnikova N, Zhang C, Teichberg D, et al. Survival of retinal pigment epithelium after exposure to prolonged oxidative injury: a detailed gene expression and cellular analysis. Invest Ophthalmol Vis Sci. 2004;45:3767-3777.
    71. Suner IJ E-HD, Pereira-Simon S. Cigarette smoke increases severity of experimental choroidal neovascularization (CNV): role of inflammation. Invest Ophthalmol Vis Sci. 2005;46:3507.
    72. Heeschen C, Weis M, Aicher A, Dimmeler S, Cooke JP. A novel angiogenic pathway mediated by non-neuronal nicotinic acetylcholine receptors. J Clin Invest. 2002;110:527-536.
    73. Ng MK, Wu J, Chang E, et al. A central role for nicotinic cholinergic regulation of growth factor-induced endothelial cell migration. Arterioscler Thromb Vasc Biol. 2007;27:106-112.
    74. Villablanca AC. Nicotine stimulates DNA synthesis and proliferation in vascular endothelial cells in vitro. J Appl Physiol. 1998;84:2089-2098.
    75. Mousa S, Mousa SA. Cellular and molecular mechanisms of nicotine's pro-angiogenesis activity and its potential impact on cancer. J Cell Biochem. 2006;97:1370-1378.
    76. Jain RK. Clearing the smoke on nicotine and angiogenesis. Nat Med. 2001;7:775-777.
    77. Heeschen C, Jang JJ, Weis M, et al. Nicotine stimulates angiogenesis and promotes tumor growth and atherosclerosis. Nat Med. 2001;7:833-839.
    78. Serobyan N, Schraufstatter IU, Strongin A, Khaldoyanidi SK. Nicotinic acetylcholine receptor-mediated stimulation of endothelial cells results in the arrest of haematopoieticprogenitor cells on endothelium. Br J Haematol. 2005;129:257-265.
    79. Suner IJ E-HD, Tafur CM. The effects of nicotine in choroidal neovascularization: characterization of nicotinic receptors in choroid and reduction of choroidal neovascularization size and severity via receptor blockade. Invest Ophthalmol Vis Sci. 2003;44:3933.
    80. Wang Y, Wang L, Ai X, et al. Nicotine could augment adhesion molecule expression in human endothelial cells through macrophages secreting TNF-alpha, IL-1beta. Int Immunopharmacol. 2004;4:1675-1686.
    81. Wang X, Zhu J, Chen J, Shang Y. Effects of nicotine on the number and activity of circulating endothelial progenitor cells. J Clin Pharmacol. 2004;44:881-889.
    82. Heeschen C, Chang E, Aicher A, Cooke JP. Endothelial progenitor cells participate in nicotine-mediated angiogenesis. J Am Coll Cardiol. 2006;48:2553-2560.
    83. Avogaro A, Fadini GP. The Janus face of nicotinic angiogenesis. J Am Coll Cardiol. 2006;48:2561-2563.
    84. Tuo J, Bojanowski CM, Chan CC. Genetic factors of age-related macular degeneration. Prog Retin Eye Res. 2004;23:229-249.
    85. Afzal A, Shaw LC, Ljubimov AV, Boulton ME, Segal MS, Grant MB. Retinal and choroidal microangiopathies: therapeutic opportunities. Microvasc Res. 2007;74:131-144.
    86. Gage FH. Cell therapy. Nature. 1998;392:18-24.
    87. Stappenbeck TS, Miyoshi H. The role of stromal stem cells in tissue regeneration and wound repair. Science. 2009;324:1666-1669.
    88. Kinnaird T, Stabile E, Burnett MS, Epstein SE. Bone-marrow-derived cells for enhancing collateral development: mechanisms, animal data, and initial clinical experiences. Circ Res. 2004;95:354-363.
    89. Studeny M, Marini FC, Champlin RE, Zompetta C, Fidler IJ, Andreeff M. Bone marrow-derived mesenchymal stem cells as vehicles for interferon-beta delivery into tumors. Cancer Res. 2002;62:3603-3608.
    90. Discher DE, Mooney DJ, Zandstra PW. Growth factors, matrices, and forces combine and control stem cells. Science. 2009;324:1673-1677.
