神经生长因子缓释微球、神经干细胞联合治疗老年性痴呆的实验研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
老年性痴呆(Senile Dementia,SD),亦称阿尔茨海默病(Alzheimer Disease,AD)是一种以进行性认知障碍和记忆能力损害为主的中枢神经系统退变性疾病,伴有不同程度的运动、认知、语言和人格等多方面的异常。AD的发病率随着人口年龄的增大而增高。目前,我国正步入老龄社会,老年人口不断增多,AD患者大幅度增加。AD发病率高,病程长,又缺乏有效的治疗方法,众多的AD病人给家庭和社会带来严重的经济和精神负担。因此,寻找有效的AD治疗方法已成为当今医学界的紧迫课题。
     基底前脑胆碱能神经元的营养匮乏或者丢失是AD的一个重要病理变化。神经营养因子能够很好地阻止或者减少神经元的营养匮乏或者丢失;在众多的神经营养因子家族中,神经生长因子是最典型的,迄今为止研究最深入的神经营养因子。基底前脑中胆碱能神经元表达低亲和力的p75~(NTF)和TrkA受体,神经生长因子通过这些受体来提高基底前脑胆碱能神经元的功能,神经生长因子已被广泛地应用于老年性痴呆的实验性治疗。神经生长因子是大分子蛋白类物质,很难透过血脑屏障;生物半衰期很短。将神经生长因子成功地投递到基底前脑仍是一个很大的难题。神经生长因子的应用多数采用埋管后连续的侧脑室灌注。虽然埋管后连续的侧脑室灌注显示出一定的疗效,长期的侧脑室灌注引起很大的副作用。一种行之有效的方法是用微球的方式将神经生长因子投递到脑内。
     脑组织细胞移植对AD模型鼠的学习记忆能力有一定的改善作用,但临床供体问题难以解决。神经干细胞在中枢神经系统中的存在并培养成功,尤其是神经干细胞系的建立,解决了脑细胞移植供体不足的难题,成为细胞移植的理想供体。神经干细胞在体内和体外的成活、分化都离不开神经营养因子的支持和诱导;在中枢神经系统中的成活、迁移和分化离不开局部微环境中各种营养的支持。神经干细胞能够在特定的脑组织微环境的作用下分化成这一区域特定的神经细胞;其中神经营养因子对神经干细胞的成活、迁移和分化起着至关重要的作用。
     本文就神经生长因子缓释微球和神经干细胞联合治疗AD进行了实验研究。一方面神经干细胞分化成成熟的神经细胞补充溃变的神经元;另一方面,神经生长因子缓释微球所提供的神经生长因子可促进幸存的神经细胞的存活和功能的恢复,同时为神经干细胞的成活、迁移和分化提供营养的支持。
     本课题研究内容包括三个方面:1、神经生长因子缓释微球的制备及评价;2、神经干细胞移植后在AD模型鼠基底前脑内的迁移和分化;3、神经生长因子缓释微球、神经干细胞联合治疗AD模型鼠。
     第一章神经生长因子缓释微球的制备及评价
     一、材料和方法
     采用水/油/水(W1/O/W2)的双乳化技术来制备神经生长因子缓释微球。将5mg蛋白(rhNGF/FITC-BSA 1/2000,w/w)溶于100μl去离子水中,100mg PLGA溶于4ml二氯甲烷/丙酮(3∶1,v/v)的混合溶液中,将蛋白溶液加到PLGA溶液中,在冰浴条件下超声1min(超声功率为20W),即形成油包水(W1/O)型的乳液。然后,将蛋白和PLGA的混合溶液加入25ml 1%PVA的水溶液中,在冰浴的条件下匀速搅拌5min(1000rpm),即形成水包油包水(W1/O/W2)型的复乳液。在常压下,磁力搅拌3-4h,挥发有机溶剂,固化的微球通过离心(12000rpm,10min)收集,用去离子水清洗三遍除掉微球表面的PVA和没有包进的药物,最后冷冻干燥24h,去除微球中的水分。并研究不同条件和添加剂对蛋白包封率的影响。将神经生长因子缓释微球注射入鼠的基底前脑,观察其在体内的释放。
     二、结果
     蛋白/多聚物比率越高,载药量也越高,但是蛋白包封率则越低;当蛋白/PLGA比率由5/100(w/w)增加到15/100(w/w)时,包封率则由89.1%下降到56.5%。乳化前将水溶性添加剂加入内水相能够明显提高蛋白的包封率。内水相中加入聚乙二醇能够将包封率从89.1%提高到97.5%。神经生长因子缓释微球能够持续释放有生物学活性的神经生长因子达35天,且初时爆破释放较低。神经生长因子缓释微球能够在基底前脑持续释放神经生长因子达4周以上。
     第二章神经干细胞移植后在AD模型鼠基底前脑内的迁移和分化
     一、材料和方法
     新生的SD大鼠的基底前脑,机械分离后胰酶消化,制成单细胞悬液,加入含2%B27、表皮生长因子(EGF)和碱性成纤维生长因子(FGF-2)(终浓度均为10ng/ml)的DMEM/F12培养基,放置在37℃的二氧化碳培养箱内培养。待原代克隆形成后机械分离成单细胞悬液,按上述条件继续培养。以后每5~7天分离克隆传代一次,方法同前。用10%的小牛血清诱导贴壁的神经干细胞分化。用BrdU(6μg/ml)标记传代培养的细胞。用免疫细胞化学方法,行Nestin、NF、GFAP、BrdU染色,鉴定培养细胞的增殖能力和分化潜能。雄性SD大鼠(250-300g),单侧切断穹隆—海马伞(FF),以制备隔-海马通路损伤的AD模型。大鼠切断FF,将神经干细胞4ul(2.5x10~4个/μl)移植入基底前脑,坐标为:前囟+0.6mm,外侧+0.6mm插入,腹侧-5.5mm。分别在1、2、3、4周检测神经干细胞在体内的存活、迁移和分化情况。
     二、结果
     神经干细胞能够在EGF和FGF-2的刺激下分裂增殖,1周左右形成由数十到数百个细胞组成的细胞球。传代培养的神经干细胞具有与原代培养的神经干细胞相同的生物学特性,并能够分化为神经元和神经胶质细胞,分别呈NF、GFAP阳性。克隆细胞球染色,显示Nestin和BrdU标记阳性反应。免疫荧光标记检测,贴壁的细胞为DAPI+BrdU+GFAP阳性反应或者DAPI+BrdU+NF阳性反应。移植后1周,大部分移植细胞聚集在移植的针道附近,结果显示Nestin阳性,细胞胞体较小,向四周伸出突起。移植后2周,细胞向附近的脑组织迁移,部分细胞迁移到相对较远的地方,部分细胞与周围组织整合,Nestin阳性细胞数明显减少。移植后3周、4周,看不到Nestin阳性细胞;免疫荧光双标染色显示,移植的针道附近以及基底前脑散在有许多抗BrdU+抗NF和抗BrdU+抗GFAP免疫荧光双标阳性细胞。
     第三章、神经生长因子缓释微球、神经干细胞联合移植治疗AD模型鼠
     一、材料和方法
     青年健康SD雄性大鼠,体重250-310克,共分为3组:①正常对照组8只;②FF损伤组即模型组8只;③神经生长因子缓释微球治疗组12只;④NSC移植治疗组12只;⑤神经生长因子缓释微球+NSC移植治疗组12只。单侧切断FF,以toFF通路损伤的AD模型。动物模型的建立后,即刻行同侧神经干细胞移植。接着进行同侧神经生长因子缓释微球注射。对内侧隔核、斜角带核的胆碱能神经元进行计数和形态学参数的测定。并用SPSS统计软件包进行统计学处理。
     二、结果
     1、神经生长因子缓释微球、神经干细胞联合移植治疗对AD模型鼠基底前脑胆碱能神经元的影响
     穹窿-海马伞切断4周后,损伤组损伤侧的MS和VDB的胆碱能阳性神经元都大量减少。神经生长因子缓释微球治疗组损伤侧的胆碱能神经元得到明显的保护,明显高于损伤组损伤侧的胆碱能神经元存活数(p<0.01),与正常组相比,低于正常组(p<0.05)。NSC移植组损伤侧的胆碱能神经元得到补充和保护,明显高于损伤组损伤侧的胆碱能神经元存活数(p<0.01),与正常组相比,低于正常组(p<0.05)。神经生长因子缓释微球、NSC移植联合治疗组损伤侧的胆碱能神经元得到最为明显的保护和补充,明显高于神经生长因子缓释微球组或者NSC移植组损伤侧的胆碱能神经元存活数(p<0.01),基本达到正常组水平(P>0.05)。
     2、神经生长因子缓释微球、神经干细胞联合移植治疗对AD模型鼠空间学习记忆能力的影响
     Y型迷宫测试5组大鼠空间学习记忆能力,结果显示损伤组大鼠空间学习能力明显下降(P<0.05),记忆能力显著下降(P<0.01);神经生长因子缓释微球治疗组空间学习能力得到改善(P<0.05),记忆能力得到显著提高(P<0.01);NSC移植组空间学习能力得到改善(P<0.05),记忆能力得到显著提高(P<0.01);神经生长因子缓释微球、NSC移植联合治疗组空间学习能力得到显著改善(P<0.05),记忆能力得到显著提高(P<0.01);与正常组相比未见差异(P>0.05)。
     总之,本实验结果显示,采用水/油/水(W1/O/W2)的双乳化技术来制备神经生长因子缓释微球,蛋白/多聚物比率越高,载药量也越高,蛋白包封率则越低。乳化前将水溶性添加剂加入内水相能够明显提高蛋白的包封率。神经生长因子缓释微球能够持续释放有生物学活性的神经生长因子达35天,且初时爆破释放较低。体内注射结果显示神经生长因子缓释微球能够在基底前脑持续释放神经生长因子达4周以上。神经干细胞能够利用无血清的培养基在EGF和FGF-2刺激下从基底前脑组织中培养获得,并且能够在宿主脑内存活、迁移和分化,较好地与宿主脑组织整合。单侧切断FF后,基底前脑MS、VDB的胆碱能阳性神经元大量丢失。MS、VDB的胆碱能神经元的丢失与AD模型大鼠的空间学习记忆障碍密切相关。神经生长因子缓释微球和神经干细胞单独或者联合应用能够对AD模型鼠基底前脑胆碱能神经元有明显的补充和保护作用;联合应用的疗效明显优于单独应用。神经生长因子缓释微球和神经干细胞单独或者联合应用能够显著提高AD模型鼠的空间学习记忆能力;联合应用疗效更佳。
Alzheimer's disease (AD) is an irreversible, progressive disorder due to brain cells (neurons) deterioration, resulting in cognitive impairment, such as abnormal primarily memory, judgment and reasoning, movement coordination, and pattern recognition. A lot of investigations have showed that the prevalence of AD is strongly associated with aging. As the life span of human beings increases and more people live beyond the age of 65, the number of people with AD is also progressively increased. Contrasting with the enormous toll that this disease puts on patient, his or her caregivers, society as a whole is the lack of an effective therapy at present.
     One of the neuropathological characteristics of AD is atrophy or loss of cholinergic neuron observed in the basal forebrain (BF). Neurotrophic factors are a good strategy to prevent or reduce the neuronal atrophy or loss. Among the neurotrophic factor family, nerve growth factor (NGF) is the most highly characterized neurotrophic factor for peripheral sympathetic neurons and a subpopulation of sensory neurons. The cholinergic neurons of BF express both the low affinity receptor (P75~(NTF)) and TrkA receptor, and respond to NGF by increased activity levels of the choline acetyltransferase (CHAT). NGF is widely used for therapeutic studies in the experimental models of AD. However, NGF is a large molecular protein that does not easily cross the blood-brain barrier and has a short biologic half-life. Although infusing NGF solution into the cerebroventricular space via osmotic minipumps showed some therapeutic effects, long-term ICV NGF administration may cause negative side effects. The delivery of NGF to the BF poses a major challenge. New approach for drug formulation is required. One possibility is using controlled release formulations for the targeted delivery of NGF to brain.
     Neural stem cells have been successfully cultured, which solved the clinical problem of human fetal donors. Neural stem cells can generate neurons, astroglia and oligodendroglia in response to environmental signals, such as neurotrophic factors, retinoic acid and growth factors. The survival and differentiation of neural stem cells are associated with the inducement of the neurotrophic factors in vitro and in vivo. In central nervous system (CNS), the survival, migration and differentiation of neural stem cells is not lack of the microenvironment of the brain. Neural stem cells can differentiate specific neurons of specific areas of the brain under the inducement of the microenvironment.
     In this study, we treated the rat model of AD by combining rhNGF microspheres with neural stem cells. On the one hand, neural stem cells will differentiate into neurons, which will supplie the degenerative neurons. On the other hand, the rhNGF released from the rhNGF microspheres will promote an/or ameliorate the survival and function of degenerative neurons. Furthermore, the released rhNGF from the microspheres will supply the nourishment for the survival, migration and differentiation of neural stem cells.
     The following three parts were included in this study:
     1. The preparation and evaluation of rhNGF microspheres. Microspheres, containing rhNGF and BSA, were prepared by (water-inooil)-in-water (W/O/W) emulsion and solvent evaporation technique with some modification. Briefly, 5 mg protein mixture of rhNGF and BSA (1/2000, w/w) in 100μl distilled water was emulsified in Poly (D, L-lactic-co-glycolic acid) (PLGA) (100mg) solution (3 ml of methylene dichloride and 1 ml of acetone) using sonication for lmin at 30 W over ice-bath. The first w/o emulsion was added to 25 ml of 1% PVA aqueous solution and homogenized at 1000rpm for 5 min over ice-bath. The resulting w/o/w double emulsion was stirred in a hood for 3-4 h to evaporate the organic solution at room temperature. The microspheres were collected by centrifugation, washed three times with distilled water and freeze-dried to obtain a free flowing powder. The rhNGF and FITC-BSA (1/2000, w/w) microspheres were prepared in the same way for in vivo studies. The following variables were designed to assess the effect of technical parameters on particle size, protein loading, and encapsulation efficiency. (1) Microspheres were prepared with a series of protein/PLGA ratios of 5%, 10% and 15% respectively. (2) Microspheres were prepared by adding different additives such as sucrose, PEG, and glycerol in the inner aqueous phase at concentrations of 5 and 10% (w/w) respectively. The results showed rhNGF in PLGA microspheres provided a sustained release formulation with low initial burst (11.4 %) for at least 35 days in vitro. The results showed that the higher the protein/polymer ratio, the higher the protein loading into the microspheres, and the lower the efficiency of protein encapsulation in the microspheres. The encapsulation efficiency could be increased with adding water-soluble additives in the inner aqueous layer prior to the emulsification. The efficiency of protein encapsulation was about 89.7 % without using additive and increased to 97.5 % with the using of PEG. The microencapsulation technique allowed an entrapment of biologically active rhNGF. This is the first report so far of rhNGF-loaded microspheres implanted into BF. The biodegradable rhNGF-loaded microspheres maintained a sustained release of rhNGF for at least 4 weeks in brain tissue.
