Generation of porcine induced pluripotent stem cells and their differentiation into cardiac lineages

108 404 0
Generation of porcine induced pluripotent stem cells and their differentiation into cardiac lineages

Đang tải... (xem toàn văn)

Tài liệu hạn chế xem trước, để xem đầy đủ mời bạn chọn Tải xuống

Thông tin tài liệu

.. .GENERATION OF PORCINE INDUCED PLURIPOTENT STEM CELLS AND THEIR DIFFERENTIATION INTO CARDIAC LINEAGES TAN WAN CHIU GRACE (B.Eng.(Hons.), NTU) A THESIS SUBMITTED FOR THE DEGREE OF MASTER OF. .. generate porcine induced pluripotent stem cells (IPSCs) from porcine fibroblast cells and differentiate such cells into cardiac lineages to provide a model to study the development of cardiac cells. .. Pluripotent Stem Cells …………………………………………………………………… 45 4.9 Culture of cells transitioning from fibroblasts to Induced Pluripotent Stem Cells 45 iv 4.10 Culture of Porcine Induced Pluripotent Stem

GENERATION OF PORCINE INDUCED PLURIPOTENT STEM CELLS AND THEIR DIFFERENTIATION INTO CARDIAC LINEAGES TAN WAN CHIU GRACE (B.Eng.(Hons.), NTU) NATIONAL UNIVERSITY OF SINGAPORE 2014 GENERATION OF PORCINE INDUCED PLURIPOTENT STEM CELLS AND THEIR DIFFERENTIATION INTO CARDIAC LINEAGES TAN WAN CHIU GRACE (B.Eng.(Hons.), NTU) A THESIS SUBMITTED FOR THE DEGREE OF MASTER OF ENGINEERING DEPARTMENT OF BIOENGINEERING NATIONAL UNIVERSITY OF SINGAPORE 2014 Declaration I hereby declare that this thesis is my original work and it has been written by me in its entirety I have duly acknowledged all the sources of information which have been used in the thesis This thesis has also not been submitted for any degree in any university previously Tan Wan Chiu Grace July 2014 i ACKNOWLEDGEMENTS I would like to thank the following people who have helped me complete this master’s thesis Firstly, I would like to thank A/ Prof Tong Yen Wah for his support in the entire Master’s programme I would like to thank him for his patience throughout the course I would also like to thank Dr Winston Shim and Dr Wei Heming for their guidance throughout my time at National Heart Centre They have provided great help in the design of the experiments and the technical aspects of the project I would also like to thank my colleagues at National Heart Centre who have helped me in one way or another ii Table of Contents Introduction 13 Aims and objectives 14 Literature Review 15 3.1 Human Embryonic Stem Cells (hESCs) 15 3.2 Porcine Embryonic Stem Cells 16 3.3 Generation of Induced Pluripotent Stem Cells 17 3.3.1 Retroviral/ Lentiviral Reprogramming 18 3.3.2 Episomal Reprogramming 20 3.3.3 mRNA reprogramming 21 3.3.4 Chemical reprogramming 21 3.3.5 Genes that regulate pluripotency in Human Embryonic Stem Cells………………… 22 3.3.6 OCT4………………………………………………………………23 3.3.7 SOX2………… 24 3.3.8 NANOG 24 3.3.9 KLF4…………… 25 3.3.10 c-myc……… 26 3.3.11 Lin28………… 26 3.4 Cardiac differentiation of Human IPS cell lines 27 3.5 Generation of Porcine Induced Pluripotent Stem Cells 29 3.5.1 Factors used for reprogramming of porcine cells 29 3.5.2 Reprogramming techniques of porcine cells 29 3.5.3 Retroviral and Lentiviral reprogramming 30 3.5.4 Episomal reprogramming of Porcine IPS cells 30 3.5.5 Reprogramming with decreased number of transcription factors………… 31 iii 3.6 Characterization and Definition of Porcine IPS cell lines 32 3.7 Difference in culture methods of Porcine IPSCs 34 3.7.1 Culture media 34 3.7.2 Culture on Matrigel 34 3.8 Differentiation protocol that has been published for pigs 35 3.8.1 Neural………… 35 3.8.2 Liver………… 36 3.8.3 Cardiac differentiation protocol 36 Materials and Methods 37 4.1 Culture of Mouse Embryonic Fibroblast (MEF) cells as feeder cells for hESCs 37 4.2 Culture of Human Embryonic Stem Cells (hESCs) 38 4.2.1 Culture of Human Embryonic Stem Cells on MEF feeders 38 4.2.2 Feeder- free Culture of Human Embryonic Stem Cells 39 4.2.3 Coating of Matrigel ™ 40 4.2.4 Enzymatic passaging of Human IPSCs 40 4.3 Differentiation of Human IPS cells into cardiomyocytes 40 4.4 Patch clamp recordings of human IPS derived cardiomyocytes 42 4.5 Culture of Porcine Fibroblast Cells 42 4.6 Preparation of Plasmids for transfection 42 4.7 Packaging of Lentiviruses for the induction of Pluripotent Stem Cells………………… 44 4.8 Infection of fibroblast cells to form Induced Pluripotent Stem Cells …………………………………………………………………… 45 4.9 Culture of cells transitioning from fibroblasts to Induced Pluripotent Stem Cells 45 iv 4.10 Culture of Porcine Induced Pluripotent Stem Cells 45 4.10 Characterization of Porcine Induced Pluripotent Stem Cells 46 4.10.1 Immunocytochemical staining for Porcine IPSCs 47 4.10.2 Polymerase Chain Reaction for Porcine IPSCs 49 4.10.3 Teratoma formation 50 4.10.4 Karyotyping 50 4.10.5 Alkaline Phosphatase Staining 50 4.11 Differentiation 51 4.11.1 In vitro teratoma 51 4.11.2 Differentiation into fat and cardiomyocytes 51 4.11.3 Staining of fat globules 52 Results……… 53 5.1 Morphology of hESC 53 5.2 Morphology of iPSCs 54 5.3 Differentiation of hESCs / hIPSCs into cardiomyocytes 55 5.4 Action potential recording of IPS derived cardiomyocytes 56 5.5 Culture of porcine fibroblast cells 57 5.6 Reprogramming process of porcine fibroblast cells 58 5.6 Formation of Porcine IPS colonies and the continuous culture of the colonies 60 5.7 Characterization of Porcine IPS cells 65 5.7.1 Immunocytochemical Staining of Porcine IPS colonies 65 5.7.2 PCR for exogenous genes in Porcine IPS 72 5.7.3 PCR for endogenous genes in porcine IPS 75 5.7.4 Teratoma formation 76 5.7.5 Karyotyping of IPS clones 77 v 5.7.6 Alkaline Phosphatase Staining of Porcine IPS colonies 78 5.8 Withdrawal of bFGF from the culture media 79 5.9 In vitro teratoma formation 82 5.10 Differentiation of porcine IPS cells into fat and cardiomyocytes 84 Discussion 89 6.1 Morphology of porcine IPS cells 89 6.2 Expression of pluripotent genes in porcine IPS cells 89 6.3 Function of LIF in the culture of porcine IPS cells 90 6.4 Importance of doxycycline in the culture media 91 6.5 Differentiation of porcine IPS cells into cardiomyocytes 92 6.6 Use of porcine cells for other downstream work 92 Future work 94 Conclusion 95 References ……………………………………………………………96 vi SUMMARY Heart disease is a major cause of death worldwide As a result of myocardial infarction, functional cardiomyocytes are lost and this results in a heart that does not contract properly The pig model is a good model to study cardiac functions in humans, as the size of the heart in the pig is similar to that of the human In this project, we aim to convert porcine fibroblast cells into porcine cardiomyocytes, in the hope of creating new functional cardiomyocytes to replace the ones that are no longer functional after myocardial infarction As no porcine embryonic stem cells have been isolated, induced pluripotent stem cells for the pig must be used as a source of pluripotent cells Hence, fibroblasts from the pig is taken, and induced into pluripotent cells These cells are in turn differentiated into cardiomyocytes, where more downstream work can be done to characterize them and also to use these cells as form of therapy for the failing heart In this project, we isolated fibroblasts from the pig’s thigh region, and used lentiviruses to induce the cells to pluripotent cells These pluripotent cells are studied and characterized The pluripotent stem cells exhibited morphology and genomic markers of pluripotency These cells were also finally differentiated