    91. Studeny M, Marini FC, Dembinski JL, et al. Mesenchymal stem cells: potential precursors for tumor stroma and targeted-delivery vehicles for anticancer agents. J Natl Cancer Inst. 2004;96:1593-1603.
    92. Kyriakou CA, Yong KL, Benjamin R, et al. Human mesenchymal stem cells (hMSCs) expressing truncated soluble vascular endothelial growth factor receptor (tsFlk-1) following lentiviral-mediated gene transfer inhibit growth of Burkitt's lymphoma in a murine model. J Gene Med. 2006;8:253-264.
    93. Huang SD, Lu FL, Xu XY, et al. Transplantation of angiogenin-overexpressing mesenchymal stem cells synergistically augments cardiac function in a porcine model of chronic ischemia. J Thorac Cardiovasc Surg. 2006;132:1329-1338.
    94. Guerin E, Sheridan C, Assheton D, et al. SDF1-alpha is associated with VEGFR-2 in humanchoroidal neovascularisation. Microvasc Res. 2008;75:302-307.
    95. Okuyama H, Krishnamachary B, Zhou YF, Nagasawa H, Bosch-Marce M, Semenza GL. Expression of vascular endothelial growth factor receptor 1 in bone marrow-derived mesenchymal cells is dependent on hypoxia-inducible factor 1. J Biol Chem. 2006;281:15554-15563.
    96. Yu WH, Dong FT, Zhang ZQ, Huo DM, Du H. Differentiation of rat mesenchymal stem cells in the microenvironment of choroidal neovascularization. Zhonghua Yan Ke Za Zhi. 2007;43:146-150.
    97. Ding X, Patel M, Chan CC. Molecular pathology of age-related macular degeneration. Prog Retin Eye Res. 2009;28:1-18.
    98. Harris JR, Brown GA, Jorgensen M, et al. Bone marrow-derived cells home to and regenerate retinal pigment epithelium after injury. Invest Ophthalmol Vis Sci. 2006;47:2108-2113.
    99. Arnhold S, Heiduschka P, Klein H, et al. Adenovirally transduced bone marrow stromal cells differentiate into pigment epithelial cells and induce rescue effects in RCS rats. Invest Ophthalmol Vis Sci. 2006;47:4121-4129.
    100. Tamaki Y. Novel approach for management of age-related macular degeneration--antiangiogenic therapy and retinal regenerative therapy. Nippon Ganka Gakkai Zasshi. 2007;111:232-268; discussion 269.
    101. Stender S, Murphy M, O'Brien T, et al. Adeno-associated viral vector transduction of human mesenchymal stem cells. Eur Cell Mater. 2007;13:93-99; discussion 99.
    102. Bosch P, Stice SL. Adenoviral transduction of mesenchymal stem cells. Methods Mol Biol. 2007;407:265-274.
    103. Ball SG, Shuttleworth CA, Kielty CM. Mesenchymal stem cells and neovascularization: role of platelet-derived growth factor receptors. J Cell Mol Med. 2007;11:1012-1030.
    104. Ma Y, Xu Y, Xiao Z, et al. Reconstruction of chemically burned rat corneal surface by bone marrow-derived human mesenchymal stem cells. Stem Cells. 2006;24:315-321.
    105. Reich SJ, Bennett J. Gene therapy for ocular neovascularization: a cure in sight. Curr Opin Genet Dev. 2003;13:317-322.
    106. Breitbach M, Bostani T, Roell W, et al. Potential risks of bone marrow cell transplantation into infarcted hearts. Blood. 2007;110:1362-1369.
    107. Docheva D, Popov C, Mutschler W, Schieker M. Human mesenchymal stem cells in contact with their environment: surface characteristics and the integrin system. J Cell Mol Med. 2007;11:21-38.
    108. Eichler W, Yafai Y, Wiedemann P, Fengler D. Antineovascular agents in the treatment of eye diseases. Curr Pharm Des. 2006;12:2645-2660.
    109. Oshima Y, Oshima S, Nambu H, et al. Increased expression of VEGF in retinal pigmented epithelial cells is not sufficient to cause choroidal neovascularization. J Cell Physiol. 2004;201:393-400.
    110. Nishijima K, Ng YS, Zhong L, et al. Vascular endothelial growth factor-A is a survival factorfor retinal neurons and a critical neuroprotectant during the adaptive response to ischemic injury. Am J Pathol. 2007;171:53-67.