     2. The migration and differentiation of neural stem cells in the basal forebrain of the rat model of AD. The single cell suspension from brain tissues of the basal brain of new-born (less than 1 day) rats by gentle mechanical dissociation with the use of trypsin were cultured in DMEM/F12 medium containing 2% B27, EGF and FGF-2 (10 ng/ml). The cell were maintained for 5-7 day at 37℃in 5% CO_2. After 5-7 days, neurospheres were generated and floated in the culture. Unilateral fimbria-fornix (FF) of SD rats was transected to simulate the impairment of cholineric neurons of AD by the lesion of the pathway of septohippocampus. Neural stem cells (25,000 cells/μl×4μl) were injected into rats brain: AP+0.6 ram, LL+0.6 ram, DV-5.5 mm。The results showed neural stem cells could divide and proliferate under the inducement of EGF and FGF-2. After 5-7 d in vitro, large self-renewing and expandable spheres were generated. The proliferating and multipotential differentiation properties of neural stem cells were identified by immunochemistry. BrdU labeling and immunochemistry test were used to confirm the proliferation potential. In vivo studies, after 1 week, most of grafted cells stayed in the needle pathway, which survived well in the host brain, expressing Nestin antigen. After 2 weeks, most of grafted cells migrated neighbouring brain tissue, less of grafted cells expressing Nestin antigen. After 3 or 4 weeks, non-grafted cells express Nestin antigen, most of grafted cells migrated into whole basal forebrain and showed BrdU+NF or BrdU+GFAP.
     3. The therapeutic effect of combining rhNGF microspherewith neural stem cells on rat AD model. Adult male SD rats (12-20 weeks old, 250-310g) were used in this study. The rats were divided into five groups:①Normal control groups rats (Normal group, n=8);②FF-lesion groups rats (LES group, n=8);③RhNGF microspheres group rats (MIC group, n=12);④Neural stem cells group rats (NSC group, n=12);⑤The combination of rhNGF microspheres and neural stem cells group rats (MIC+NSC group, n=12). Neural stem cells (25,000 cells/μl×4μl) were injected into rats brain: AP+0.6mm, LL+0.6mm, DV-5.5mm。Three milligrams of rhNGF microspheres (suspended in 10μl of dispersing medium) were stereotaxically implanted into the BF (coordinates: AP +0.6 mm, LL +0.6 mm, DV-5.5 mm from bregma). The rats survived for four week after FF-lesion, discrimination learning and memory in Y-maze were observed. The morphologic data and percentages of cholinergic neurons of medial septum (MS) and vertical diagonal branch (VDB) were analyzed statistically by SPSS. The results showed after FF lesion, the percentages of cholinergic neurons at the lesion side to the intact side of MS and VDB were significantly lost. In MIC group, the percentages of cholinergic neurons at the lesion side to the intact side of MS and VDB were protected, which were significantly higher than that in the lesion group (p<0.01). In NSC group, the percentages of cholinergic neurons at the lesion side to the intact side of MS and VDB were protected, which were significantly higher than that in the lesion group (p<0.01). In MIC+NSC group, the percentages of cholinergic neurons at the lesion side to the intact side of MS and VDB were protected, which were significantly higher than that in MIC or NSC group (p<0.01). In the behavioral changes, the learning and memory of rats were significantly improved in the MIC+NSC group.
     Summary: The rhNGF-containing PLGA microspheres were preparaed by a W/O/W emulsion and solvent evaporation technique with some modifications. Using higher protein/polymer ratios in primary emulsions resulted in higher protein contents in the microspheres. The encapsulation efficiency could be increased with adding water-soluble additives in the inner aqueous layer prior to the emulsification. The in vitro rhNGF release lasted for more than 5 weeks. The rhNGF- microspheres maintained a sustained release of rhNGF for at least 4 weeks in brain. Neural stem cells can be obtained from basal forebrain by using free-serum medium adding EGF and FGF-2 (10 ng/ml). After FF lesion the percentages of cholinergic neurons at the lesion side to the intact side of MS and VDB were significantly lost. The loss of cholinergic neurons was associated with the impairment of learning and memory of the rat AD model. The combinative therapy of rhNGF microspheres and neural stem cells could significantly ameliorate, rescue and supply the degenerative neurons of the basal forebrain, and significantly improve the spatial learning of the rat AD model.
引文
[1] [History and syndromes of the disease, named after Alois Alzheimer]. Pflege Z, 1999, 52:685-686.
    [2] Small, DH and Cappai, R. Alois Alzheimer and Alzheimer's disease: a centennial perspective. J Neurochem, 2006, 99:708-710.
    [3] Braak, E, Griffing, K, Arai, K, et al. Neuropathology of Alzheimer's disease: what is new since A. Alzheimer? Eur Arch Psychiatry Clin Neurosci, 1999,249 Suppl 3:14-22.
    [4] Hansen, LA. The neuropathology of aging, mild cognitive impairment, and Alzheimer disease. Arch Neurol, 2006, 63:647-648.
    [5] Wilkins, CH, Grant, EA, Schmitt, SE, et al. The neuropathology of Alzheimer disease in African American and white individuals. Arch Neurol, 2006,63:87-90.
    [6] The neuropathology of Alzheimer disease. Proceedings of a workshop presented by the John Douglas French Foundation for Alzheimer's Disease. November 6-7, 1986. Abstracts. Bull Clin Neurosci, 1987, 52:1-33.
    [7] Cork, LC. Neuropathology of Down syndrome and Alzheimer disease. Am J Med Genet Suppl, 1990, 7:282-286.
    [8] Wolf, DS, Gearing, M, Snowdon, DA, et al. Progression of regional neuropathology in Alzheimer disease and normal elderly: findings from the Nun study. Alzheimer Dis Assoc Disord, 1999, 13:226-231.
    [9] Cras, P, Kawai, M, Lowery, D, et al. Senile plaque neurites in Alzheimer disease accumulate amyloid precursor protein. Proc Natl Acad Sci U S A, 1991, 88:7552-7556.
    [10] Ohyagi, Y. [Mechanism of senile plaque formation in Alzheimer disease]. Fukuoka Igaku Zasshi, 1999, 90:113-117.
    [11] Perry, G, Friedman, R, Shaw, G, et al. Ubiquitin is detected in neurofibrillary tangles and senile plaque neurites of Alzheimer disease brains. Proc Natl Acad Sci U S A, 1987, 84:3033-3036.
    [12] Ujiie, M, Dickstein, DL, Carlow, DA, et al. Blood-brain barrier permeability precedes senile plaque formation in an Alzheimer disease model. Microcirculation, 2003, 10:463-470.
    [13] Baum, L, Hansen, L, Masliah, E, et al. Glycogen synthase kinase 3 alteration in Alzheimer disease is related to neurofibrillary tangle formation. Mol Chem Neuropathol, 1996, 29:253-261.
    [14] Farber, NB, Rubin, EH, Newcomer, JW, et al. Increased neocortical neurofibrillary tangle density in subjects with Alzheimer disease and psychosis. Arch Gen Psychiatry, 2000, 57:1165-1173.
    [15] Jellinger, KA and Attems, J. Neurofibrillary tangle-predominant dementia: comparison with classical Alzheimer disease. Acta Neuropathol (Berl), 2006.
    [16] Sheng, JG, Mrak, RE and Griffin, WS. Progressive neuronal DNA damage associated with neurofibrillary tangle formation in Alzheimer disease. J Neuropathol Exp Neurol, 1998, 57:323-328.
    [17] Burns, JM, Galvin, JE, Roe, CM, et al. The pathology of the substantia nigra in Alzheimer disease with extrapyramidal signs. Neurology, 2005, 64:1397-1403.
    [18] Lyness, SA, Zarow, C and Chui, HC. Neuron loss in key cholinergic and aminergic nuclei in Alzheimer disease: a meta-analysis. Neurobiol Aging, 2003, 24:1-23.
    [19] Roberson, ED, Hesse, JH, Rose, KD, et al. Frontotemporal dementia progresses to death faster than Alzheimer disease. Neurology, 2005, 65:719-725.
    [20] Stadelmann, C, Bruck, W, Bancher, C, et al. Alzheimer disease: DNA fragmentation indicates increased neuronal vulnerability, but not apoptosis. J Neuropathol Exp Neurol, 1998, 57:456-464.
    [21] Miller, MA. Regulation of galanin in memory pathways. Ann N Y Acad Sci, 1998, 863:323-341.
    [22] Alenda, A and Nunez, A. Sensory-interference in rat primary somatosensory cortical neurons. Eur J Neurosci, 2004, 19:766-770.
    [23] Mufson, EJ, Ginsberg, SD, Ikonomovic, MD, et al. Human cholinergic basal forebrain: chemoanatomy and neurologic dysfunction. J Chem Neuroanat, 2003, 26:233-242.
    [24] Dreyfus, CF, Bernd, P, Martinez, HJ, et al. GABAergic and cholinergic neurons exhibit high-affinity nerve growth factor binding in rat basal forebrain. Exp Neurol, 1989, 104:181-185.
    [25] Gao, B, Hornung, JP and Fritschy, JM. Identification of distinct GAB AA-receptor subtypes in cholinergic and parvalbumin-positive neurons of the rat and marmoset medial septum-diagonal band complex. Neuroscience, 1995,65:101-117.
    [26] Ingham, CA, Bolam, JP and Smith, AD. GABA-immunoreactive synaptic boutons in the rat basal forebrain: comparison of neurons that project to the neocortex with pallidosubthalamic neurons. J Comp Neurol, 1988,273:263-282.
    [27] Roth, KA. Caspases, apoptosis, and Alzheimer disease: causation, correlation, and confusion. J Neuropathol Exp Neurol, 2001, 60:829-838.
    [28] Macaya, A. [Apoptosis in the nervous system]. Rev Neurol, 1996, 24:1356-1360.
    [29] Athan, ES, Lee, JH, Arriaga, A, et al. Polymorphisms in the promoter of the human APP gene: functional evaluation and allele frequencies in Alzheimer disease. Arch Neurol, 2002, 59:1793-1799.
    
