猪诱导性多潜能干细胞的建立
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
猪诱导性多潜能干细胞的建立
     诱导性多潜能干细胞(iPS细胞)是指通过在分化的体细胞中表达特定的转录因子来诱导其发生重编程而获得的可不断自我更新和具有多向分化潜能的细胞。iPS细胞技术的产生和发展,是干细胞领域乃至整个生物学领域的一项重大发现,具有广阔的应用前景。本研究旨在应用限定因子建立猪iPS细胞诱导技术,为将来深入研究应用猪iPS细胞奠定基础。论文由三个试验组成,其内容如下:
     试验一:携带限定因子的慢病毒表达载体的构建
     以胎猪原始生殖嵴和囊胚为材料,用RT-PCR方法克隆出猪Sox2、Klf4和c-Myc基因的开放阅读框序列,人OCT4基因序列直接从质粒pMX-hOCT4中扩增获取。经限制性内切酶酶切,与逆转录病毒载体pLEGFP-N1连接,分别构建出限定因子EGFP融合蛋白逆转录病毒表达载体。再从重组逆转录病毒载体中扩增出CMV启动子,限定因子和EGFP序列,同时通过酶切从pLL3.7质粒中移除U6启动子、CMV启动子和EGFP序列,最后把扩增出的CMV启动子,限定因子和EGFP序列与改造后的pLL3.7进行重组,分别构建出限定因子EGFP融合蛋白慢病毒表达载体pLL-hOCT4/pSox2/pKlf4/pMyc -EGFP。重组慢病毒载体质粒转染293T细胞后,在荧光显微镜下观察限定因子表达情况。结果显示,猪Sox2、Klf4和c-Myc基因开放阅读框序列分别与已发表的猪Sox2、Klf4和c-Myc基因序列高度同源;重组慢病毒载体质粒转染293T细胞后表现为限定因子EGFP融合蛋白定位于细胞核表达,符合限定因子表达特点。这些结果证实,本试验的限定因子EGFP融合蛋白慢病毒载体构建正确。
     试验二:慢病毒载体法诱导猪iPS细胞
     采用组织块培养法从57d胎龄的杜洛克-长白-大约克三元杂交猪胎儿分离培养建立7株猪胎儿成纤维细胞系。通过磷酸钙法转染四种限定因子EGFP融合蛋白慢病毒表达载体质粒和包装辅助质粒,包装产生的四种限定因子慢病毒经浓缩后感染猪胎儿成纤维细胞,在含1000U/mL LIF和4ng/mL bFGF的干细胞培养液培养条件下进行培养传代,逐步分离培养出集落边缘界限清晰的细胞克隆,高倍显微镜下集落中的单个细胞呈现细胞核较大,核质比例较高。按1∶4比例每3~4 d传代一次,细胞集落生长稳定,核型正常,AP检测为阳性,免疫细胞化学和RT-PCR检测有关键性多潜能相关基因Oct4和Nanog的表达。此外,细胞集落中SSEA-1蛋白表达检测阳性,SSEA-3/4和TRA-1-61/81检测阴性。将其注射到BALB/cA裸鼠背侧皮下可形成含有外胚层、中胚层和内胚层组织来源的畸胎瘤。以上结果证实,本试验所获得的猪的细胞克隆具有ES细胞样特征,即成功地从猪胎儿成纤维细胞诱导出iPS细胞。
     试验三:纯化蛋白穿膜法诱导猪iPS细胞
     由于慢病毒携带外源基因的随机稳定整合会带来潜在的细胞癌化畸变等危害,因此根据慢病毒介导外源限定因子EGFP融合蛋白的试验结果,构建携带细胞穿膜肽外源限定因子的融合蛋白用于猪iPS细胞的诱导,将会为猪iPS细胞的安全生产、应用奠定基础。将体外表达、纯化的携带有细胞穿膜肽——九聚精氨酸(R_9)的限定因子-R_9融合蛋白添加到培养的猪胎儿成纤维细胞培养液中尝试建立猪iPS细胞。结果显示,在荧光显微镜下可观察到限定因子-R_9原核蛋白能进入猪胎儿成纤维细胞,且蛋白能定位于细胞核;在干细胞培养液培养条件下同时用限定因子-R_9原核蛋白处理6个周期后,能逐渐分离培养出集落边缘界限清晰的细胞克隆,重构克隆中的单个细胞呈现细胞核较大,核质比例较高,AP为阳性,免疫细胞化学检测ES细胞特异性分子标记Oct4和Nanog蛋白表达阳性。这些结果证实本试验的限定因子-R_9原核蛋白表达载体构建正确;细胞穿膜肽R_9可转导限定因子重组蛋白进入细胞核;重组限定因子-R_9原核蛋白可使猪胎儿成纤维细胞发生重构,且重构细胞具有ES细胞样形态,可表达AP、Oct4和Nanog蛋白。
     综合以上三个试验的研究结果,可以得出如下结论:
     1.运用RT-PCR技术可以从胎猪的原始生殖嵴和囊胚细胞克隆出猪Sox2、Klf4和c-Myc限定因子基因的开放阅读框序列;用基因工程技术将这些限定因子基因序列(包括人Oct4基因序列)与逆转录病毒载体pLEGFP-N1连接,再进行基因重组,能成功地构建限定因子EGFP融合蛋白慢病毒表达载体。应用这些重组限定因子EGFP融合蛋白可有效追踪掌握限定因子在细胞重编程过程中的作用机制和特点。
     2.用限定因子EGFP融合蛋白慢病毒能诱导猪胎儿成纤维细胞重编程,建立猪iPS细胞。通过限定因子EGFP融合蛋白追踪发现,外源限定因子在猪iPS细胞集落中大多数不表达或低水平表达,少数细胞高表达,提示体细胞完全重编程形成iPS细胞后外源基因不表达,不完全重编程的体细胞外源基因仍处于高表达状态;不同的猪胎儿成纤维细胞可能需要不同的时间去完成重编程进程,或者不同的细胞可能是在不同的时间点上启动重编程进程。
     3.将携带有细胞穿膜肽——九聚精氨酸(R_9)的限定因子重组蛋白用于猪胎儿成纤维细胞的重编程,获得了具有ES细胞样形态、可表达AP、Oct4和Nanog蛋白的重构细胞,为建立诱导猪体细胞直接重编程的新方法提供了新的思路。
     本论文的创新点:首次应用含有猪源性限定因子EGFP融合蛋白慢病毒成功建立猪iPS细胞。首次尝试应用限定因子重组蛋白直接重编程猪胎儿成纤维细胞。
Generation of Induced Pluripotent Stem Cells from Porcine Fetal Fibroblasts by Defined Factors
     Pluripotent cells, which derived from any differentiated cell type through ectopic expression of transcription factors, were designated as induced pluripotent stem (iPS) cells, exhibiting morphology and growth properties indistinguishible to ES cells and expressing ES cell marker genes. With the ability to differentiate into all types of cells, iPS cells technology has enormous potential values in generation of human disease models, drug screening, toxicology and regenerative medicine. The current research was attempted to generate porcine iPS by defined factors EGFP fusion protein, which would be of great help to better our understanding of nuclear reprogramming mechanisms, and enrich our means of examine safety and effectiveness of iPS cells in human, as well as to extend the applications of swine iPS cells.