    111. Verheul HM, Pinedo HM. Possible molecular mechanisms involved in the toxicity of angiogenesis inhibition. Nat Rev Cancer. 2007;7:475-485.
    112. Grisanti S, Tatar O. The role of vascular endothelial growth factor and other endogenous interplayers in age-related macular degeneration. Prog Retin Eye Res. 2008;27:372-390.
    113. van Wijngaarden P, Qureshi SH. Inhibitors of vascular endothelial growth factor (VEGF) in the management of neovascular age-related macular degeneration: a review of current practice. Clin Exp Optom. 2008;91:427-437.
    114. Tunon J, Ruiz-Moreno JM, Martin-Ventura JL, Blanco-Colio LM, Lorenzo O, Egido J. Cardiovascular risk and antiangiogenic therapy for age-related macular degeneration. Surv Ophthalmol. 2009;54:339-348.
    115. Wong TY. Age-related macular degeneration and cardiovascular disease in the era of anti-vascular endothelial growth factor therapies. Am J Ophthalmol. 2009;148:327-329.
    116. Barnstable CJ, Tombran-Tink J. Neuroprotective and antiangiogenic actions of PEDF in the eye: molecular targets and therapeutic potential. Prog Retin Eye Res. 2004;23:561-577.
    117. Bhutto IA, McLeod DS, Hasegawa T, et al. Pigment epithelium-derived factor (PEDF) and vascular endothelial growth factor (VEGF) in aged human choroid and eyes with age-related macular degeneration. Exp Eye Res. 2006;82:99-110.
    118. Bhutto IA, Uno K, Merges C, Zhang L, McLeod DS, Lutty GA. Reduction of endogenous angiogenesis inhibitors in Bruch's membrane of the submacular region in eyes with age-related macular degeneration. Arch Ophthalmol. 2008;126:670-678.
    119. Holekamp NM, Bouck N, Volpert O. Pigment epithelium-derived factor is deficient in the vitreous of patients with choroidal neovascularization due to age-related macular degeneration. Am J Ophthalmol. 2002;134:220-227.
    120. Matsuoka M, Ogata N, Otsuji T, Nishimura T, Takahashi K, Matsumura M. Expression of pigment epithelium derived factor and vascular endothelial growth factor in choroidal neovascular membranes and polypoidal choroidal vasculopathy. Br J Ophthalmol. 2004;88:809-815.
    121. Tatar O, Adam A, Shinoda K, et al. Expression of VEGF and PEDF in choroidal neovascular membranes following verteporfin photodynamic therapy. Am J Ophthalmol. 2006;142:95-104.
    122. Becerra SP. Focus on Molecules: Pigment epithelium-derived factor (PEDF). Exp Eye Res. 2006;82:739-740.
    123. Sarojini H, Estrada R, Lu H, et al. PEDF from mouse mesenchymal stem cell secretome attracts fibroblasts. J Cell Biochem. 2008;104:1793-1802.
    124. Becerra SP, Fariss RN, Wu YQ, Montuenga LM, Wong P, Pfeffer BA. Pigment epithelium-derived factor in the monkey retinal pigment epithelium and interphotoreceptor matrix: apical secretion and distribution. Exp Eye Res. 2004;78:223-234.
    125. Notari L, Miller A, Martinez A, et al. Pigment epithelium-derived factor is a substrate formatrix metalloproteinase type 2 and type 9: implications for downregulation in hypoxia. Invest Ophthalmol Vis Sci. 2005;46:2736-2747.
    126. Aymerich MS, Alberdi EM, Martinez A, Becerra SP. Evidence for pigment epithelium-derived factor receptors in the neural retina. Invest Ophthalmol Vis Sci. 2001;42:3287-3293.
    127. Notari L, Baladron V, Aroca-Aguilar JD, et al. Identification of a lipase-linked cell membrane receptor for pigment epithelium-derived factor. J Biol Chem. 2006;281:38022-38037.
    128. Gehlbach P, Demetriades AM, Yamamoto S, et al. Periocular injection of an adenoviral vector encoding pigment epithelium-derived factor inhibits choroidal neovascularization. Gene Ther. 2003;10:637-646.