    [30] Ghetti, B, Murrell, J, Benson, MD, et al. Spectrum of amyloid beta-protein immunoreactivity in hereditary Alzheimer disease with a guanine to thymine missense change at position 1924 of the APP gene. Brain Res, 1992, 571:133-139.
    
    [31] Li, Y, Hollingworth, P, Moore, P, et al. Genetic association of the APP binding protein 2 gene (APBB2) with late onset Alzheimer disease. Hum Mutat, 2005, 25:270-277.
    
    [32] Nishiwaki, Y, Kamino, K, Yoshiiwa, A, et al. Mutational screening of APP gene in patients with early-onset Alzheimer disease utilizing mismatched PCR-RFLP. Clin Genet, 1996,49:119-123.
    
    [33] Wong, L, Liang, Y, Jiang, L, et al. Mutation of the gene for the human lysosomal serine protease cathepsin G is not the cause of aberrant APP processing in familial Alzheimer disease. Neurosci Lett, 1993, 152:96-98.
    
    [34] Brooks, WS, Kwok, JB, Kril, JJ, et al. Alzheimer's disease with spastic paraparesis and 'cotton wool' plaques: two pedigrees with PS-1 exon 9 deletions. Brain, 2003, 126:783-791.
    
    [35] Ikeda, K, Urakami, K, Isoe, K, et al. High concentrations of PS-1 mRNA in skin fibroblasts of patients with Down's syndrome. J Neurol Neurosurg Psychiatry, 1998, 64:690-691.
    
    [36] Prihar, G, Verkkoniem, A, Perez-Tur, J, et al. Alzheimer disease PS-1 exon 9 deletion defined. Nat Med, 1999,5:1090.
    
    [37] Sandbrink, R, Zhang, D, Schaeffer, S, et al. Missense mutations of the PS-1/S182 gene in German early-onset Alzheimer's disease patients. Ann Neurol, 1996,40:265-266.
    
    [38] Takano, T, Sahara, N, Yamanouchi, Y, et al. Assignment of Alzheimer's presenilin-2 (PS-2) gene to 1q42.1 by fluorescence in situ hybridization. Neurosci Lett, 1997, 221:205-207.
    
    [39] Takano, T, Yamanouchi, Y, Sahara, N, et al. Assignment of Alzheimer's presenilin-1 (PS-1) gene to 14q24.3 by fluorescence in situ hybridization. Neurosci Lett, 1996, 214:69-71.
    
    [40] Danik, M, Champagne, D, Petit-Turcotte, C, et al. Brain lipoprotein metabolism and its relation to neurodegenerative disease. Crit Rev Neurobiol, 1999, 13:357-407.
    
    [41] Baum, L, Chen, L, Masliah, E, et al. Lipoprotein lipase mutations and Alzheimer's disease. Am J Med Genet, 1999, 88:136-139.
    
    [42] Beffert, U, Arguin, C and Poirier, J. The polymorphism in exon 3 of the low density lipoprotein receptor-related protein gene is weakly associated with Alzheimer's disease. Neurosci Lett, 1999, 259:29-32.
    [43] D'Introno, A, Solfrizzi, V, Colacicco, AM, et al. Polymorphisms in the oxidized low-density lipoprotein receptor-1 gene and risk of Alzheimer's disease. J Gerontol A Biol Sci Med Sci, 2005, 60:280-284.
    [44] Gafvels, ME, Caird, M, Britt, D, et al. Cloning of a cDNA encoding a putative human very low density lipoprotein/apolipoprotein E receptor and assignment of the gene to chromosome 9pter-p23. Somat Cell Mol Genet, 1993, 19:557-569.
    [45] Kamboh, MI, Ferrell, RE and DeKosky, ST. Genetic association studies between Alzheimer's disease and two polymorphisms in the low density lipoprotein receptor-related protein gene. Neurosci Lett, 1998, 244:65-68.
    [46] Kang, DE, Saitoh, T, Chen, X, et al. Genetic association of the low-density lipoprotein receptor-related protein gene (LRP), an apolipoprotein E receptor, with late-onset Alzheimer's disease. Neurology, 1997, 49:56-61.
    [47] Shihabuddin, LS, Palmer, TD and Gage, FH. The search for neural progenitor cells: prospects for thetherapy of neurodegenerative disease. Mol Med Today, 1999, 5:474-480.
    [48] Wehr, H, Parnowski, T, Puzynski, S, et al. Apolipoprotein E genotype and lipid and lipoprotein levels in dementia. Dement Geriatr Cogn Disord, 2000, 11:70-73.
    [49] Russ, C, Powell, JF, Zhao, J, et al. The microtubule associated protein Tau gene and Alzheimer'sdisease-an association study and meta-analysis. Neurosci Lett, 2001, 314:92-96.
    [50] Strittmatter, WJ, Saunders, AM, Goedert, M, et al. Isoform-specific interactions of apolipoprotein E with microtubule-associated protein tau: implications for Alzheimer disease. Proc Natl Acad Sci U S A, 1994, 91:11183-11186.
    [51] Vitali, A, Piccini, A, Borghi, R, et al. Soluble amyloid beta-protein is increased in frontotemporal dementia with tau gene mutations. J Alzheimers Dis, 2004, 6:45-51.
    [52] Dickson, DW and Rogers, J. Neuroimmunology of Alzheimer's disease: a conference report. Neurobiol Aging, 1992, 13:793-798.
    [53] Marcus, C. UBC researcher tracks cause of Alzheimer's dementia. Can Med Assoc J, 1985, 132:65-67.
    [54] Gahtan, E and Overmier, JB. Inflammatory pathogenesis in Alzheimer's disease: biological mechanisms and cognitive sequeli. Neurosci Biobehav Rev, 1999, 23:615-633.
    [55] Novak, M. [Neuroimmunology of Alzheimer's disease]. Bratisl Lek Listy, 1997, 98:303-314.
    [56] Arispe, N, Pollard, HB and Rojas, E. beta-Amyloid Ca(2+)-channel hypothesis for neuronal death in Alzheimer disease. Mol Cell Biochem, 1994, 140:119-125.
    
    [57] Branconnier, RJ, Branconnier, ME, Walshe, TM, et al. Blocking the Ca(2+)-activated cytotoxic mechanisms of cholinergic neuronal death: a novel treatment strategy for Alzheimer's disease. Psychopharmacol Bull, 1992,28:175-181.
    
    [58] Ito, E, Oka, K, Etcheberrigaray, R, et al. Internal Ca2+ mobilization is altered in fibroblasts from patients with Alzheimer disease. Proc Natl Acad Sci U S A, 1994, 91:534-538.
    
    [59] Mori, Y, Inagaki, C, Kuno, M, et al. [Ionic mechanisms underlying the regulation of cell proliferation, differentiation and death]. Nippon Yakurigaku Zasshi, 2003, 122:201-214.
    
    [60] Nieoullon, A. [Cellular bases of neurodegenerative processes]. Therapie, 1998, 53:21-29.
    
    [61] Sheng, JG, Mrak, RE, Jones, RA, et al. Neuronal DNA damage correlates with overexpression of interleukin-1 beta converting enzyme in APPV717F mice. Neurobiol Aging, 2001, 22:895-902.
    
    [62] Greenamyre, JT and Maragos, WF. Neurotransmitter receptors in Alzheimer disease. Cerebrovasc Brain Metab Rev, 1993,5:61-94.
    
    [63] Mann, DM, Yates, PO and Marcyniuk, B. Monoaminergic neurotransmitter systems in presenile Alzheimer's disease and in senile dementia of Alzheimer type. Clin Neuropathol, 1984, 3:199-205.
    
    [64] Whitehouse, PJ. Neurotransmitter receptor alterations in Alzheimer disease: a review. Alzheimer Dis Assoc Disord, 1987, 1:9-18.
    
    [65] Zhong, ZG, Liu, MC and Lai, SL. [Effect of bushen yizhi recipe on neurotransmitter release in A beta segment neurotoxin induced NG108-15 cellular model of Alzheimer disease]. Zhongguo Zhong Xi Yi Jie He ZaZhi, 2002, 22:50-53.
    
    [66] Pailler, FM, Bequet, D, Corbe, H, et al. [Aluminum, hypothetic cause of Alzheimer disease]. Presse Med, 1995,24:489-490.
    
    [67] Farrar, G, Altmann, P, Welch, S, et al. Defective gallium-transferrin binding in Alzheimer disease and Down syndrome: possible mechanism for accumulation of aluminium in brain. Lancet, 1990, 335:747-750.
    
    [68] Grundke-Iqbal, I and Iqbal, K. Neuronal cytoskeleton in the biology of Alzheimer disease. Prog Clin Biol Res, 1989,317:745-753.
    
    [69] Perry, G, Rizzuto, N, Autilio-Gambetti, L, et al. Paired helical filaments from Alzheimer disease patients contain cytoskeletal components. Proc Natl Acad Sci U S A, 1985, 82:3916-3920.
    [70] Samuel, W, Crowder, R, Hofstetter, CR, et al. Neuritic plaques in the Lewy body variant of Alzheimer disease lack paired helical filaments. Neurosci Lett, 1997, 223:73-76.
    