     Experiment 1. To construct lentiviral expression vector with defined factors gene
     Open reading frame (ORF) of porcine Sox2, Klf4 and c-Myc gene was successfully amplified respectively by RT-PCR from the primordial genital ridges and blastula. Human OCT4 gene was amplified directly by PCR from plasmid pMX-hOCT4. After confirmation by DNA sequencing, such correct sequences of defined factors were inserted individually into retroviral vector pLEGFP-N1. Recombinant retroviral vector pLEGFP-hOCT4/pSox2/pKlf4/pMyc was obtained respectively. Next, DNA sequences containing CMV promoter, defined factors and EGFP were amplified respectively from pLEGFP-hOCT4/pSox2/pMyc/pKlf4 by PCR. The lentiviral vector pLL3.7 was modified by removing U6 promoter, CMV promoter and EGFP sequences. Lastly, modified pLL3.7 was ligated to DNA sequence containing CMV promoter, defined factors and EGFP by T4 DNA ligase. Thus, recombinant lentiviral vector pLL-hOCT4/pSox2/pKlf4/pMyc-EGFP was constructed well respectively. Then 293T cells were transfected with recombinant lentiviral plasmids respectively. The results showed that the cloned sequences of pig Sox2, Klf4 and c-Myc gene were highly homologous to corresponding genes ORF published in genebank respectively. All the four fusion proteins hOCT4/pSox2/pKlf4/pMyc-EGFP was located in the nucleus of 293T cells as their natural localization. This confirmed that recombinant lentiviral vector pLL-hOCT4/pSox2/pKlf4/pMyc-EGFP was constructed correctly.
     Experiment 2. To generate pig iPS cells by lentiviral vector with defined factors
     Seven porcine fetal fibroblasts (PFFs) cell lines were established from 7 porcine fetuses of Duroc-Landrace-Yorkshire crossbred at 57d of gestation by using explants-seeding method. Lentiviral plasmids harboring four defined factors and packaging helper plasmids were co-transfected into 293T cells by calcium phosphate transfection method. Four defined factors lentivirus generated by 293T were concentrated to reprogram PFFs. PPFs, expressed exogenous defined factors genes, were then sub-cultured in stem cell medium supplemented with 1000U/mL LIF and 4ng/mL bFGF, and as a consequence the clear-cut cell colonies were gradually derived. The cells in such colonies had large translucent nuclei and a high nucleocytoplasmic ratio. All of the cells grew at similar rates, requiring sub-culture at a roughly 1:4 ratio every 3~4 days. They exhibited normal karyotype, and expressed alkaline phosphatase (AP), Oct4 and Nanog. Unlike human ES cells, such porcine cell colonies were positive for SSEA-1, but negative for SSEA-3/4 and Tra-1-61/81. Teratomas containing tissues of ectoderm, mesoderm, and endoderm formed gradually after subcutaneous injection of these cells into BALB/cA nude mice dorsal skin. Taken together, our results demonstrated that these derived cell colonies can exhibit very similar morphology and characteristics to ES cells, could be termed as pig iPS cells.
     Experiment 3. To generate pig iPS cells by defined factors recombinant proteins
     As exogenous genes were randomly inserted into the target cells genome, it possibly brought the potential hazard of insertional mutagenesis. Therefore it is necessary to seek for some new methods to induce somatic cell reprogramming without virus. In view of above study results, we attempted to use defined factors recombinant proteins carring cell-penetrating peptide (CPP) for generation of porcine iPS cells, which would be of tremendous benefits for the safe applications of iPS cells. Defined factors genes were amplified by PCR with specific primers of 9 arginines (R_9) from recombinant plasmid pLL-hOCT4/pSox2/pMyc/pKlf4-EGFP and inserted into prokaryotic expression vector pET-28a-EGFP. After DNA sequencing confirmation, the four recombinant plasmids were then transformed into BL21 (Escherichia coli) strain respectively. After IPTG induction, hOCT4/pSox2/pMyc/pKlf4-R_9-EGFP fusion proteins were purified using Novagen His-Bind kit and confirmed by SDS-PAGE respectively. Then defiend factors recombinant proteins were added into PFFs medium every 48h to generate pig iPS cells. Our results showed that such purified hOCT4/pSox2/pMyc/pKlf4-R_9-EGFP fusion proteins could enter into PFFs efficiently, and most of them located in nuclei. PPFs were sub-cultured in stem cell medium condition and treated with defined factors recombinant proteins for 6 cycles simultaneously; the clear-cut cell colonies were gradually derived. These cells had large translucent nuclei and a high nucleocytoplasmic ratio. They were positive for AP, Oct4 and Nanog. It confirmed that recombinant prokaryotic expression vector for the hOCT4/pSox2/pKlf4/pMyc-R_9-EGFP was constructed correctly. And the transduction activity of hOCT4/pSox2/pKlf4/pMyc-R_9-EGFP fusion protein to PFFs was proved respectively. Recombinants proteins hOCT4/pSox2/pKlf4/pMyc-R_9-EGFP can reprogram PFFs, and reprogrammed cells exhibited the morphology of ES cells and express ES cell-specific genes, such as AP, Oct4 and Nanog.
     Form above results, we concluded that:
     1. The open reading frame (ORF) of pig Sox2, Klf4 and c-Myc genes were obtained from the primordial genital ridges and blastula by RT-PCR. Lentiviral vectors of defined factors (including pig Sox2, Klf4 and c-Myc genes, human OCT4) EGFP fusion protein were constructed successfully by genetic engineering technology. The mechanism and characteristics of defined factors in cells reprogramming could be tracked effectively by constructed defined factors EGFP fusion protein.
     2. Pig iPS cells were generated successfully from PFFs by defined factors EGFP fusion proteins lentivirus. Some cells in pig iPS colonies expressed defined factors EGFP fusion proteins, the others showed there was no or weak expression of defined factors EGFP fusion proteins under fluorescence microscope. It suggested that different PFFs may require different time to complete reprogramming process, or different cells may start reprogramming process at different time points.
     3. Reprogrammed cells exhibited the morphology of ES cells and expressed ES cell marker genes, such as AP, Oct4 and Nanog, were generated from PFFs by defined factors recombinant proteins with cell penetrating peptide------R_9. This would provide new ideas for the induction of porcine somatic cells reprogramming.
     The innovation of the present study was for the first time, to our knowledge, to establish pig iPS cells by defined factors (including pig Sox2, Klf4 and c-Myc genes, human OCT4) EGFP fusion proteins lentivirus. Moreover, first attempt to reprogram directly PFFs by defined factors recombinant proteins.