    129. Demetriades AM, Deering T, Liu H, et al. Trans-scleral delivery of antiangiogenic proteins. J Ocul Pharmacol Ther. 2008;24:70-79.
    130. Apte RS, Barreiro RA, Duh E, Volpert O, Ferguson TA. Stimulation of neovascularization by the anti-angiogenic factor PEDF. Invest Ophthalmol Vis Sci. 2004;45:4491-4497.
    131. Mori K, Gehlbach P, Ando A, et al. Intraocular adenoviral vector-mediated gene transfer in proliferative retinopathies. Invest Ophthalmol Vis Sci. 2002;43:1610-1615.
    132. Saishin Y, Silva RL, Kachi S, et al. Periocular gene transfer of pigment epithelium-derived factor inhibits choroidal neovascularization in a human-sized eye. Hum Gene Ther. 2005;16:473-478.
    133. Mori K, Duh E, Gehlbach P, et al. Pigment epithelium-derived factor inhibits retinal and choroidal neovascularization. J Cell Physiol. 2001;188:253-263.
    134. Mori K, Gehlbach P, Ando A, McVey D, Wei L, Campochiaro PA. Regression of ocular neovascularization in response to increased expression of pigment epithelium-derived factor. Invest Ophthalmol Vis Sci. 2002;43:2428-2434.
    135. Rasmussen H, Chu KW, Campochiaro P, et al. Clinical protocol. An open-label, phase I, single administration, dose-escalation study of ADGVPEDF.11D (ADPEDF) in neovascular age-related macular degeneration (AMD). Hum Gene Ther. 2001;12:2029-2032.
    136. Mori K, Gehlbach P, Yamamoto S, et al. AAV-mediated gene transfer of pigment epithelium-derived factor inhibits choroidal neovascularization. Invest Ophthalmol Vis Sci. 2002;43:1994-2000.
    137. Semkova I, Kreppel F, Welsandt G, et al. Autologous transplantation of genetically modified iris pigment epithelial cells: a promising concept for the treatment of age-related macular degeneration and other disorders of the eye. Proc Natl Acad Sci U S A. 2002;99:13090-13095.
    138. Campochiaro PA, Nguyen QD, Shah SM, et al. Adenoviral vector-delivered pigment epithelium-derived factor for neovascular age-related macular degeneration: results of a phase I clinical trial. Hum Gene Ther. 2006;17:167-176.
    139. Zhang SX, Wang JJ, Gao G, Parke K, Ma JX. Pigment epithelium-derived factor downregulates vascular endothelial growth factor (VEGF) expression and inhibitsVEGF-VEGF receptor 2 binding in diabetic retinopathy. J Mol Endocrinol. 2006;37:1-12.
    140. Yamagishi S, Abe R, Jinnouchi Y, Matsui T, Imaizumi T, Inoue H. Pigment epithelium-derived factor inhibits vascular endothelial growth factor-induced vascular hyperpermeability both in vitro and in vivo. J Int Med Res. 2007;35:896-899.
    141. Zamiri P, Masli S, Streilein JW, Taylor AW. Pigment epithelial growth factor suppresses inflammation by modulating macrophage activation. Invest Ophthalmol Vis Sci. 2006;47:3912-3918.
    142. Kent D, Sheridan C. Choroidal neovascularization: a wound healing perspective. Mol Vis. 2003;9:747-755.
    143. Ohno-Matsui K. Molecular mechanism for choroidal neovascularization in age-related macular degeneration. Nippon Ganka Gakkai Zasshi. 2003;107:657-673.
    144. Asahara T, Murohara T, Sullivan A, et al. Isolation of putative progenitor endothelial cells for angiogenesis. Science. 1997;275:964-967.
    145. Espinosa-Heidmann DG, Suner I, Hernandez EP, Frazier WD, Csaky KG, Cousins SW. Age as an independent risk factor for severity of experimental choroidal neovascularization. Invest Ophthalmol Vis Sci. 2002;43:1567-1573.
    146. Dawn B, Bolli R. Adult bone marrow-derived cells: regenerative potential, plasticity, and tissue commitment. Basic Res Cardiol. 2005;100:494-503.
