用户名: 密码: 验证码:
核因子-κB亚基p50和p65对永生化神经前体细胞存活的影响及机制研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
研究背景和目的
     缺血缺氧性脑损伤是严重威胁人类生命健康的常见疾病之一,其导致神经组织缺失,传统的药物治疗疗效难以令人满意。神经干细胞或前体细胞的发现为组织再生修复带来了希望。神经干细胞或前体细胞是一类具有自我更新能力、增殖能力和多向分化潜能的未成熟细胞。它在神经系统发育、正常脑功能的维护和脑损伤的修复中发挥了重要的作用。但是研究发现,由于局部缺血缺氧性微环境的改变,内源性迁移或外源性移植的神经干细胞或前体细胞存活数量少、存活时间短,难以发挥有效的修复作用。核因子-κB (NF-κB)是一种核转录因子,能调控一系列基因的表达。它由5种亚基组成,p65和p50是其中最重要的两个亚基。NF-κB在神经系统中广泛表达,在缺血状态下,缺血灶周围组织中NF-κB被激活,但其发挥保护作用还是促凋亡作用,目前尚存争议。以上这些现象使我们关注到这样一个问题,缺血灶周围NF-κB激活对神经干细胞或前体细胞的存活有无影响?干预NF-κB能否成为改善神经前体细胞脑损伤修复作用的新策略?
     本课题拟以永生化神经前体细胞株(INPC)为细胞模型,通过NF-κB亚基p50、p65基因转染探讨其对神经前体细胞存活的影响及其机制,进而明确NF-κB在神经发生学和缺血缺氧性神经前体细胞损伤中的作用,为进一步探讨神经前体细胞修复治疗缺血性脑损伤奠定理论基础。
     研究方法
     1.介导外源基因导入永生化神经前体细胞株非病毒载体的选择
     用非病毒载体阳离子脂质体Lipofectamine 2000,TRANSfection或阳离子聚合物Sofast分别负载质粒EGFP-C1转染INPC,转染后24 h检测转染效率。对转染效率最高的一种载体,观察其转染后12、24、48和72 h的转染效率,确定EGFP表达最高峰。用台盼蓝排斥试验检测转染和未转染细胞的活力。质粒EGFP-C1转染INPC后,经新霉素类似物G418筛选,挑选稳定细胞克隆INPC/EGFP,并观察其EGFP阳性率。应用巢蛋白(Nestin)抗体鉴定INPC/EGFP。胎牛血清诱导INPC/EGFP分化,观察分化后细胞的形态及EGFP表达。免疫细胞化学检测脂质体Lipofectamine 2000介导RcCMV-p65质粒瞬时转染INPC后p65的表达。
     2.NF-κB亚基p50、p65基因修饰的永生化神经前体细胞株的构建
     采用脂质体Lipofectamine 2000将p50和p65编码质粒RcCMV-p50、RcCMV-p65和对照空质粒Rc/CMV分别转染永生化大鼠神经前体细胞株(INPC)。经G418筛选,挑选阳性克隆。采用有限稀释法分离单细胞克隆并扩大培养。采用免疫细胞化学和免疫印迹法挑选p50或p65表达量最高的单细胞克隆。瞬时转染RcCMV-p50质粒于已挑选出的p65表达量最高的单细胞克隆。RT-PCR检测新霉素(Neo)基因、p50或p65基因转录水平的表达。免疫印迹法检测p50或p65基因蛋白的表达。
     3.转染NF-κB亚基后不同二聚体的形成、分布及转录活性
     将NF-κB亚基p50、p65基因修饰后的不同永生化神经前体细胞株进行凝胶电泳迁移实验(EMSA)检测胞核内NF-κB的DNA结合活性。免疫印迹分析胞浆NF-κB抑制蛋白(IκBα)的表达。免疫细胞化学法检测p50和p65分别在细胞中的含量及胞浆胞核的分布。运用荧光素酶报告系统检测各细胞株NF-κB转录活性。
     4.NF-κB对永生化神经前体细胞株存活的影响及相关作用机理
     在正常及缺氧缺糖1 h或3 h条件下,荧光素酶报告系统检测各细胞株的NF-κB转录活性,Annexin V和碘化丙锭( PI)双标记法流式细胞仪检测各细胞株凋亡率。缺氧缺糖6 h后,双苯甲亚胺Hoechst 33342染细胞核,观察细胞形态学改变,PI单染流式细胞术检测细胞凋亡率。缺氧缺糖12h后,四甲基偶氮唑盐比色试验(MTT试验)测定细胞存活率。在正常及缺氧缺糖6 h条件下,免疫印迹分析各细胞株NF-κB凋亡相关靶基因Bcl-2, Bax, Bcl-Xs/l, FAS-L,p53, XIAP和Actin蛋白的表达,通过Amersham公司ImageQuant TL软件分析条带的光密度值,计算各细胞株Bax/Bcl-2比值。
     5.统计学分析
     采用SPSS11.5统计软件分析数据,计量资料以均数±标准差(±s)表示。组间多个样本均数的比较采用重复测量的双因素方差分析或单因素方差分析,P<0.05为差异有统计学意义。
     研究结果
     1.Lipofectamine 2000,TRANSfection和Sofast转染INPC后24 h,转染效率分别为(25.5±2.9)%,(4.0±1.7)%,(7.9±1.4)%,Lipofectamine 2000的转染效率最高。其转染后12、24、48和72 h的转染效率分别为(17.1±0.7)%,(25.5±2.9)%,(19.4±0.9)%,(15.6±1.4)%,EGFP在转染后24 h表达最高。INPC/EGFP中,EGFP阳性率约为95%。INPC/EGFP巢蛋白表达阳性。其分化后呈神经元或星形胶质细胞样,且胞体及突起中仍可见绿色荧光。Lipofectamine 2000介导RcCMV-p65质粒的瞬时转染后,部分细胞呈p65阳性染色,阳性率约为15%。
     2.通过稳定筛选和有限稀释法分离得到稳定转染p65的单细胞克隆A12、A13、B11、C21、C22和E2。其p65免疫染色和免疫印迹分析均呈阳性,但C21的p65表达强于其它克隆。将C21克隆命名为INPC/p65细胞株,进行后续实验,并再次瞬时转染RcCMV-p50质粒,获得INPC/p50p65细胞株。同样得到稳定转染RcCMV-p50、RcCMV质粒的单细胞克隆,分别命名为INPC/p50和INPC/CMV细胞株。Neo基因RT-PCR显示,对照空质粒已成功转染至INPC/CMV。RT-PCR和免疫印迹分析显示,p50和p65基因均能在稳定转染相应质粒的细胞株中高效表达。
     3.EMSA表明,INPC/p50、INPC/p65和INPC/p50p65细胞胞核中分别形成p50同源二聚体、p65同源二聚体、p50p65异源二聚体和p50同源二聚体。INPC/p65和INPC/p50p65细胞株中,IκBα在胞浆中表达升高。免疫细胞化学显示INPC/p50细胞株中主要为强的胞核p50阳性染色,然而INPC/p65细胞株中主要为胞浆p65阳性染色,但在一些细胞中也可见胞核p65阳性染色。NF-κB转录活性在INPC/p50细胞株中轻度增高,在INPC/p65、INPC/p50p65细胞株中明显的升高,其中以INPC/p65细胞株最高(均P<0.05)。
     4.κB依赖性的荧光素酶活性在不同的NF-κB亚基转染细胞株间及不同的缺氧缺糖处理组间(对照组,缺氧缺糖1 h组,缺氧缺糖3 h组)有显著性差异。Annexin V-FITC凋亡检测发现,INPC/p65和INPC/p50p65细胞株发生了自发性的细胞凋亡,而且对缺氧缺糖刺激更为敏感。缺氧缺糖6 h后,各细胞株可观察到凋亡形态学改变,流式细胞术检测证实INPC/p65和INPC/p50p65细胞株的凋亡率升高(P<0.05),并且缺氧缺糖12 h后,细胞存活率低于其它细胞株(P<0.05)。在正常及缺氧缺糖处理6 h条件下, Bax和Bcl-2蛋白的表达及两者的比值在INPC/p65和INPC/p50p65细胞株中升高(P<0.05)。
     研究结论
     1.Lipofectamine 2000可高效简便地转染INPC。这为探讨转NF-κB基因对永生化神经前体细胞的生物学特性影响奠定了实验基础。
     2.成功构建了NF-κB亚基p50、p65基因修饰的永生化大鼠神经前体细胞株。
     3.p50、p65的高表达可逃逸胞浆内内源性IκBα的阻滞作用,导致胞核内形成不同的NF-κB二聚体,并可直接升高NF-κB转录活性。