引文
[1] Wilmut I, Schnieke AE, McWhir J, et al. Viable offspring derived from fetal and adult mammalian cells. Nature, 1997, 385(6619):810~813
    [2] Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell, 2006, 126(4): 663~676
    [3] Nagy A, Gócza E, Diaz E M, et al. Embryonic stem cells alone are able to support fetal development in the mouse. Development, 1990, 110(3): 815~821
    [4] Huang J, Deng K, Wu H, et al. Efficient Production of Mice from Embryonic Stem Cells Injected into Four- or Eight-Cell Embryos by Piezo Micromanipulation. Stem Cells, 2008, 26 (7): 1883~1890
    [5] Evans MJ, Kaufman MH. Establishment in culture of pluripotential cells from mouse embryos. Nature, 1981, 292(5819):154~156
    [6] Thomson JA, Itskovitz EJ, Shapiro S S, et al. Embryonic stem cell lines derived from human blastocysts. Science, 1998, 282: 1145~1147
    [7] Shamblott MJ, Axelman J, Wang S, et al. Derivation of pluripotent stem cells from cultured human primordial germ cell. Proc Natl Acad Sci USA,1998 ,95 (23) :13726~13731
    [8] Kfoury C. Therapeutic cloning: promises and issues. Mcgill J Med. 2007, 10(2): 112~120
    [9] Rideout WM 3rd, Eggan K, Jaenisch R. Nuclear cloning and epigenetic reprogramming of the genome. Science, 2001, 293(5532):1093~1098
    [10] Smith SL, Everts RE, Tian XC, et al. Global gene expression profiles reveal significant nuclear reprogramming by the blastocyst stage after cloning. Proc Natl Acad Sci U S A, 2005, 102(49): 17582~17587
    [11] Kohda T, Inoue K, Ogonuki N, et al. Variation in gene expression and aberrantly regulated chromosome regions in cloned mice. Biol Reprod, 2005, 73(6): 1302~1311
    [12] Grinnemo KH, Sylvén C, Hovatta O, et al. Immunogenicity of human embryonic stem cells. Cell Tissue Res, 2008, 331(1): 67~78
    [13] Miller RA, Ruddle FH. Pluripotent teratocarcinoma-thymus somatic cell hybrids. Cell, 1976, 9(1): 45~55
    [14] Tada M, Tada T, Lefebvre L, et al. Embryonic germ cells induce epigenetic reprogramming of somatic nucleus in hybrid cells. EMBO J, 1997, 16(21): 6510~6520
    [15] Tada M, Morizane A, Kimura H, et al. Pluripotency of reprogrammed somatic genomes in embryonic stem hybrid cells. Dev Dyn, 2003, 227(4): 504~510
    [16] Cowan CA, Atienza J, Melton DA, et al. Nuclear reprogramming of somatic cells after fusion with human embryonic stem cells. Science, 2005, 309(5739): 1369~1373
    [17] Collas P, Taranger CK. Epigenetic reprogramming of nuclei using cell extracts. Stem Cell Rev. 2006, 2(4): 309~317
    [18] Silva J, Chambers I, Pollard S, et al. Nanog promotes transfer of pluripotency after cell fusion. Nature, 2006, 441(7096): 997~1001
    [19] Tada M, Takahama Y, Abe K, et al. Nuclear reprogramming of somatic cells by in vitro hybridization with ES cells. Curr Biol, 2001, 11(19):1553~1558
    [20] Matsumura H, Tada M, Otsuji T, et al. Targeted chromosome elimination from ES-somatic hybrid cells. Nat Methods. 2007, 4(1): 23~25
    [21] Hansis C, Barreto G, Maltry N, et al. Nuclear reprogramming of human somatic cells by xenopus egg extract requires BRG1. Curr Biol, 2004, 14(16): 1475~1480
    [22] Neri T, Monti M, Rebuzzini P, et al. Mouse fibroblasts are reprogrammed to Oct-4 and Rex-1 gene expression and alkaline phosphatase activity by embryonic stem cell extracts. Cloning Stem Cells. 2007, 9(3): 394~406
    [23] Bru T, Clarke C, McGrew MJ, et al. Rapid induction of pluripotency genes after exposure of human somatic cells to mouse ES cell extracts. Exp Cell Res. 2008 , 314(14):2634~2642
    [24] Collas P, Taranger CK, Boquest AC, et al. On the way to reprogramming cells to pluripotency using cell-free extracts. Reproductive Bio Medicine, 2006, 12(6): 762~770
    [25] Hochedlinger K, Jaenisch R. Nuclear reprogramming and pluripotency. Nature, 2006, 441(7097):1061~1067
    [26] Yu J, Vodyanik MA, Smuga-Otto K, et al. Induced pluripotent stem cell lines derived from human somatic cells. Science, 2007, 318(5858): 1917~1920
    [27]刘爽,段恩奎.诱导产生多能性干细胞(iPS细胞)的研究进展.科学通报, 2008, 53(4): 377~385
    [28] Tokuzawa Y, Kaiho E, Maruyama M, et al. Fbx15 is a novel target of Oct3/4 but is dispensable for embryonic stem cell self-renewal and mouse development. Mol Cell Biol, 2003, 23: 2699~2708
    [29] Yamanaka S. Induction of pluripotent stem cells from mouse fibroblasts by four transcription factors. Cell Prolif, 2008, 41 Suppl 1:51~56
    [30] Yu J, Vodyanik M A, He P, et al. Human embryonic stem cells reprogram myeloid precursors following cell-cell fusion. Stem Cells, 2006, 24(1): 168~176
    [31] Maherali N, Sridharan R, Wei Xie, et al. Directly Reprogrammed Fibroblasts Show Global Epigenetic Remodeling and Widespread Tissue Contribution. Cell Stem Cell, 2007, 1(1): 55~70
    [32] Okita K, Ichisaka T, Yamanaka S. Generation of germline-competent induced pluripotent stem cells. Nature, 2007, 448(7151): 313~317
    [33] Park IH, Zhao R, West JA, et al. Reprogramming of human somatic cells to pluripotency with defined factors. Nature, 2008, 451(7175): 141~146
    [34] Liu H, Zhu F, Yong J, et al., Generation of induced pluripotent stem cells from adult rhesusmonkey fibroblasts. Cell Stem Cell, 2008, 3(6):587~590
    [35] Liao J, Cui C, Chen S, et al., Generation of induced pluripotent stem cell lines from adult rat cells. Cell Stem Cell, 2009, 4(1):11~15
    [36] Esteban MA, Xu J, Yang J, et al., Generation of induced pluripotent stem cell lines from Tibetan miniature pig. J Biol Chem, 2009, 284(26):17634~17640
    [37] Wu Z, Chen J, Ren J, et al., Generation of Pig-Induced Pluripotent Stem Cells with a Drug-Inducible System. J Mol Cell Biol, 2009, 1(1):46~54
    [38] Ezashi T, Telugu BP, Alexenko AP, et al. Derivation of induced pluripotent stem cells from pig somatic cells. Proc Natl Acad Sci U S A, 2009, 106(27):10993~10998
    [39] Blelloch R, Venere M, Yen J, et al. Generation of induced pluripotent stem cells in the absence of drug selection. Cell Stem Cell. 2007, 1(3):245~247
    [40] Nakagawa M, Koyanagi M, Tanabe K, et al. Generation of induced pluripotent stem cells without Myc from mouse and human fibroblasts. Nat Biotechnol, 2008, 26 (1): 101~106
    [41] Wernig M, Meissner A, Cassady JP, et al. c-Myc is dispensable for direct reprogramming of mouse fibroblasts. Cell Stem Cell, 2008a, 2(1): 10~12
