用户名: 密码: 验证码:
应用小鼠胚胎干细胞(ESCs裂解液逆转化胎儿成纤维细胞(MEFs)为诱导性多能干细胞(iPSCs)的研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
诱导已分化体细胞发生去分化并形成类似于ESCs多能性细胞特征的过程称作“体细胞重编程”。从ESCs裂解液中提取活性物质诱导已分化体细胞重编程为多能干细胞的过程,既没有诱导性干细胞基因组染色质的直接参与,也没有被诱导体细胞基因组DNA结构序列的变化;研究ESCs提取物中有活性细胞因子的作用,有利于发现和识别与重编程相关的细胞因子及其表达调控机制,并进而研究其相互作用关系;因为ESCs的提取物直接诱导靶细胞使其发生了生物学变化,而没有引起靶细胞基因组碱基序列发生结构学变化,不会使靶细胞发生遗传学变异,因此,应用这项技术对体细胞重编程所诱导形成的多能干细胞具有更好的生物安全性;以上种种优势赋予这项研究内容在生物医学领域具有重要的理论价值和广阔的应用前景。
     本研究通过优化小鼠体内受精囊胚、杂合二倍体孤雌囊胚、人工重组异期卵母细胞核二倍体孤雌囊胚(含有速激核成熟新生鼠生长初期卵母细胞与成熟卵母细胞基因组二倍体小鼠孤雌囊胚)的获取途径,获得不同囊胚来源的ESCs。采用形态学观察法、碱性磷酸酶(AKP)染色法、核型分析法、Oct-4和SSEA-1免疫细胞化学检测法、Oct-4基因和Sox-2基因表达检测法、类胚体(EB)分化能力检测等方法对其ESCs的特性进行鉴定。用于被诱导的受体MEFs,采用曲古抑菌素A(TSA)和5-氮杂-2’-脱氧胞苷(5-aza-dc)预处理,然后再经过适宜浓度链球菌溶血素O(SLO)渗透化处理;用于起诱导作用的不同囊胚来源的小鼠ESCs,采用液氮反复冻融和离心的方法,得到不同类型ESCs的无细胞裂解液;将受体细胞孵育在无细胞裂解液的培养液中进行重编程逆转化性诱导培养,获得来源于体细胞的多能干细胞。对诱导所得多能干细胞进行多能干细胞特性的鉴定。借助双重PCR性别鉴定的方法,分选出来源于雌性的有性生殖ESCs,孤雌胚胎干细胞(pESCs)和重构pESCs,三种雌性胚胎干细胞分别通过冻融和离心的方法,获得三种不同类型雌性ESCs(供体细胞)的无细胞裂解液,将此作为诱导剂重编程逆转化雄性MEFs(受体细胞),获得多能干细胞后,依据供体细胞和受体细胞的性别差异,采用双重PCR性别鉴定的方法确定重编程所得多能干细胞集落的来源。最后以鸡成纤维细胞作为重编程的受体细胞,探讨小鼠ESCs裂解液进行种间体细胞重编程、逆转化鸡胚成纤维细胞形成多能干细胞的可行性,并通过核型分析鉴定其所得多能干细胞的来源。
     通过上述研究,旨在比较不同来源ESCs裂解液介导体细胞重编程逆转化为多能干细胞的可能性及其效率,筛选适宜的共孵育方式,验证重编程体系的可靠性,比较有性生殖ESCs和无性生殖pESCs裂解液的作用效果,进一步研究父本基因缺失情况下对体细胞重编程逆转化效率的作用和对异种动物体细胞诱导形成多能干细胞的可能性。这项研究结果将为今后进一步探索ESCs和pESCs裂解液中重编程有效成分的筛选、鉴定和物种间多能干细胞的研究奠定基础。研究结果如下:
     1.用于分离小鼠ESCs(?)勺三种囊胚有效生成途径研究
     1.1小鼠体内受精囊胚内细胞团(ICM)集落获取的条件优化
     本研究首先通过比较不同胎龄MEFs的生长状况和连续传代过程中细胞的生长情况,研究影响MEFs分离和培养效率的因素,旨在为早期胚胎共培养体系选择更加有效的MEFs的分离培养体系,用于MEFs饲养层的制备、ESCs的分离培养和被诱导受体细胞的筛查处理。然后,采集3.5d、4d和4.5d胎龄的胚胎,分别移入DMEM培养液、DMEM+白血病抑制因子(LIF)培养液和包含有MEFs贴壁生长细胞的共培养体系中进行体外培养,比较三种培养体系中ICM孵出的时间、孵化囊胚贴壁比率和ICM集落形成比率,旨在筛选获取优质ICM集落的实验方法,为ESCs(?)勺分离培养奠定基础。结果表明:采用12.5-14.5d胎龄的小鼠胎儿分禺MEFs,传代培养后选取第3代MEFs制备饲养层,采集4d的小鼠囊胚置于其MEFs饲养层的共培养体系中,经体外培养3-4d,可以获取优质的ICM,这为体内受精囊胚来源ESCs的分离培养奠定了基础。
     1.2小鼠杂合二倍体孤雌囊胚生成途径的优化
     本研究采用5种化学激活方法即乙醇、乙醇+6-二甲基氨基嘌呤(6-DMAP)、乙醇+6-DMAP+细胞松弛素B (CB)、SrCl2、SrCl2+CB处理小鼠成熟卵母细胞,通过观察激活卵母细胞第二极体排出情况和核型分析,探讨不同激活方法所得激活卵母细胞染色体倍性的倾向类型,比较不同染色体倍性激活卵母细胞的体外发育能力,筛选有利于形成杂合二倍体孤雌囊胚的激活方法。结果表明:在含有10%乙醇培养液中激活卵母细胞5min的基础上,添加6-DMAP有利于抑制第二极体的排出,促进双原核(2PN)杂合二倍体孤雌胚胎的形成,获取更高的过二细胞阻滞率和桑葚胚/囊胚发育率;采用含有10mM SrCl2和CB处理卵母细胞4h,也能够获取较多的双原核(2PN)杂合二倍体和较高的桑葚胚/囊胚发育率,因此,以上两种孤雌激活方法适宜用于获取小鼠杂合二倍体孤雌囊胚。
     1.3人工重组异期卵母细胞核二倍体孤雌囊胚(含有速激核成熟新生小鼠生长初期卵母细胞和M Ⅱ期卵母细胞基因组的二倍体孤雌胚胎)的构建
     本研究分别研究了CB对生发泡(GV)期卵母细胞去核效果的影响、去除成年小鼠MⅠ期卵母细胞GV的方法选择、新生小鼠生长初期卵母细胞与去核GV期卵母细胞黏合体系的筛选、电融合条件选择以及重构胚化学激活方法的筛选等,旨在构建含有速激核成熟新生小鼠生长初期卵母细胞和MⅡ期卵母细胞基因组的二倍体孤雌胚胎,用于ESCs的分离。结果表明:预先采用10μg/mL CB溶液处理GV期卵母细胞,0.5%链霉蛋白酶去除GV期卵母细胞的透明带,显微操作仪上去除生发泡,将去除生发泡的GV期卵母细胞与新生小鼠生长初期卵母细胞移入自制凹窝内进行黏合,选择1.5KV/cm-2.0KV/cm的电场强度、40μS的脉冲宽度对黏合细胞进行电击,发生电融合和电激活,获得排出了第一极体的速激核成熟重构卵母细胞。利用显微操作仪将重构卵母细胞的核区移植到去除第一极体的MⅡ期成熟卵母细胞的透明带下,通过电融合使两者发生融合,形成含有两个MⅡ期卵母细胞核的重构卵细胞,再经过对其进行7%乙醇(5min)联合2.5mmol/L6-DMAP (4h)(?)勺激活后用于体外培养,可以构建出含有速激核成熟新生小鼠生长初期卵母细胞和MⅡ期卵母细胞基因组的二倍体孤雌胚胎。
     2.小鼠ESCs、pESCs和重构pESCs(?)勺分离、培养与鉴定
     2.1小鼠体内受精囊胚ESCs的分离、培养与鉴定
     本研究采集13.5d胎龄的胎儿分离MEFs,选取第3代MEFs制备饲养层;分别采用全胚法和免疫外科法从4d胚龄的囊胚中获取ICM,将ICM小团块移到饲养层上进行增殖培养,观察集落形态,并对细胞集落进行多能性鉴定。结果表明:采用全胚法和免疫外科法均能分离得到小鼠ESCs集落,其集落形成率差异不显著(30.26±2.77%vs32.92±4.37%,P>0.05)。两种方法所得ESCs集落具有典型的ESCs形态特征,AKP染色阳性,ESCs的染色体为二倍体,具有正常核型,Oct-4和SSEA-1免疫细胞化学检测为阳性,RT-PCR检测表明ESCs表达多能性基因:Oct-4和Sox-2, RT-PCR检测显示EB中三个胚层的标记基因(Fgf-5、Bra T和Afp)均为阳性。
     2.2昆明小鼠孤雌囊胚来源pESCs的分离、培养与鉴定
     鉴于孤雌囊胚于体外培养时间过长,胚胎难以突破透明带引起囊胚孵化率和贴壁率降低的情况,本研究选用免疫外科法分离单倍体pESCs(源自SrCl2激活卵母细胞4h所得囊胚)和二倍体pESCs(源自10mM SrCl2+5μg/mL CB激活卵母细胞4h所得囊胚),并对所得细胞进行多能性鉴定,旨在探讨孤雌囊胚染色体倍性对pESCs分离和培养效率的影响,筛选适宜倍性的孤雌囊胚获取pESCs。(?)吉果表明:本研究所得到的pESCs集落具有与体内受精囊胚来源ESCs类似的多能性鉴定效果。从二倍体孤雌囊胚分离所得pESCs的集落形成率显著高于单倍体孤雌囊胚处理组(13.54±4.43%vs7.17±4.45%,P<0.05),宜选择有利于提高二倍体孤雌囊胚形成率的激活方法进行pESCs的分离。
     2.3异期二倍染色体重构胚胎来源pESCs的分离、培养与鉴定
     本研究通过构建包含有新生小鼠生长初期卵母细胞和MⅡ期卵母细胞的二倍染色体重构核型孤雌胚胎,经体外培养获得囊胚,采用全胚法分离pESCs,并对所得重构pESCs进行多能性鉴定。结果表明:将融合后发育至桑葚胚期和囊胚期的180枚胚胎转移到MEFs饲养层上,48h后从透明带中孵出并贴壁于饲养层上,96h后ICM长出并开始增殖,5d后ICM增殖成柱状,挑出消化离散成细胞小团块后,5d后可见形成了26个pESCs集落,其集落形成率为14.43±4.36%。所得集落具有与体内受精囊胚来源ESCs类似的多能性鉴定效果。
     3.应用小鼠ESCs裂解液重编程逆转化MEFs为多能干细胞的研究
     3.1不同浓度SLO对MEFs生长状态的影响
     本试验采用不同浓度SLO处理MEFs,通过比较SLO处理下MEFs的细胞密度、存活率和贴壁率,并绘制适宜浓度SLO处理后MEFs的生长曲线,研究不同浓度SLO对MEFs生长状态的影响,探讨SLO对体细胞的损伤是否具有剂量依赖性,以便筛选适宜浓度的SLO用于重编程体细胞的渗透化处理。结果表明:25USLO处理组的细胞密度和经CaCl2封膜后继续培养的细胞贴壁率分别与10U SLO处理组之间差异不显著(p>0.05),分别极显著地高于50U和100U SLO处理组(p<0.01)。第3代MEFs经25USLO渗透化处理后细胞仍具有正常分裂增殖的生长模式,说明该浓度的SLO对MEFs的分裂增殖影响较小。因此,宜选用25U SLO对重编程MEFs进行渗透化处理。
     3.2昆明小鼠ESCs裂解液的制备
     本研究比较了两种机械性裂解法即超声波处理法和液氮反复冻融法对ESCs裂解液制备效果的影响,并对所得裂解液进行体外培养试验和加入未经SLO渗透化处理的MEFs后的Oct-4免疫细胞化学染色,尽可能地排除ESCs完整细胞或细胞碎片残留等造成裂解液诱导体细胞重编程的假阳性,旨在探讨适宜的ESCs裂解方法,以获得用于体细胞重编程的高效ESCs裂解液。结果表明:液氮反复冻融7次可使ESCs细胞绝大部分发生裂解,获得的总蛋白质浓度显著高于超声波裂解细胞处理组(35.6mg/mL vs14.3mg/mL)。裂解液经体外培养后未见MEFs饲养层上生长有ESCs集落,AKP染色也未见红棕色细胞,Oct-4免疫细胞化学染色MEFs为阴性反应。因此,宜选择液氮反复冻融7次的方法制备用于体细胞重编程的ESCs裂解液
     3.3小鼠ESCs裂解液与MEFs共孵育体系的研究
     本研究首先探讨了供、受体细胞的性别属性对ESCs裂解液诱导体细胞重编程效率的影响,结果表明供、受体细胞性别对ESCs裂解液诱导MEFs重编程所得多能干细胞集落数量无显著差异。然后,比较了三种不同来源囊胚分离所得雌性ESCs裂解液重编程雄性MEFs或经过TSA和5-aza-dC预处理的雄性MEFs为多能干细胞的效率,以雄性MEFs与其自身裂解液共孵育和未经裂解液处理的雄性MEFs作为对照组,并对所得多能干细胞进行鉴定,旨在探讨适宜的裂解液诱导体细胞重编程体系。结果表明:体内受精囊胚来源并经双重PCR性别鉴定为雌性的ESCs裂解液、孤雌囊胚ESCs裂解液和重构囊胚来源ESCs裂解液均能够重编程雄性MEFs为多能干细胞,这些多能干细胞集落具有与体内受精囊胚来源ESCs类似的多能性鉴定指标表现。在重编程所得多能干细胞集落数量上,体内受精囊胚来源雌性ESCs裂解液处理组(64.33±3.7个、80.67±2.08个)>重构囊胚来源ESCs裂解液处理组(45.00±4.36个、53.67±3.06个)>孤雌囊胚处理组(35.67±2.52个47.67±4.16个);TSA和5-Aza-dC的预处理有助于促进雄性MEFs的重编程,但是,在没有ESCs裂解液的诱导作用下,仅采用TSA和5-Aza-dC预处理雄性MEFs并不能获得多能干细胞集落;无ESCs裂解液参与的单纯重编程操作过程和雄性的MEFs裂解液并不能使雄性MEFs发生重编程,因此,促使雄性MEFs发生重编程的物质来源于ESCs的裂解液
     4. ESCs裂解液诱导MEFs重编程所得多能干细胞集落的来源鉴定