into cardiomyocytes Even though the differentiated cells did not beat, there were cardiac genes found to be expressed in the form of mRNA This shows that the differentiation protocol is successful to a certain extent More work can be done to streamline to protocol to achieve greater efficiency in the production of porcine cardiomyocytes vii List of Tables Table 3.1 The applications, reprogramming methods and the advantages of using each method for somatic cell reprogramming (Gonzalez, Boue, & Izpisua Belmonte, 2011) 18 Table 3.2 Various pluripotent characteristics which defined porcine IPS cells 33 Table 4.1 Materials required for complete media 37 Table 4.2 Materials required for hESC media 39 Table 4.3 Materials required for CARM media 41 Table 4.4 Materials required for Mouse ESC media 46 Table 4.5 Primary Antibodies used for immunocytochemical staining 47 Table 4.6 Secondary Antibodies used for Immunocytochemical Staining 48 Table 4.7 Protocol for Polymerase Chain Reaction 49 Table 5.1 PCR primers for exogenous transcription factors 73 Table 5.2 PCR primers for endogenous transcription factors 75 Table 5.3 Primers used for determination of the presence of germ layers 83 Table 5.4 PCR primers to determine the presence of cardiomyocytes 87 viii of pluripotent cells The activation of endogenous genes were also observed through the use of RT-PCR Although the continuous expression of exogenous genes was necessary for the pluripotency of the porcine IPS cells, there was also the activation of endogenous genes However, we postulate that the expression of the selected endogenous genes may not be sufficient to sustain the cells for prolonged culture This may be due to the lack of other critical growth factors in the culture media Further work may be necessary to define the culture conditions best suited to porcine IPS cells 6.5 Differentiation of porcine IPS cells into cardiomyocytes Thus far, we were not able to differentiate the porcine IPSC to beating cardiomyocytes in our culture However, we were able to detect cardiac markers that were present in the culture The expression of these cardiac markers could come from precursors of cardiomyocytes and hence are cells that may not beat This may explain why no beating cells were observed Also, there may be the possibility of the presence of beating cardiomyocytes, but the efficiency of the production of cardiomyocytes was extremely low, and the beating cells were too few to be observed visually Though their observed differentiation towards fat cells suggested that mesodermal differentiation was initiated, but directing the lineage towards cardio-mesoderm and subsequent cardiomyocytes remains to be elucidated 6.6 Use of porcine cells for other downstream work Although the porcine IPS cells were not able to differentiate into cardiomyocytes, it is possible to consider the use of the porcine IPS cells for other downstream experiments, such as the differentiation of the cells into mesenchymal stem cells Studies 92 have shown that through the differentiation of IPS cells, MSCs can also be obtained alongside the IPS cells (Wei et al., 2012) Differentiated cells which not exhibit cardiomyocte behavior can be harvested and characterized to determine the nature of such cells 93 Future work The understanding of the developmental biology of porcine embryonic stem cells will enable us to understand the signaling pathway of such cells and the culture conditions to maintain the pluripotency of porcine pluripotent stem cells There have been several groups that each have their own definition of porcine pluripotent cells, and it is important to standardize such standards to ensure that the porcine IPS cell lines generated are of