     4.转染NF-κB不同亚基后升高的NF-κB转录活性导致神经前体细胞发生自发性凋亡,并对凋亡性刺激更加敏感,而这一现象可能通过Bcl-2家族依赖性途径介导。
     研究总结
     本课题通过脂质体基因转染技术构建了NF-κB亚基p50、p65基因修饰的永生化神经前体细胞株,并证实p50、p65的高表达可逃逸胞浆内内源性IκBα的阻滞作用,导致胞核内形成不同的NF-κB二聚体,并直接升高NF-κB转录活性,从而观察到升高的NF-κB转录活性导致神经前体细胞自发凋亡,并对凋亡性刺激更加敏感,这一现象可能通过Bcl-2家族依赖性途径介导。上述研究有助于我们明确NF-κB在神经发生学和缺血缺氧性神经干细胞或前体细胞损伤中的作用,抑制NF-κB活性可能成为改善神经前体细胞脑损伤修复作用的新策略。
     目的研究人胚不同胚龄或脑区神经前体细胞(NPC)体外培养及增殖分化特性。方法取人胚脑组织原代细胞分为小胚龄全脑组、较大胚龄全脑组、较大胚龄新皮质组、较大胚龄纹状体组、较大胚龄间脑组、较大胚龄中脑组、较大胚龄后脑组和较大胚龄延髓组8组,悬浮培养。鉴定细胞球巢蛋白抗原的表达,分化及自我更新能力。观察各组培养细胞的生长、增殖状况。运用免疫荧光细胞化学法比较小胚龄全脑组、较大胚龄全脑组、较大胚龄新皮质组、较大胚龄纹状体组及较大胚龄间脑组神经球分化后,神经元及星形胶质细胞的比例。结果各组培养出的悬浮细胞球巢蛋白抗原阳性,可分化为微管相关蛋白2(MAP2)或胶质纤维酸性蛋白(GFAP)阳性细胞,且5-溴-2-脱氧尿苷(BrdU)掺入实验阳性。培养一周,较大胚龄纹状体组的神经球数目最多,形态最规则,其次分别为较大胚龄间脑组、小胚龄全脑组、较大胚龄全脑组、较大胚龄新皮质组,其它组仅见个别神经球。采用有限稀释法可从较大胚龄纹状体组挑选单细胞克隆球。小胚龄全脑组、较大胚龄全脑组、较大胚龄新皮质组、较大胚龄纹状体组及较大胚龄间脑组NPC诱导分化后,MAP2或GFAP阳性细胞率组间比较差异无显著性。结论从不同胚龄和脑区的中枢神经系统来源的人神经前体细胞均能在体外扩增,针对不同胚龄可采用不同的原代取材方法,小胚龄可取全脑组织培养,而大胚龄则可取纹状体等特定脑区进行培养。不同胚龄或脑区来源的NPC星形胶质细胞及神经元分化比例一致。
Background and objective
     Ischemic-hypoxic brain damage is one of the most common diseases that threaten the health of human. Because the ischemia and hypoxia induce the loss of neural tissue, the traditional drug treatment method is not satisfied. The neural stem cells or progenitor cells bring the hope for the repair and regeneration of the brain tissue. Neural stem cells are functionally defined as an immature cell with the capacity for self-renewal, proliferation and multi-potential, generating the main classes of neural cell types (glia and neurons). The potential of these cells suggests that they may play an important role not only in development of nervous system but also in cell replacement for maintaining the normal brain or repairing injured brain. In the ischemic-hypoxic brain damage, the endogenous migrated or exogenous transplanted neural stem cells are all under the ischemic or hypoxic microenvironment. The cell survival count is too low, or the survival time is too short to have an effective repair in the damaged brain. The nuclear factor-κB, NF-κB, in mammals, consists of five members: RelA(p65), RelB, c-Rel, p105/p50 and p100/p52, which can regulate the expression of a series of genes. The p65 and p50 proteins are considered as the most important two subunits. NF-κB is widely expressed in the nervous system. A controversy exists in the fact that the activation of NF-κB in the peri-lesion zones of cerebral ischemia is illustrated to be involved in promoting neural cell apoptosis as well as survival. All these phenomena have attracted our attention in regard to the role of increased NF-κB activity in the survival of neural progenitor cells. And could the intervention of NF-κB activity be a novel therapeutic strategy for improving the effect of neural progenitor cells for neural regeneration? We previously have established an immortalized neural progenitor cell strain (INPC) by transfection of the simian virus 40 large T antigen gene into the primary cultured neural progenitor cells of newborn rat. The purpose of this study is to explore the potential role of NF-κB in the survival of the immortalized neural progenitor cell strain and the relative mechanism by NF-κB subunits p50, p65 gene transfection. These researches will certainly help us to understand the role of NF-κB in the central nervous system development and the regulation of neural progenitor cell death induced by ischemia or hypoxia. And these findings will provide experimental basis for the novel therapeutic strategies for neuroprotection.