     采用双重PCR性别鉴定方法对体内受精囊胚来源经性别鉴定为雌性ESCs裂解液、孤雌囊胚来源pESCs裂解液和重构孤雌囊胚来源ESCs裂解液诱导雄性MEFs重编程所得到的多能干细胞进行性别鉴定,鉴定所得多能干细胞的来源,判定细胞提取物诱导的重编程体系的可靠性。结果表明:体内受精囊胚来源经性别鉴定为雌性ESCs的裂解液、孤雌囊胚来源pESCs裂解液和重构孤雌囊胚来源的ESCs裂解液均能够诱导雄性MEFs重编程为多能干细胞,所得多能干细胞的基因组DNA中具有250bp的Sry基因序列片段和339bp的ZFX基因序列片段,为雄性细胞。因此,pESCs裂解液重编程所得具有多能性特征的细胞是源自于雄性MEFs,它们所表现出的多能性检测指标的阳性反应并不是由于裂解液制备过程中的ESCs污染所致。
     5. ESCs裂解液诱导种间体细胞重编程的可行性研究
     本研究以鸡胚成纤维细胞作为重编程的受体细胞,采用小鼠体内受精囊胚来源ESCs裂解液与其进行共孵育,通过形态学观察,研究其对鸡胚成纤维细胞的逆转化效果,对所获得干细胞集落进行多能性检测,旨在探讨小鼠ESCs裂解液作用于异种体细胞后,使其重编程逆转化为多潜能干细胞的可行性。结果表明:与小鼠ESCs裂解液共孵育的鸡胚成纤维细胞均变为圆形细胞,未见梭形的贴壁细胞,形成了42.33±4.5个细胞集落。细胞集落经AKP染色和Oct-4和SSEA-1免疫细胞化学染色,均呈阳性反应。表明小鼠ESCs裂解液重编程鸡胚成纤维细胞后,所获得的细胞集落表现出一定的多能干细胞特性,经核型分析表明这些细胞来源于鸡胚成纤维细胞。单独的重编程操作过程和与鸡胚成纤维细胞裂解液共孵育,均无法将鸡胚成纤维细胞重编程为具有ESCs多能性特征的干细胞,说明参与重编程鸡胚成纤维细胞的物质来源于小鼠ESCs的裂解液。结果表明,ESCs裂解液诱导体细胞重编程逆转化为多能干细胞的作用也能够在小鼠和鸡的种间发生。
     总结论:通过共孵育(或者共培养)途径应用ESCs的无细胞裂解液作为诱导剂,能够在较短的时间内(处理后1天内呈现细胞形态学显著变化成为圆形细胞;4天内形成边界清晰的细胞集落;共孵育后第3天细胞集落表现AKP活性,第7天细胞表达Oct-4、Sox-2和Nanog基因)逆转化经过SLO、TSA和5-Aza-dC预处理的MEFs成为多能干细胞;应用这个逆转化体细胞成为多能干细胞的细胞培养体系,使用同样的预处理体细胞的方法,也能够诱导异种鸡胚成纤维细胞成为多能干细胞。
The process of de-differentiating somatic cells to a pluripotent state whereby they adopt characteristics of embryonic stem cells is referred to as'cellular reprogramming' The advantages of cell extracts-mediated reprogramming of somatic cells are:(1) Chromosome reprogramming of imported pluripotent cells does not exist,(2) Manipulating cellular composition of the extract to identify cytokines which are relevant to reprogramming,(3) Since the extract is derived from cells, its role is biological, rather than chemical, so security is higher. More and more researchers pay attention to this method, which is very worthwhile to study and develop in depth, and there are bright prospects of application.
     Embryonic stem cells were isolated from the high-quality inner cell mass (ICM) from blastocysts derived from in vivo fertilization, parthenogenetic activation, and reconstruction of embryo using somatic cell nuclear transfer, and were identified with morphological observation, alkaline phosphatase assay, karyotype analysis, immunocytochemistry staining for Oct-4and SSEA-1, RT-PCR analysis for Oct-4and Sox-2, and examination the in vivo differentiation of embryoid body. MEFs were pretreated with trochostatin A (TSA) and5-Aza-deoxycytidine (5-Aza-dc) and permeabilized with streptolysin O, then those cells were incubated with reprogramming cell-free extracts which were prepared by repeatedly freezing and thawing ESCs in liquid nitrogen. Reprogrammed MEFs were continually cultured in ESCs medium and were identified whether those cells were reprogrammed to became a pluripotential stem cells or not. To rule out the possibility of ESCs contamination during the preparation of ESCs extracts, further pluripotential stem cells generation were performed using male MEFs which were identified by dual PCR for sex identification system of a different gender from extract-donor ESCs. The donors of ESCs-derived extract were the parthenogenetic embryonic stem cells (pESCs) from the oocytes which been parthenogenetic activated and female ESCs with the identified by dual PCR for sex identification system from the blastocystosts derived from in vivo fertilization. Therefore, we can decided the origin of the pluripotential stem cells according to their gender using dual PCR for sex identification system. Finally, chicken embryo fibroblasts which being act as the target cells of reprogramming and were incubated with mouse ESCs extracts, to explore the feasibility of reprogramming between the different species, the origin of cells resulted from reprogramming mediated by mouse ESCs extracts was identified by karyotype analysis.
     The aim of this study was to screening the appropriate incubation method to improve the efficiencies of reprogramming which being mediated by ESCs extracts, to assess the reliability of the reprogramming system, and to explore the reprogramming mechanism about the effects of paternal gene on the efficiencies of reprogramming and The feasibility of reprogramming which be induced with the effective components in the ESCs extracts between the different species etc. The main results in this paper were as follows:
     1. Effective generation pathways of blastocysts which were used to isolate the ESCs
     1.1Optimization of generation pathways of ICM from mouse blastocysts which produced from in vivo fertilization
     The effects of different gestational age and continuous passage on isolation, culture and growth status of MEFs were explored in this study in order to establish a effective system for the isolation and culture of MEFs, and to obtain the high-quality MEFs for preparing the feeder layer and acting as the target cells of reprogramming.3.5dpc (days post coitum),4dpc and4.5dpc mouse embryos were collected and cultured in different culture system including DMEM, DMEM+LIF and co-culture with MEFs respectively, the hatching time of ICM, attachment efficiencies of hatching blastocysts and formation efficiencies of ICM outgrowth were compared, in order to find out an effective method for obtaining high-quality ICM outgrowth for the isolation of the ESCs. Results indicated that12.5dpc to14.5dpc mouse embryos were appropriate for isolating the primary MEFs, the third generation MEFs were suitable for preparing the feeder layer.4dpc embryos were collected and co-cultured with MEFs for3days to4days, high-quality ICM outgrowth could be obtained for the isolation of ESCs.