good quality and of good differentiation potential The differentiation of porcine IPS cells into cardiomyocytes has only been shown by one group The results have not been reproduced by other laboratories and we similarly failed to differentiate the porcine IPSC following that published protocol Further investigations will need to be done to improve the cardiac differentiation protocol The potential divergent of signaling pathways for cardiomyocytes differentiation in pig and human should be investigated in detail as cardiac differentiation protocols that worked in human IPSC failed to elicit similar outcome from the porcine IPSC 94 Conclusion We have successfully generated multiple lines of porcine IPS cells with pluripotent differentiation potential These cell lines serve to provide valuable bench-top model for studying mesoderm differentiation and cardiomesoderm commitment for better understanding of the divergent differentiation cascade between human and porcine system 95 References Ameen, C., Strehl, R., Bjorquist, P., Lindahl, A., Hyllner, J., & Sartipy, P (2008) Human embryonic stem cells: current technologies and emerging industrial applications Crit Rev Oncol Hematol, 65(1), 54-80 doi: S1040-8428(07)00136-9 [pii] 10.1016/j.critrevonc.2007.06.012 Amit, M., Carpenter, M K., Inokuma, M S., Chiu, C P., Harris, C P., Waknitz, M A., Thomson, J A (2000) Clonally derived human embryonic stem cell lines maintain pluripotency and proliferative potential for prolonged periods of culture Dev Biol, 227(2), 271-278 doi: 10.1006/dbio.2000.9912 S0012-1606(00)99912-3 [pii] Aravalli, R N., Cressman, E N., & Steer, C J (2012) Hepatic differentiation of porcine induced pluripotent stem cells in vitro Vet J, 194(3), 369-374 doi: 10.1016/j.tvjl.2012.05.013 Avilion, A A., Nicolis, S K., Pevny, L H., Perez, L., Vivian, N., & Lovell-Badge, R (2003) Multipotent cell lineages in early mouse development depend on SOX2 function Genes Dev, 17(1), 126140 doi: 10.1101/gad.224503 Bergmann, O., Bhardwaj, R D., Bernard, S., Zdunek, S., BarnabeHeider, F., Walsh, S., Frisen, J (2009) Evidence for cardiomyocyte renewal in humans Science, 324(5923), 98-102 doi: 10.1126/science.1164680 Blau, H M., & Rossi, F M (1999) Tet B or not tet B: advances in tetracycline-inducible gene expression Proc Natl Acad Sci U S A, 96(3), 797-799 Brambrink, T., Foreman, R., Welstead, G G., Lengner, C J., Wernig, M., Suh, H., & Jaenisch, R (2008) Sequential expression of pluripotency markers during direct reprogramming of mouse 96 somatic cells Cell Stem Cell, 2(2), 151-159 doi: 10.1016/j.stem.2008.01.004 Brevini, T A., Antonini, S., Cillo, F., Crestan, M., & Gandolfi, F (2007) Porcine embryonic stem cells: Facts, challenges and hopes Theriogenology, 68 Suppl 1, S206-213 doi: 10.1016/j.theriogenology.2007.05.043 Brook, F A., & Gardner, R L (1997) The origin and efficient derivation of embryonic stem cells in the mouse Proc Natl Acad Sci U S A, 94(11), 5709-5712 Chambers, I., Colby, D., Robertson, M., Nichols, J., Lee, S., Tweedie, S., & Smith, A (2003) Functional expression cloning of Nanog, a pluripotency sustaining factor in embryonic stem cells Cell, 113(5), 643-655 doi: S0092867403003921 [pii] Chen, J., Lu, Z., Cheng, D., Peng, S., & Wang, H (2011) Isolation and characterization of porcine amniotic fluid-derived multipotent stem cells PLoS One, 6(5), e19964 doi: 10.1371/journal.pone.0019964 Cheng, D., Guo, Y., Li, Z., Liu, Y., Gao, X., Gao, Y., Wang, H (2012) Porcine induced pluripotent stem cells require LIF and maintain their developmental potential in early stage of embryos PLoS One, 7(12), e51778 doi: 10.1371/journal.pone.0051778 Cheng, D., Li, Z., Liu, Y., Gao, Y., & Wang, H (2012) Kinetic analysis