     Methods
     1. An effective method of non-viral vectors mediated exogenous genes transfection in the immortalized neural progenitor cells
     The plasmid EGFP-C1 was transfected into INPC by three different non-viral vectors respectively, including Lipofectamine 2000, TRANSfection and Sofast. The expression of EGFP and the transfection efficiency of the 3 vectors were measured at 24 h after transfection. The transfection efficiency of the vector which was the most efficient, was detected 12, 24, 48 and 72 h after transfection to determine the expression peak of EGFP. The viability of transfected and non-transfected cells was measured by trypan blue rejection test. After the plasmid EGFP-C1 was transfected into INPC, the stable cell clone designated INPC/EGFP was isolated by G418 selection. The positive rate of EGFP was observed in INPC/EGFP. And the specific molecular marker of neural progenitor cells, nestin, was detected using immunocytochemistry in INPC/EGFP. After INPC/EGFP was induced by fetal bovine serum, the cell morphology and the expression of EGFP were observed in the differentiated cells. The level of p65 expression was determined in plasmid RcCMV-p65 transiently transfected INPC mediated by Lipofectamine 2000 using immunocytochemistry.
     2. Construction of immortalized neural progenitor cell strain genetically modified by NF-κB subunits p50, p65 genes
     The control vector RC/CMV and the expression vectors RcCMV-p50 and RcCMV-p65, containing the coding regions of NF-κB subunits p50 and p65 genes, were transfected into immortalized neural progenitor cell strain (INPC) by Lipofectamine 2000, respectively. Stably transfected clones were screened out following G418 selection. Individual clones were screened and expanded into clonal cell strains using limited dilution method. The individual clone in which the level of p50 or p65 was expressed mostly was screened out by immunocytochemistry and Western Blotting. Subsequently, plasmid RcCMV-p50 was transiently transfected into clonal cell strain which expressed p65 mostly. The transcription of Neo, p50 or p65 gene was detected by reverse transcription-polymerase chain reaction (RT-PCR). The translation of p50 or p65 gene was detected by Western Blotting.
     3. The formation, distribution and transcriptive activity of NF-κB dimer following transfection of NF-κB subunits in INPC
     The NF-κB DNA binding activity was measured by electrophoresis mobility shift assay (EMSA) in the cell nuclear extracts from immortalized neural progenitor cell strain genetically modified by NF-κB subunits p50, p65 genes. The expression of IκBαin cytoplasm was detected by Western Blot. The content and cellular distribution of p50 or p65 were determined by an indirect immunocytochemistry study. And the NF-κB transcriptive activity was measured by luciferase reporter gene assay in each cell strain.
     4. The role of NF-κB in the survival of immortalized neural progenitor cells and the underlying mechanism
     Under normal and oxygen and glucose deprivation for 1 h or 3 h conditions, NF-κB transcriptive activity in each cell strain was detected by luciferase assays. Under normal and oxygen and glucose deprivation for 3 h conditions, the apoptotic rate in each cell strain was determined by Annexin V assays. After oxygen and glucose deprivation for 6 h, the cell morphological changes were observed by bisBenzimide Hoechst 33342 staining, and the cell apoptotic rate was measured by flow cytometry (FCM) using PI staining only. Following oxygen and glucose deprivation for 12 h, the cell survival rate was measured by MTT assay. Each cell strain before and after 6 h OGD insult were harvested to detect bcl-2 and bcl-x, bax, p53, Fas ligand, XIAP and Actin genes expressions by western blotting. The band densities were analysis through ImageQuant TL software to obtain the Bax/Bcl-2 ratio.
     5. Statistical analysis
     Data were presented as mean±standard deviation. The means among the groups were analyzed using two-way ANOVA with repeated measures and one-way ANOVA. Statistical significance was accepted if P < 0.05.
     Results
     1. The transfection efficiency of Lipofectamine 2000, TRANSfection and Sofast was (25.5±2.9)%, (4.0±1.7)%, (7.9±1.4)% respectively, at 24 h after transfection. And Lipofectamine 2000 was the most efficient. Its transfection efficiency at 12, 24, 48 and 72 h after transfection was (17.1±0.7)%, (25.5±2.9)%, (19.4±0.9)%, (15.6±1.4)%, respectively. The expression of EGFP was the highest at 24 h after transfection. The positive rate of EGFP was 95% in INPC/EGFP. And INPC/EGFP was still nestin positive. The differentiated cells presented as neurons or astrocytes, and EGFP was still found in their somas and processes. After transient transfection of plasmid RcCMV-p65 by Lipofectamine 2000, some cells presented as p65 positive staining, the positive rate was 15%.
     2. The individual clones of stable transfection of p65 including A12, A13, B11, C21, C22 and E2 were obtained following G418 selection and limited dilution method. All of them were positive for p65 immunostaining and western blotting, but the level of p65 expression was significantly higher in cultured C21. C21 was designated as INPC/p65. Subsequently, plasmid RcCMV-p50 was transiently transfected into INPC/p65, and the INPC/p50p65 cell strain was obtained. As the same, the individual clone of stable transfection of p50 in which the level of p50 was expressed mostly was designated as INPC/p50. The control vector Rc/CMV had been transfected into INPC/CMV determined by RT-PCR of Neo gene. p50 or p65 gene was transcripted or translated correctly and efficiently in the cell strains which had been transfected with the corresponding plasmids.