     1.2Optimization of generation pathways of mouse heterozygous diploid blastocysts
     Oocytes were activated with five methods of parthenogenetic activation, including ethanol, ethanol+6-DMAP, ethanol+6-DMAP+CB, SrCl2, SrCl2+CB. Then, activated oocytes were observed under the inverted microscope and karyotype analysis of activated oocytes were done to identify the parthenogenetic activation types of activated oocytes, including uniform haploid, heterozygous diploid with one pronucleus, mosaic haploid, and heterozygous diploid with two pronuclei. The development rate of morula and blastocyst from activated oocytes with different parthenogenetic activation types were compared to screen the appropriate activated method to obtain heterozygous diploid blastocysts. Results indicated that when oocytes were treated with10%alcohol for5min united with6-DMAP (6-dimethylaminopyridine) or with lOmM SrCl2and5μg/mL CB for4h, the extrusion of second polar body would be inhibitted, higher rate of diploid parthenogenetic embryos, overcome the two-cell block and generation of morula and blastocyst were produced. But, the morula and blastocyst development rate of oocytes were treated with lOmM SrCl2and5u g/ml CB for4h was higher than that treated with10%alcohol for5min united with6-DMAP. Therefore, those two parthenogenetic activation methods are the appropriate method to obtain the higher rate of mouse heterozygous diploid blastocysts.
     1.3Reconstruction of mouse diploid parthenogenetic embryo contained genetic materials from primary oocyte with quickly activated and matured nucleus of new-born mouse and MⅡ oocyte
     The selection of suitable method for enucleating the germinal vesicle (GV) of MⅠ oocyte, GV oocyte being treated with cytochalasin B (CB) at appropriate concentrations before being enucleated, adhesion system between primary oocyte of new-born mouse and GV oocyte without GV, electrofusion parameteres for fusion of reconstructed oocyte and chemical activation of reconstructed embryo were explored in this study, in order to investigate the method for reconstructing mouse diploid parthenogenetic embryo contained genetic materials from primary oocyte with quickly activated and matured nucleus of new-born mouse and MⅡ oocyte. Results indicated that GV oocyte was pretreated with10μg/mL CB, GV oocyte was treated with0.5%pronase for removing the zona pellucida, GV oocyte without germinal vesicle was adhesived to primary oocyte of new-born mouse in the hollowness culture system, the adhesived oocyte was treated with the appropriate parameters (electric field strength was between1.5KV/cm and2.0KV/cm and the pulse width was40μs). The nuclear region of reconstructed oocyte which had been expelled the first polar body was transferred into the perivitelline space of MⅡ oocyte which the first polar body had been enucleated. The nuclear transfer embryo was fused by electrofusion, reconstructed embryo was activated with7%ethanol for5min united with2.5mmol/L6-DMAP for4h, then we can obtain the mouse diploid parthenogenetic embryo contained genetic materials from primary oocyte with quickly activated and matured nucleus of new-born mouse and MⅡ oocyte.
     2. Isolation, culture and identification of ESCs from blastocysts derived from in vivo fertilization, parthenogenetic activation, and reconstruction of embryo
     2.1Isolation, culture and identification of ESCs from blastocysts derived from in vivo fertilization
     13.5dpc mouse embryos were collected for the isolation of MEFs. Mitomycin C-treated MEFs at the third passages for preparing the feeder layer. ICMs were isolated from4dpc mouse embryos with intact embryo method and immunosurgury method, then they were transferred on the feeder layer and cultured in ESCs medium, finally cell colonies on the feeder layer were identified. Results indicated that there were no significantly differences in colony formation rate between intact embryo method and immunosurgury method (30.26±2.77%vs32.92±4.37%, P>0.05). The pluripotent nature of ESCs isolated with intact embryo method and immunosurgury method was identified by typical cellular and colonial morphology of ESCs, positive AKP staining, normal karyotype, positive Oct-4and SSEA-1immunocytochemistry staining, expression of Oct-4and Sox-2. These ESCs could also spontaneously differentiate into embryoid bodies which could express Fgf-5、Bra T and Afp.
     2.2Isolation, culture and identification of ESCs from blastocysts derived from parthenogenetic activation
     Considering it was not easy for parthenogenetic blastocyst to hatch out from zona pellucida because of long time operation in vitro, immunosurgury method was used to isolated pESCs from uniform haploid blastocysts which developed from oocytes activated with SrCl2for4h, and pESCs from heterozygous diploid blastocysts which developed from oocytes activated with lOmM SrCl2and5μg/mL CB for4h, then cell colonies on the feeder layer were identified. Isolation efficiencies of pESCs from uniform haploid blastocysts and heterozygous diploid blastocysts were compared in order to select parthenogenetic blastocysts with suitable chromosome ploidy for the isolation of pESCs. Results indicated that the pluripotent nature of pESCs obtained in this study was identified by the criteria as listed in the identification of ESCs from blastocysts derived from in vivo fertilization. The colonies formation rate of pESCs from heterozygous diploid blastocysts was significantly higher than that from uniform haploid blastocysts (13.54±4.43%vs7.17±4.45%, P<0.05)。 Therefore, we should select the parthenogenetic activation method which could result in higher formation rate of heterozygous diploid blastocysts for the isolation of pESCs.
     2.3Isolation, culture and identification of pESCs from blastocysts derived from reconstructed embryos
     Intact embryo method was used to isolated pESCs from mouse diploid parthenogenetic blastocysts contained genetic materials from primary oocyte with quickly activated and matured nucleus of new-born mouse and MII oocyte. Results indicated that180morula and blastocyst derived from reconstructed embryos, after being cultured for48h morula and blastocyst developed into hatched blastocyst, after being cultured for96h ICM outgrowth were generated on the MEFs feeder layer,26pESCs colonies were observed after the ICM were dissociated into clusters of cells and transferred on the MEFs feeder layer, the pESCs colonies formation rate was14.43±4.36%. The pluripotent nature of pESCs obtained in this study was identified by the criteria as listed in the identification of ESCs from blastocysts derived from in vivo fertilization.
     3. Cell-free ESCs extracts or-mediated derivation of multipotent stem cells from MEFs
     3.1Effects of SLO at different concentration on growth state of MEFs
     The aim of this study was to explore the comparatively suitable SLO concentration for reversible permeabilization of MEFs before those cells were used to reprogram into pluripotent stem cells using cell-free extracts, MEFs were incubated in PBS without Ca2+and Mg2+supplemented with SLO which was added to a final concentration of10U,25U,50U and100U SLO, cells density, survival rate and adherence rate of MEFs were measured. Results were as follows:No significant difference was found in cells density of MEFs between25U SLO treated group and10U SLO treated group (p>0.05), but the cells density of MEFs permeabilized with10U SLO or25U SLO was significantly higher than those permeabilized with50U SLO or100U SLO respectively (p<0.01). There was no significantly difference in the survival rate of MEFs permeabilized with25U SLO compared with the0U or10U SLO group respectively (p>0.05), but was significantly higher than those permeabilized with50U SLO (p<0.05). The adherence rate of MEFs permeabilized with25U SLO and resealed in2mM CaCl2was significantly higher than that with50U or100U SLO (p<0.01). Proliferation of MEFs permeabilized with25U SLO was normal. Therefore,25U SLO was better suited to permeabilize MEFs considering both improving the efficiency of permeabilization and reducing the damage to MEFs.
     3.2Preparation of ESCs extract from Kunming mouse blastocysts derived from in vivo fertilization
     Effects of ultrasonic waves treatment and repeatedly freezing and thawing in liquid nitrogen on preparation of ESCs extract were explored in this study to obtain the high-efficiency lysis method to prepare the ESCs extract for programming of somatic cells. In order to detect and reduce the possibility of ESCs contamination during the preparation of ESCs extracts, ESCs extracts were transferred on the feeder layer and cultured in vitro, and MEFs without being permeablizated with streptolysin O were added to the ESCs extracts and were used to perform immunocytochemistry staining for Oct-4. Results were as follows:the vast majority of ESCs could be lysed after being repeatedly frozen and thawed in liquid nitrogen for seven times, the protein concentration were higher than that in ultrasonic waves treatment group (35.6mg/ml vs14.3mg/ml). There were no ESCs colonies observed on the feeder layer, MEFs without being permeablizated with streptolysin O revealed negative reaction to Oct-4immunocytochemstry staining. Therefore, ESCs were lysed and intracellular substances were destroyed as less as possible after ESCs were repeatedly freezing and thawing in liquid nitrogen for seven times, it provided a incubated culture system for reprogramming of somatic cells.
     3.3Studies on incubated system for mouse ESCs extract and MEFs
     Efficiencies of male MEFs which being pretreated with TSA and5-aza-dC or not being reprogrammed into multipotent stem cells by three kinds of female ESCs extracts respectively were compared in this study, male MEFs treated with male MEFs extracts and male MEFs without being treated with extracts were acted as controls, the pluripotent nature of multipotent stem cells obtained in this study was identified by the criteria as listed in the identification of ESCs from blastocysts derived from in vivo fertilization, the aim of the study was to optimize the reprogramming system which mediated by ESCs extracts. Results were as follows:male MEFs could be reprogrammed into multipotent stem cells by the female ESCs which be identified by dual PCR for sex identification system extracts from blastocysts derived from in vitro fertilization, parthenogenetic activation, and reconstruction of embryo respectively. The pluripotent nature of multipotent stem cells obtained in this study was identified by the criteria as listed in the identification of ESCs from blastocysts derived from in vivo fertilization. The number of colonies of multipotent stem cells from male MEFs which being treated with female ESCs extracts from blastocysts derived from in vivo fertilizationwere largest (64.33±3.7、80.67±2.08), following that of blastocysts derived from reconstruction of embryo (45.00±4.36、53.67±3.06), and that of blastocysts derived from parthenogenetic activation (35.67±2.52、47.67±4.16). Reprogramming of male MEFs could be improved by the pretreatment with TSA and5-Aza-dC, but it could not obtain the colonies of multipotent stem cells from male MEFs only pretreated with TSA and5-Aza-dC. Colony of multipotent stem cells was not be observed in control groups. Therefore, components which could reprogram the male MEFs into multipotent stem cells were from the ESCs extracts.
     4. Identification of the origin of multipotent stem cells from MEFs which been reprogrammed by ESCs extracts
     To rule out the possibility of ESCs contamination during the preparation of ESCs extracts, and assess the reliability of the reprogramming system, further pluripotential stem cells generation were performed using male MEFs which were identified by dual PCR for sex identification system(which can amplify250bp Sry gene and399bp ZFX gene) of a different gender from extract-donor ESCs. The donors of ESCs-derived extract were a parthenogenetic embryonic stem cells (pESCs) from the oocytes (which can only amplify399bp ZFX gene) which been parthenogenetic activated and the female ESCs identified with dual PCR for sex identification system from the blastocyst produced from in vivo fertilization. Therefore, we can decided the origin of the pluripotential stem cells according to their gender using dual PCR for sex identification system. Results indicated that40colonies of multipotent stem cells from8batch reprogramming (5colonies of multipotent stem cells were randomly selected in every batch) which mediated by pESCs extracts, pESCs from the reconstructed embryos extract and female extract from in vivo fertilization were used to perform dual PCR for sex identification. Multipotent stem cells which had been amplified250bp Sry gene and399bp ZFX gene, that was there were male colonies. Therefore, the origin of multipotent stem cells obtained in our study were male MEFs, ESCs extracts were not the contaminated by donor ESCs during the preparation of pESCs extracts.