of porcine fibroblast reprogramming toward pluripotency by defined factors Cell Reprogram, 14(4), 312-323 doi: 10.1089/cell.2012.0025 Dang, D T., Pevsner, J., & Yang, V W (2000) The biology of the mammalian Kruppel-like family of transcription factors Int J Biochem Cell Biol, 32(11-12), 1103-1121 doi: S1357- 2725(00)00059-5 [pii] 97 Doyon, Y., & Cote, J (2004) The highly conserved and multifunctional NuA4 HAT complex Curr Opin Genet Dev, 14(2), 147-154 doi: 10.1016/j.gde.2004.02.009 Dvash, T., & Benvenisty, N (2004) Human embryonic stem cells as a model for early human development Best Pract Res Clin Obstet Gynaecol, 18(6), 929-940 doi: 10.1016/j.bpobgyn.2004.06.005 Evans, M J., & Kaufman, M H (1981) Establishment in culture of pluripotential cells from mouse embryos Nature, 292(5819), 154156 Ezashi, T., Telugu, B P., Alexenko, A P., Sachdev, S., Sinha, S., & Roberts, R M (2009) Derivation of induced pluripotent stem cells from pig somatic cells Proc Natl Acad Sci U S A, 106(27), 10993-10998 doi: 10.1073/pnas.0905284106 Ezashi, T., Telugu, B P., & Roberts, R M (2012) Induced pluripotent stem cells from pigs and other ungulate species: an alternative to embryonic stem cells? Reprod Domest Anim, 47 Suppl 4, 92-97 doi: 10.1111/j.1439-0531.2012.02061.x Fujishiro, S H., Nakano, K., Mizukami, Y., Azami, T., Arai, Y., Matsunari, H., Hanazono, Y (2013) Generation of naive-like porcine-induced pluripotent stem cells capable of contributing to embryonic and fetal development Stem Cells Dev, 22(3), 473-482 doi: 10.1089/scd.2012.0173 Gao, Y., Guo, Y., Duan, A., Cheng, D., Zhang, S., & Wang, H (2013) Optimization of Culture Conditions for Maintaining Porcine Induced Pluripotent Stem Cells DNA Cell Biol doi: 10.1089/dna.2013.2095 Gao, Y., Hyttel, P., & Hall, V J (2011) Dynamic changes in epigenetic marks and gene expression during porcine epiblast specification Cell Reprogram, 13(4), 345-360 doi: 10.1089/cell.2010.0110 98 Gonzalez, F., Boue, S., & Izpisua Belmonte, J C (2011) Methods for making induced pluripotent stem cells: reprogramming a la carte Nat Rev Genet, 12(4), 231-242 doi: 10.1038/nrg2937 Graichen, R., Xu, X., Braam, S R., Balakrishnan, T., Norfiza, S., Sieh, S., Davidson, B P (2008) Enhanced cardiomyogenesis of human embryonic stem cells by a small molecular inhibitor of p38 MAPK Differentiation, 76(4), 357-370 doi: 10.1111/j.14320436.2007.00236.x Hall, V J., Christensen, J., Gao, Y., Schmidt, M H., & Hyttel, P (2009) Porcine pluripotency cell signaling develops from the inner cell mass to the epiblast during early development Dev Dyn, 238(8), 2014-2024 doi: 10.1002/dvdy.22027 Hall, V J., Kristensen, M., Rasmussen, M A., Ujhelly, O., Dinnyes, A., & Hyttel, P (2012) Temporal repression of endogenous pluripotency genes during reprogramming of porcine induced pluripotent stem cells Cell Reprogram, 14(3), 204-216 doi: 10.1089/cell.2011.0089 Hanna, L A., Foreman, R K., Tarasenko, I A., Kessler, D S., & Labosky, P A (2002) Requirement for Foxd3 in maintaining pluripotent cells of the early mouse embryo Genes Dev, 16(20), 2650-2661 doi: 10.1101/gad.1020502 Harley, V R., Lovell-Badge, R., & Goodfellow, P N (1994) Definition of a consensus DNA binding site for SRY Nucleic Acids Res, 22(8), 1500-1501 Hay, D C., Sutherland, L., Clark, J., & Burdon, T (2004) Oct-4 knockdown induces similar patterns of endoderm and trophoblast differentiation markers in human and mouse embryonic stem cells Stem Cells, 22(2), 225-235 Hilbert, S L., & Ferrans, V J (1992) Porcine aortic valve bioprostheses: morphologic and functional considerations J Long Term Eff Med Implants, 2(2-3), 99-112 99 Hombria, J C., & Lovegrove, B (2003) Beyond homeosis HOX function in morphogenesis and