     3. In EMSA, INPC/p50, INPC/p65 and INPC/p50p65 all gave rise to NF-κB specific bands, which were composed of p50 homodimer, p65 homodimer, and p50 p65 heterodimer and p50 homodimer, respectively. The expression of IκBαwas increased significantly in the cytoplasm of INPC/p65 and INPC/p50p65. A strong, predominantly anti-p50 nuclear immunocytochemistry staining was obtained in INPC/p50. However, in INPC/p65 predominantly anti-p65 cytoplasm staining was obtained, and in several cells nuclear staining also could be seen. The NF-κB transcriptive activity slightly increased in INPC/p50, while obviously increased in INPC/p65 and INPC/p50p65 cell strain, more in former (all P<0.05).
     4. TheκB dependent luciferase activity was significantly different among the cell strains or treatment groups (control group, OGD 1 h group, OGD 3 h group). The spontaneous cell apoptosis and the deteriorating cell apoptosis under OGD condition were resulted from enhanced transfected NF-κB transcriptive activity in INPC/p65 and INPC/p50p65 cell strains by Annexin V assays. After oxygen and glucose deprivation for 6 h, the apoptotic morphological changes could be seen in all cell strains. And the apoptotic rate increased significantly in INPC/p65 and INPC/p50p65 cell strains, compared with INPC, INPC/CMV and INPC/p50 cell strains (P<0.05). After oxygen and glucose deprivation for 12 h, the survival rate was significantly reduced in INPC/p65 and INPC/p50p65 compared with other cell strains (P<0.05). Under normal and oxygen and glucose deprivation for 6 h conditions, transfected NF-κB transcriptive activity in INPC/p65 and INPC/p50p65 cell strains up regulated the level of Bax and Bcl-2 and the ratio of Bax to Bcl-2.
     Conclusions
     1. Exogenous genes can be effectively and conveniently transfected into INPC by Lipofectamine 2000. These provide a good basis for the further research on the biological characteristics of immortalized neural progenitor cells transfected by NF-κB genes.
     2. Immortalized rat neural progenitor cell strains genetically modified by NF-κB subuint p50 or p65 genes has been constructed successfully.
     3. Different NF-κB dimers could be directly found in the nuclear of the cell strains transfected with p50 or p65 genes, escaping cytoplasmic retention by endogenous IκBαproteins, resulting in increased NF-κB transcriptive activity.
     4. The nuclear NF-κB transcriptive activity induced by NF-κB subunits transfection results in spontaneous cell apoptosis, furthermore aggravates the cell apoptosis under OGD condition in an immortalized neural progenitor cell line, which is partly carried out through the Bcl-2 family dependent pathway.
     Summary
     In these researches, immortalized neural progenitor cell strains genetically modified by NF-κB subunits p50, p65 genes have been successfully constructed by liposome transfection technology. And we confirm overexpression of p50, p65 results in different NF-κB dimers in the cellular nuclear, escaping cytoplasmic retention by endogenous IκBαproteins, and leads to increased NF-κB transcriptive activity. Then we observe the upregulated NF-κB transcriptive activity results in spontaneous cell apoptosis, furthermore increases the vulnerability to injury in the immortalized neural progenitor cell. Finally, we demonstrate this is partly carried out through the Bcl-2 family dependent pathway. These researches will help us to understand the role of NF-κB during central nervous system development and in the regulation of ischemic or hypoxic neural progenitor cell death. And these findings will provide experimental basis for the novel therapeutic strategies for neuroprotection.
     Objective To investigate the culture, proliferation and differentiation of human neural progenitor cells derived from embryonic brains of different age or region. Methods The free-floating cells were cultured as 6-9 week embryonic whole brains group, 14-17 week embryonic whole brain group, 14-17 week embryonic neocortex group, 14-17 week embryonic striatum group, 14-17 week embryonic diencephalons group, 14-17 week embryonic mesencephalon group, 14-17 week embryonic metencephalon group and 14-17 week embryonic myelencephalon group. The expression of nestin, self-renew and multipotential property of the cell clusters was identified. The growth and proliferation of the cell aggregations of each group were observed. After the neurospheres from 6-9 week embryonic whole brains group, 14-17 week embryonic whole brain group, 14-17 week embryonic neocortex group, 14-17 week embryonic striatum group and 14-17 week embryonic diencephalons group were induced, the percentage of neuron or astrocyte was investigated by immunocytochemical staining. Results Nestin and Brdu immunochemistry staining were positive in the cell aggregations of each group. And they could differentiate into MAP2 or GFAP positive cells. One week in vitro, there were the most neurospheres in 14-17 week embryonic striatum group. And there was a decreased trend in the numbers of neurospheres of 14-17 week embryonic diencephalons group, 6-9 week embryonic whole brains group, 14-17 week embryonic whole brain group, 14-17 week embryonic neocortex group. Only few neurosphere was observed in other groups. Single cell from 14-17 week embryonic striatum group could form clone. After differentiation of neural progenitor cells from 6-9 week embryonic whole brains group, 14-17 week embryonic whole brain group, 14-17 week embryonic neocortex group, 14-17 week embryonic striatum group and 14-17 week embryonic diencephalons group, the percentage of MAP2 or GFAP positive cells have no significant difference among them. Conclusion In vitro, neural progenitor cells can be isolated from embryonic brains of different age or region. Different region should be obtained as primary culture according to the different age embryonic. Under the same culture conditions, the percentage of differentiated neuron or astrocyte has no difference in the neural progenitor cells from embryonic brains of different age or region.
引文
1. Bull ND, Bartlett PF. The adult mouse hippocampal progenitor is neurogenic but not a stem cell. J Neurosci. 2005, 25(47):10815-10821.
    2. Reynolds BA, Rietze RL. Neural stem cells and neurospheres--re-evaluating the relationship. Nat Methods, 2005, 2(5):333-336.
    3. Temple S. The development of neural stem cells. Nature. 2001, 414(6859): 112-117.
    4.桂伶俐,刘志恒,张传汉,等.人胚不同脑区神经前体细胞的分离培养及分化特性的比较.中国组织化学与细胞化学杂志, 2007, 16(2):129-133.
    5. Gobbel GT, Choi SJ, Beier S, et al. Long-term cultivation of multipotential neural stem cells from adult rat subependyma. Brain Res, 980(2):221-32.
    6. Felling RJ, Snyder MJ, Romanko MJ, et al. Neural stem/progenitor cells participate in the regenerative response to perinatal hypoxia/ischemia. J Neurosci, 2006, 26(16):4359-69.