     5. Feasibility of interspecies somatic cells reprogramming-mediated by ESCs extracts
     Chicken embryonic fibroblasts which being act as the target cells of reprogramming and were incubated with mouse ESCs extracts from Kunming mouse blastocysts derived from in vivo fertilization, morphology changes were observed, and the pluripotent nature of cells obtained from chicken embryonic fibroblasts after being reprogrammed by mouse ESCs extracts were identified, in order to explore the feasibility of reprogramming between the different species. Results indicated that after be incubated with mouse ESCs extracts, reprogrammed chicken embryonic fibroblasts turned to be round, fusiform adherent cells were not observed, and42.33±4.5colonies were generated. The pluripotent nature of the colonies was identified by typical cellular and colonial morphology of ESCs, positive AKP staining, and positive Oct-4and SSEA-1immunocytochemistry staining, and results from karyotype analysis indicated that those cells were derived from chicken embryonic fibroblasts. Chicken embryonic fibroblasts incubated with chicken embryonic fibroblasts extracts or without incubated with any extracts were not appeared the pluripotent nature, Therefore, components which could reprogram the chicken embryonic fibroblasts into multipotent stem cells were from the mouse ESCs extracts. Interspecies somatic cells reprogramming between mouse ESCs and chicken embryonic fibroblasts could be mediated by ESCs extracts.
     Conclusions:MEFs pretreated with SLO, TSA and5-Aza-dC could be reprogram ed into multipotent stem cells with cell-free ESCs extract in a shorter period. One day after treated with ESCs extract, MEFs formed the round cells, colonies with defined edges developed and resembled ESCs colonied within4days, these colonies showed the AKP activity on day3and they express Oct-4, Sox2and Nanog on day7. Chicken embryonic fibroblasts could be reprogrammed into multipotent stem cells using the reprogramming system mediated by mouse ESCs extract.
引文
[1]Briggs R, King T J. Transplantation of living nuclei from blastula cells into enucleated Frogs' eggs[J]. Proc Natl Acad Sci USA,1952,38(5):455
    [2]Gurdon J B, Elsdale T R, Fischberg M. Sexually mature individuals of xenopus laevis from the transplantation of single somatic nuclei [J]. Nature,1958,182 (4627):64-65
    [3]Campbell K H, Mc Whir J, Ritchie W A, et al. Sheep cloned by nuclear transfer from a cultured cell line[J]. Nature,1996,380(6569):64-66
    [4]Hochedlinger K, Jaenisch R. Monoclonal mice generated by nuclear transfer from mature B and T donor cells[J]. Nature,2002,415(6875):1035-1038
    [5]Rideout W M, Eggan K, Jaenisch R. Nuclear cloning and epigenetic reprogramming of the genome[J]. Science,2001,293,1093-1098
    [6]Rideout W M, Wckayama T, Wutz A, et al. Generation of mice from wild-type and targeted ES cells by nuclear cloning[J]. Nature Genet,2000,24,109-110
    [7]Humpherys D, Eggan K, Akutsu H, et al. Epigenetic instability in ES cells and cloned mice[J]. Science,2001,293,95-97
    [8]Wakayama T, Yanagimachi R. Mouse cloning with nucleus donor cells of different age and type[J]. Mol Reprod Dev,2001,58:376-383
    [9]Weissman I L. Stem cells:units of development, units of regeneration, and units in evolution[J]. Cell,2000,100:157-168
    [10]Liu L. Cloning efficiency and differentiation[J]. Nature Biotechnol,2001,19:406
    [11]Kubota C, Yamakuchi H, Todoroki J, et al. Six cloned calves produced from adult fibroblast cells after long-term culture[J]. Proc Natl Acad Sci USA,2000,97(3):990-995
    [12]Behboodi E, Memili E, Melican D T, et al. Viable transgenic goats derived from skin cells [J]. Transgenic Res,2004,13(3):215-224
    [13]Polejaeva I A, Chen S H, Vaught T D, et al Clones pigs produced by nuclear transfer from adult somatic cell[J]. Nature,2000,407(6800):86-90
    [14]Li S, Chen X, Fang Z, et al. Rabbits generated from fibroblasts through nuclear transfer[J]. Reproduction,2006,131(6):1085-1090
    [15]Lagutina I, Lazzari G, Duchi R, et al. Somatic cell nuclear transfer in horses:effect of oocyte morphology, embryo reconstruction method and donor cell type[J]. Reproduction,2005,130(4): 559-567
    [16]Takahashi S, Ito Y. Evaluation of meat products from cloned cattle:Biological and biochemical properties[J]. Cloning Stem Cells,2004,6(2):165-171
    [17]Tome D, Dubarry M, Fromentin G. Nutritional value of milk and meat products derived from cloning[J]. Cloning Stem Cells.2004,6(2):172-177
    [18]Rudenko L, Matheson J C. The US FDA and animal cloning:Risk and regulatory approach[J]. Theriogenology,2007,67(1):198-206
    [19]Rudenko L, Matheson J C, Sundlof S F. Animal cloning and the FDA-the risk assessment paradigm under public scrutiny[J]. Nat Biotechnol,2007,25(1):39-43
    [20]Kawase E, Yamazaki Y, Yagi T, et al. Mouse embryonic stem (ES) cell lines established from neuronal cell-derived cloned blastocysts[J]. Genesis,2000,28:156-163
    [21]Wakayama T, Tabar V, Rodriguez I, et al. Differentiation of embryonic stem cell lines generated from adult somatic cells by nuclear transfer[J]. Science,292,2001,740-743
    [22]Colman A, Kind A. Therapeutic cloning:concepts and practicalities [J]. Trends Biotechnol,2000, 18:192-196
    [23]Rideout WM, Hochedlinger K, Kyba M, et al. Correction of a genetic defect by nuclear transplantation and combined cell and gene therapy[J]. Cell,2002,109(1):17-27
    [24]Sumer H, Liu J, Tat PA, et al. Somatic cell nuclear transfer[J]. Pros & cons. J Stem Cells,2009, 4(2):85-93
    [25]French A J, Adams C A, Anderson L S, et al. Development of human cloned blastocysts following somatic cell nuclear transfer with adult fibroblasts[J]. Stem Cells,2008,26(2): 485-93
    [26]Li J, Liu X, Wang H, et al. Human embryos derived by somatic cell nuclear transfer using an alternative enucleation approach[J]. Cloning Stem Cells,2009,11(1):39-50
    [27]Stojkovic M, Stojkovic P, Leary C, et al. Derivation of a human blastocyst after heterologous nuclear transfer to donated oocytes[J]. Reprod Biomed Online,2005,11(2):226-231
    [28]Cibelli J B, Campbell K H, Seidel G E, et al. The health profile of cloned animals [J]. Nat Biotechnol,2002,20 (1):13-14
    [29]Niwa H, Toyooka Y, Shimosato D, et al. Interaction between Oct3/4 and Cdx 2 determines trophectoderm differentiation [J]. Cell,2005,123 (5):917-929
    [30]Han J, Sidhu KS. Current concepts in reprogramming somatic cells to pluripotent state[J]. Curr Stem Cell Res Ther,2008,3(1):66-74
    [31]Miller R A, Ruddle F H. Pluripotent teratocarcinoma-thymus somatic cell hybrids [J]. Cell,1976, 9(1):45-55
    [32]Miller R A, Ruddle F H. Teratocarcinoma X friend erythroleukemia cell hybrids resemble their pluripotent embryonal carcinoma parent[J]. Dev Biol,1977,56(1):157-173
    [33]Miller R A, Ruddle F H. Properties of teratocarcinoma-thymus somatic cell hybrids[J]. Somatic Cell Genet,1977,3(3):247-261
    [34]Andrews P W, Goodfellow P N. Antigen expression by somatic cell hybrids of a murine embryonal carcinoma cell with thymocytes and L cells[J]. Somatic Cell Genet,1980,6(2): 271-284
    [35]Forejt J, Gregorova S, Dohnal K, et al. Gene expression of differentiated parent in teratocarcinoma cell hybrids. Repression or reprogramming[J]? Cell Differ,1984,15(2-4): 229-234
    [36]Flasza M, Shering AF, Smith K, et al. Reprogramming in inter-species embryonal carcinoma-somatic cell hybrids induces expression of pluripotency and differentiation markers[J]. Cloning Stem Cells,2003,5(4):339-354
    [37]Hajkova P, Erhardt S, Lane N, et al. Epigenetic reprogramming in mouse primordial germ cells [J]. Mech Dev,2002,117(1-2):15-23
    [38]Tada M, Tada T, Lefebvre L, et al. Embryonic germ cells induce epigenetic reprogramming of somatic nucleus in hybrid cells[J]. EMBO J,1997,16(21):6510-6520
    [39]Matveeva N M, Shilov A G, Kaftanovskaya E M, et al. In vitro and in vivo study of pluripotency in intraspecific hybrid cells obtained by fusion of murine embryonic stem cells with splenocytes[J]. Mol Reprod Dev,1998,50(2):128-138
    [40]Tada M, Takahama Y, Abe K, et al. Nuclear reprogramming of somatic cells by in vitro hybridization with ES cells[J]. Curr Biol,2001,11(19):1553-1558
    [41]Cowan C A, Atienza J, Melton D A, et al. Nuclear reprogramming of somatic cells after fusion with human embryonic stem cells [J]. Science,2005,309 (5739):1369-1373
    [42]Silva J, Chambers I, Pollard S, et al. Nanog promotes transfer of pluripotency after cell fusion [J]. Nature,2006,441 (7096):997-1001
    [43]Yu J, Vodyanik M A, He P, et al. Human embryonic stem cells reprogram myeloid precursors following cell-cell fusion[J]. Stem Cells,2006,24(1):168-176
    [44]Ma D K, Chiang C H, Ponnusamy K, et al. G9a and Jhdm-a regulate embryonic stem cell fusion-induced reprogramming of adult neural stem cells [J]. Stem Cells,2008,26(8): 2131-2141