    [71] Sparkman, DR, Goux, WJ, Jones, CM, et al. Alzheimer disease paired helical filament core structures contain glycolipid. Biochem Biophys Res Commun, 1991, 181:771-779.
    
    [72] Vincent, IJ and Davies, P. A protein kinase associated with paired helical filaments in Alzheimer disease. Proc Natl Acad Sci U S A, 1992, 89:2878-2882.
    
    [73] Skausig, OB. [Drug therapy of Alzheimer disease]. Ugeskr Laeger, 2004, 166:4497-4498; author reply 4498.
    
    [74] Skolnick, AA. Old Chinese herbal medicine used for fever yields possible new Alzheimer disease therapy. Jama, 1997,277:776.
    
    [75] Bennett, DA and Holtzman, DM. Immunization therapy for Alzheimer disease? Neurology, 2005,64:10-12.
    
    [76] Oken, RJ, Wisniewski, HM, Kieras, FJ, et al. Alzheimer disease (AD): aspirin prophylaxis and therapy. Alzheimer Dis Assoc Disord, 1992, 6:53-54.
    
    [77] Owens, CT. Estrogen replacement therapy for Alzheimer disease in postmenopausal women. Ann Pharmacother, 2002, 36:1273-1276.
    
    [78] Zandi, PP, Carlson, MC, Plassman, BL, et al. Hormone replacement therapy and incidence of Alzheimer disease in older women: the Cache County Study. Jama, 2002, 288:2123-2129.
    
    [79] Tuszynski, MH, U, HS, Alksne, J, et al. Growth factor gene therapy for Alzheimer disease. Neurosurg Focus, 2002, 13:e5.
    
    [80] Stromberg, I, Wetmore, CJ, Ebendal, T, et al. Rescue of basal forebrain cholinergic neurons after implantation of genetically modified cells producing recombinant NGF. J Neurosci Res, 1990, 25:405-411.
    
    [81] Balse, E, Lazarus, C, Kelche, C, et al. Intrahippocampal grafts containing cholinergic and serotonergic fetal neurons ameliorate spatial reference but not working memory in rats with fimbria-fornix/cingular bundle lesions. Brain Res Bull, 1999,49:263-272.
    
    [82] Hartig, W, Bauer, A, Brauer, K, et al. Functional recovery of cholinergic basal forebrain neurons under disease conditions: old problems, new solutions? Rev Neurosci, 2002, 13:95-165.
    
    [83] Segal, M, Bjorklund, A and Gage, FH. Transplanted septal neurons make viable cholinergic synapses with a host hippocampus. Brain Res, 1985, 336:302-307.
    [84] Tarricone, BJ, Simon, JR, Li, YJ, et al. Neural grafting of cholinergic neurons in the hippocampal formation. Behav Brain Res, 1996, 74:25-44.
    [85] Brodski, C, Schnurch, H and Dechant, G. Neurotrophin-3 promotes the cholinergic differentiation of sympathetic neurons. Proc Natl Acad Sci U S A, 2000, 97:9683-9688.
    [86] Nonner, D, Barrett, EF and Barrett, IN. Neurotrophin effects on survival and expression of cholinergic properties in cultured rat septal neurons under normal and stress conditions. J Neurosci, 1996, 16:6665-6675.
    [87] Nonner, D, Barrett, EF, Kaplan, P, et al. Bone morphogenetic proteins (BMP6 and BMP7) enhance theprotective effect of neurotrophins on cultured septal cholinergic neurons during hypoglycemia. J Neurochem , 2001, 77:691-699.
    [88] Aloe, L. Rita Levi-Montalcini and the discovery of nerve growth factor: past and present studies. Arch ItalBiol, 2003, 141:65-83.
    [89] Calissano, P, Levi, A, Alema, S, et al. Proceedings: Studies on the interaction of nerve growth factor with tubulin and actin. Hoppe Seylers Z Physiol Chem, 1975, 356:382-383.
    [90] Hamburger, V. The history of the discovery of the nerve growth factor. J Neurobiol, 1993, 24:893-897.
    [91] Levi, A and Alema, S. The mechanism of action of nerve growth factor. Annu Rev Pharmacol Toxicol, 1991,31:205-228.
    [92] Connor, B, Young, D, Yan, Q, et al. Brain-derived neurotrophic factor is reduced in Alzheimer's disease. Brain Res Mol Brain Res, 1997,49:71-81.
    [93] Laske, C, Stransky, E, Leyhe, T, et al. Decreased brain-derived neurotrophic factor (BDNF)- and beta-thromboglobulin (beta-TG)- blood levels in Alzheimer's disease. Thromb Haemost, 2006, 96:102-103.
    [94] Pezet, S and Malcangio, M. Brain-derived neurotrophic factor as a drug target for CNS disorders. Expert Opin Ther Targets, 2004, 8:391-399.
    [95] Mrzljak, L and Goldman-Rakic, PS. Low-affinity nerve growth factor receptor (p75NGFR)- and choline acetyltransferase (ChAT)-immunoreactive axons in the cerebral cortex and hippocampus of adult macaque monkeys and humans. Cereb Cortex, 1993, 3:133-147.
    [96] Hefti, F, Hartikka, J, Salvatierra, A, et al. Localization of nerve growth factor receptors in cholinergic neurons of the human basal forebrain. Neurosci Lett, 1986, 69:37-41.
    [97] Mufson, EJ, Ma, SY, Dills, J, et al. Loss of basal forebrain P75(NTR) immunoreactivity in subjects with mild cognitive impairment and Alzheimer's disease. J Comp Neurol, 2002,443:136-153.
    [98] Hagg, T, Fass-Holmes, B, Vahlsing, HL, et al. Nerve growth factor (NGF) reverses axotomy-induced decreases in choline acetyltransferase, NGF receptor and size of medial septum cholinergic neurons. Brain Res, 1989,505:29-38.
    [99] Will, B and Hefti, F. Behavioural and neurochemical effects of chronic intraventricular injections of nerve growth factor in adult rats with fimbria lesions. Behav Brain Res, 1985, 17:17-24.
    [100] Vinores, S and Guroff, G. Nerve growth factor: mechanism of action. Annu Rev Biophys Bioeng, 1980, 9:223-257.
    [101] Otto, D, Frotscher, M and Unsicker, K. Basic fibroblast growth factor and nerve growth factor administered in gel foam rescue medial septal neurons after fimbria fornix transection. J Neurosci Res, 1989, 22:83-91.
    [102] Grothe, C, Otto, D and Unsicker, K. Basic fibroblast growth factor promotes in vitro survival and cholinergic development of rat septal neurons: comparison with the effects of nerve growth factor. Neuroscience, 1989,31:649-661.
    [103] Powell, EM, Sobarzo, MR and Saltzman, WM. Controlled release of nerve growth factor from a polymeric implant. Brain Res, 1990, 515:309-311.
    [104] Houweling, DA, Lankhorst, AJ, Gispen, WH, et al. Collagen containing neurotrophin-3 (NT-3) attracts regrowing injured corticospinal axons in the adult rat spinal cord and promotes partial functional recovery. ExpNeurol, 1998, 153:49-59.
    [105] Emerich, DF, Lindner, MD, Winn, SR, et al. Implants of encapsulated human CNTF-producing fibroblasts prevent behavioral deficits and striatal degeneration in a rodent model of Huntington's disease. J Neurosci, 1996, 16:5168-5181.
    [106] Winn, SR, Lindner, MD, Lee, A, et al. Polymer-encapsulated genetically modified cells continue to secrete human nerve growth factor for over one year in rat ventricles: behavioral and anatomical consequences. Exp Neurol, 1996, 140:126-138.
    [107] Maa, YF and Hsu, CC. Effect of primary emulsions on microsphere size and protein-loading in the double emulsion process. J Microencapsul, 1997, 14:225-241.
    [108] Schrier, JA and DeLuca, PP. Recombinant human bone morphogenetic protein-2 binding and incorporation in PLGA microsphere delivery systems. Pharm Dev Technol, 1999,4:611-621.
    [109] Tracy, MA. Development and scale-up of a microsphere protein delivery system. Biotechnol Prog, 1998, 14:108-115.
    
    [110] Woo, BH, Jiang, G, Jo, YW, et al. Preparation and characterization of a composite PLGA and poly(acryloyl hydroxyethyl starch) microsphere system for protein delivery. Pharm Res, 2001, 18:1600-1606.
    
    [111] Camarata, PJ, Suryanarayanan, R, Turner, DA, et al. Sustained release of nerve growth factor from biodegradable polymer microspheres. Neurosurgery, 1992, 30:313-319.
    
    [112] Maysinger, D, Jalsenjak, I and Cuello, AC. Microencapsulated nerve growth factor: effects on the forebrain neurons following devascularizing cortical lesions. Neurosci Lett, 1992, 140:71-74.
    
    [113] Maysinger, D, Filipovic-Grcic, J and Cuello, AC. Effects of coencapsulated NGF and GM1 in rats with cortical lesions. Neuroreport, 1993, 4:971-974.
    
    [114] Cao, X and Schoichet, MS. Delivering neuroactive molecules from biodegradable microspheres for application in central nervous system disorders. Biomaterials, 1999, 20:329-339.
    
    [115] Menei, P, Pean, JM, Nerriere-Daguin, V, et al. Intracerebral implantation of NGF-releasing biodegradable microspheres protects striatum against excitotoxic damage. Exp Neurol, 2000, 161:259-272.
    
    [116] Pean, JM, Menei, P, Morel, O, et al. Intraseptal implantation of NGF-releasing microspheres promote the survival of axotomized cholinergic neurons. Biomaterials, 2000, 21:2097-2101.
    
    [117] Maysinger, D, Krieglstein, K, Filipovic-Grcic, J, et al. Microencapsulated ciliary neurotrophic factor: physical properties and biological activities. Exp Neurol, 1996, 138:177-188.
    
    [118] Maysinger, D, Berezovskaya, O and Fedoroff, S. The hematopoietic cytokine colony stimulating factor 1 is also a growth factor in the CNS: (II). Microencapsulated CSF-1 and LM-10 cells as delivery systems. Exp Neurol, 1996, 141:47-56.
    
    [119] Gouhier, C, Chalon, S, Aubert-Pouessel, A, et al. Protection of dopaminergic nigrostriatal afferents by GDNF delivered by microspheres in a rodent model of Parkinson's disease. Synapse, 2002, 44:124-131.
    
    [120] Aubert-Pouessel, A, Venier-Julienne, MC, Clavreul, A, et al. In vitro study of GDNF release from biodegradable PLGA microspheres. J Control Release, 2004, 95:463-475.
    
    [121] Jollivet, C, Aubert-Pouessel, A, Clavreul, A, et al. Striatal implantation of GDNF releasing biodegradable microspheres promotes recovery of motor function in a partial model of Parkinson's disease. Biomaterials, 2004, 25:933-942.
    