    7. Dempsey RJ, Sailor KA, Bowen KK, et al. Stroke induced progenitor cell proliferation in adult spontaneously hypertensive rat brain:effect of exogenous IGF-1 and GDNF. J Neurochem, 2003, 87(3):586-597.
    8. Jin K, Sun Y, Xie L, et al. Directed migration of neuronal precursors into the ischemic cerebral cortex and striatum. Mol Cell Neurosci, 2003, 24(1):171-189.
    9. Widera D, Mikenberg I, Kaltschmidt B, et al. Potential role of NF-kappaB in adult neural stem cells: the underrated steersman? Int J Dev Neurosci, 2006, 24(2-3):91-102.
    10. Tamatani M, Che YH, Matsuzaki H, et al. Tumor necrosis factor induces Bcl-2 and Bcl-x expression through NFkappaB activation in primary hippocampal neurons. J Biol Chem, 1999, 274(13):8531-8538.
    11. Gabriel C, Justicia C, Camins A, et al. Activation of nuclear factor-κB in the rat brain after transient focal ischemia. Brain Res Mol Brain Res, 1999, 65: 6l-69.
    12. Irving EA, Hadingham SJ, Roberts J,et al. Decreased nuclear factor-κB DNA binding activity followorg permanent focal cerebral ischemia in the rat. Neurosci Lett, 2000,288: 45-48.
    13. Stephenson D, Ym T, SmaLstig FR, et al. Transcription factor nuclear factor-κB is activated in neurons after focal cerebral ischemia. J Cereb Blood Flow Metab, 2000, 20: 592-603.
    14. Shen W, Zhang C, Zhang G. Nuclear factorκB activation is mediated by NMDA and non-NMDA receptor and L-type voltage-gated Ca2+ channel following severe global ischemia in rat hippocampus. Brain Res, 2002, 933: 23-30.
    15. Domanska-JanikΚ, Bronisz-Κowalczyk A, Zajac H, et al. Interrelations between nuclear-factorκB activation, glial response and neuronal apoptosis in gerbil hippocampus after ischemia. Acta Neurobiol Exp, 2001,61:45-51.
    16. Seegers H, Grillon E, Trioullier Y, et al. Nuclear factor-κB activation in permanent intraluminal focal cerebral ischemia in the rat. Neurosci Lett, 2000, 288:241-245.
    17. Qiu J, Grafe MR, Schmura SM, et al. Differential NF-κB regulation of bcl-x gene expression of hippocampus and basal forebrain in response to hypoxia. J Neurosci Res, 2001, 64:223-234.
    18. Qin ZH, Chen RW, Wang Y, et al. Nuclear factorκB nuclear translocation upregulates c-Myc and p53 expression during NMDA receptor-mediated apoptosis in rat striatum. J Neurosci, 1999. l9: 4023-4033.
    19. Bhakar AL, Tannis LL, Zeindler C, et al. Constitutive nuclear factor-κB activity is required for central neuron survival. J Neurosci, 2002, 22:8466-8475.
    20. Botchkina GI, Geimonen E, Bilof ML, et al. Loss of NF-κB activity during cerebral ischemia and TNF cytotoxicity. Mol Med, 1999, 5: 372-381.
    21. Yu Z, Zhou D, Bruce-Κeller AJ, et al. Lack of the p50 subunit of nuclear factor-κB increases the vulnerability of hippocampal neurons to excitotoxic injury. J Neurosci, 1999, 19:8856-8865.
    22. rving EA, Hadingham SJ, Roberts J,et al. Decreased nuclear factor-κB DNA binding activity followorg permanent focal cerebral ischemia in the rat. Neurosci Lett, 2000, 288: 45-48.
    23. Widera D, Mikenberg I, Elvers M, et al. Tumor necrosis factorαtriggers proliferation of adult neural stem cells via IKK/NF-κB signaling. BMC Neurosci, 2006, 7:64.
    24. Sabolek M, Herborg A, Schwarz J, et al. Dexamethasone blocks astroglial differentiation from neural precursor cells. Neuroreport, 2006, 17(16): 1719-1723.
    25. Piotrowska MJ, Widera D, Kaltschmidt B, et al. Mathematical model for NF-kappaB-driven proliferation of adult neural stem cells. Cell Prolif, 2006, 39(6):441-455.
    26. Widera D, Mikenberg I, Kaus A, et al. Nuclear Factor-kappaB controls the reaggregation of 3D neurosphere cultures in vitro. Eur Cell Mater, 2006, 11:76-84; discussion 85.
    27. Young KM, Bartlett PF, Coulson EJ. Neural progenitor number is regulated by nuclear factor-kappaB p65 and p50 subunit-dependent proliferation rather than cell survival. J Neurosci Res, 2006, 83(1):39-49.
    28.高峰,田玉科,杨辉,等.猿肾病毒40大T抗原基因永生化大鼠神经前体细胞株的构建.中华麻醉学杂志, 2005, 25(8): 597-600.
    29. Rosenqvist N, Hard Af Segerstad C, Samuelsson C, et al. Activation of silenced transgene expression in neural precursor cell lines by inhibitors of histone deacetylation. J Gene Med, 2002, 4(3):248-57.
    30.姚文龙,张传汉,钱巍,等.局灶性脑缺血大鼠永生化神经前体细胞移植的可行性.中国临床康复, 2006, 10(45):36-39.
    31. Lundberg C, Winkler C, Whittemore SR, et al. Conditionally immortalized neural progenitor cells grafted to the striatum exhibit site-specific neuronal differentiation and establish connections with the host globus pallidus. Neurobiol Dis, 1996, 3:33-50.
    32. Lundberg C, Martinez-Serrano A, Cattaneo E, et al. Survival, integration, and differentiation of neural stem cell lines after transplantation to the adult rat striatum. Exp Neurol, 1997, 145: 342-360.
    1. Rosenqvist N, Hard Af Segerstad C, Samuelsson C, et al. Activation of silenced transgene expression in neural precursor cell lines by inhibitors of histone deacetylation. J Gene Med, 2002, 4(3):248-257.
    2.高峰,田玉科,杨辉,等.增强型绿色荧光蛋白基因在永生化大鼠神经前体细胞株的表达.第四军医大学学报, 2006, 26(3):215-217.
    3.高峰,田玉科,杨辉,等.猿肾病毒40大T抗原基因永生化大鼠神经前体细胞株的构建.中华麻醉学杂志, 2005, 25(8): 597-600.