organogenesis Differentiation, 71(8), 461-476 doi: 07108004 [pii] Hori, Y., Rulifson, I C., Tsai, B C., Heit, J J., Cahoy, J D., & Kim, S K (2002) Growth inhibitors promote differentiation of insulinproducing tissue from embryonic stem cells Proc Natl Acad Sci U S A, 99(25), 16105-16110 doi: 10.1073/pnas.252618999 252618999 [pii] Hou, P., Li, Y., Zhang, X., Liu, C., Guan, J., Li, H., Deng, H (2013) Pluripotent stem cells induced from mouse somatic cells by small-molecule compounds Science, 341(6146), 651-654 doi: 10.1126/science.1239278 Itskovitz-Eldor, J., Schuldiner, M., Karsenti, D., Eden, A., Yanuka, O., Amit, M., Benvenisty, N (2000) Differentiation of human embryonic stem cells into embryoid bodies compromising the three embryonic germ layers Mol Med, 6(2), 88-95 Kaczynski, J., Cook, T., & Urrutia, R (2003) Sp1- and Kruppel-like transcription factors Genome Biol, 4(2), 206 Kehat, I., Kenyagin-Karsenti, D., Snir, M., Segev, H., Amit, M., Gepstein, A., Gepstein, L (2001) Human embryonic stem cells can differentiate into myocytes with structural and functional properties of cardiomyocytes J Clin Invest, 108(3), 407-414 doi: 10.1172/JCI12131 Kim, J H., Auerbach, J M., Rodriguez-Gomez, J A., Velasco, I., Gavin, D., Lumelsky, N., McKay, R (2002) Dopamine neurons derived from embryonic stem cells function in an animal model of Parkinson's disease Nature, 418(6893), 50-56 doi: 10.1038/nature00900 nature00900 [pii] Laflamme, M A., Chen, K Y., Naumova, A V., Muskheli, V., Fugate, J A., Dupras, S K., Murry, C E (2007) 100 Cardiomyocytes derived from human embryonic stem cells in prosurvival factors enhance function of infarcted rat hearts Nat Biotechnol, 25(9), 1015-1024 doi: 10.1038/nbt1327 Lin, C H., Jackson, A L., Guo, J., Linsley, P S., & Eisenman, R N (2009) Myc-regulated microRNAs attenuate embryonic stem cell differentiation EMBO J, 28(20), 3157-3170 doi: 10.1038/emboj.2009.254 Liu, K., Ji, G., Mao, J., Liu, M., Wang, L., Chen, C., & Liu, L (2012) Generation of porcine-induced pluripotent stem cells by using OCT4 and KLF4 porcine factors Cell Reprogram, 14(6), 505513 doi: 10.1089/cell.2012.0047 Loh, Y H., Wu, Q., Chew, J L., Vega, V B., Zhang, W., Chen, X., Ng, H H (2006) The Oct4 and Nanog transcription network regulates pluripotency in mouse embryonic stem cells Nat Genet, 38(4), 431-440 doi: ng1760 [pii] 10.1038/ng1760 Masuda, S., Wu, J., Hishida, T., Pandian, G N., Sugiyama, H., & Izpisua Belmonte, J C (2013) Chemically induced pluripotent stem cells (CiPSCs): a transgene-free approach J Mol Cell Biol, 5(5), 354-355 doi: 10.1093/jmcb/mjt034 Meissner, A., Wernig, M., & Jaenisch, R (2007) Direct reprogramming of genetically unmodified fibroblasts into pluripotent stem cells Nat Biotechnol, 25(10), 1177-1181 doi: nbt1335 [pii] 10.1038/nbt1335 Mitsui, K., Tokuzawa, Y., Itoh, H., Segawa, K., Murakami, M., Takahashi, K., Yamanaka, S (2003) The homeoprotein Nanog is required for maintenance of pluripotency in mouse epiblast and ES cells Cell, 113(5), 631-642 doi: S0092867403003933 [pii] Montserrat, N., Bahima, E G., Batlle, L., Hafner, S., Rodrigues, A M., Gonzalez, F., & Izpisua Belmonte, J C (2011) Generation of 101 pig iPS cells: a model for cell therapy J Cardiovasc Transl Res, 4(2), 121-130 doi: 10.1007/s12265-010-9233-3 Montserrat, N., de Onate, L., Garreta, E., Gonzalez, F., Adamo, A., Eguizabal, C., Izpisua Belmonte, J C (2012) Generation of feeder-free pig induced pluripotent stem cells without Pou5f1 Cell Transplant, 21(5), 815-825 doi: 10.3727/096368911X601019 Mummery, C., Ward-van Oostwaard, D., Doevendans, P., Spijker, R., van den Brink, S., Hassink, R., Tertoolen, L (2003) Differentiation of human embryonic stem cells to cardiomyocytes: role of