    [122] Carrascosa, C, Torres-Aleman, I, Lopez-Lopez, C, et al. Microspheres containing insulin-like growth factor I for treatment of chronic neurodegeneration. Biomaterials, 2004,25:707-714.
    [123] Isacson, O, Deacon, TW, Pakzaban, P, et al. Transplanted xenogeneic neural cells in neurodegenerative disease models exhibit remarkable axonal target specificity and distinct growth patterns of glial and axonal fibres. Nat Med, 1995, 1:1189-1194.
    [124] Jacoby, DB, Lindberg, C, Ratliff, J, et al. Fetal pig neural cells as a restorative therapy for neurodegenerative disease. Artif Organs, 1997, 21:1192-1198.
    [125] Arnhold, S, Lenartz, D, Kruttwig, K, et al. Differentiation of green fluorescent protein-labeled embryonic stem cell-derived neural precursor cells into Thy-1-positive neurons and glia after transplantation into adult rat striatum. J Neurosurg, 2000,93:1026-1032.
    [126] Kimura, H, Yoshikawa, M, Matsuda, R, et al. Transplantation of embryonic stem cell-derived neural stem cells for spinal cord injury in adult mice. Neurol Res, 2005,27:812-819.
    [127] Nonaka, M, Yoshikawa, M, Nishimura, F, et al. Intraventricular transplantation of embryonic stem cell-derived neural stem cells in intracerebral hemorrhage rats. Neurol Res, 2004, 26:265-272.
    [128] Svendsen, CN and Caldwell, MA. Neural stem cells in the developing central nervous system: implications for cell therapy through transplantation. Prog Brain Res, 2000, 127:13-34.
    [129] Modo, M, Cash, D, Mellodew, K, et al. Tracking transplanted stem cell migration using bifunctional, contrast agent-enhanced, magnetic resonance imaging. Neuroimage, 2002, 17:803-811.
    [130] Modo, M, Rezaie, P, Heuschling, P, et al. Transplantation of neural stem cells in a rat model of stroke: assessment of short-term graft survival and acute host immunological response. Brain Res, 2002,958:70-82.
    [131] Ogawa, Y, Sawamoto, K, Miyata, T, et al. Transplantation of in vitro-expanded fetal neural progenitor cells results in neurogenesis and functional recovery after spinal cord contusion injury in adult rats. J Neurosci Res, 2002, 69:925-933.
    [132] Fukunaga, A, Kawase, T and Uchida, K. Functional recovery after simultaneous transplantation with neuro-epithelial stem cells and adjacent mesenchymal tissues into infarcted rat brain. Acta Neurochir (Wien), 2003, 145:473-480, discussion 480-471.
    [133] Fukunaga, A, Uchida, K, Hara, K, et al. Differentiation and angiogenesis of central nervous system stem cells implanted with mesenchyme into ischemic rat brain. Cell Transplant, 1999, 8:435-441.
    [134] Rubio, F, Kokaia, Z, Arco, A, et al. BDNF gene transfer to the mammalian brain using CNS-derived neural precursors. Gene Ther, 1999, 6:1851-1866.
    [135] Philips, MF, Mattiasson, G, Wieloch, T, et al. Neuroprotective and behavioral efficacy of nerve growth factor-transfected hippocampal progenitor cell transplants after experimental traumatic brain injury. J Neurosurg, 2001, 94:765-774.
    [136] Mayer, RJ, Landon, M, Laszlo, L, et al. Protein processing in lysosomes: the new therapeutic target in neurodegenerative disease. Lancet, 1992, 340:156-159.
    [137] Nemeth, H, Toldi, J and Vecsei, L. Kynurenines, Parkinson's disease and other neurodegenerative disorders: preclinical and clinical studies. J Neural Transm Suppl, 2006:285-304.
    [138] Ringheim, GE and Conant, K. Neurodegenerative disease and the neuroimmune axis (Alzheimer's and Parkinson's disease, and viral infections). J Neuroimmunol, 2004, 147:43-49.
    [139] Sattin, A, Muhoberac, BB, Aprison, MH, et al. Tetrahydroaminoacridine (THA) as a pharmacological probe in Alzheimer's disease (AD) and other neurodegenerative disorders. Med Hypotheses, 1989, 29:155-159.
    [140] Wishart, TM, Parson, SH and Gillingwater, TH. Synaptic vulnerability in neurodegenerative disease. J Neuropathol Exp Neurol, 2006, 65:733-739.
    [141] Riekkinen, PJ, Laulumaa, V, Sirvio, J, et al. Recent progress in the research of Alzheimer's disease. Med Biol, 1987, 65:83-88.
    [142] Nabeshima, T. [Nerve growth factor strategy and preparation of animal model for Alzheimer-type senile dementia]. Yakugaku Zasshi, 1995, 115:499-512.
    [143] Menei, P, Benoit, JP, Boisdron-Celle, M, et al. Drug targeting into the central nervous system by stereotactic implantation of biodegradable microspheres. Neurosurgery, 1994, 34:1058-1064; discussion 1064.
    [144] Mahoney, MJ and Saltzman, WM. Controlled release of proteins to tissue transplants for the treatment of neurodegenerative disorders. J Pharm Sci, 1996, 85:1276-1281.
    [145] Konturek, SJ, Pawlik, W, Mysh, W, et al. Comparison of organ uptake and disappearance half-time of human epidermal growth factor and insulin. Regul Pept, 1990, 30:137-148.
    [146] Zhu, G, Mallery, SR and Schwendeman, SP. Stabilization of proteins encapsulated in injectable poly (lactide- co-glycolide). Nat Biotechnol, 2000, 18:52-57.
    [147] Cohen, S, Yoshioka, T, Lucarelli, M, et al. Controlled delivery systems for proteins based on poly(lactic/glycolic acid) microspheres. Pharm Res, 1991, 8:713-720.
    [148] Menei, P, Montero-Menei, C, Venier, MC, et al. Drug delivery into the brain using poly(lactide-co-glycolide) microspheres. Expert Opin Drug Deliv, 2005, 2:363-376.
    [149] Jiang, W, Gupta, RK, Deshpande, MC, et al. Biodegradable poly(lactic-co-glycolic acid) microparticles for injectable delivery of vaccine antigens. Adv Drug Deliv Rev, 2005, 57:391-410.
    [150] Haller, MF and Saltzman, WM. Nerve growth factor delivery systems. J Control Release, 1998, 53:1-6.
    [151] Haller, MF and Saltzman, WM. Localized delivery of proteins in the brain: can transport be customized? Pharm Res, 1998, 15:377-385.
    [152] Ruan, G, Ng, JK and Feng, SS. Effects of polymer, organic solvent and mixing strength on integrity of proteins and liposomes encapsulated in polymeric microspheres fabricated by the double emulsion process. J Microencapsul, 2004, 21:399-412.
    [153] Obeidat, WM and Price, JC. Viscosity of polymer solution phase and other factors controlling the dissolution of theophylline microspheres prepared by the emulsion solvent evaporation method. J Microencapsul, 2003, 20:57-65.
    [154] Kang, F, Jiang, G, Hinderliter, A, et al. Lysozyme stability in primary emulsion for PLGA microsphere preparation: effect of recovery methods and stabilizing excipients. Pharm Res, 2002, 19:629-633.
    [155] Estey, T, Kang, J, Schwendeman, SP, et al. BSA degradation under acidic conditions: a model for protein instability during release from PLGA delivery systems. J Pharm Sci, 2006, 95:1626-1639.
    [156] van de Weert, M, Hennink, WE and Jiskoot, W. Protein instability in poly(lactic-co-glycolic acid) microparticles. Pharm Res, 2000, 17:1159-1167.
    [157] Carrascosa, C, Espejo, L, Torrado, S, et al. Effect of gamma-sterilization process on PLGA microspheres loaded with insulin-like growth factor-I (IGF-I). J Biomater Appl, 2003, 18:95-108.
    [158] Meinel, L, Illi, OE, Zapf, J, et al. Stabilizing insulin-like growth factor-I in poIy(D,L-lactide-co-glycolide) microspheres. J Control Release, 2001, 70:193-202.
    [159] Yuksel, E, Weinfeld, AB, Cleek, R, et al. Increased free fat-graft survival with the long-term, local delivery of insulin, insulin-like growth factor-I, and basic fibroblast growth factor by PLGA/PEG microspheres. Plast Reconstr Surg, 2000, 105:1712-1720.
    [160] Jeffery, H, Davis, SS and O'Hagan, DT. The preparation and characterization of poly(lactide-co-glycolide) microparticles. II. The entrapment of a model protein using a (water-in-oil)-in-water emulsion solvent evaporation technique. Pharm Res, 1993, 10:362-368.
    [161] Greene, LA and Shooter, EM. The nerve growth factor: biochemistry, synthesis, and mechanism of action. Annu Rev Neurosci, 1980, 3:353-402.
    [162] Kojima, M, Takahashi, N, Ikeuchi, T, et al. Nerve growth factor (NGF)-mediated up-regulation of low-affinity NGF receptor gene expression in cultured basal forebrain cholinergic neurons from postnatal 3-day-old rats. Brain Res Mol Brain Res, 1992, 16:267-273.
    [163] Batchelor, PE, Armstrong, DM, Blaker, SN, et al. Nerve growth factor receptor and choline acetyltransferase colocalization in neurons within the rat forebrain: response to fimbria-fornix transection. J Comp Neurol, 1989, 284:187-204.
    [164] Mufson, EJ, Bothwell, M, Hersh, LB, et al. Nerve growth factor receptor immunoreactive profiles in the normal, aged human basal forebrain: colocalization with cholinergic neurons. J Comp Neurol, 1989, 285:196-217.
    [165] Woolf, NJ, Gould, E and Butcher, LL. Nerve growth factor receptor is associated with cholinergic neurons of the basal forebrain but not the pontomesencephalon. Neuroscience, 1989, 30:143-152.
    [166] Hefti, F. Nerve growth factor promotes survival of septal cholinergic neurons after fimbrial transections. JNeurosci, 1986,6:2155-2162.
    [167] Krewson, CE, Dause, R, Mak, M, et al. Stabilization of nerve growth factor in controlled release polymers and in tissue. J Biomater Sci Polym Ed, 1996, 8:103-117.
    [168] Zhu, W, Mao, Y, Zhao, Y, et al. Transplantation of vascular endothelial growth factor-transfected neural stem cells into the rat brain provides neuroprotection after transient focal cerebral ischemia. Neurosurgery, 2005, 57:325-333; discussion 325-333.
    [169] Mark Saltzman, W and Baldwin, SP. Materials for protein delivery in tissue engineering. Adv Drug Deliv Rev, 1998,33:71-86.
    [170] Benoit, JP, Faisant, N, Venier-Julienne, MC, et al. Development of microspheres for neurological disorders: from basics to clinical applications. J Control Release, 2000,65:285-296.
    [171] Ogawa, Y, Okada, H, Yamamoto, M, et al. In vivo release profiles of leuprolide acetate from microcapsules prepared with polylactic acids or copoly(lactic/glycolic) acids and in vivo degradation of these polymers. Chem Pharm Bull (Tokyo), 1988, 36:2576-2581.
    [172] Gertz, HJ, Cervos-Navarro, J and Ewald, V. The septo-hippocampal pathway in patients suffering from senile dementia of Alzheimer's type. Evidence for neuronal plasticity? Neurosci Lett, 1987, 76:228-232.
    [173] Blanco-Prieto, MJ, Leo, E, Delie, F, et al. Study of the influence of several stabilizing agents on the entrapment and in vitro release of pBC 264 from poly(lactide-co-glycolide) microspheres prepared by a W/O/W solvent evaporation method. Pharm Res, 1996, 13:1127-1129.
    [174] Takada, S, Yamagata, Y, Misaki, M, et al. Sustained release of human growth hormone from microcapsules prepared by a solvent evaporation technique. J Control Release, 2003, 88:229-242.
    [175] Pean, JM, Boury, F, Venier-Julienne, MC, et al. Why does PEG 400 co-encapsulation improve NGF stability and release from PLGA biodegradable microspheres? Pharm Res, 1999, 16:1294-1299.