    4. De Smedt SC, Demeester J, Hennink WE. Cationic polymer based gene delivery systems. Pharm Res, 2000, 17(2):113-126.
    5. Ewert K, Ahmad A, Evans HM, et al. Cationic lipid-DNA complexes for non-viral gene therapy: relating supramolecular structures to cellular pathways. Expert Opin Biol Ther, 2005, 5(1):33-53.
    6. Heim R, Cubitt AB, Heim RA, et al. Improved green fluorescence. Nature, 1995, 373(6516):663-664.
    7.李美山,张成,陈松林,等.绿色荧光蛋白和成肌调节因子在骨髓基质干细胞中的共表达.第四军医大学学报, 2006, 26(9):813-817.
    1.司徒镇强,吴军正,主编.细胞培养:西安世界图书出版公司, 1996, 227-229.
    2. Widera D, Mikenberg I, Kaltschmidt B, et al. Potential role of NF-kappaB in adult neural stem cells: the underrated steersman? Int J Dev Neurosci, 2006, 24(2-3):91-102.
    3. Ziegler-Heitbrock HW, Wedel A, Schraut W, et al. Tolerance to lipopolysaccharide involves mobilization of nuclear factor kappa B with predominance of p50 homodimers. J Biol Chem, 1994, 269(25):17001-17004.
    4. Ten RM, Paya CV, Israel N, et al. The characterization of the promoter of the gene encoding the p50 subunit of NF-kappa B indicates that it participates in its own regulation. EMBO J, 1992, 11(1):195-203.
    5. Cogswell PC, Scheinman RI, Baldwin AS Jr. Promoter of the human NF-kappa B p50/p105 gene. Regulation by NF-kappa B subunits and by c-REL. J Immunol, 1993, 150(7):2794-2804.
    6. Chen LF, Greene WC. Shaping the nuclear action of NF-kappaB. Nat Rev Mol Cell Biol, 2004, 5(5):392-401.
    7. Lin L, DeMartino GN, Greene WC. Cotranslational biogenesis of NF-kappaB p50 by the 26S proteasome. Cell, 1998, 92(6):819-828.
    8. Schmitz ML, Baeuerle PA. The p65 subunit is responsible for the strong transcription activating potential of NF-kappa B. EMBO J, 1991, 10(12): 3805-3817.
    1.高峰,田玉科,杨辉,等.猿肾病毒40大T抗原基因永生化大鼠神经前体细胞株的构建.中华麻醉学杂志, 2005,25:597-600.
    2. Pahl HL, Baeuerle PA. Expression of influenza virus hemagglutinin activates transcription factor NF-kappa B. J Virol, 1995,69: 1480-1484.
    3. Hayden MS, Ghosh S. Signaling to NF-kappaB. Genes Dev, 2004,18:2195-2224.
    4. Schmitz ML, Baeuerle PA. The p65 subunit is responsible for the strong transcription activating potential of NF-kappa B. EMBO J, 1991, 10(12):3805-3817.
    5. Jacobs MD Harrison SC. Structure of an IκBα/NF-κB complex. Cell, 1998, 95: 749-758.
    6. Bitar R, Beauparlant P, Lin R, et al. Retrovirus-mediated transfer of nuclear factor-kappa B subunit genes modulates I kappa B alpha and interferon beta expression. Cell Growth Differ, 1995, 6:965-976.
    7. Vacca A, Felli MP, Palermo R, et al. Notch3 and pre-TCR interaction unveils distinct NF-kappaB pathways in T-cell development and leukemia. EMBO J, 2006, 25(5):1000-1008.
    8. Ishikawa H, Claudio E, Dambach D, et al. Chronic inflammation and susceptibility to bacterial infections in mice lacking the polypeptide (p)105 precursor (NF-kappaB1) but expressing p50. J Exp Med, 1998, 187(7):985-996.
    9. Zuniga-Pflucker JC, Schwartz HL, Lenardo MJ. Gene transcription in differentiating immature T cell receptor(neg) thymocytes resembles antigen-activated mature T cells. J Exp Med, 1993, 178(4):1139-1149.
    1. Gabriel C, Justicia C, Camins A, et al. Activation of nuclear factor-κB in the rat brain after transient focal ischemia. Brain Res Mol Brain Res, 1999, 65: 6l-69.
    2. Irving EA, Hadingham SJ, Roberts J,et al. Decreased nuclear factor-κB DNA binding activity followorg permanent focal cerebral ischemia in the rat. Neurosci Lett, 2000, 288: 45-48.
    3. Stephenson D, Ym T, SmaLstig FR, et al. Transcription factor nuclear factor-κB is activated in neurons after focal cerebral ischemia. J Cereb Blood Flow Metab, 2000, 20: 592-603.
    4. Shen W, Zhang C, Zhang G. Nuclear factorκB activation is mediated by NMDA and non-NMDA receptor and L-type voltage-gated Ca2+ channel following severe global ischemia in rat hippocampus. Brain Res, 2002, 933: 23-30.
    5. Domanska-JanikΚ, Bronisz-Κowalczyk A, Zajac H, et al. Interrelations between nuclear-factorκB activation, glial response and neuronal apoptosis in gerbil hippocampus after ischemia. Acta Neurobiol Exp, 2001, 61:45-51.
    6. Seegers H, Grillon E, Trioullier Y, et al. Nuclear factor-κB activation in permanent intraluminal focal cerebral ischemia in the rat. Neurosci Lett, 2000, 288:241-245.
    7. Qiu J, Grafe MR, Schmura SM, et al. Differential NF-κB regulation of bcl-x gene expression of hippocampus and basal forebrain in response to hypoxia. J Neurosci Res, 2001, 64:223-234.
    8. Qin ZH, Chen RW, Wang Y, et al. Nuclear factorκB nuclear translocation upregulates c-Myc and p53 expression during NMDA receptor-mediated apoptosis in rat striatum. J Neurosci, 1999, l9: 4023-4033.
    9. Bhakar AL, Tannis LL, Zeindler C, et al. Constitutive nuclear factor-κB activity is required for central neuron survival. J Neurosci, 2002, 22:8466-8475.
    10. Botchkina GI, Geimonen E, Bilof ML, et al. Loss of NF-κB activity during cerebral ischemia and TNF cytotoxicity. Mol Med, 1999, 5: 372-381.
    11. Yu Z, Zhou D, Bruce-Κeller AJ, et al. Lack of the p50 subunit of nuclear factor-κB increases the vulnerability of hippocampal neurons to excitotoxic injury. J Neurosci, 1999, 19:8856-8865.