    [42] Kim JB, Zaehres H, Wu G, et al. Pluripotent stem cells induced from adult neural stem cells by reprogramming with two factors. Nature. 2008, 454(7204):646~650
    [43] Wernig M, Meissner A, Foreman R, et al. In vitro reprogramming of fibroblasts into a pluripotent ES-cell-like state. Nature, 2007, 448(7151): 318~324
    [44] Yoshida Y, Takahashi K, Okita K, et al. Hypoxia enhances the generation of induced pluripotent stem cells. Cell Stem Cell, 2009, 5(3):237~241
    [45] Huangfu D, Maehr R., Guo W, et al. Induction of pluripotent stem cells by defined factors is greatly improved by small-molecule compounds. Nat. Biotechnol. 2008a, 26, 795~797
    [46] Huangfu D, Osafune K, Maehr R, et al. Induction of pluripotent stem cells from primary human fibroblasts with only OCT4 and Sox2. Nat. Biotechnol. 2008b, 26, 1269~1275
    [47] Shi Y, Do JT, Desponts C, et al. A combined chemical and genetic approach for the generation of induced pluripotent stem cells. Cell Stem Cell, 2008a, 2(6):525~528
    [48] Shi Y, Desponts C, Do JT, et al. Induction of pluripotent stem cells from mouse embryonic fibroblasts by Oct4 and Klf4 with small-molecule compounds. Cell Stem Cell. 2008b, 3(5):568~574
    [49] Mikkelsen TS, Hanna J, Zhang X, et al. Dissecting direct reprogramming through integrative genomic analysis. Nature, 2008, 454(7200):49~55
    [50] Marson A, Foreman R, Chevalier B, et al. Wnt signaling promotes reprogramming of somatic cells to pluripotency. Cell Stem Cell, 2008, 3(2):132~135
    [51] Zhao Y, Yin X, Qin H, et al. Two supporting factors greatly improve the efficiency of human iPSC generation. Cell Stem Cell, 2008, 3(5):475~479
    [52] Takenaka C, Nishishita N, Takada N, et al. Effective generation of iPS cells from CD34+ cord blood cells by inhibition of p53. Exp Hematol, 2010, 38(2):154~162
    [53] Silva J, Barrandon O, Nichols J, et al. Promotion of reprogramming to ground state pluripotency by signal inhibition. PLoS Biol. 2008, 6(10):e253
    [54] Li W, Wei W, Zhu S, et al. Generation of rat and human induced pluripotent stem cells by combining genetic reprogramming and chemical inhibitors. Cell Stem Cell, 2009, 4(1):16~19
    [55] Ichida JK, Blanchard J, Lam K et al. A small-molecule inhibitor of tgf-Beta signaling replaces sox2 in reprogramming by inducing nanog. Cell Stem Cell, 2009, 5(5):491~503
    [56] Lyssiotis CA, Foreman RK, Staerk J. Reprogramming of murine fibroblasts to induced pluripotent stem cells with chemical complementation of Klf4. Proc Natl Acad Sci U S A, 2009, 106(22):8912~8917
    [57] Heng JC, Feng B, Han J, et al. The nuclear receptor Nr5a2 can replace OCT4 in the reprogramming of murine somatic cells to pluripotent cells. Cell Stem Cell. 2010, 6(2):167~174
    [58] Esteban MA, Wang T, Qin B, et al., Vitamin C enhances the generation of mouse and human induced pluripotent stem cells. Cell Stem Cell, 2010, 6(1):71~79
    [59] Mali P, Chou BK, 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. Stem Cells, 2010, 28(4):713~20
    [60] Maherali N, Hochedlinger K. Guidelines and techniques for the generation of induced pluripotent stem cells. Cell Stem Cell, 2008, 3(6):595~605
    [61] Brambrink T, Foreman R, Welstead GG, et al. Sequential expression of pluripotency markers during direct reprogramming of mouse somatic cells. Cell Stem Cell, 2008, 2(2):151~159
    [62] Maherali N, Ahfeldt T, Rigamonti A, et al. A high-efficiency system for the generation and study of human induced pluripotent stem cells. Cell Stem Cell, 2008, 3(3):340~345
    [63] Stadtfeld M, Nagaya M, Utikal J,et al. Induced pluripotent stem cells generated without viral integration. Science, 2008a, 322(5903):945~949
    [64] Okita K, Nakagawa M, Hyenjong H, et al. Generation of mouse induced pluripotent stem cells without viral vectors. Science, 2008, 322(5903):949~953
    [65] Yu J, Hu K, Smuga-Otto K, et al., Human induced pluripotent stem cells free of vector and transgene sequences. Science, 2009, 324(5928):797~801
    [66] Woltjen K, Michael IP, Mohseni P, et al., piggyBac transposition reprograms fibroblasts to induced pluripotent stem cells. Nature, 2009, 458(7239):766~770
    [67] Zhou H, Wu S, Joo JY, et al. Generation of induced pluripotent stem cells using recombinant proteins. Cell Stem Cell, 2009, 4(5):381~384
    [68] Kim D, Kim CH, Moon JI, et al., Generation of human induced pluripotent stem cells by direct delivery of reprogramming proteins. Cell Stem Cell, 2009, 4(6):472~476
    [69] Stadtfeld M, Maherali N, Breault DT, et al. Defining molecular cornerstones during fibroblast to iPS cell reprogramming in mouse. Cell Stem Cell, 2008b, 2(3):230~240
    [70] Soldner F, Hockemeyer D, Beard C, et al. Parkinson's disease patient-derived inducedpluripotent stem cells free of viral reprogramming factors. Cell, 2009, 136(5):964~977
    [71] Carey BW, Markoulaki S, Hanna J, et al. Reprogramming of murine and human somatic cells using a single polycistronic vector. Proc Natl Acad Sci U S A, 2009, 106(1):157~162
    [72] Kaji K, Norrby K, Paca A, et al. Virus-free induction of pluripotency and subsequent excision of reprogramming factors. Nature, 2009, 458(7239):771~775
    [73] Yusa K, Rad R, Takeda J, et al. Generation of transgene-free induced pluripotent mouse stem cells by the piggyBac transposon. Nat Methods, 2009, 6(5):363~369
    [74] Jia F, Wilson KD, Sun N, et al. A nonviral minicircle vector for deriving human iPS cells. Nat Methods, 2010, 7(3):197~199
    [75] Wolf D, Goff SP. TRIM28 mediates primer binding site-targeted silencing of murine leukemia virus in embryonic cells. Cell, 2007, 131(1):46~57
    [76] Miller DG, Adam MA, Miller AD. Gene transfer by retrovirus vectors occurs only in cells that are actively replicating at the time of infection. Mol Cell Biol, 1990, 10(8):4239~4242
    [77] Naldini L, Bl?mer U, Gallay P, et al. In vivo gene delivery and stable transduction of nondividing cells by a lentiviral vector. Science, 1996, 272(5259):263~267
    [78] Lois C, Hong EJ, Pease S, et al. Germline transmission and tissue-specific expression of transgenes delivered by lentiviral vectors. Science, 2002, 295(5556):868~872
    [79] Hockemeyer D, Soldner F, Cook EG, et al. A drug-inducible system for direct reprogramming of human somatic cells to pluripotency. Cell Stem Cell, 2008, 3(3):346~353