coculture with visceral endoderm-like cells Circulation, 107(21), 2733-2740 doi: 10.1161/01.CIR.0000068356.38592.68 Nakagawa, M., Koyanagi, M., Tanabe, K., Takahashi, K., Ichisaka, T., Aoi, T., Yamanaka, S (2008) Generation of induced pluripotent stem cells without Myc from mouse and human fibroblasts Nat Biotechnol, 26(1), 101-106 doi: 10.1038/nbt1374 Nichols, J., Zevnik, B., Anastassiadis, K., Niwa, H., KleweNebenius, D., Chambers, I., Smith, A (1998) Formation of pluripotent stem cells in the mammalian embryo depends on the POU transcription factor Oct4 Cell, 95(3), 379-391 doi: S00928674(00)81769-9 [pii] Niwa, H., Burdon, T., Chambers, I., & Smith, A (1998) Selfrenewal of pluripotent embryonic stem cells is mediated via activation of STAT3 Genes Dev, 12(13), 2048-2060 Niwa, H., Miyazaki, J., & Smith, A G (2000) Quantitative expression of Oct-3/4 defines differentiation, dedifferentiation or self-renewal of ES cells Nat Genet, 24(4), 372-376 doi: 10.1038/74199 Notarianni, E., Laurie, S., Moor, R M., & Evans, M J (1990) Maintenance and differentiation in culture of pluripotential embryonic cell lines from pig blastocysts J Reprod Fertil Suppl, 41, 51-56 102 Odorico, J S., Kaufman, D S., & Thomson, J A (2001) Multilineage differentiation from human embryonic stem cell lines Stem Cells, 19(3), 193-204 doi: 10.1634/stemcells.19-3-193 Okita, K., Ichisaka, T., & Yamanaka, S (2007) Generation of germline-competent induced pluripotent stem cells Nature, 448(7151), 313-317 doi: 10.1038/nature05934 Pan, G J., Chang, Z Y., Scholer, H R., & Pei, D (2002) Stem cell pluripotency and transcription factor Oct4 Cell Res, 12(5-6), 321329 doi: 10.1038/sj.cr.7290134 Pasumarthi, K B., & Field, L J (2002) Cardiomyocyte cell cycle regulation Circ Res, 90(10), 1044-1054 Pesce, M., & Scholer, H R (2001) Oct-4: gatekeeper in the beginnings of mammalian development Stem Cells, 19(4), 271278 Pevny, L H., & Lovell-Badge, R (1997) Sox genes find their feet Curr Opin Genet Dev, 7(3), 338-344 doi: S0959-437X(97)80147-5 [pii] Piedrahita, J A., Anderson, G B., & Bondurant, R H (1990) On the isolation of embryonic stem cells: Comparative behavior of murine, porcine and ovine embryos Theriogenology, 34(5), 879901 Ramalho-Santos, M., Yoon, S., Matsuzaki, Y., Mulligan, R C., & Melton, D A (2002) "Stemness": transcriptional profiling of embryonic and adult stem cells Science, 298(5593), 597-600 doi: 10.1126/science.1072530 1072530 [pii] Reubinoff, B E., Pera, M F., Fong, C Y., Trounson, A., & Bongso, A (2000) Embryonic stem cell lines from human blastocysts: somatic differentiation in vitro Nat Biotechnol, 18(4), 399-404 doi: 10.1038/74447 103 Rodriguez, A., Contreras, D A., & Alberio, R (2013) Isolation and culture of pig epiblast stem cells Methods Mol Biol, 1074, 97-110 doi: 10.1007/978-1-62703-628-3_8 Roger, V L., Go, A S., Lloyd-Jones, D M., Adams, R J., Berry, J D., Brown, T M., Stroke Statistics, Subcommittee (2011) Heart disease and stroke statistics 2011 update: a report from the American Heart Association Circulation, 123(4), e18-e209 doi: 10.1161/CIR.0b013e3182009701 Rossant, J (2007) Stem cells and lineage development in the mammalian blastocyst Reprod Fertil Dev, 19(1), 111-118 Ruan, W., Han, J., Li, P., Cao, S., An, Y., Lim, B., & Li, N (2011) A novel strategy to derive iPS cells from porcine fibroblasts Sci China Life Sci, 54(6), 553-559 doi: 10.1007/s11427-011-4179-5 Sinclair, A H., Berta, P., Palmer, M S., Hawkins, J R., Griffiths, B L., Smith, M J., Goodfellow, P N (1990) A gene from the human sex-determining region encodes a protein with homology to a conserved DNA-binding motif Nature, 346(6281), 240-244 doi: 10.1038/346240a0 Symons, J A., Alcami, A., & Smith, G L (1995) Vaccinia