    [45]Bunn C L, Eisenstadt J M. Cybrid formation in mouse L cells:the influence of cytoplast-to-cell ratio[J]. Somatic Cell Genet,1977,3(3):335-341
    [46]Strelchenko N, Kukharenko V, Shkumatov A, et al. Reprogramming of human somatic cells by embryonic stem cell cytoplast[J]. Reprod Biomed Online,2006,12(1):107-111
    [47]Pralong D, Lim ML, Vassiliev I, et al. Tetraploid embryonic stem cells contribute to the inner cell mass of mouse blastocysts[J]. Cloning Stem Cells,2005,7(4):272-278
    [48]Pralong D, Mrozik K, Occhiodoro F, et al. A novel method for somatic cell nuclear transfer to mouse embryonic stem cells[J]. Cloning Stem Cells,2005,7(4):265-271
    [49]Matsumura H, Tada M, Otsuji T, et al. Targeted chromosome elimination from ES-somatic hybrid cells [J]. Nat Methods,2007,4(1):23-25
    [50]Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors [J]. Cell,2006,126(4):663-676
    [51]Hanna J, Wernig M, Markoulaki S, et al. Treatment of sickle cell anemia mouse model with iPS cells generated from autologous skin[J]. Science,2007,318(5858):1920-1923
    [52]Okita K, Ichisaka T, Yamanaka S. Generation of germline-competent induced pluripotent stem cells [J]. Nature,2007,448:313-317
    [53]Sumer H, Jones KL, Liu J, et al. Reprogramming of somatic cells after fusion with induced pluripotent stem cells and nuclear transfer embryonic stem cells[J]. Stem Cells Dev,2010,19(2): 239-246
    [54]Liu J, Sumer H, Leung J, et al. Late passage human fibroblasts induced to pluripotency are capable of directed neuronal differentiation[J]. Cell Transplant,2011,20(2):193-203
    [55]Tat PA, Sumer H, Jones KL, et al. The efficient generation of induced pluripotent stem (iPS) cells from adult mouse adipose tissue-derived and neural stem cells[J]. Cell Transplant,2010, 19(5):525-536
    [56]Takahashi K, Tanabe K, Ohnuki M, et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors[J]. Cell,2007,131(5):861-872
    [57]Yu J, Vodyanik M A, Smuga-Otto K, et al. Induced pluripotent stem cell lines derived from human somatic cells[J]. Science,2007,318(5858):1917-1920
    [58]Liu H, Zhu F, Yong J, et al. Generation of induced pluripotent stem cell from adult rhesus monkey fibroblast[J]. Cell Stem Cell,2008,3(6):587-590
    [59]Liao J, Cui C, Chen S, et al. Generation of induced pluripotent stem cell lines from adult rat cells[J]. Cell Stem Cell,2009,4(1):11-15
    [60]Ezashi T, Telugu B P, Alexenko A P, et al. Derivation of induced pluripotent stem cells from pig somatic cells[J]. Proc Natl Acad Sci U S A,2009,106(27):10993-10998
    [61]MeGann C J, Odelberg S J, Keating M T. Mammalian myotube dedifferentiation induced by newt regeneration extract [J]. Proc Natl Acad Sci USA,2001,98 (24):13699-13704
    [62]Hansis C, Barreto G, Maltry N, et al. Nuclea r reprogramming of human somatic cells by xenopus egg extract requires BRG1[J]. CUIT Biol,2004,14(16):1475-1480
    [63]Taranger C K., Noer A, Serensen A L, et al. Induction of dedifferentiation, genomewide transcriptional programming, and epigenetic reprogramming by extracts of carcinoma and embryonic stem cells[J]. Mol Biol Cell,2005,16(12):5719-5735
    [64]Pewsey E, Bruce C, Georgiou AS, et al. Proteomics analysis of epithelial cells reprogrammed in cell-free extract[J]. Mol Cell Proteomics,2009,8(6):1401-1412
    [65]Zhang X M, Li Q M, Su D J, et al. RA induces the neural-like cells generated from epigenetic modified NIH/3T3 cells[J]. Mol. Biol. Rep,2010,37:1197-1202
    [66]Zhang X M, Wang N, Li D M, et al. Induction of epigenetic reprogramming in fibroblast by extracts of carcinoma [J]. African Journal of Biotechnology,2012,11(12):2855-2861
    [68]Collas P, Taranger C K. Epigenetic reprogramming of nuclei using cell extracts [J]. Stem Cell Rev,2006,2 (4):309-317
    [69]Bru T, Clarke C, McGrew M J, et al. Rapid induction of pluripotency genes after exposure of human somatic cells to mouse ES cell extracts [J]. Exp Cell Res,2008,314 (14):2634-2642
    [70]Oswald J, Engemann S, Lane N, et al. Active demethylation of the paternal genome in the mouse zygote[J]. Curr Biol,2000,10:475-478
    [71]Mayer W, Niveleau A, Walter J, et al. Demethylation of the zygotic paternal genome[J]. Nature, 2000,403:501-502
    [72]Santos F, Hendrich B, Reik W, et al. Dynamic reprogramming of DNA methylation in the early mouse embryo[J]. Dev Biol,2002,241:172-182
    [73]Olek A, Walter J. The pre-implantation ontogeny of the H19 methylation imprint[J]. Nat Genet, 1997,17:275-276
    [74]Dean W, Santos F, Stojkovic M, et al. Conservation of methylation reprogramming in mammalian development:aberrant reprogramming in cloned embryos [J]. Proc Natl Acad Sci USA,2001,98:13734-13738
    [75]Ram P T, Schultz R M. Reporter gene expression in G2 of the 1-cell mouse embryo [J]. Dev Biol,1993,156:552-556
    [76]Reik W, Walter J. Evolution of imprinting mechanisms:the battle of the sexes begins in the zygote[J]. Nat Genet,2001,27:255-256
    [77]Howlett S K, Reik W. Methylation levels of maternal and paternal genomes during preimplantation development [J]. Development,1991,113:119-127
    [78]Carlson L L, Page A W, Bestor T H. Properties and localization of DNA methyltransferase in preimplantation mouse embryos:implications for genomic imprinting [J]. Genes Dev,1992,6: 2536-2541
    [79]Rougier N, Bourc'his D, Gomes DM, et al. Chromosome methylation patterns during mammalian preimplantation development[J]. Genes Dev,1998,12:2108-2113
    [80]Howell C Y, Bestor T H, Ding F, et al. Genomic imprinting disrupted by a maternal effect mutation in the Dnmtl gene[J]. Cell,2001,104:829-838
    [81]Jackson M, Krassowska A, Gilbert N, et al. Severe global DNA hypomethylation blocks differentiation and induces histone hyperacetylation in embryonic stem cells [J]. Mol Cell Biol, 2004,24:8862-8871
    [82]Surface L E, Thornton S R, Boyer L A. Polycomb group proteins set the stage for early lineage commitment[J]. Cell Stem Cell,2010,7:288-298
    [83]Meissner A, Mikkelsen T S, Gu H, et al. Genome-scale DNA methylation maps of pluripotent and differentiated cells [J]. Nature,2008,454:766-770
    [84]Kapranov P, Cheng J, Dike S, et al. RNA maps reveal new RNA classes and a possible function for pervasive transcription[J]. Science,2007,316:1484-1488
    [85]Mallanna S K, RizzinoA. Emerging roles of microRNAs in the control of embryonic stem cells and the generation of induced pluripotent stem cells [J]. Dev Biol,2010,344:16-25
    [86]Suh M R, Lee Y, Kim J Y, et al. Human embryonic stem cells express a unique set of microRNAs[J]. Dev Biol,2004,270(2):488-498
    [87]Li X, Jin P. Roles of small regulatory RNAs in determining neuronal identity[J]. Nat Rev Neurosci,2010,11:329-338
    [88]Chen C Z, Li L, Lodish H F, et al. MicroRNAs modulate hematopoietic lineage differentia-tion[J]. Science,2004,30 (5654):83-86
    [89]Krichevsky A M, Sonntag K C, Isacson O, et al. Specific microRNAs modulate embryonic stem cell-derived neurogenesis[J]. Stem Cells,2006,24(4):857-864
    [90]Boyer L A, Lee T I, Cole M F, et al. Core transcriptional regulatory circuitry in human embryonic stem cells[J]. Cell,2005,122(6):947-956
    [91]Mikkelsen T S, Ku M, Jaffe D B, et al. Genomewide maps of chromatin state in pluripotent and lineage-committed cells [J]. Nature,2007,448:553-560
    [92]Hawkins R D, Hon G C, Lee L K,et al. Distinct epigenomic landscapes of pluripotent and lineage-committed human cells[J]. Cell Stem Cell,2010,6:479-491
    [93]Wen B, Wu H, Shinkai Y, et al. Large histone H3 lysine 9 dimethylated chromatin blocks distinguish differentiated from embryonic stem cells[J]. Nat. Genet.2009,41:246-250
    [94]Filion G J, Van Steensel B. Reassessing the abundance of H3K9me2 chromatin domains in embryonic stem cells[J]. Nat Genet,2010,42(4):5-6
    [95]Wen B, Wu H, Shinkai Y, et al. Reply to [ldquo] Reassessing the abundance of H3K9me2 chromatin domains in embryonic stem cells[J]. Nat. Genet,2010,42:5-6
    [96]Humburg P, Bulger D, Stone G. Parameter estimation for robust HMM analysis of ChIP-chip data[J]. BMC Bioinformatics,2008,9:343
    [97]Dodge J E, Kang Y K, Beppu H, et al. Histone H3-K9 methyltransferase ESET is essential for early development[J]. Mol Cell Biol,2004,24:2478-2486
    [98]Pasini D, Bracken A P, Hansen J B, et al. The polycomb group protein Suz12 is required for embryonic stem cell differentiation [J]. Mol Cell Biol,2007,27:3769-3779
    [99]Shi Y, Desponts C, Do J T, et al. Induction of pluripotent stem cells from mouse embryonic fibroblasts by Oct4 and Klf4 with small-molecule compounds[J]. Cell Stem Cell,2008,3: 568-574
    [100]Shi Y, Do J T, Desponts C, et al. A combined chemical and genetic approach for the generation of induced pluripotent stem cells[J]. Cell Stem Cell,2008b,2:525-528