    12. Irving EA, Hadingham SJ, Roberts J,et al. Decreased nuclear factor-κB DNA binding activity followorg permanent focal cerebral ischemia in the rat. Neurosci Lett, 2000, 288: 45-48.
    13.高峰,田玉科,杨辉,等.猿肾病毒40大T抗原基因永生化大鼠神经前体细胞株的构建.中华麻醉学杂志, 2005,25: 597-600.
    14. De Cristobal J, Cardenas A, Lizasoain I, et al. Inhibition of glutamate release via recovery of ATP levels accounts for a neuroprotective effect of aspirin in rat cortical neurons exposed to oxygen-glucose deprivation. Stroke, 2002, 33(1): 261-267.
    15. Sheehy AM, Schlissel MS. Overexpression of RelA causes G1 arrest and apoptosis in a pro-B cell line, J Biol Chem, 1999, 274(13):8708-8716.
    16. Chen J, Simon R. Ischemic tolerance in the brain. Neurology. 1997, 48(2): 306-311.
    17. Nurmi A, Lindsberg PJ, Koistinaho M, et al. Nuclear factor-kappaB contributes to infarction after permanent focal ischemia. Stroke, 2004, 35:987-991.
    18. Rosenqvist N, Hard Af Segerstad C, Samuelsson C, et al. Activation of silenced transgene expression in neural precursor cell lines by inhibitors of histone deacetylation. J Gene Med, 2002, 4(3):248-257.
    19. Osorio LM, De Santiago A, Aguilar-Santelises M, et al. CD6 ligation modulates the Bcl-2/Bax ratio and protects chronic lymphocytic leukemia B cells from apoptosis induced by anti-IgM. Blood, 1997, 89(8):2833-2841.
    20. Miyashita T, Reed JC. Tumor suppressor p53 is a direct transcriptional activator of the human bax gene. Cell, 1995, 80(2):293-299.
    21. Tamatani M, Che YH, Matsuzaki H, et al. Tumor necrosis factor induces Bcl-2 and Bcl-x expression through NFkappaB activation in primary hippocampal neurons. J Biol Chem, 1999, 274(13):8531-8538.
    22. Caba E, Bahr BA. Biphasic NF-kappaB activation in the excitotoxic hippocampus. Acta Neuropathol (Berl), 2004, 108(3):173-182.
    23. Schelman WR, Andres RD, Sipe KJ, et al. Glutamate mediates cell death and increases the Bax to Bcl-2 ratio in a differentiated neuronal cell line. Brain Res Mol Brain Res, 2004, 128(2):160-169.
    24. Cheng XR, Zhang L, Hu JJ, et al. Neuroprotective effects of t etramethylpyrazine on hydrogen peroxide-induced apoptosis in PC12 cells. Cell Biol Int, 2006, 18: [Epub ahead of print].
    25. Russell JC, Szuflita N, Khatri R, et al. Transgenic expression of human FGF-1 protects against hypoxic-ischemic injury in perinatal brain by intervening at caspase-XIAP signaling cascades. Neurobiol Dis, 2006, 22(3):677-690.
    26. Yonekura I, Takai K, Asai A, et al. p53 potentiates hippocampal neuronal death caused by global ischemia. J Cereb Blood Flow Metab, 2006, 26(10):1332-1340.
    27. Zhang W, Potrovita I, Tarabin V, et al. Neuronal activation of NF-kappaB contributes to cell death in cerebral ischemia. J Cereb Blood Flow Metab, 2005, 25:30-40.
    1. Piao JH, Odeberg J, Samuelsson EB, et al. Cellular composition of long-term human spinal cord- and forebrain-derived neurosphere cultures. J Neurosci Res, 2006, 84:471–482.
    2.祝畅,张传汉,刘志恒,等.人胚神经干细胞的长期贴壁培养和鉴定.中国康复医学杂志, 2006, 21(4):322-324.
    3. Berninger B, Hack MA, Gotz M. Neural stem cells: on where they hide, in which disguise, and how we may lure them out. Handb Exp Pharmacol, 2006, (174):319-360.
    4. Reynold BA, Tetzlaff W, Weiss S. A multipotent EGF-responsive striatal embryonic progenitor cell produces neurons and astrocytes. J Neurosci, 1992, 12(11):4565-4574.
    5. Carpenter M K, Cui X, Hu Z, et al. In vitro expansion of a multipotent populaion of human neural stem cells. Exp Neurol, 1999, 158:265-278.
    6. Tarasenko YI, Yu Y, Jordan PM, et al. Effect of growth factors on proliferation and phenotypic differentiation of human fetal neural stem cells. J Neurosci Res, 2004, 78 (5): 625-636.
    7. Piao JH, Odeberg J, Samuelsson EB, et al. Cellular composition of long-term human spinal cord- and forebrain-derived neurosphere cultures. J Neurosci Res, 2006, 84:471-482.
    8. Kelly CM, Tyers P, Borg MT, et al. EGF and FGF-2 responsiveness of rat and mouse neural precursors derived from the embryonic CNS. Brain Res Bull, 2005, 68(1-2):83-94.
    9. Horiguchi S, Takahashi J, Kishi Y, et al. Neural precursor cells derived from human embryonic brain retain regional specificity. J Neurosci Res, 2004, 75(6):817-824.
    10. Kim HT, Kim IS, Lee IS, et al. Human neurospheres derived from the fetal central nervous system are regionally and temporally specified but are not committed. Exp Neurol, 2006, 199(1):222-235.
    11.刘士勇,张可成,杨辉,等. IL-1β和胎牛血清对大鼠神经干细胞分化的影响.中国组织化学与细胞化学杂志, 2000, 9(4):432-435.
    12. Hulspas R, Tiarks C, Reilly J, et al. In vitro cell density-dependent clonal growth of EGF-responsive murine neural progenitor cells under serum-free conditions. Exp Neurol, 1997, 148:147-156.
    1. Saccani S, Pantano S, Natoli G. Modulation of NF-kappaB activity by exchange of dimers. Mol Cell. 2003, 11(6):1563-1574.
    2. Hayden MS, Ghosh S. Genes Dev. Signaling to NF-kappaB. 2004,18(18): 2195-2224.
    3. Nurmi A, Lindsberg PJ, et al. Nuclear factor-kappaB contributes to infarction after permanent focal ischemia. Stroke, 2004, 35(4):987-991.