    147. Suda T, Arai F, Shimmura S. Regulation of stem cells in the niche. Cornea. 2005;24:S12-S17.
    148. Sakurai E, Anand A, Ambati BK, van Rooijen N, Ambati J. Macrophage depletion inhibits experimental choroidal neovascularization. Invest Ophthalmol Vis Sci. 2003;44:3578-3585.
    149. Rafii S, Lyden D. Therapeutic stem and progenitor cell transplantation for organ vascularization and regeneration. Nat Med. 2003;9:702-712.
    150. Nakamura T, Ishikawa F, Sonoda KH, et al. Characterization and distribution of bone marrow-derived cells in mouse cornea. Invest Ophthalmol Vis Sci. 2005;46:497-503.
    151. Ozerdem U, Alitalo K, Salven P, Li A. Contribution of bone marrow-derived pericyte precursor cells to corneal vasculogenesis. Invest Ophthalmol Vis Sci. 2005;46:3502-3506.
    152. Rakic JM, Lambert V, Devy L, et al. Placental growth factor, a member of the VEGF family, contributes to the development of choroidal neovascularization. Invest Ophthalmol Vis Sci. 2003;44:3186-3193.
    153. Carmeliet P. Angiogenesis in life, disease and medicine. Nature. 2005;438:932-936.
    154. Li Y, Reca RG, Atmaca-Sonmez P, et al. Retinal pigment epithelium damage enhances expression of chemoattractants and migration of bone marrow-derived stem cells. Invest Ophthalmol Vis Sci. 2006;47:1646-1652.
    155. Butler JM, Guthrie SM, Koc M, et al. SDF-1 is both necessary and sufficient to promote proliferative retinopathy. J Clin Invest. 2005;115:86-93.
    156. Lindahl P, Johansson BR, Leveen P, Betsholtz C. Pericyte loss and microaneurysm formation in PDGF-B-deficient mice. Science. 1997;277:242-245.
    157. Song S, Ewald AJ, Stallcup W, Werb Z, Bergers G. PDGFRbeta+ perivascular progenitorcells in tumours regulate pericyte differentiation and vascular survival. Nat Cell Biol. 2005;7:870-879.
    158. Quesenberry PJ, Becker PS. Stem cell homing: rolling, crawling, and nesting. Proc Natl Acad Sci U S A. 1998;95:15155-15157.
    159. Zubilewicz A, Hecquet C, Jeanny JC, Soubrane G, Courtois Y, Mascarelli F. Two distinct signalling pathways are involved in FGF2-stimulated proliferation of choriocapillary endothelial cells: a comparative study with VEGF. Oncogene. 2001;20:1403-1413.
    160. Rosenthal R, Malek G, Salomon N, et al. The fibroblast growth factor receptors, FGFR-1 and FGFR-2, mediate two independent signalling pathways in human retinal pigment epithelial cells. Biochem Biophys Res Commun. 2005;337:241-247.
    161. Shen WY, Yu MJ, Barry CJ, Constable IJ, Rakoczy PE. Expression of cell adhesion molecules and vascular endothelial growth factor in experimental choroidal neovascularisation in the rat. Br J Ophthalmol. 1998;82:1063-1071.
    162. Ogata N, Matsushima M, Takada Y, et al. Expression of basic fibroblast growth factor mRNA in developing choroidal neovascularization. Curr Eye Res. 1996;15:1008-1018.
    163. Urbich C, Aicher A, Heeschen C, et al. Soluble factors released by endothelial progenitor cells promote migration of endothelial cells and cardiac resident progenitor cells. J Mol Cell Cardiol. 2005;39:733-742.
    164. Ferrara N, Kerbel RS. Angiogenesis as a therapeutic target. Nature. 2005;438:967-974.
    165. Kamizuru H, Kimura H, Yasukawa T, Tabata Y, Honda Y, Ogura Y. Monoclonal antibody-mediated drug targeting to choroidal neovascularization in the rat. Invest Ophthalmol Vis Sci. 2001;42:2664-2672.
    166. Constable I, Shen WY, Rakoczy E. Emerging biological therapies for age-related macula degeneration. Expert Opin Biol Ther. 2005;5:1373-1385.
    167. Natori T, Sata M, Washida M, Hirata Y, Nagai R, Makuuchi M. Nicotine enhances neovascularization and promotes tumor growth. Mol Cells. 2003;16:143-146.