    [101]Wendt K S, Yoshida K, Itoh T, et al. Cohesin mediates transcriptional insulation by CCCTC-binding factor[J]. Nature,2008,451:796-801
    [102]Onder T T, Kara N, Cherry A, et al. Chromatin-modifying enzymes as modulators of reprogramming[J]. Nature,2012,483(7391):598-602
    [103]Hansis C, Barreto G, Maltry N, et al. Nuclear reprogramming of human somatic cells by Xenopus egg extract requires BRG1 [J]. Curr Biol,2004,14(16):1475-1480
    [104]Miyamoto K, Tsukiyama T, Yang Y, et al. Cell-free extracts from mammalian oocytes partially induce nuclear reprogramming in somatic cells[J]. Biol Repro,2009,80(5):935-943
    [105]Rathbone A J, Fisher P A, Lee J H, et al. Reprogramming of ovine somatic cells with Xenopus laevis oocyte extract prior to SCNT improves live birth rate[J]. Cell Reprogram,2010,12(5): 609-616
    [106]Rajasingh J, Lambers E, Hamada H, et al. Cell-free embryonic stem cell extract-mediated derivation of multipotent stem cells from NIH3T3 fibroblasts for functional and anatomical ischemic tissue repair[J]. Circulation Research,2008,16(11):e107-e117
    [107]Xu Y N, Guan N, Wang Z D, et al. ES cell extract-induced expression of pluripotent factors in somatic cells [J]. The Anatomical Record,2009,292(8):1229-1239
    [108]Han J, Sachdev P S, Sidhu K S. A combined epigenetic and non-genetic approach for reprogramming human somatic cells[J]. PLoS One,2010,19; 5(8):e12297
    [109]Zhan W J, Liu Z P, Liu Y, et al. Modulation of rabbit corneal epithelial cells fate using embryonic stem cell extract[J]. Molecular Vision,2010,16:1154-1161
    [110]Hakelien A M, Landsverk H B, Robl J M, et al. Reprogramming fibroblasts to express T-cell functions using cell extract [J]. Nat Biotechnol,2002,20(5):460-466
    [111]Hakelien A M, Gaustad K G, Collas P. Transient alteration of cell fate using a nuclear and cytoplasmic extract of an insulinoma cell line[J]. Biochem Biophys Res Commun,2004, 316(3):834-841
    [112]Gaustad K G, Boquest A C, Anderson B E, et al. Differentiation of human adipose tissue stem cells using extract of rat cardiomyocytes[J]. Biochem Biophys Res Commun,2004,314(2): 420-427
    [113]Schimrosczyk K, Song Y H, Vykoukal J, et al. Liposome-mediated transfection with extract from neonatal rat cardiomyocytes induces transdifferentiation of human adipose-derived stem cells into cardiomyocytes [J]. Scand J Clin Lab Invest,2008,68(6):464-472
    [114]Labovsky V, Hofer E L, Feldman L, et al. Cardiomyogenic differentiation of human bone marrow mesenchymal cells:Role of cardiac extract from neonatal rat cardiomyocytes [J]. Differentiation,2010,79(2):93-101
    [115]Peran M, Marchal JA, Lopez E, et al. Human cardiac tissue induces transdifferentiation of adult stem cells towards cardiomyocytes [J]. Cytotherapy,2010,12(3):332-337
    [116]Allegrucci C, Rushton M D, Dixon J E, et al. Epigenetic reprogramming of breast cancer cells with oocyte extracts[J]. Mol Cancer,2011,10(1):7-21
    [117]Peran M, Sanchez-Ferrero A, Tosh D,er al. Ultrastructural and molecular analyzes of insulin-producing cells induced from human hepatoma cells[J]. Cytotherapy,2011,13(2): 193-200
    [118]Vojdani Z, Tavakolinejad S, Talaei-Khozani T, et al. Cardiomyocyte marker expression in a human lymphocyte cell line using mouse cardiomyocyte extract[J]. Hum cell,2011,24(1): 35-42
    [119]Tang X, Sheng L, Xie F, et al. Differentiation of bone marrow-derived mesenchymal stem cells into chondrocytes using chondrocyte extract[J]. Mol Med Report,2012,6(4):745-749
    [120]Xu Y, Shi Y, Ding S. A chemical approach to stem-cell biology and regenerative medicine[J]. 2008, Nature,453:338-344
    [121]Mikkelsen T S, Hanna J, Zhang X L, et al. Dissecting direct reprogramming through integrative genomic analysis[J]. Nature,2008,454(3):49-56
    [122]张喜梅,郭铁云,闫晓飞,等.5-aza-dC和TSA对NIH/3T3细胞多能性基因表达作用的实验研究[J].解剖科学进展,2010,16(5):405-408
    [123]Talaei-Khozani T, Kharazinejad E, Rohani L, et al. Expression of pluripotency markers in human granulosa cells after embryonic stem cell extract exposure and epigenetic modification[J]. Iran J Reprod Med,2012,10(3):193-200
    [124]Huangfu D, Maehr R, Guo W, et al. Induction of pluripotent stem cells by defined factors is greatly improved by small-molecule compounds[J]. Nat Biotechnol,2008,26,795-797
    [125]Huangfu D, Osafune K, Maehr R, et al. Induction of pluripotent stem cells from primary human fibroblasts with only Oct4 and Sox2[J]. Nat. Biotechnol,2008,26:1269-1275
    [126]Kishigami S, Bui H T, Wakayama S, et al. Successful mouse cloning of an outbred strain by trichostatin A treatment after somatic nuclear transfer[J]. J Reprod Dev,2007,53:165-170
    [127]Zhao J, Hao Y, Ross J W, et al. Histone deacetylase inhibitors improve in vitro and in vivo developmental competence of somatic cell nuclear transfer porcine embryos [J]. Cell Reprogram,2010,12:75-83
    [128]Akagi S, Matsukawa K, Mizutani E, et al. Treatment with a histone deacetylase inhibitor inhibitor after nuclear transfer improves the preimplantation development of cloned bovine embryos[J]. J Reprod Dev,2011,57:120-126
    [129]Shi L H, Ai J S, Ouyang Y C, et al. Trichostatin a and nuclear reprogramming of cloned rabbit embryos [J]. J Anim Sci,2008,86(5):1106-1113
    [130]Enright B P, Kubota C, Yang X, et a.l Epigenetic characteristics and developmentofembryos cloned from donorcells treated by trichostatin A or 5-aza-2'-deoxycytidine [J]. Biol Reprod, 2003,69:896-901
    [131]张东,杨鹭,王勇胜,等.TSA处理供体细胞对组蛋白乙酰化和核重编程效果的影响[J].畜牧兽医学报,2009,40(7):1007-1012
    [132]Huangfu D, Maehr R, Guo W, et al. Induction of pluripotent stem cells by defined factors is greatly improved by small-molecule compounds[J]. Nat Biotechnol,2008,26,795-797
    [133]Shi W, Hoeflich A, Flaswinkel H, et al. Induction of a senescent-like phenotype does not confer the ability of bovine mimortal cells to support the development of nuclear transfer embryos[J]. BiolReprod,2003,69:301-309
    [134]Mali P, Chou B, Yen J, et al. Butyrate greatly enhances derivation of human induced pluripotent stem cells by promoting epigenetic remodeling and the expression of pluripotency-associated genes[J]. Stem Cells,2010,28(4):713-720
    [135]Martinez-Iglesias O, Ruiz-Llorente L, Sanchez-Martinez R, et al. Histone deacetylase inhibitors:mechanism of action and therapeutic use in cancer[J]. Clin Transl Oncol,2008, 10(7):395-398
    [136]Bolden J E, Peart M J, Johnstone R W. Anticancer activities of histone deacetylase inhibitors[J]. Nat Rev Drug Discov,2006,5(9):769-784
    [137]Pandian G N, Shinohara K, Ohtsuki A, et al. Synthetic small molecules for epigenetic activation of pluripotency genes in mouse embryonic fibroblasts[J]. Chembiochem,2011, 12(18):2822-2828
    [138]Pandian G N, Nakano Y, Sato S, et al. A synthetic small molecule for rapid induction of multiple pluripotency genes in mouse embryonic fibroblasts[J]. Scientific Reports,2012,2: 544
    [139]Feldman N, Gerson A, Fang J, et al. G9a-mediated irreversible epigenetic inactivation ofOct-3/4 during early embryogenesis[J]. Nat Cell Biol,2006,8:188-194
    [140]Epsztejn-Litman S, Feldman N, Abu-Remaileh M, et al. De novo DNA methylation promoted by G9a prevents reprogramming of embryonically silenced genes[J]. Nat Struct Mol Biol, 2008,15:1176-1183
    [141]Mezentseva N V, Yang J, Kaur K, et al. The histone methyltransferase inhibitor BIX01294 enhances the cardiac potential of bone marrow cells [J]. Stem Cells Dev,2012 Sep 20, doi: 10.1089/scd.2012.0181
    [142]Feng B, Ng J, Heng J D, et al. Molecules that promote or enhance reprogramming of somatic cells to induced pluripotent stem cells [J]. Cell StemCell,2009,4(4):301-312
    [143]Ying Q L, Nichols J, Chambers I, et al. BMP induction of Id proteins suppresses differentiation and sustains embryonic stem cell self-renewal in collaboration with STAT3[J]. Cell,2003,115:281-292
    [144]Ying Q L, Wray J, Nichols J, et al. The ground state of embryonic stem cell self-renewal [J]. Nature,2008,453:519-523
    [145]Buehr M, Meek S, Blair K, et al. Capture of authentic embryonic stem cells from rat blastocysts[J]. Cell,2008,135:1287-1298
    [146]Kunath T, Saba-El-Leil M K, Almousailleakh M, et al. FGF stimulation of the Erkl/2 signalling cascade triggers transition of pluripotent embryonic stem cells from self-renewal to lineage commitment[J]. Development,2007,134:2895-2902
    [147]Sato N, Meijer L, Skaltsounis L, et al. Maintenance of pluripotency in human and mouse embryonic stem cells through activation of Wnt signaling by a pharmacological GSK-3-specific inhibitor[J]. Nat Med,2004,10:55-63
    [148]Silva J, Barrandon O, Nichols J, et al. Promotion of reprogramming to ground state pluripotency by signal inhibition[J]. PLoS Biol,2008,6(10):2237-2247