    [80] Markoulaki S, Hanna J, Beard C, et al. Transgenic mice with defined combinations of drug-inducible reprogramming factors. Nat Biotechnol, 2009, 27(2):169~171
    [81] Hochedlinger K, Yamada Y, Beard C, et al. Ectopic expression of Oct-4 blocks progenitor-cell differentiation and causes dysplasia in epithelial tissues. Cell, 2005, 121(3):465~477
    [82] Shao L, Feng W, Sun Y, et al. Generation of iPS cells using defined factors linked via the self-cleaving 2A sequences in a single open reading frame. Cell Res, 2009, 19(3):296~306
    [83] Kustikova O, Fehse B, Modlich U, et al. Clonal dominance of hematopoietic stem cells triggered by retroviral gene marking. Science, 2005, 308(5725):1171~1174
    [84] Aoi T, Yae K, Nakagawa M,et al. Generation of pluripotent stem cells from adult mouse liver and stomach cells. Science, 2008, 321(5889):699~702
    [85] Varas F, Stadtfeld M, de Andres-Aguayo L, et al. Fibroblast-derived induced pluripotent stem cells show no common retroviral vector insertions. Stem Cells, 2009, 27(2):300~306
    [86] Szymczak AL, Vignali DA. Development of 2A peptide-based strategies in the design of multicistronic vectors. Expert Opin Biol Ther, 2005, 5(5):627~638
    [87] Noguchi H, Matsushita M, Okitsu T, et al. A new cell-permeable peptide allows successful allogeneic islet transplantation in mice. Nat Med, 2004,10(3):305~309
    [88] Wakayama T, Perry AC, Zuccotti M, et al. Full-term development of mice from enucleated oocytes injected with cumulus cell nuclei. Nature, 1998, 394(6691):369~374
    [89] Stadtfeld M, Brennand K, Hochedlinger K. Reprogramming of pancreatic beta cells intoinduced pluripotent stem cells. Curr Biol, 2008c, 18(12):890~894
    [90] Hanna J, Saha K, Pando B, et al. Direct cell reprogramming is a stochastic process amenable to acceleration. Nature, 2009, 462(7273):595~601
    [91] Hanna J, Markoulaki S, Schorderet P, et al. Direct reprogramming of terminally differentiated mature B lymphocytes to pluripotency. Cell, 2008, 133(2):250~264
    [92] Eminli S, Utikal J, Arnold K, et al., Reprogramming of neural progenitor cells into induced pluripotent stem cells in the absence of exogenous Sox2 expression.Stem Cells, 2008, 26(10):2467~2474
    [93] Aasen T, Raya A, Barrero MJ, et al. Efficient and rapid generation of induced pluripotent stem cells from human keratinocytes. Nat Biotechnol, 2008, 26(11):1276~1284
    [94] Aoki T, Ohnishi H, Oda Y, et al. Generation of Induced Pluripotent Stem Cells from Human Adipose-Derived Stem Cells Without c-MYC. Tissue Eng Part A. 2010 Apr 6. [Epub ahead of print]
    [95] Niwa H, Burdon T, Chambers I, et al. Self-renewal of pluripotent embryonic stem cells is mediated via activation of STAT3. Genes Dev, 1998, 12(13):2048~2060
    [96] Kopp JL, Ormsbee BD, Desler M, et al. Small increases in the level of Sox2 trigger the differentiation of mouse embryonic stem cells. Stem Cells, 2008, 26(4):903~911
    [97] Li C, Zhou J, Shi G, et al. Pluripotency can be rapidly and efficiently induced in human amniotic fluid-derived cells. Hum Mol Genet, 2009, 18(22):4340~4349
    [98] Rowland BD, Peeper DS. KLF4, p21 and context-dependent opposing forces in cancer. Nat Rev Cancer, 2006, 6(1):11~23
    [99] Cheng J, Dutra A, Takesono A, et al. Improved generation of C57BL/6J mouse embryonic stem cells in a defined serum-free media. Genesis, 2004, 39(2):100~104
    [100] Ying QL, Wray J, Nichols J et al. The ground state of embryonic stem cell self-renewal. Nature, 2008, 453(7194):519~523
    [101] Amit M, Itskovitz-Eldor J. Maintenance of human embryonic stem cells in animal serum- and feeder layer-free culture conditions. Methods Mol Biol, 2006, 331:105~113
    [102] Wernig M, Lengner CJ, Hanna J, et al. A drug-inducible transgenic system for direct reprogramming of multiple somatic cell types. Nat Biotechnol, 2008b, 26(8):916~924
    [103] Watanabe K, Ueno M, Kamiya D, et al. A ROCK inhibitor permits survival of dissociated human embryonic stem cells. Nat Biotechnol, 2007, 25(6):681~686
    [104] Park IH, Lerou PH, Zhao R, et al. Generation of human-induced pluripotent stem cells. Nat Protoc, 2008, 3(7):1180~1186
    [105] Meissner A, Wernig M, Jaenisch R. Direct reprogramming of genetically unmodified fibroblasts into pluripotent stem cells. Nat Biotechnol, 2007, 25(10):1177~1181
    [106] Qin D, Li W, Zhang J, et al. Direct generation of ES-like cells from unmodified mouse embryonic fibroblasts by Oct4/Sox2/Myc/Klf4. Cell Res, 2007, 17(11):959~962
    [107] Lowry WE, Richter L, Yachechko R, et al., Generation of human induced pluripotent stem cells from dermal fibroblasts. Proc Natl Acad Sci U S A, 2008, 105(8):2883~2888
    [108] Bernstein BE, Mikkelsen TS, Xie X, et al. A bivalent chromatin structure marks key developmental genes in embryonic stem cells. Cell, 2006, 125(2):315~326
    [109] Jaenisch R, Young R. Stem cells, the molecular circuitry of pluripotency and nuclear reprogramming. Cell, 2008, 132(4):567~582
    [110] Zhao XY, Li W, Lv Z, et al., iPS cells produce viable mice through tetraploid complementation. Nature, 2009, 461(7260):86~90
    [111] Kang L, Wang J, Zhang Y, et al., iPS cells can support full-term development of tetraploid blastocyst-complemented embryos. Cell Stem Cell, 2009, 5(2):135~138
    [112] Gertow K, Przyborski S, Loring JF, et al. Isolation of human embryonic stem cell-derived teratomas for the assessment of pluripotency. Curr Protoc Stem Cell Biol, 2007, Chapter 1:Unit1B.4
    [113] Kroon E, Martinson LA, Kadoya K, et al. Pancreatic endoderm derived from human embryonic stem cells generates glucose-responsive insulin-secreting cells in vivo. Nat Biotechnol, 2008, 26(4):443~452
    [114] Draper JS, Smith K, Gokhale P, et al. Recurrent gain of chromosomes 17q and 12 in cultured human embryonic stem cells. Nat Biotechnol, 2004, 22(1):53~54
    [115] Yamanaka S. Elite and stochastic models for induced pluripotent stem cell generation. Nature, 2009, 460(7251):49~52
    [116] Blelloch R, Wang Z, Meissner A,et al. Reprogramming efficiency following somatic cell nuclear transfer is influenced by the differentiation and methylation state of the donor nucleus. Stem Cells, 2006, 24(9):2007~2013
    [117] Toma JG, Akhavan M, Fernandes KJ, et al. Isolation of multipotent adult stem cells from the dermis of mammalian skin. Nat Cell Biol, 2001, 3(9):778~784