    168. Ambati J, Ambati BK, Yoo SH, Ianchulev S, Adamis AP. Age-related macular degeneration: etiology, pathogenesis, and therapeutic strategies. Surv Ophthalmol. 2003;48:257-293.
    169. Suner IJ, Tafur, C.M., Espinosa-Heidmann, D.G., Legra, J.M., Arango, K., Hernandez, E., Monroy, D., Csaky, K.G., Cousins, S.W. Nicotine increases severity of choroidal neovascularization. Invest Ophthalmol Vis Sci. 2002;43:1292.
    170. Suner IJ, Espinosa-Heidmann, D.G., Pereira-Simon, S., Pin? a, Y., Cousins, S.W. Cigarette smoke increases severity of experimental choroidal neovascularization (CNV): role of inflammation. Invest Ophthalmol Vis Sci. 2005;46:3507.
    171. Jacobi J, Jang JJ, Sundram U, Dayoub H, Fajardo LF, Cooke JP. Nicotine accelerates angiogenesis and wound healing in genetically diabetic mice. Am J Pathol. 2002;161:97-104.
    172. Wong HP, Yu L, Lam EK, Tai EK, Wu WK, Cho CH. Nicotine promotes colon tumor growth and angiogenesis through beta-adrenergic activation. Toxicol Sci. 2007;97:279-287.
    173. Cloez-Tayarani I, Changeux JP. Nicotine and serotonin in immune regulation and inflammatory processes: a perspective. J Leukoc Biol. 2007;81:599-606.
    174. Aicher A, Heeschen C, Mohaupt M, Cooke JP, Zeiher AM, Dimmeler S. Nicotine strongly activates dendritic cell-mediated adaptive immunity: potential role for progression of atherosclerotic lesions. Circulation. 2003;107:604-611.
    175. Nouri-Shirazi M, Tinajero R, Guinet E. Nicotine alters the biological activities of developing mouse bone marrow-derived dendritic cells (DCs). Immunol Lett. 2007;109:155-164.
    176. Grossniklaus HE, Ling JX, Wallace TM, et al. Macrophage and retinal pigment epithelium expression of angiogenic cytokines in choroidal neovascularization. Mol Vis. 2002;8:119-126.
    177. Heeschen C, Weis M, Cooke JP. Nicotine promotes arteriogenesis. J Am Coll Cardiol. 2003;41:489-496.
    178. Zhang S, Day I, Ye S. Nicotine induced changes in gene expression by human coronary artery endothelial cells. Atherosclerosis. 2001;154:277-283.
    179. Albaugh G, Bellavance E, Strande L, Heinburger S, Hewitt CW, Alexander JB. Nicotine induces mononuclear leukocyte adhesion and expression of adhesion molecules, VCAM and ICAM, in endothelial cells in vitro. Ann Vasc Surg. 2004;18:302-307.
    180. Wang Y, Wang Z, Zhou Y, et al. Nicotine stimulates adhesion molecular expression via calcium influx and mitogen-activated protein kinases in human endothelial cells. Int J Biochem Cell Biol. 2006;38:170-182.
    181. Tutka P, Mosiewicz J, Wielosz M. Pharmacokinetics and metabolism of nicotine. Pharmacol Rep. 2005;57:143-153.
    182. Pittenger MF, Mackay AM, Beck SC, et al. Multilineage potential of adult human mesenchymal stem cells. Science. 1999;284:143-147.
    183. Lima e Silva R, Shen J, Hackett SF, et al. The SDF-1/CXCR4 ligand/receptor pair is an important contributor to several types of ocular neovascularization. FASEB J. 2007;21:3219-3230.
    184. Spaide RF. Rationale for combination therapies for choroidal neovascularization. Am J Ophthalmol. 2006;141:149-156.
    185. Nolan DJ, Ciarrocchi A, Mellick AS, et al. Bone marrow-derived endothelial progenitor cells are a major determinant of nascent tumor neovascularization. Genes Dev. 2007;21:1546-1558.
    186. Fukuchi T, Takahashi K, Uyama M, Matsumura M. Comparative study of experimental choroidal neovascularization by optical coherence tomography and histopathology. Jpn J Ophthalmol. 2001;45:252-258.
    187. Prockop DJ. Marrow stromal cells as stem cells for nonhematopoietic tissues. Science. 1997;276:71-74.

© 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