    [176] Sun, SW, Jeong, YI, Jung, SW, et al. Characterization of FITC-albumin encapsulated poly(DL-lactide-co-glycolide) microspheres and its release characteristics. J Microencapsul, 2003, 20:479-488.
    [177] Yang, YY, Chung, TS and Ng, NP. Morphology, drug distribution, and in vitro release profiles of biodegradable polymeric microspheres containing protein fabricated by double-emulsion solvent extraction/evaporation method. Biomaterials, 2001, 22:231-241.
    [178] Leo, E, Pecquet, S, Rojas, J, et al. Changing the pH of the external aqueous phase may modulate protein entrapment and delivery from poly(lactide-co-glycolide) microspheres prepared by a w/o/w solvent evaporation method. J Microencapsul, 1998,15:421-430.
    [179] Ogawa, Y, Yamamoto, M, Okada, H, et al. A new technique to efficiently entrap leuprolide acetate into microcapsules of polylactic acid or copoly(lactic/glycolic) acid. Chem Pharm Bull (Tokyo), 1988, 36:1095-1103.
    [180] Ogawa, Y, Yamamoto, M, Takada, S, et al. Controlled-release of leuprolide acetate from polylactic acid or copoly(lactic/glycolic) acid microcapsules: influence of molecular weight and copolymer ratio of polymer. Chem Pharm Bull (Tokyo), 1988, 36:1502-1507.
    [181] Bodmeier, R and McGinity, JW. The preparation and evaluation of drug-containing poly(dl-lactide) microspheres formed by the solvent evaporation method. Pharm Res, 1987, 4:465-471.
    [182] Perez, C, Castellanos, IJ, Costantino, HR, et al. Recent trends in stabilizing protein structure upon encapsulation and release from bioerodible polymers. J Pharm Pharmacol, 2002, 54:301-313.
    [183] Hoffman, D, Wahlberg, L and Aebischer, P. NGF released from a polymer matrix prevents loss of ChAT expression in basal forebrain neurons following a fimbria-fornix lesion. Exp Neurol, 1990, 110:39-44.
    [184] Reynolds, BA and Weiss, S. Generation of neurons and astrocytes from isolated cells of the adult mammalian central nervous system. Science, 1992, 255:1707-1710.
    [185] McKay, RD. Stem cell biology and neurodegenerative disease. Philos Trans R Soc Lond B Biol Sci, 2004,359:851-856.
    [186] Fukushima, N, Yokouchi, K, Kawagishi, K, et al. Differential neurogenesis and gliogenesis by local and migrating neural stem cells in the olfactory bulb. Neurosci Res, 2002,44:467-473.
    [187] Zhang, B, Wang, RZ, Yao, Y, et al. Proliferation and differentiation of neural stem cells in adult rats after cerebral infarction. Chin Med Sci J, 2004, 19:73-77.
    [188] Eriksson, PS, Perfilieva, E, Bjork-Eriksson, T, et al. Neurogenesis in the adult human hippocampus. Nat Med, 1998,4:1313-1317.
    [189] Suzuki, T, Ooto, S, Akagi, T, et al. Effects of prolonged delivery of brain-derived neurotrophic factor on the fate of neural stem cells transplanted into the developing rat retina. Biochem Biophys Res Commun, 2003, 309:843-847.
    [190] Calza, L, Giuliani, A, Fernandez, M, et al. Neural stem cells and cholinergic neurons: regulation by immunolesion and treatment with mitogens, retinoic acid, and nerve growth factor. Proc Natl Acad Sci USA, 2003, 100:7325-7330.
    [191] Pera, MF, Andrade, J, Houssami, S, et al. Regulation of human embryonic stem cell differentiation by BMP-2 and its antagonist noggin. J Cell Sci, 2004, 117:1269-1280.
    [192] Taupin, P and Gage, FH. Adult neurogenesis and neural stem cells of the central nervous system in mammals. J Neurosci Res, 2002, 69:745-749.
    [193] Gage, FH. Mammalian neural stem cells. Science, 2000, 287:1433-1438.
    [194] Gregg, CT, Shingo, T and Weiss, S. Neural stem cells of the mammalian forebrain. Symp Soc Exp Biol, 2001:1-19.
    [195] Merkle, FT and Alvarez-Buylla, A. Neural stem cells in mammalian development. Curr Opin Cell Biol, 2006, 18:704-709.
    [196] Morshead, CM, Reynolds, BA, Craig, CG, et al. Neural stem cells in the adult mammalian forebrain: a relatively quiescent subpopulation of subependymal cells. Neuron, 1994, 13:1071-1082.
    [197] Ciccolini, F and Svendsen, CN. Fibroblast growth factor 2 (FGF-2) promotes acquisition of epidermal growth factor (EGF) responsiveness in mouse striatal precursor cells: identification of neural precursors responding to both EGF and FGF-2. J Neurosci, 1998, 18:7869-7880.
    [198] Deleyrolle, L, Marchal-Victorion, S, Dromard, C, et al. Exogenous and fibroblast growth factor 2/epidermal growth factor-regulated endogenous cytokines regulate neural precursor cell growth and differentiation. Stem Cells, 2006, 24:748-762.
    [199] Kojima, A and Tator, CH. Epidermal growth factor and fibroblast growth factor 2 cause proliferation of ependymal precursor cells in the adult rat spinal cord in vivo. J Neuropathol Exp Neurol, 2000, 59:687-697.
    [200] Kuhn, HG, Winkler, J, Kempermann, G, et al. Epidermal growth factor and fibroblast growth factor-2 have different effects on neural progenitors in the adult rat brain. J Neurosci, 1997, 17:5820-5829.
    [201] Ohta, Y, Nagai, M, Nagata, T, et al. Intrathecal injection of epidermal growth factor and fibroblast growth factor 2 promotes proliferation of neural precursor cells in the spinal cords of mice with mutant human SOD1 gene. J Neurosci Res, 2006, 84:980-992.
    [202] Grirti, A, Frolichsthal-Schoeller, P, Galli, R, et al. Epidermal and fibroblast growth factors behave as mitogenic regulators for a single multipotent stem cell-like population from the subventricular region of the adult mouse forebrain. J Neurosci, 1999, 19:3287-3297.
    [203] Bick, MD and Devine, EA. Interaction of chromosomal proteins with BrdU substituted DNA as determined by chromatin-DNA competition. Nucleic Acids Res, 1977,4:3687-3700.
    [204] Kumar, G, Sharma, AK and Jayaraman, K. Incorporation of BrdU in a DNA fragment may affect protein-DNA interactions in a site-dependent manner. Oncogene, 1992, 7:1453-1455.
    [205] Pera, F and Mattias, P. Labelling of DNA and differential sister chromatid staining after BrdU treatment in vivo. Chromosoma, 1976, 57:13-18.
    [206] Souleil, C and Panijel, J. Effect of antigen dose on the presence of a particular BrdU-labelled DNA fraction in lymph node cells. Exp Cell Res, 1973, 78:462-466.
    [207] Lendahl, U, Zimmerman, LB and McKay, RD. CNS stem cells express a new class of intermediate filament protein. Cell, 1990, 60:585-595.
    [208] Barcellona, ML, Gammon, S, Hazlett, T, et al. Polarized fluorescence correlation spectroscopy of DNA-DAPI complexes. Microsc Res Tech, 2004,65:205-217.
    [209] De Castro, LF and Zacharias, M. DAPI binding to the DNA minor groove: a continuum solvent analysis. J Mol Recognit, 2002, 15:209-220.
    [210] Kamo, KK and Griesbach, RJ. Evaluation of DAPI as a fluorescent probe for DNA in viable Petunia protoplasts. Biotech Histochem, 1993, 68:350-359.
    [211] Kapuscinski, J. DAPI: a DNA-specific fluorescent probe. Biotech Histochem, 1995, 70:220-233.
    [212] Larsen, TA, Goodsell, DS, Cascio, D, et al. The structure of DAPI bound to DNA. J Biomol Struct Dyn, 1989,7:477-491.
    [213] Li, M, Wu, RS and Tsai, JS. DAPI derivative: a fluorescent DNA dye that can be covalently attached to biomolecules. Bioorg Med Chem Lett, 2003, 13:4351-4354.
    [214] Otto, F and Tsou, KC. A comparative study of DAPI, DIPI, and Hoechst 33258 and 33342 as chromosomal DNA stains. Stain Technol, 1985, 60:7-11.
    [215] Chan, CT, Li, SH and Verma, S. Nocturnal hemodialysis is associated with restoration of impaired endothelial progenitor cell biology in end-stage renal disease. Am J Physiol Renal Physiol, 2005, 289:F679-684.
    [216] Goldman, SA. Directed mobilization of endogenous neural progenitor cells: the intersection of stem cell biology and gene therapy. Curr Opin Mol Ther, 2004, 6:466-472.
    [217] Mehler, MF and Kessler, JA. Progenitor cell biology: implications for neural regeneration. Arch Neurol, 1999,56:780-784.
    [218] Walenta, K, Friedrich, EB, Sehnert, F, et al. In vitro differentiation characteristics of cultured human mononuclear cells-implications for endothelial progenitor cell biology. Biochem Biophys Res Commun, 2005, 333:476-482.
    [219] Weissman, IL. Translating stem and progenitor cell biology to the clinic: barriers and opportunities. Science, 2000, 287:1442-1446.
    [220] Martinez-Serrano, A, Rubio, FJ, Navarro, B, et al. Human neural stem and progenitor cells: in vitro and in vivo properties, and potential for gene therapy and cell replacement in the CNS. Curr Gene Ther, 2001, 1:279-299.
    [221] Martinez-Serrano, A, Lundberg, C, Horellou, P, et al. CNS-derived neural progenitor cells for gene transfer of nerve growth factor to the adult rat brain: complete rescue of axotomized cholinergic neurons after transplantation into the septum. J Neurosci, 1995, 15:5668-5680.
    [222] Iwai, M, Abe, K, Kitagawa, H, et al. Gene therapy with adenovirus-mediated glial cell line-derived neurotrophic factor and neural stem cells activation after ischemic brain injury. Hum Cell, 2001, 14:27-38.
    [223] Kim, HT, Kim, IS, Lim, SE, et al. Gene and cell replacement via neural stem cells. Yonsei Med J, 2004, 45 Suppl:32-40.
    [224] Ourednik, V, Ourednik, J, Park, KI, et al. Neural stem cells -- a versatile tool for cell replacement and gene therapy in the central nervous system. Clin Genet, 1999, 56:267-278.
    [225] Ourednik, V, Ourednik, J, Park, KI, et al. Neural stem cells are uniquely suited for cell replacement and gene therapy in the CNS. Novartis Found Symp, 2000,231:242-262; discussion 262-249, 302-246.
    [226] Park, KI, Himes, BT, Stieg, PE, et al. Neural stem cells may be uniquely suited for combined gene therapy and cell replacement: Evidence from engraftment of Neurotrophin-3-expressing stem cells in hypoxic-ischemic brain injury. Exp Neurol, 2006, 199:179-190.
    [227] Villa, A, Snyder, EY, Vescovi, A, et al. Establishment and properties of a growth factor-dependent, perpetual neural stem cell line from the human CNS. Exp Neurol, 2000, 161:67-84.
    [228] Ladeby, R, Wirenfeldt, M, Dalmau, I, et al. Proliferating resident microglia express the stem cell antigen CD34 in response to acute neural injury. Glia, 2005, 50:121-131.
    [229] Mueller, D, Shamblott, MJ, Fox, HE, et al. Transplanted human embryonic germ cell-derived neural stem cells replace neurons and oligodendrocytes in the forebrain of neonatal mice with excitotoxic brain damage. J Neurosci Res, 2005, 82:592-608.
    [230] Kulbatski, I, Mothe, AJ, Nomura, H, et al. Endogenous and exogenous CNS derived stem/progenitor cell approaches for neurotrauma. Curr Drug Targets, 2005,6:111-126.
    [231] Doering, LC and Snyder, EY. Cholinergic expression by a neural stem cell line grafted to the adult medial septum/diagonal band complex. J Neurosci Res, 2000, 61:597-604.
    [232] Baetge, EE. Neural stem cells for CNS transplantation. Ann N Y Acad Sci, 1993, 695:285-291.