    4. Schmitz ML, Bacher S, Kracht M. I kappa B-independent control of NF-κB activity by modulatory phosphorylations. Trends Biochem Sci, 2001, 26:186-190.
    5. Dixit V, Mak TW. NF-kappaB signaling. Many roads lead to madrid. Cell, 2002, 111(5):615-619.
    6. Doetsch F, Caille I, et al. Subventricular zone astrocytes are neural stem cells in the adult mammalian brain. Cell, 1999, 97(6):703-716.
    7. Johansson CB, Momma S, et al. Identification of a neural stem cell in the adult mammalian central nervous system. Cell, 1999, 96(1):25-34.
    8. Lendahl U, Zimmerman LB, et al. CNS stem cells express a new class of intermediate filament protein. Cell, 1990, 60(4):585-595.
    9. Pevny LH, Sockanathan S, et al. A role for SOX1 in neural determination. Development, 1998, 125(10):1967-1978.
    10. Brazel CY, Limke TL, et al. Sox2 expression defines a heterogeneous population of neurosphere-forming cells in the adult murine brain. Aging Cell, 2005, 4(4):197-207.
    11. Marzesco AM, Janich P, et al. Release of extracellular membrane particles carrying the stem cell marker prominin-1 (CD133) from neural progenitors and other epithelial cells. J Cell Sci, 2005, 118(13): 2849-2858.
    12. Sawamoto K, Nakao N, et al. Generation of dopaminergic neurons in the adult brain from mesencephalic precursor cells labeled with a nestin-GFP transgene. J Neurosci, 2001, 21(11):3895-3903.
    13. Sakakibara S, Imai T, et al. Mouse-Musashi-1, a neural RNA-binding protein highlyenriched in the mammalian CNS stem cell. Dev Biol, 1996, 176(2):230-242.
    14. Reynolds BA, Weiss S. Generation of neurons and astrocytes from isolated cells of the adult mammalian central nervous system. Science, 1992, 255(5052):1707-1710.
    15. Gage FH. Mammalian neural stem cells. Science, 2000, 287(5457): 1433-1438.
    16. Zhu LL, Wu LY, et al. Effects of hypoxia on the proliferation and differentiation of NSCs. Mol Neurobiol, 2005, 31(1–3):231-242.
    17. Limoli CL, Rola R, et al. Cell-density-dependent regulation of neural precursor cell function. Proc Natl Acad Sci U.S.A., 2004, 101(45): 16052-16057.
    18. Shingo T, Sorokan ST, et al. Erythropoietin regulates the in vitro and in vivo production of neuronal progenitors by mammalian forebrain neural stem cells. J Neurosci, 2001, 21(24):9733-9743.
    19. Digicaylioglu M, Bichet S, et al. Localization of specific erythropoietin binding sites in defined areas of the mouse brain. Proc Natl Acad Sci U.S.A., 1995, 92(9):3717-3720.
    20. Hang CH, Shi JX, et al. Up-regulation of intestinal nuclear factor kappa B and intercellular adhesion molecule-1 following traumatic brain injury in rats. World J Gastroenterol, 2005, 11(8):1149-1154.
    21. Haughey NJ, Nath A, et al. Disruption of neurogenesis by amyloid beta-peptide, and perturbed neural progenitor cell homeostasis, in models of Alzheimer’s disease. J Neurochem, 2002, 83(6):1509-1524.
    22. Kaltschmidt B, Uherek M, et al. Inhibition of NF-kB potentiates amyloid-b-mediated neuronal apoptosis. Proc Natl Acad Sci U.S.A., 1999, 96:9409-9414.
    23. Ferguson KL, Callaghan SM, et al. The Rb-CDK 4/6 signaling pathway is critical in neural precursor cell cycle regulation. J Biol Chem, 2000, 275(43): 33593-33600.
    24. Guttridge DC, Albanese C, et al. NF-kappaB controls cell growth and differentiation through transcriptional regulation of cyclin D1. Mol Cell Biol, 1999, 19(8):5785-5799.
    25. Aarum J, Sandberg K, et al. Migration and differentiation of neural precursor cells can be directed by microglia. Proc Natl Acad Sci U.S.A., 2003, 100(26):15983-15988.
    26. Arvidsson A, Collin T, et al. Neuronal replacement from endogenous precursors in theadult brain after stroke. Nat Med, 2002, 8(9):963-970.
    27. Parent JM, Valentin VV, et al. Prolonged seizures increase proliferating neuroblasts in the adult rat subventricular zone-olfactory bulb pathway. J Neurosci, 2002, 22(8):3174-3188.
    28. Widera D, Holtkamp W, et al. MCP-1 induces migration of adult neural stem cells. Eur J Cell Biol, 2004, 83(8):381-387.
    29. Tran PB, Ren D, et al. Chemokine receptors are expressed widely by embryonic and adult neural progenitor cells. J Neurosci Res, 2004, 76(1): 20-34.
    30. Sun L, Lee J, et al. Neuronally expressed stem cell factor induces neural stem cell migration to areas of brain injury. J Clin Invest, 2004, 113(9):1364-1374.
    31. Imitola J, Raddassi K, et al. Directed migration of neural stem cells to sites of CNS injury by the stromal cell-derived factor 1alpha/CXC chemokine receptor 4 pathway. Proc Natl Acad Sci U.S.A., 2004, 101(52): 18117-18122.
    32. Rostasy K, Gorgun G, et al. Tumor necrosis factor alpha leads to increased cell surface expression of CXCR4 in SK-N-MC cells. J Neurovirol, 2005, 11(3):247-255.
    33. Mondal D, Pradhan L, et al. Signal transduction pathways involved in the lineage-differentiation of NSCs: can the knowledge gained from blood be used in the brain? Cancer Invest, 2004, 22(6):925-943.
    34. Fan Z, Bau B, et al. IL-1beta induction of IL-6 and LIF in normal articular human chondrocytes involves the ERK, p38 and NFkappaB signaling pathways. Cytokine, 2004, 28(1):17-24.
    35. Feng Z, Porter AG. NF-kappaB/Rel proteins are required for neuronal differentiation of SH-SY5Y neuroblastoma cells. J Biol Chem, 1999, 274(43):30341-30344.
    36. Goncalves MB, Boyle J, et al. Timing of the retinoid-signalling pathway determines the expression of neuronal markers in neural progenitor cells. Dev Biol, 2005, 278(1):60-70.

© 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