    [149]Li W, Wei W, Zhu S, et al. Generation of rat and human induced pluripotent stem cells by combining genetic reprogramming and chemical inhibitors[J]. Cell StemCell,2009,4(1): 16-19
    [150]Yu J, Thomson J A. Pluripotent stem cell lines[J]. Genes Dev,2008,22:1987-1997
    [151]Lin T, Ambasudhan R, Yuan X, et al. A chemical platform for improved induction of human iPSCs[J]. Nat Methods,2009,6 (11):805-808
    [152]Ichida J K, Blanchard J, Lam K, et al. A Small-molecule inhibitor of Tgf-β signaling replaces Sox2 in reprogramming by inducing Nanog[J]. Cell Stem Cell,2009,5(5): 491-503
    [153]Grigoryan T, Wend P, Klaus A, et al. Deciphering the function of canonical Wnt signals in development and disease:conditional loss- and gain-of-function mutations of beta-catenin in mice[J]. Genes Dev,2008,22:2308-2341
    [154]Marson A, Foreman R, Chevalier B, et al. Wnt signaling promotes reprogramming of somatic cells to pluripotency[J]. Cell Stem Cell,2008,3:132-135
    [155]Lluis F, Pedone E, Pepe S, et al. Periodic activation of Wnt/beta-catenin signaling enhances somatic cell reprogramming mediated by cell fusion[J]. Cell Stem Cell,2008,3:493-507
    [156]Cole M F, Johnstone S E, Newman J J, et al. Tcf3 is an integral component of the core regulatory circuitry of embryonic stem cells[J]. Genes Dev,2008,22:746-755
    [157]Tam W L, Lim C Y, Han J, et al. T-cell factor 3 regulates embryonic stem cell pluripotency and self-renewal by the transcriptional control of multiple lineage pathways [J]. Stem Cells, 2008,26,2019-2031
    [158]Yi F, Pereira L, Merrill B J. Tcf3 functions as a steady-state limiter of transcriptional programs of mouse embryonic stem cell self-renewal [J]. Stem Cells,2008,26:1951-1960
    [159]Yang W, Wei W, Shi C, et al. Pluripotin combined with LIF greatly promotes the derivation of ES cell lines from refractory strains [J]. Stem Cells,2009,27(2):383-389
    [160]Geens M, Mateizel I, Sermon K, et al. Human embryonic stem cell lines derived from single blastomeres of two 4-cell stage embryos[J]. Hum Reprod,2009,24(11):2709-2717
    [161]Li H, Lam A, Xu A M, et al. High dosage of Exendin-4 increased early insulin secretion in differentiated beta cells from mouse embryonic stem cells[J]. Acta Pharmacol Sin,2010,31(5): 570-577
    [162]Hovatta O, Jaconi M, Tohonen V, et al. A teratocarcinoma-like human embryonic stem cell (hESC) line and four hESC lines reveal potentially oncogenic genomic changes [J]. PLoS One, 2010,5(4):e10263
    [163]周结学,刘东,郑克立,等.鼠胚成纤维细胞的培养条件及滋养层制备[J].中国组织工程研究与临床康复,2008,12(34):6607-6611
    [164]Pitto L, Rizzo M, Simili M, et al. miR-290 acts as a physiological effector of senescence in mouse embryo fibroblasts[J]. Physiol Genomics,2009,39(3):210-218
    [165]Wang T, Chen K S, Zeng X M, et al. The histone demethylases Jhdm la/lb enhance somatic cell reprogramming in a vitamin-C-dependent matter [J]. Cell Stem Cell,2011,9(6):575-587
    [166]Williams R L, Hilton D J, Pease S, et al. Myeloid leukemia inhibitory factor maintains the development potential of embryonic stem cell[J]. Nature,1988,336:684-688
    [167]Smith Ag, Heath J K, Donaldson D D, et al. Inhibition of pluripopential embryonic stem cell differentiation by purified polypetides[J]. Nature,1988,336:688-671
    [168]Bhatt H, Rrunet L J, Steward C L. Uterine expression of luekaemia inhibitory factory coincides with onset of blastocyst implantation[J]. Proc Natl Acad Sci USA,1991,88: 11408-11412
    [169]Nachtigall M J, Kliman H J, Feinberg R F, et al. The effect of leukemia inhibitory factor (LIF) on trophoblast differentiation:a potential role in human implantation [J]. J Clin Endocrinol Metab,1996,81(2):801-806
    [170]Steward C L, Kaspar P, Bruner L J, et al. Blastocyst implantation depends on maternal expression of leukaemia inhibitory factor[J]. Nature,1992,359(6390):76-79
    [171]Ruck P, Marzusch K, Kaiserling E, et al. Distribution of cell adhesion molecules in decidua of early human pregnancy[J]. An immunohistochemical study. Lab Invest,1994,71(1):94-101
    [172]张炜,周剑萍,张俊慧,等.LIF对体外培养小鼠孕早期胚胎发育及ICAM-1表达的影响[J].上海医科大学学报,1999,26(4):255-257,264
    [173]刘红林.哺乳动物胚胎植入前发育的基因表达[J].国外医学遗传学分册,1995,18(2)90-93
    [174]刘红林,范必勤,宋卉.小鼠胎胚干细胞孤雌集落的建立[J].动物学报,1998,44(2):112-114
    [175]Prather R S, Mayes M A, Murphy C N. Parthenogenetic activation of pig eggs by exposure to protein Kinase inhibitors [J]. Rep rod Ferti 1Dev,1997,9 (5):539-544;
    [176]刘红林,汪河海,范必勤.乙醇激活诱导小鼠孤雌胚的发育与核型分析[J].畜牧与兽医,2000,32(1):7-8;
    [177]Ilyin V, Pavker I. Effects of alcohols on responses evoked by inositol trisphosphate in Xenopus oocytes[J]. Physiol,1992,448:339-54
    [178]陈大元主编.受精生物学——受精机制与生殖工程[M].北京:科学出版社,2000
    [179]Sagata N. Meiotic metaphase arrestin animal oocyte Its mechanism and biological significance[J]. Cell Biolog,1996,6:22-28
    [180]邓满齐,范必勤.小鼠卵激活过程中胞质游离Ca2+的变化及孤雌发育研究[J].实验生物学报,1994,27(3):289-297
    [181]Cuthbertson K S R, Whittingham D G, Cobbold P H. Free Ca2+ increase in exponential phases During mouse oocytes activation[J]. Nature,1981,294:754-757
    [182]兰国成,王子玉,马所峰.乙醇及6-DMAP对小鼠卵母细胞孤雌激活的研究[J].细胞生物学杂志,2002,24:307-409
    [183]Fan B Q. Oocyte meiotic maturation and development of parthenogentic and reconstituted embryos in mammal [J]. Jiangsu J of Agr sci,1999,15(2):116-126
    [184]刘红林,范必勤,陈宜峰.小鼠卵母细胞的乙醇激活[J].南京农业大学学报,1996,19:56-60
    [185]Hideki S, Masahiko I, Moritoshi S, et al. Measurement of intracellular IP3 during Ca2+ osicillations in mouse eggs with GFP-based FRET probe[J]. Biochemical and Biophysical Research Communication,2006,345:781-788
    [186]何志全,窦忠英.不同化学激活剂对小鼠卵母细胞的激活效果[J].中国兽医科学,2006,36(12):1013-1018
    [187]Liu L Yang X Z. Interplay of maturation-promoting factor and mitogen-activated protein kinase inactivation during metaphase-to interphase transition of activated bovine oocytes[J]. Biol Reprod,1999,61:1-7
    [188]Winston N J, Maro B. Calmodulin-dependent protein kinase II is activated transiently in ethanol-stimulated mouse oocytes[J]. Dev Biol.1995,170:350-352
    [189]刘红林,范必勤,汪河海.小鼠孤雌胚的体内体外发育研究[J].畜牧兽医学报,1999,30(2):103-109
    [190]Fan H Y, Sun Q Y. Involvement of mitogen-activated protein kinase cascade during oocyte maturation and fertilization in mammals[J]. Biol Reprod,2004,70:535-547
    [191]Sun Q Y, Lax Y, Rubinstein S, et al. Mitogen-activated protein kinase and cell cycle progression during mouse egg activation induced by various stimuli [J]. Z Naturforsch,1999, 54:285-294
    [192]Szollosi M S, Kubiak J Z, Debey P, at al. Inhibition of protein kinases by 6-dimethylamino-purine accelerates the transition to interphase in activated mouse oocytes[J]. J.Cell Sci.,1993, 104:861-872
    [193]Czolowaka R, Waksmundaka M, Kubiak J Z, et al. Chromosome condensation activity in ovulated metaphase Ⅱ mouse oocyte assayed by fusion with interphase blastomeres [J]. J Cell Sci,1986,84:129-133
    [194]Kono T, Obata Y, Yoshimizu T, et al. Epigenetic modifications during oocyte growth correlates with extended parthenogenetic development in the mouse[J]. Nature Genet,13,91-94.
    [195]白照岱,刘凯,邴鲁军.小鼠卵母细胞透明带膨胀辅助去核方法的研究.生殖医学杂志,2005,14(1):31-35
    [196]邵华,茫烈,其木格,等.去除卵丘细胞的小鼠卵母细胞体外成熟过程中的超微结构变化[J].畜牧与饲料科学,2006,27(2):17-19
    [197]Bronson R A, Cooper G. W, Rosenfeld D L. et al. The effect of an IgAl protease on immunoglobulins bound to the sperm surface and sperm cervical mucus penetrating ability[J]. Fertil Steril,1987,47(6):985-991
    [198]Levanduski M J, Westhusln M E. Efect of cytoskeletal inhibitor on fusion and development of bovine nuclear transfer embryos[J]. Theriogenolgy,1990,33:273-280
    [199]Tan X W, Ma S F, Yu J N, et al. Effects of species and cellular activity of oviductal epithelial cells on their dialogue with COcultured mouse embryos[J]. Cell Tissue Res.2007,327(1): 55-66
    [200]Wang M K, Liu J L, Li G P, et al. Sucrose pretreatment for enucleation:an efficient and non-damage method for removing the spindle of the mouse M II oocyte[J]. Mol Reprod Dev, 2001,58(4):432-436
    [201]朱秀萍,朱淑文,张菁,等.细胞松弛素B对猪卵母细胞去核的影响[J].上海交通大学学报(农业科学版),2005,23(3):253-255
    [202]朱德生,谢松涛,杨贵忠.细胞松弛素B浓度对小鼠卵母细胞去核效果的影响[J].中国兽医学报,2004,24(2):195-196
    [203]高立功,山灵,刘凤军,等.CCB、蔗糖及成熟时间对山羊卵母细胞去核效果的影响[J].西北农林科技大学学报(自然科学版),2007,35(5):6-8,13
    [204]Downing S L, Allen S K J. Induced triploidy in the Pacific oysters, Crassostrea gigas, optimal treatments with cytochalasin B depend on temperature [J]. Aquaculture,1987,61:1-15
    [205]张瑞强,杨军,金庆辉,等.细胞电融合技术及最新进展[J].中国生物工程杂志,2008,28(9):124-129
    [206]Wilmut I, Schnieke A E, Mcwhir J, et al. Viable offspring derived from fetal and adult mammalian cells [J]. Nature,1997,385:810-813
    [207]Baguisi A, Behboodi E, Melican D T, et al. Production of goats by somatic cell nuclear transfer [J]. Nat Biotechnol,1999,17:456-461
    [208]Wells D N, Misica P M, Tervit H R. Production of cloned calves following nuclear transfer with cultured adult mural granulose cells [J]. Biol Reprod,1999,60:996-100