    [233] Chu, K, Kim, M, Jeong, SW, et al. Human neural stem cells can migrate, differentiate, and integrate after intravenous transplantation in adult rats with transient forebrain ischemia. Neurosci Lett, 2003, 343:129-133.
    [234] Svendsen, CN, Caldwell, MA and Ostenfeld, T. Human neural stem cells: isolation, expansion and transplantation. Brain Pathol, 1999, 9:499-513.
    [235] Wang, Q, Matsumoto, Y, Shindo, T, et al. Neural stem cells transplantation in cortex in a mouse model of Alzheimer's disease. J Med Invest, 2006, 53:61-69.
    [236] Iwanami, A, Kaneko, S, Nakamura, M, et al. Transplantation of human neural stem cells for spinal cord injury in primates. J Neurosci Res, 2005, 80:182-190.
    [237] Nakamura, M, Toyama, Y and Okano, H. [Transplantation of neural stem cells for spinal cord injury]. Rinsho Shinkeigaku, 2005, 45:874-876.
    [238] Pallini, R, Vitiani, LR, Bez, A, et al. Homologous transplantation of neural stem cells to the injured spinal cord of mice. Neurosurgery, 2005, 57:1014-1025; discussion 1014-1025.
    [239] Deng, J, Petersen, BE, Steindler, DA, et al. Mesenchymal stem cells spontaneously express neural proteins in culture and are neurogenic after transplantation. Stem Cells, 2006, 24:1054-1064.
    [240] Revishchin, AV, Aleksandrova, MA, Podgomyi, OV, et al. Human fetal neural stem cells in rat brain: effects of preculturing and transplantation. Bull Exp Biol Med, 2005, 139:213-216.
    [241] Shindo, T, Matsumoto, Y, Wang, Q, et al. Differences in the neuronal stem cells survival, neuronal differentiation and neurological improvement after transplantation of neural stem cells between mild and severe experimental traumatic brain injury. J Med Invest, 2006, 53:42-51.
    [242] Lacza, Z, Horvath, E and Busija, DW. Neural stem cell transplantation in cold lesion: a novel approach for the investigation of brain trauma and repair. Brain Res Brain Res Protoc, 2003, 11:145-154.
    [243] Lacza, Z, Horvath, EM, Komjati, K, et al. PARP inhibition improves the effectiveness of neural stem cell transplantation in experimental brain trauma. Int J Mol Med, 2003, 12:153-159.
    [244] Gao, J, Prough, DS, McAdoo, DJ, et al. Transplantation of primed human fetal neural stem cells improves cognitive function in rats after traumatic brain injury. Exp Neurol, 2006, 201:281 -292.
    [245] Kuznetsov, SL, Nikolaeva, LR, Spivak, IA, et al. Effect of transplantation of cultured human neural stem and progenitor cells on regeneration of the cornea after chemical burn. Bull Exp Biol Med, 2006, 141:129-132.
    [246] Englund, U, Bjorklund, A, Wictorin, K, et al. Grafted neural stem cells develop into functional pyramidal neurons and integrate into host cortical circuitry. Proc Natl Acad Sci U S A, 2002, 99:17089-17094.
    [247] Englund, U, Bjorklund, A and Wictorin, K. Migration patterns and phenotypic differentiation of long-term expanded human neural progenitor cells after transplantation into the adult rat brain. Brain Res Dev Brain Res, 2002, 134:123-141.
    [248] Englund, U, Fricker-Gates, RA, Lundberg, C, et al. Transplantation of human neural progenitor cells into the neonatal rat brain: extensive migration and differentiation with long-distance axonal projections. Exp Neurol, 2002,173:1-21.
    [249] Jain, M, Armstrong, RJ, Elneil, S, et al. Migration and differentiation of transplanted human neural precursor cells. Neuroreport, 2003, 14:1257-1262.
    [250] Jeong, SW, Chu, K, Jung, KH, et al. Human neural stem cell transplantation promotes functional recovery in rats with experimental intracerebral hemorrhage. Stroke, 2003, 34:2258-2263.
    [251] Levi-Montalcini, R and Aloe, L. Tropic, trophic, and transforming effects of nerve growth factor. Adv Biochem Psychopharmacol, 1980, 25:3-15.
    [252] Thoenen, H and Barde, YA. Physiology of nerve growth factor. Physiol Rev, 1980, 60:1284-1335.
    [253] Varon, S and Skaper, SD. Short-latency ionic effects of nerve growth factor deprivation and readministration on ganglionic cells. J Supramol Struct, 1980, 13:329-337.
    [254] Kiss, J, Shooter, EM and Patel, AJ. A low-affinity nerve growth factor receptor antibody is internalized and retrogradely transported selectively into cholinergic neurons of the rat basal forebrain. Neuroscience, 1993, 57:297-305.
    [255] Kojima, M, Ikeuchi, T and Hatanaka, H. Nerve growth factor induces trkA mRNA expression in cultured basal forebrain cholinergic neurons from 17-day fetal rats. Neurosci Lett, 1994, 169:47-50.
    [256] Kramer, BM, Van der Zee, CE and Hagg, T. P75 nerve growth factor receptor is important for retrograde transport of neurotrophins in adult cholinergic basal forebrain neurons. Neuroscience, 1999, 94:1163-1172.
    [257] Steininger, TL, Wainer, BH, Klein, R, et al. High-affinity nerve growth factor receptor (Trk) immunoreactivity is localized in cholinergic neurons of the basal forebrain and striatum in the adult rat brain. Brain Res, 1993,612:330-335.
    [258] Koliatsos, VE, Applegate, MD, Knusel, B, et al. Recombinant human nerve growth factor prevents retrograde degeneration of axotomized basal forebrain cholinergic neurons in the rat. Exp Neurol, 1991, 112:161-173.
    [259] Koliatsos, VE, Clatterbuck, RE, Nauta, HJ, et al. Human nerve growth factor prevents degeneration of basal forebrain cholinergic neurons in primates. Ann Neurol, 1991, 30:831-840.
    [260] Koliatsos, VE, Nauta, HJ, Clatterbuck, RE, et al. Mouse nerve growth factor prevents degeneration of axotomized basal forebrain cholinergic neurons in the monkey. J Neurosci, 1990, 10:3801-3813.
    [261] Eriksdotter Jonhagen M, Nordberg A, Amberla K, et al. Intracerebroventricular infusion of nerve growth factor in three patients with Alzheimer's disease. Dement Geriatr Cogn Disord., 1998, 9:246-257.
    [262] Pean, JM, Venier-Julienne, MC, Boury, F, et al. NGF release from poly(D,L-lactide-co-glycolide) microspheres. Effect of some formulation parameters on encapsulated NGF stability. J Control Release, 1998, 56:175-187.
    [263] Brion, S, Plas, J and Masse, G. [Alzheimer's disease and Meynert's nucleus basalis. An anatomo- clinical point of view]. Encephale, 1986, 12:111-114.
    [264] Burns, A, Whitehouse, P, Arendt, T, et al. Alzheimer's disease in senile dementia: loss of neurones in the basal forebrain. Whitehouse, P., Price, D., Struble, R., Clarke, A., Coyle, J. and Delong, M. Science (1982), 215, 1237-1239. Int J Geriatr Psychiatry, 1997, 12:7-10.
    [265] Gartner, U, Hartig, W, Brauer, K, et al. Immunofluorescence and immunoelectron microscopic evidence for differences in myelination of GABAergic and cholinergic septohippocampal fibres. Int J Dev Neurosci, 2001, 19:347-352.
    [266] German, DC, Yazdani, U, Speciale, SG, et al. Cholinergic neuropathology in a mouse model of Alzheimer's disease. J Comp Neurol, 2003,462:371-381.
    [267] Whitehouse, PJ, Price, DL, Struble, RG, et al. Alzheimer's disease and senile dementia: loss of neurons in the basal forebrain. Science, 1982, 215:1237-1239.
    [268] Kiss, J, Patel, AJ and Freund, TF. Distribution of septohippocampal neurons containing parvalbumin or choline acetyltransferase in the rat brain. J Comp Neurol, 1990, 298:362-372.
    [269] Bender, R, Plaschke, M, Naumann, T, et al. Development of cholinergic and GABAergic neurons in the rat medial septum: different onset of choline acetyltransferase and glutamate decarboxylase mRNA expression. J Comp Neurol, 1996, 372:204-214.
    [270] Brauer, K, Seeger, G, Hartig, W, et al. Electron microscopic evidence for a cholinergic innervation of GABAergic parvalbumin-immunoreactive neurons in the rat medial septum. J Neurosci Res, 1998, 54:248-253.
    [271] Pang, KC, Nocera, R, Secor, AJ, et al. GABAergic septohippocampal neurons are not necessary for spatial memory. Hippocampus, 2001, 11:814-827.
    [272] Alvarez-Buylla, A and Lois, C. Neuronal stem cells in the brain of adult vertebrates. Stem Cells, 1995, 13:263-272.
    [273] Picard-Riera, N, Nait-Oumesmar, B and Baron-Van Evercooren, A. Endogenous adult neural stem cells: limits and potential to repair the injured central nervous system. J Neurosci Res, 2004, 76:223-231.
    [274] Uchida, N, Buck, DW, He, D, et al. Direct isolation of human central nervous system stem cells. Proc Natl Acad Sci U S A, 2000, 97:14720-14725.
    [275] Yu, RK and Yanagisawa, M. Glycobiology of neural stem cells. CNS Neurol Disord Drug Targets, 2006, 5:415-423.
    [276] Kamitori, K, Machide, M, Tomita, K, et al. Cell-type-specific expression of protein tyrosine kinase-related receptor RYK in the central nervous system of the rat. Brain Res Mol Brain Res, 2002, 104:255-266.
    [277] Kawaguchi, A, Miyata, T, Sawamoto, K, et al. Nestin-EGFP transgenic mice: visualization of the self-renewal and multipotency of CNS stem cells. Mol Cell Neurosci, 2001, 17:259-273.
    [278] Fujimura, J, Ogawa, R, Mizuno, H, et al. Neural differentiation of adipose-derived stem cells isolated from GFP transgenic mice. Biochem Biophys Res Commun, 2005, 333:116-121.
    [279] Roberts, TJ, Price, J, Williams, SC, et al. Preservation of striatal tissue and behavioral function after neural stem cell transplantation in a rat model of Huntington's disease. Neuroscience, 2006, 139:1187-1199.
    [280] Okano, H, Ogawa, Y, Nakamura, M, et al. Transplantation of neural stem cells into the spinal cord after injury. Semin Cell Dev Biol, 2003, 14:191-198.
    [281] Ziv, Y, Avidan, H, Pluchino, S, et al. Synergy between immune cells and adult neural stem/progenitor cells promotes functional recovery from spinal cord injury. Proc Natl Acad Sci U S A, 2006, 103:13174-13179.
    [282] Scott, DE and Hansen, SL. Post-traumatic regeneration, neurogenesis and neuronal migration in the adult mammalian brain. Va Med Q, 1997, 124:249-261.
    [283] Okano, H, Okada, S, Nakamura, M, et al. Neural stem cells and regeneration of injured spinal cord. Kidney Int, 2005, 68:1927-1931.
    [284] Vaccarino, FM, Ganat, Y, Zhang, Y, et al. Stem cells in neurodevelopment and plasticity. Neuropsychopharmacology, 2001, 25:805-815.
    [285] Svendsen, CN, Clarke, DJ, Rosser, AE, et al. Survival and differentiation of rat and human epidermal growth factor-responsive precursor cells following grafting into the lesioned adult central nervous system. Exp Neurol, 1996, 137:376-388.
    [286] Muraoka, K, Shingo, T, Yasuhara, T, et al. The high integration and differentiation potential of autologous neural stem cell transplantation compared with allogeneic transplantation in adult rat hippocampus. Exp Neurol, 2006, 199:311-327.
    [287] Enomoto, M, Shinomiya, K and Okabe, S. Migration and differentiation of neural progenitor cells from two different regions of embryonic central nervous system after transplantation into the intact spinal cord. Eur J Neurosci, 2003, 17:1223-1232.
    [288] Tuszynski, MH, Armstrong, DM and Gage, FH. Basal forebrain cell loss following fimbria/fornix transection. Brain Res, 1990, 508:241-248.
    