    [118] Boyer LA, Lee TI, Cole MF, et al. Core transcriptional regulatory circuitry in human embryonic stem cells. Cell, 2005, 122(6):947~956
    [119] Egli D, Birkhoff G, Eggan K. Mediators of reprogramming: transcription factors and transitions through mitosis. Nat Rev Mol Cell Biol, 2008, 9(7):505~516
    [120] Rosner MH, Vigano MA, Ozato K, et al. A POU-domain transcription factor in early stem cells and germ cells of the mammalian embryo. Nature, 1990, 345(6277): 686~692
    [121] Pan G, Qin B, Liu N, et al. Identification of a nuclear localization signal in Oct4 and generation of a dominant negative mutant by its ablation. J Biol Chem, 2004, 279(35):37013~37020
    [122] Pesce M, Sch?ler HR. Oct-4: gatekeeper in the beginnings of mammalian development. Stem Cells, 2001, 19(4):271~278
    [123] Niwa H, Miyazaki J, Smith AG. Quantitative expression of Oct-3/4 defines differentiation,dedifferentiation or self-renewal of ES cells. Nat Genet, 2000, 24(4):372~376
    [124] Kehler J, Tolkunova E, Koschorz B, et al. Oct4 is required for primordial germ cell survival. EMBO Rep, 2004, 5(11):1078~1083
    [125] Gidekel S, Pizov G, Bergman Y, et al. Oct-3/4 is a dose-dependent oncogenic fate determinant. Cancer Cell, 2003, 4(5):361~370
    [126] Harley VR, Lovell-Badge R, Goodfellow PN. Definition of a consensus DNA binding site for SRY. Nucleic Acids Res, 1994, 22(8): 1500~1501
    [127] Wood HB, Episkopou V. Comparative expression of the mouse Sox1, Sox2 and Sox3 genes from pre-gastrulation to early somite stages. Mech Dev, 1999, 86(1-2): 197~201
    [128] Miyagi S, Saito T, Mizutani K, et al. The Sox-2 regulatory regions display their activities in two distinct types of multipotent stem cells. Mol Cell Biol, 2004, 24(10):4207~4220
    [129] Darr H, Benvenisty N. Factors involved in self-renewal and pluripotency of embryonic stem cells. Handb Exp Pharmacol, 2006, (174): 1~19
    [130] Avilion AA, Nicolis SK, Pevny LH, et al. Multipotent cell lineages in early mouse development depend on SOX2 function. Genes Dev, 2003, 17(1):126~140
    [131] Masui S, Nakatake Y, Toyooka Y, et al. Pluripotency governed by Sox2 via regulation of Oct3/4 expression in mouse embryonic stem cells. Nat Cell Biol, 2007, 9(6):625~635
    [132] Fong H, Hohenstein KA, Donovan PJ. Regulation of Self-renewal and Pluripotency by Sox2 in Human Embryonic Stem Cells. Stem Cells, 2008, 26(8):1931~1938
    [133] Ferri AL, Cavallaro M, Braida D, et al. Sox2 deficiency causes neurodegeneration and impaired neurogenesis in the adult mouse brain. Development, 2004, 131(15):3805~3819
    [134] Williamson KA, Hever AM, Rainger J, et al. Mutations in SOX2 cause anophthalmia-esophageal-genital (AEG) syndrome. Hum Mol Genet, 2006, 15(9):1413~1422
    [135] Kelberman D, Rizzoti K, Avilion A, et al., Mutations within Sox2/SOX2 are associated with abnormalities in the hypothalamo-pituitary-gonadal axis in mice and humans. J Clin Invest, 2006, 116(9):2442~2455
    [136] Dong C, Wilhelm D, Koopman P. Sox genes and cancer. Cytogenet Genome Res, 2004, 105(2-4):442~447
    [137] Dang DT, Pevsner J, Yang VW. The biology of the mammalian Krüppel-like family of transcription factors. Int J Biochem Cell Biol, 2000, 32(11-12):1103~1121
    [138] Conkright MD, Wani MA, Anderson KP, et al. A gene encoding an intestinal-enriched member of the Krüppel-like factor family expressed in intestinal epithelial cells. Nucleic Acids Res, 1999, 27(5):1263~1270
    [139] Segre JA, Bauer C, Fuchs E. Klf4 is a transcription factor required for establishing the barrier function of the skin. Nat Genet, 1999, 22(4):356~360
    [140] Nakatake Y, Fukui N, Iwamatsu Y, et al. Klf4 cooperates with Oct3/4 and Sox2 to activate the Lefty1 core promoter in embryonic stem cells. Mol Cell Biol, 2006, 26(20): 7772~7782
    [141] Jiang J, Chan YS, Loh YH, et al. A core Klf circuitry regulates self-renewal of embryonicstem cells. Nat Cell Biol, 2008, 10(3): 353~360
    [142] Alon U. Network motifs: theory and experimental approaches. Nat Rev Genet, 2007, 8(6): 450~461
    [143] Li Y, McClintick J, Zhong L, et al. Murine embryonic stem cell differentiation is promoted by SOCS-3 and inhibited by the zinc finger transcription factor Klf4. Blood, 2005, 105: 635~637
    [144] McConnell BB, Ghaleb AM, Nandan MO, et al. The diverse functions of Krüppel-like factors 4 and 5 in epithelial biology and pathobiology. Bioessays, 2007, 29(6):549~557
    [145] Knoepfler PS, Zhang XY, Cheng PF, et al. Myc influences global chromatin structure. EMBO J, 2006, 25(12): 2723~2734
    [146] Dominguez-Sola D, Ying CY, Grandori C, et al. Nontranscriptional control of DNA replication by c-Myc. Nature, 2007, 448(7152): 445~451
    [147] Cartwright P, McLean C, Sheppard A, et al. LIF/STAT3 controls ES cell self-renewal and pluripotency by a Myc-dependent mechanism. Development, 2005, 132: 885~896
    [148] Davis AC, Wims M, Spotts GD,et al., A null c-myc mutation causes lethality before 10.5 days of gestation in homozygotes and reduced fertility in heterozygous female mice. Genes Dev, 1993, 7(4):671~682
    [149] Chambers I, Colby D, Robertson M, et al. Functional expression cloning of Nanog, a pluripotency sustaining factor in embryonic stem cells. Cell. 2003, 113(5):643~655
    [150] Lagarkova MA, Volchkov PY, Lyakisheva AV,et al. Diverse epigenetic profile of novel human embryonic stem cell lines. Cell Cycle, 2006, 5(4):416~420
    [151] Wang J, Rao S, Chu J, et al. A protein interaction network for pluripotency of embryonic stem cells. Nature, 2006, 444(7117):364~368
    [152] Scheper W, Copray S. The molecular mechanism of induced pluripotency: a two-stage switch. Stem Cell Rev, 2009, 5(3):204~223
    [153] Smith AG, Heath JK, Donaldson DD, et al. Inhibition of pluripotential embryonic stem cell differentiation by purified polypeptides. Nature, 1988, 336(6200):688~690
    [154] Liu Y, Ji L, Ten Y, et al. The molecular mechanism of embryonic stem cell pluripotency and self-renewal. Sci China C Life Sci, 2007, 50(5):619~623
    [155] Xu RH, Chen X, Li DS, et al. BMP4 initiates human embryonic stem cell differentiation to trophoblast. Nat Biotechnol, 2002, 20(12):1261~1264
    [156] Qi X, Li TG, Hao J, et al. BMP4 supports self-renewal of embryonic stem cells by inhibiting mitogen-activated protein kinase pathways. Proc Natl Acad Sci U S A, 2004, 101(16):6027~6032