    [209]Wakayama T, Perry A C F, Zuccotti M, et al. Full-term development of mice from nucleared oocyte injected with cumulus cell nuclei [J]. Nature,1998,394:369-374
    [210]陆风花.世界首例成活的体细胞克隆水牛在广西大学诞生[J].广西农业生物科学,2005,24(1):5
    [211]李光鹏,徐立滨,蔡世勋,等.小鼠电融合胚胎发育影响因素的研究[J].东北农业大学学报,1996,27(4):391-395
    [212]Brendan G T, Kim J G, Alan O T. Electrofusion parameters for nuclear transfer predicted using isofusion contours produced with bovine embryonic cells [J]. Mol Reprod Dev,1996,43: 306-312
    [213]周琪,谭景和.哺乳动物细胞电融合技术及其影响因素的研究进展[J].生物技术,1993, 3(6):4-8
    [214]江培洲,沈新明,乔贵林,等.不同电融合条件对小鼠卵丘细胞核移植重组胚融合和早期发育的影响[J].第一军医大学学报,2005,25(2):156-159,164
    [215]李日聪,王锦,薛林涛,等.猪体细胞核移植电融合参数的研究[J].广西农业生物科学,2008,27(4):304-308
    [216]Milazzotto M P, Feitosa W B, Coutinho A R, et al. Effect of chemical or electrical activation of bovine oocytes on blastocyst developm ent and quality[J]. Reprod Dom est Anim,2008,43: 319-322
    [217]Wang Z G, Wang W, Yu S D, et al. Effects of different activation protocols on preimplantation development, apoptosis and ploidy of bovine parthenogenetic embryos[J]. Anim Reprod Sci,2008,105:292-301
    [218]Wani N A. Chemical activation of in vitro matured dromedary camel(Camelus dromedarius) oocytes:optimization of protocols[J]. Theriogenology,2008,69:591-602
    [219]Loi P, Ptak G, Barboni B, et al. Genetic rescue of an endangered mammal by cross-species nuclear transfer using post-mortem somatic cells[J]. Nat Biotechnol,2001,19:962-964
    [220]Evans M J, Kaufman M H. Establishment in culture of pluripotential cells from mouse embryos[J]. Nature,1981,292(5819):154-156
    [221]尚克刚,胡新立.小鼠囊胚的不同遗传背景对形成ES细胞集落的影响[J].北京大学学报(自然科学版),1993,29(3):196-201
    [222]叶鑫生,许田,汤锡芳,等.干细胞和发育生物学[M].北京:军事医学科学出版社,2000:185-190
    [223]Solter D, Knowles B. Immunosurgery of mouse blastocyte[J]. Proc Nat Acad Sci USA,1975, 72(12):5099-5102
    [224]Martin G R. Isolation of a pluripotent cell line from early mouse embryos cultured in medium conditioned by teratocarcinoma stem cells[J]. Proc Natl Acad Sci U S A,1981,78(12): 7634-7638
    [225]徐小明,窦忠英,华进联,等.免疫外科法分离克隆BALB/c小鼠胚胎干细胞[J].中国实验动物学报,2004,12(3):129-133
    [226]李煜,梁琳,王振飞,等.昆明白小鼠胚胎干细胞分离与体外培养[J].细胞生物学杂志,2007,29(6):885-888
    [227]邵晓云,徐绍业.昆明小鼠3.5d和4d囊胚不同分离方法的比较[J].中国组织工程研究与临床康复,2011,15(40):7533-7536
    [228]李国善,邵敬於.人类胚胎辅助孵化技术和应用概况[J].生殖与避孕,1996,16(6):408-412
    [229]Sathananthan H, Menezes J, Gunasheela S. Mechanics of human blastocyst hatching in vitro. Reprod Biomed Online[J].2003,7(2):228-234
    [230]徐瑜,孙莹璞.辅助孵化在IVF-ET中的应用进展[J].国外医学:计划生育分册,2003,22(1):34-36
    [231]陈林君,刘丽娟,李红霞,等.小鼠孤雌生殖的胚胎干细胞的初步研究[J].南京师大学报(自然科学版),2006,29(1):71-74
    [232]银益飞,孙筱放,蒋永华,等.小鼠孤雌胚胎干细胞系的建立及生物学特性鉴定[J].现代生物医学进展,2007,7(7):996-998
    [233]Baton S C, Ferguson-smith A C, Fundele R, et al. Influence of paternally imprinted gene on development[J]. Development,1991,113:679-688
    [234]Obata Y, Kono T. Maternal Primary imprinting is established at a special time for each gene through out oocyte growth[J]. BioChem,2002,277(7):5285-5259
    [235]Muza V, Mann S, Genomic M. Imprinting-defusing the ovariantime bomb[J]. Trends Genet, 1994.10:118-123
    [236]Sleutels F, Barlow D P. The origins of genomic imprinting inmammals[J]. Homology Effects Academic Press,2002,119-154
    [237]Gonzalez M R, Bischofberger M, Pernot L, et al. Bacterial pore-forming toxins:the (w)hole story? [J]. Cell Mol Life Sci,2008,65(3):493-507
    [238]乔旭,吴芬芳,苏鹏,等.CDC/MACPF家族成孔毒素研究进展[J].遗传,2010,32(11):1126-1132
    [239]Kimoto H, Fujii Y, Hirano S, et al. Genetic and biochemical properties of streptococcal NAD-glycohydrolase inhibitor [J]. J Biol Chem,2006,281(14):9181-9189
    [240]Hochedlinger K, Jaenisch R. Nuclear reprogramming and pluripotency [J]. Nature,2006, 441(7097):1061-1067
    [241]Van der Goot G (Ed). Pore-forming toxins In:Current Topics in Microbiology and Immunology,1st edition [M]. Springer Verlag, Berlin Heidelberg,2001
    [242]Qin M D, Tai G P, Collas P, et al. Cell extract-derived differentiation of embryonic stem cells [J]. Stem Cells,2005,23:712-718
    [243]Tang S, Wang Y S, Zhang D, et al. Reprogramming donor cells with oocyte extracts improves in vitro development of nuclear transfer embryos [J]. Animal Reproduction Science,2009,115: 1-9
    [244]Miyamoto K, Furusawa T, Ohnuki M, et al. Reprogramming events of mammalian somatic cells induced by Xenopus laevis egg extracts[J]. Mol Reprod Dev,2007,74:1268-1277
    [245]Eddie J S, Sriranjani K, Poothappillai K, et al. Cloned calves from chromatin remodeled in vitro [J]. Biology of Reproduction,2004,70:146-153
    [246]Mostafavi-Pour Z, Keihani S, Talaei-Khozani T, et al. Expression of a2, a5 and a6 subunits of integrin in de-differentiated NIH3T3 cells by cell-free extract of embryonic stem cells[J]. Mol Biol Rep,2012,39(7):7339-7346
    [247]Cibelli J B, GrantK A, Chapman K B, et a.l Parthenogenetic stem cells in nonhuman primates[J]. Science,2002,295 (5556):819
    [248]Lin H, Lei J, Wininger D, et al. Multilineage potential of homozygous stem cells derived from metaphase Ⅱ oocytes[J]. Stem Cells,2003,21(2):152-161
    [249]Newman-Smith E D, Werb Z. Stem cell defects in parthenogenetic peri-implantation embryos[J]. Development,1995,121(7):2069-2077
    [250]王彦,钱德俭,韦多,等.小鼠孤雌胚胎干细胞系的建立[J].组织工程与重建外科杂志,2010,6(3):132-135
    [251]Kim S J, Lee J E, Park J H, et al. Efficient derivation of new human embryonic stem cell lines [J]. Mol Cells,2005,19(1):46-53
    [252]Lanzuolo C, Orlando V. The function of the epigenome in cell reprogramming[J]. Cell Mol Life Sci,2007,64(9):1043-1062
    [253]Eilertsen K J, Power R A, Harkins L L, et al. Targeting cellular memory to reprogram the epigenome, restore potential, and improve somatic cell nuclear transfer[J]. Anim Reprod Sci, 2007,98(1-2):129-146
    [254]Schmittwolf C, Kirchhof N, Jauch A, et al. In vivo haematopoietic activity is induced in neurosphere cells by chromatin-modifying agents[J]. EMBO J,2005,24(3):554-566
    [255]Enright B P, Sung L Y, Chang C C, et al. Methylation and acetylation characteristics of cloned bovine embryos from donor cells treated with 5-aza-2'-Deoxycytidine [J]. Biol Reprod,2005, 72:944-948
    [256]Li J, Svarcova O, Villemoes K, et al. High in vitro development after somatic cell nuclear transfer and trichostatin A treatment of reconstructed porcine embryos [J]. Theriogenology, 2008,70(5):800-808
    [257]Ding X, Wang Y, Zhang D, et al. Increased pre-implantation development of cloned bovine embryos treated with 5-aza-2'-deoxycytidine and trichostatin A [J]. Theriogenology,2008, 70(4):622-630
    [258]Ruau D, Ensenat-Waser R, Dinger TC, et al. Pluripotency associated genes are reactivated by chromatin-modifying agents in neurosphere cells[J]. Stem Cells,2008; 26:920-926
    [259]刘海明,张振,杨力,等.小鼠胚胎成纤维细胞重编程为多能干细胞[J].中国组织工程研究,2012,16(32):5963-5966
    [260]Wang Q, Mou X N, Cao H H, et al. A novel xeno-free and feeder-cell-free system for human pluripotent stem cell culture[J]. Protein cell,2012,3(1):51-59
    [251]Hanna J, Saha K, Pando B, et al. Direct cell reprogramming is a stochastic process amenable to acceleration[J]. Nature,2009,462:595-601
    [262]Chan E M, Ratanasirintrawoot S, Park I H, et al. Live cell imaging distinguishes bona fide human iPS cells from partially reprogrammed cells[J]. Nat Biotechnol,2009,27:1033-1037
    [263]Cho H J, Lee C S, Kwon Y W, et al. Induction of pluripotent stem cells from adult somatic cells by protein-based reprogramming without genetic manipulation [J]. Blood,2010,116: 386-395
    [264]Esteban M A, Xu J, Yang J, et al. Generation of induced pluripotent stem cell lines from Tibetan miniature pig[J]. J Biol Chem,2009,284(26):17634-17640
    [265]Wu Z, Chen J, Ren J, et al. Generation of pig-induced pluripotent stem cells with a drug-inducible system[J]. J Mol Cell Biol,2009,1(1):46-54
    [266]殷慧群,曹鸿国,孙雪萍,等限定因子诱导胎猪成纤维细胞重编程为多能性细胞[J].生物化学与生物物理进展,2010,37(6):607-612)
    [267]张淑金,孟书燕,雷蕾,等.定量分析诱导山羊体细胞重编程过程中端粒酶的表达变化[J].生物工程学报,2010,26(12):1660-1667
    [268]Song L, Tuan R S. MicroRNAs and cell differentiation in mammalian development[J]. Birth Defects Res C Embryo Today,2006,78(2):140-149
    [269]Alvarez-garcia I, Miska EA. MicroRNA functions in animal development and human disease[J]. Development,2005,132 (21):4653-62
    [270]Stadler B M, Ruohola-baker H. Small RNAs:keeping stem cells in line[J]. Cell,2008,132(4): 563-566
    [271]Hayes B, Fagerlie S R, Ramakrishnan A, et al. Derivation, characterization, and in vitro differentiation of canine embryonic stem cells [J]. Stem Cells,2008,26(2):465-473
    [272]Choi Y J, Lin C P, Ho J J, et al. MiR-34 miRNAs provide a barrier for somatic cell reprogramming[J].Nat Cell Biol,2011,13(11):1353-1360

© 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