virus encodes a soluble type I interferon receptor of novel structure and broad species specificity Cell, 81(4), 551-560 Synnergren, J., Akesson, K., Dahlenborg, K., Vidarsson, H., Ameen, C., Steel, D., Sartipy, P (2008) Molecular signature of cardiomyocyte clusters derived from human embryonic stem cells Stem Cells doi: 2007-1033 [pii] 10.1634/stemcells.2007-1033 Takahashi, K., Tanabe, K., Ohnuki, M., Narita, M., Ichisaka, T., Tomoda, K., & Yamanaka, S (2007) Induction of pluripotent stem cells from adult human fibroblasts by defined factors Cell, 131(5), 861-872 doi: 10.1016/j.cell.2007.11.019 104 Tam, P P., & Behringer, R R (1997) Mouse gastrulation: the formation of a mammalian body plan Mech Dev, 68(1-2), 3-25 Tanabe, K., Nakamura, M., Narita, M., Takahashi, K., & Yamanaka, S (2013) Maturation, not initiation, is the major roadblock during reprogramming toward pluripotency from human fibroblasts Proc Natl Acad Sci U S A, 110(30), 12172-12179 doi: 10.1073/pnas.1310291110 Telugu, B P., Ezashi, T., & Roberts, R M (2010) Porcine induced pluripotent stem cells analogous to naive and primed embryonic stem cells of the mouse Int J Dev Biol, 54(11-12), 1703-1711 doi: 10.1387/ijdb.103200bt Telugu, B P., Ezashi, T., Sinha, S., Alexenko, A P., Spate, L., Prather, R S., & Roberts, R M (2011) Leukemia inhibitory factor (LIF)-dependent, pluripotent stem cells established from inner cell mass of porcine embryos J Biol Chem, 286(33), 28948-28953 doi: 10.1074/jbc.M111.229468 Thomson, J A., Itskovitz-Eldor, J., Shapiro, S S., Waknitz, M A., Swiergiel, J J., Marshall, V S., & Jones, J M (1998) Embryonic stem cell lines derived from human blastocysts Science, 282(5391), 1145-1147 Vejlsted, M., Du, Y., Vajta, G., & Maddox-Hyttel, P (2006) Posthatching development of the porcine and bovine embryo defining criteria for expected development Theriogenology, 65(1), in vivo and in 153-165 vitro doi: 10.1016/j.theriogenology.2005.09.021 Warren, L., Manos, P D., Ahfeldt, T., Loh, Y H., Li, H., Lau, F., Rossi, D J (2010) Highly efficient reprogramming to pluripotency and directed differentiation of human cells with synthetic modified mRNA Cell Stem Cell, 7(5), 618-630 doi: 10.1016/j.stem.2010.08.012 West, F D., Terlouw, S L., Kwon, D J., Mumaw, J L., Dhara, S K., Hasneen, K., Stice, S L (2010) Porcine induced 105 pluripotent stem cells produce chimeric offspring Stem Cells Dev, 19(8), 1211-1220 doi: 10.1089/scd.2009.0458 Wianny, F., Perreau, C., & Hochereau de Reviers, M T (1997) Proliferation and differentiation of porcine inner cell mass and epiblast in vitro Biol Reprod, 57(4), 756-764 Wu, Z., Chen, J., Ren, J., Bao, L., Liao, J., Cui, C., Xiao, L (2009) Generation of pig induced pluripotent stem cells with a drug-inducible system J Mol Cell Biol, 1(1), 46-54 doi: 10.1093/jmcb/mjp003 Yamanaka, S (2007) Strategies and new developments in the generation of patient-specific pluripotent stem cells Cell Stem Cell, 1(1), 39-49 doi: 10.1016/j.stem.2007.05.012 Yang, J Y., Mumaw, J L., Liu, Y., Stice, S L., & West, F D (2013) SSEA4-positive pig induced pluripotent stem cells are primed for differentiation into neural cells Cell Transplant, 22(6), 945-959 doi: 10.3727/096368912X657279 Yu, J., Chau, K F., Vodyanik, M A., Jiang, J., & Jiang, Y (2011) Efficient feeder-free episomal reprogramming with small molecules PLoS One, 6(3), e17557 doi: 10.1371/journal.pone.0017557 Yu, J., Vodyanik, M A., Smuga-Otto, K., Antosiewicz-Bourget, J., Frane, J L., Tian, S., Thomson, J A (2007) Induced pluripotent stem cell lines derived from human somatic cells Science, 318(5858), 1917-1920 doi: 10.1126/science.1151526 106

Ngày đăng: 30/09/2015, 10:13

Tài liệu cùng người dùng

Tài liệu liên quan