    [157] Welstead GG, Schorderet P, Boyer LA. The reprogramming language of pluripotency. Curr Opin Genet Dev, 2008, 18(2):123~129
    [158] Wu KJ, Grandori C, Amacker M, et al. Direct activation of TERT transcription by c-MYC. Nat Genet. 1999, 21(2):220~224
    [159] Rowland BD, Bernards R, Peeper DS. The KLF4 tumour suppressor is a transcriptional repressor of p53 that acts as a context-dependent oncogene. Nat Cell Biol, 2005, 7(11):1074~1082
    [160] Zhang W, Geiman DE, Shields JM, et al., The gut-enriched Kruppel-like factor (Kruppel-like factor 4) mediates the transactivating effect of p53 on the p21WAF1/Cip1 promoter. J Biol Chem, 2000, 16;275(24):18391~18398
    [161] Seoane J, Le HV, MassaguéJ. Myc suppression of the p21(Cip1) Cdk inhibitor influences the outcome of the p53 response to DNA damage. Nature, 2002, 419(6908):729~734
    [162] Yamanaka S. Strategies and new developments in the generation of patient-specific pluripotent stem cells. Cell Stem Cell, 2007, 1(1):39~49
    [163] Wei Z, Yang Y, Zhang P, et al. Klf4 interacts directly with Oct4 and Sox2 to promote reprogramming. Stem Cells, 2009, 27(12):2969~2978
    [164] Lin T, Chao C, Saito S, et al. p53 induces differentiation of mouse embryonic stem cells by suppressing Nanog expression. Nat Cell Biol, 2005, 7(2):165~171
    [165] Evans PM, Zhang W, Chen X, et al. Kruppel-like factor 4 is acetylated by p300 and regulates gene transcription via modulation of histone acetylation. J Biol Chem, 2007, 282(47):33994~34002
    [166] Miyagi S, Saito T, Mizutani K,et al. The Sox-2 regulatory regions display their activities in two distinct types of multipotent stem cells. Mol Cell Biol,2004, 24(10):4207~4220
    [167] Balasubramanian S, Babai N, Chaudhuri A, et al. Non cell-autonomous reprogramming of adult ocular progenitors: generation of pluripotent stem cells without exogenous transcription factors. Stem Cells, 2009, 27(12):3053~3062
    [168] Balzer E, Moss EG. Localization of the developmental timing regulator Lin28 to mRNP complexes, P-bodies and stress granules. RNA Biol, 2007, 4(1):16~25
    [169] Kumar MS, Erkeland SJ, Pester RE, et al. Suppression of non-small cell lung tumor development by the let-7 microRNA family. Proc Natl Acad Sci U S A, 2008, 105(10):3903~3908
    [170] Viswanathan SR, Daley GQ. Lin28: A microRNA regulator with a macro role. Cell, 2010, 140(4):445~449
    [171] Feldman N, Gerson A, Fang J, et al. G9a-mediated irreversible epigenetic inactivation of Oct-3/4 during early embryogenesis. Nat Cell Biol, 2006, 8(2):188~194
    [172] Jenuwein T, Allis CD. Translating the histone code. Science, 2001, 293(5532):1074~1080
    [173] Bernstein BE, Meissner A, Lander ES. The mammalian epigenome. Cell, 2007, 128 (4): 669~681
    [174] Bracken AP, Dietrich N, Pasini D, et al. Genome-wide mapping of Polycomb target genes unravels their roles in cell fate transitions. Genes Dev, 2006, 20 (9): 1123~1136
    [175] Boyer LA, Plath K, Zeitlinger J, et al. Polycomb complexes repress developmental regulatorsin murine embryonic stem cells. Nature, 2006, 441(7091):349~353
    [176] Silva J, Smith A. Capturing pluripotency. Cell, 2008, 132(4):532~536
    [177] Li H, Collado M, Villasante A, et al. The Ink4/Arf locus is a barrier for iPS cell reprogramming. Nature, 2009, 460(7259):1136~1139
    [178] Loh YH, Zhang W, Chen X, et al. Jmjd1a and Jmjd2c histone H3 Lys 9 demethylases regulate self-renewal in embryonic stem cells. Genes Dev, 2007, 21(20):2545~2557
    [179] Chin MH, Mason MJ, Xie W, et al. Induced pluripotent stem cells and embryonic stem cells are distinguished by gene expression signatures. Cell Stem Cell, 2009 , 5(1):111~123
    [180] Ghosh Z, Wilson KD, Wu Y et al. Persistent donor cell gene expression among human induced pluripotent stem cells contributes to differences with human embryonic stem cells. PLoS One, 2010, 5(2):e8975
    [181] Hanna J, Wernig M, Markoulaki S, et al. Treatment of sickle cell anemia mouse model with iPS cells generated from autologous skin. Science, 2007, 318 (5858): 1920~1923
    [182] Wernig M, Zhao JP, Pruszak J, et al. Neurons derived from reprogrammed fibroblasts functionally integrate into the fetal brain and improve symptoms of rats with Parkinson's disease. Proc Natl Acad Sci U S A, 2008c, 105(15): 5856~5861
    [183] Xu D, Alipio Z, Fink LM, et al. Phenotypic correction of murine hemophilia A using an iPS cell-based therapy. Proc Natl Acad Sci U S A, 2009, 106(3):808~813
    [184] Nelson TJ, Martinez-Fernandez A, Yamada S, et al. Repair of acute myocardial infarction by human stemness factors induced pluripotent stem cells. Circulation, 2009, 120(5):408~416
    [185] Dimos JT, Rodolfa KT, Niakan KK, et al. Induced pluripotent stem cells generated from patients with ALS can be differentiated into motor neurons.Science. 2008, 321(5893):1218~1221
    [186] Maehr R, Chen S, Snitow M, et al. Generation of pluripotent stem cells from patients with type 1 diabetes. Proc Natl Acad Sci U S A, 2009, 106(37):15768~15773
    [187] Tateishi K, He J, Taranova O, et al. Generation of insulin-secreting islet-like clusters from human skin fibroblasts. J Biol Chem. 2008 Nov 14;283(46):31601~31607
    [188] Zhang D, Jiang W, Liu M, et al. Highly efficient differentiation of human ES cells and iPS cells into mature pancreatic insulin-producing cells. Cell Res, 2009, 19(4):429~438
    [189] Song Z, Cai J, Liu Y, et al. Efficient generation of hepatocyte-like cells from human induced pluripotent stem cells. Cell Res, 2009, 19(11):1233~1242
    [190] Si-Tayeb K, Noto FK, Nagaoka M, et al. Highly efficient generation of human hepatocyte-like cells from induced pluripotent stem cells. Hepatology, 2010, 51(1):297~305
    [191] Gai H, Nguyen DM, Moon YJ, et al. Generation of murine hepatic lineage cells from induced pluripotent stem cells. Differentiation, 2010, 79(3):171~181
    [192] Kobayashi N, Rivas-Carrillo JD, Soto-Gutierrez A, et al. Gene delivery to embryonic stem cells. Birth Defects Res C Embryo Today, 2005,75(1):10~18
    [193] West FD, Terlouw SL, Kwon DJ, et al. Porcine Induced Pluripotent Stem Cells Produce Chimeric Offspring. Stem Cells Dev. 2010 Apr 9. [Epub ahead of print]
    [194] Hochedlinger K, Jaenisch R. Nuclear transplantation: lessons from frogs and mice. Curr Opin Cell Biol, 2002, 14:741~748.