    [289] Krol, KM, Crutcher, KA, Kalisch, BE, et al. Absence of p75(NTR) expression reduces nerve growth factor immunolocalization in cholinergic septal neurons. J Comp Neurol, 2000, 427:54-66.
    
    [290] Rende, M, Giambanco, I, Buratta, M, et al. Axotomy induces a different modulation of both low-affinity nerve growth factor receptor and choline acetyltransferase between adult rat spinal and brainstem motoneurons. J Comp Neurol, 1995, 363:249-263.
    
    [291] Armstrong, DM, Brady, R, Hersh, LB, et al. Expression of choline acetyltransferase and nerve growth factor receptor within hypoglossal motoneurons following nerve injury. J Comp Neurol, 1991, 304:596-607.
    
    [292] Kordower, JH, Bartus, RT, Bothwell, M, et al. Nerve growth factor receptor immunoreactivity in the nonhuman primate (Cebus apella): distribution, morphology, and colocalization with cholinergic enzymes. J Comp Neurol, 1988,277:465-486.
    
    [293] Ward, NL and Hagg, T. p75(NGFR) and cholinergic neurons in the developing forebrain: a re-examination. Brain Res Dev Brain Res, 1999, 118:79-91.
    
    [294] Hartig, W, Seeger, J, Naumann, T, et al. Selective in vivo fluorescence labelling of cholinergic neurons containing p75(NTR) in the rat basal forebrain. Brain Res, 1998, 808:155-165.
    
    [295] Sobreviela, T, Clary, DO, Reichardt, LF, et al. TrkA-immunoreactive profiles in the central nervous system: colocalization with neurons containing p75 nerve growth factor receptor, choline acetyltransferase, and serotonin. J Comp Neural, 1994, 350:587-611.
    
    [296] Strada, O, Hirsch, EC, Javoy-Agid, F, et al. Does loss of nerve growth factor receptors precede loss of cholinergic neurons in Alzheimer's disease? An autoradiographic study in the human striatum and basal forebrain. J Neurosci, 1992, 12:4766-4774.
    
    [297] Springer, JE, Koh, S, Tayrien, MW, et al. Basal forebrain magnocellular neurons stain for nerve growth factor receptor: correlation with cholinergic cell bodies and effects of axotomy. J Neurosci Res, 1987, 17:111-118.
    
    [298] Birthelmer, A, Lazaris, A, Riegert, C, et al. Does the release of acetylcholine in septal slices originate from intrinsic cholinergic neurons bearing p75(NTR) receptors? A study using 192 IgG-saporin lesions in rats. Neuroscience, 2003, 122:1059-1071.
    
    [299] Clavreul, A, Sindji, L, Aubert-Pouessel, A, et al. Effect of GDNF-releasing biodegradable microspheres on the function and the survival of intrastriatal fetal ventral mesencephalic cell grafts. Eur J Pharm Biopharm, 2006,63:221-228.
    [300] Lam, XM, Duenas, ET and Cleland, JL. Encapsulation and stabilization of nerve growth factor into poly(lactic-co-glycolic) acid microspheres. J Pharm Sci, 2001, 90:1356-1365.
    [301] Sakic, B, Kirkham, DL, Ballok, DA, et al. Proliferating brain cells are a target of neurotoxic CSF in systemic autoimmune disease. J Neuroimmunol, 2005, 169:68-85.
    [302] Meissner, KK, Kirkham, DL and Doering, LC. Transplants of neurosphere cell suspensions from aged mice are functional in the mouse model of Parkinson's. Brain Res, 2005, 1057:105-112.
    [303] Fisher, LJ. Neural precursor cells: applications for the study and repair of the central nervous system. Neurobiol Dis, 1997,4:1-22.
    [304] Suhonen, JO, Peterson, DA, Ray, J, et al. Differentiation of adult hippocampus-derived progenitors into olfactory neurons in vivo. Nature, 1996, 383:624-627.
    [305] Shihabuddin, LS. Adult rodent spinal cord derived neural stem cells. Isolation and characterization.Methods Mol Biol, 2002, 198:67-77.
    [306] Shihabuddin, LS, Ray, J and Gage, FH. FGF-2 is sufficient to isolate progenitors found in the adult mammalian spinal cord. Exp Neurol, 1997, 148:577-586.
    [307] Surendran, S, Shihabuddin, LS, Clarke, J, et al. Mouse neural progenitor cells differentiate into oligodendrocytes in the brain of a knockout mouse model of Canavan disease. Brain Res Dev Brain Res, 2004, 153:19-27.
    [308] Zhou, Y, Fang, F, Dong, Y, et al. An improved method for directional differentiation and efficient production of neurons from embryonic stem cells in vitro. J Huazhong Univ Sci Technolog Med Sci, 2005, 25:13-16.
    [309] Zhou, YF, Fang, F, Dong, YS, et al. [Inhibitory effect of murine cytomegalovirus infection on neural stem cells' differentiation and its mechanisms]. Zhonghua Er Ke Za Zhi, 2006, 44:505-508.
    [310] Zhou, C, Wen, ZX, Shi, DM, et al. Muscarinic acetylcholine receptors involved in the regulation of neural stem cell proliferation and differentiation in vitro. Cell Biol Int, 2004, 28:63-67.
    [311] Zhou, L, Del Villar, K, Dong, Z, et al. Neurogenesis response to hypoxia-induced cell death: map kinase signal transduction mechanisms. Brain Res, 2004, 1021:8-19.
    [312] Pencea, V, Bingaman, KD, Wiegand, SJ, et al. Infusion of brain-derived neurotrophic factor into the lateral ventricle of the adult rat leads to new neurons in the parenchyma of the striatum, septum, thalamus, and hypothalamus. J Neurosci, 2001, 21:6706-6717.
    [313] Wang, TT, Jing, AH, Luo, XY, et al. Neural stem cells: isolation and differentiation into cholinergic neurons. Neuroreport, 2006, 17:1433-1436.
    [314] Wu, P, Tarasenko, YI, Gu, Y, et al. Region-specific generation of cholinergic neurons from fetal human neural stem cells grafted in adult rat. Nat Neurosci, 2002, 5:1271-1278.
    [315] Ingram, DK, London, ED and Goodrick, CL. Age and neurochemical correlates of radial maze performance in rats. Neurobiol Aging, 1981,2:41-47.
    [316] Lippa, AS, Pelham, RW, Beer, B, et al. Brain cholinergic dysfunction and memory in aged rats. Neurobiol Aging, 1980, 1:13-19.
    [317] Zeisel, S, Reinstein, D, Corkin, S, et al. Cholinergic neurones and memory. Nature, 1981,293:187-188.
    [318] Emerich, DF, Black, BA, Kesslak, JP, et al. Transplantation of fetal cholinergic neurons into the hippocampus attenuates the cognitive and neurochemical deficits induced by AF64A. Brain Res Bull, 1992, 28:219-226.
    [319] Roskoden, T, Linke, R and Schwegler, H. Transient early postnatal corticosterone treatment of rats leads to accelerated aquisition of a spatial radial maze task and morphological changes in the septohippocampal region. Behav Brain Res, 2005, 157:45-53.
    [320] Winters, BD and Dunnett, SB. Selective lesioning of the cholinergic septo-hippocampal pathway does not disrupt spatial short-term memory: a comparison with the effects of fimbria-fornix lesions. Behav Neurosci, 2004,118:546-562.
    [321] Kristensen, MO. [Neurotransmitters in Alzheimer's disease]. Ugeskr Laeger, 1990, 152:2165-2168.
    [322] Everitt, BJ and Robbins, TW. Central cholinergic systems and cognition. Annu Rev Psychol, 1997, 48:649-684.
    [323] Addy, N A, Pocivavsek, A and Levin, ED. Reversal of clozapine effects on working memory in rats with fimbria-fornix lesions. Neuropsychopharmacology, 2005, 30:1121-1127.
    [324] Calhoun, ME, Mao, Y, Roberts, JA, et al. Reduction in hippocampal cholinergic innervation is unrelated to recognition memory impairment in aged rhesus monkeys. J Comp Neurol, 2004,475:238-246.
    [325] Schwegler, H and Crusio, WE. Correlations between radial-maze learning and structural variations of septum and hippocampus in rodents. Behav Brain Res, 1995, 67:29-41.
    [326] Nilsson, OG, Brundin, P and Bjorklund, A. Amelioration of spatial memory impairment by intrahippocampal grafts of mixed septal and raphe tissue in rats with combined cholinergic and serotonergic denervation of the forebrain. Brain Res, 1990, 515:193-206.
    [327] Eagle, KS, Chalmers, GR, Clary, DO, et al. Axonal regeneration and limited functional recovery following hippocampal deafferentation. J Comp Neurol, 1995, 363:377-388.
    [328] Durkin, T. Central cholinergic pathways and learning and memory processes: presynaptic aspects. Comp Biochem Physiol A, 1989, 93:273-280.
    [329] Cassel, JC, Duconseille, E, Jeltsch, H, et al. The fimbria-fornix/cingular bundle pathways: a review of neurochemical and behavioural approaches using lesions and transplantation techniques. Prog Neurobiol, 1997, 51:663-716.
    [330] Gage, FH, Bjorklund, A, Stenevi, U, et al. Intrahippocampal septal grafts ameliorate learningimpairments in aged rats. Science, 1984,225:533-536.
    [331] Winblad, B, Hardy, J, Backman, L, et al. Memory function and brain biochemistry in normal aging and in senile dementia. Ann N Y Acad Sci, 1985, 444:255-268.
    [332] Modo, M, Hopkins, K, Virley, D, et al. Transplantation of neural stem cells modulates apolipoprotein E expression in a rat model of stroke. Exp Neurol, 2003, 183:320-329.
    [333] Kayama, Y, Takagi, M and Ogawa, T. Cholinergic influence of the laterodorsal tegmental nucleus on neuronal activity in the rat lateral geniculate nucleus. J Neurophysiol, 1986, 56:1297-1309.

© 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