    [195] Niemann H, Kues W, Carnwath JW. Transgenic farm animals: present and future. Rev Sci Tech, 2005, 24(1):285~298
    [196] Fujimura T, Takahagi Y, Shigehisa T, et al. Production of alpha 1,3-galactosyltransferase gene-deficient pigs by somatic cell nuclear transfer: a novel selection method for gal alpha 1,3-Gal antigen-deficient cells. Mol Reprod Dev, 2008, 75(9):1372~1378
    [197] Osafune K, Caron L, Borowiak M, et al., Marked differences in differentiation propensity among human embryonic stem cell lines. Nat Biotechnol, 2008, 26(3):313~315
    [198] Carpenter MK, Frey-Vasconcells J, Rao MS. Developing safe therapies from human pluripotent stem cells. Nat Biotechnol, 2009, 27(7):606~613
    [199] Stadtfeld M, Apostolou E, Akutsu H, et al. Aberrant silencing of imprinted genes on chromosome 12qF1 in mouse induced pluripotent stem cells. Nature, 2010, 465(7295):175~181
    [200] Kou Z, Kang L, Yuan Y, et al. Mice Cloned from Induced Pluripotent Stem Cells (iPSC). Biol Reprod. 2010 Apr 28. [Epub ahead of print]
    [201] Keefer CL, Pant D, Blomberg L, et al. Challenges and prospects for the establishment of embryonic stem cell lines of domesticated ungulates. Anim Reprod Sci, 2007, 98(1-2):147~168
    [202] Kim HS, Son HY, Kim S, et al. Isolation and initial culture of porcine inner cell masses derived from in vitro-produced blastocysts. Zygote, 2007, 15(1):55~63
    [203] Evans MJ, Notarianni E, Laurie S, et al. Derivation and preliminary characterization of pluripotent cell lines from porcine and bovine blastocysts. Theriogenology, 1990, 33(1):125~128
    [204] Strojek RM, Reed MA, Hoover JL,et al. A method for cultivating morphologically undifferentiated embryonic stem cells from porcine blastocysts. Theriogenology, 1990, 33(4):901~913
    [205] Piedrahita JA, Anderson GB, Bondurant RH. On the isolation of embryonic stem cells: Comparative behavior of murine, porcine and ovine embryos. Theriogenology, 1990, 34(5):879~901
    [206]杨玉艾,华进联,窦忠英.绿色荧光蛋白在胚胎干细胞研究中的应用.草食家畜, 2004, 6(2):23~27
    [207] J.萨姆布鲁克, D.W.拉塞尔.分子克隆实验指南.北京:科学出版社, 2002
    [208]薛奕杰.猪慢病毒载体pLL-Myc和pLL-Klf4的构建: [硕士学位论文].合肥:安徽农业大学, 2009
    [209]张卫琴.猪Sox2和Nanog慢病毒表达载体的构建: [硕士学位论文].合肥:安徽农业大学, 2009
    [210]成志军,陶勇,章孝荣.胎猪卵巢发生过程中的组织学变化.细胞生物学杂志, 2008, 30: 352~356
    [211] Goel S, Sugimoto M, Minami N, et al. Identification, isolation, and in vitro culture of porcine gonocytes. Biol Reprod, 2007, 77(1):127~137
    [212] Jiang P, Yamauchi K, YangM, et al. Tumor cells genetically labeled with GFP in the nucleus and RFP in the cytoplasm for imaging cellular dynamics. Cell Cycle, 2006, 5 (11): 1198~1201
    [213] Hall V. Porcine embryonic stem cells: a possible source for cell replacement therapy. Stem Cell Rev, 2008, 4(4):275~282
    [214]薛庆善.体外培养的原理与技术.北京:科学出版社, 2001, 87~101
    [215] Wakayama T, Perry AC, Zuccotti M, et al. Full-term development of mice from enucleated oocytes injected with cumulus cell nuclei. Nature, 1998, 394(6691):369-374
    [216] Mali P, Ye Z, Hommond HH, et al. Improved efficiency and pace of generating induced pluripotent stem cells from human adult and fetal fibroblasts. Stem Cells, 2008, 26(8):1998~2005
    [217] Brevini TA, Antonini S, Cillo F, et al. Porcine embryonic stem cells: Facts, challenges and hopes. Theriogenology, 2007, 68 Suppl 1:S206~213
    [218] Vackova I, Ungrova A, Lopes F. Putative embryonic stem cell lines from pig embryos. J Reprod Dev, 2007, 53(6):1137~1149
    [219] Moore K, Piedrahita JA. The effects of human leukemia inhibitory factor (hLIF) and culture medium on in vitro differentiation of cultured porcine inner cell mass (pICM). In Vitro Cell Dev Biol Anim, 1997, 33(1):62~71
    [220] Amit M, Carpenter MK, Inokuma MS, et al. Clonally derived human embryonic stem cell lines maintain pluripotency and proliferative potential for prolonged periods of culture. Dev Biol, 2000, 227(2):271~278
    [221] Evans MJ, Kaufman MH. Establishment in culture of pluripotential cells from mouse embryos. Nature, 1981, 292(5819):154~156
    [222] Tiscornia G, Singer O, Verma IM. Production and purification of lentiviral vectors. Nat Protoc, 2006, 1(1):241~245
    [223] Maherali N, Sridharan R, Xie W, et al. Directly reprogrammed fibroblasts show global epigenetic remodeling and widespread tissue contribution. Cell Stem Cell, 2007, 1(1): 55~70
    [224] Poueymirou WT, Auerbach W, Frendewey D, et al., F0 generation mice fully derived from gene-targeted embryonic stem cells allowing immediate phenotypic analyses. Nat Biotechnol, 2007, 25(1):91~99
    [225] Park TS, Galic Z, Conway AE, et al. Derivation of primordial germ cells from human embryonic and induced pluripotent stem cells is significantly improved by co-culture withhuman fetal gonadal cell. Stem Cells, 2009, 27: 783~795
    [226] Duinsbergen D, Salvatori D, Eriksson M,et al. Tumors originating from induced pluripotent stem cells and methods for their prevention. Ann N Y Acad Sci, 2009, 1176:197~204
    [227] Green M, Loewenstein PM. Autonomous functional domains of chemically synthesized human immunodeficiency virus tat trans-activator protein. Cell, 1988, 55(6):1179~1188
    [228] Frankelf AD, Pabo CO. Cellular uptake of the tat protein from human immunodeficiency virus. Cell, 1988, 55(6):1189~1193
    [229] Fonseca SB, Pereira MP, Kelley SO. Recent advances in the use of cell-penetrating peptides for medical and biological applications. Adv Drug Deliv Rev, 2009, 61(11):953~964
    [230] Vives E, Brodin P, Lebleu B. A truncated HIV-1 Tat protein basic domain rapidly translocates through the plasma membrane and accumulates in the cell nucleus. J Biol Chem, 1997, 272(25):16010~16017
    [231] Wender PA, Mitchell DJ, Pattabiraman K, et al. The design, synthesis, and evaluation of molecules that enable or enhance cellular uptake: peptoid molecular transporters. Proc Natl Acad Sci U S A, 2000, 97(24):13003~13008
    [232] Futaki S. Arginine-rich peptides: potential for intracellular delivery of macromolecules and the mystery of the translocation mechanisms. Int J Pharm, 2002, 245(1-2):1~7
    [233] Veach RA, Liu D, Yao S, et al. Receptor/Transporter-independent Targeting of Functional Peptides across the Plasma Membrane. J. Biol. Chem., 2004, 279(12): 11425~11431
    [234] Melikov K, Chernomordik LV. Arginine-rich cell penetrating peptides: from endosomal uptake to nuclear delivery. Cell Mol Life Sci, 2005, 62(23):2739~2749
    [235] Futaki S, Nakase I, Tadokoro A, et al. Arginine-rich peptides and their internalization mechanisms. Biochem Soc Trans. 2007, 35(Pt 4):784~787
    [236] Herce HD, Garcia AE, Litt J, et al. Arginine-rich peptides destabilize the plasma membrane, consistent with a pore formation translocation mechanism of cell-penetrating peptides. Biophys J, 2009, 97(7):1917~1925
    [237] Caron NJ, Quenneville SP, Tremblay JP. Endosome disruption enhances the functional nuclear delivery of Tat-fusion proteins. Biochem Biophys Res Commun, 2004, 319(1):12~20
    [238]胡笳,郭燕婷,李艳梅.蛋白质翻译后修饰.科学通报, 2005, 6(11): 1061~1072
    [239] Alberio R, Croxall N, Allegrucci C. Pig Epiblast Stem Cells Depend on Activin/Nodal Signaling for Pluripotency and Self Renewal. Stem Cells Dev, 2010 Mar 8. [Epub ahead of print]

© 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