Figure 6 Real-time relative quantitation analysis of gene expression in CAG-miPSC and teratomas. Statistically significant differences are indicated with asterisks.

Discussion
Here, we investigated whether four porcine reprogramming transcription factors were able to successfully reprogram MEFs to pluripotency. The amino acid sequences for Oct4, Sox2, c-myc, and Klf4 are evolutionary highly conserved, although not fully identical between mouse and pig (88%, 98%, 93%, and 94%, respectively; amino acid sequence alignments are shown on Figure S2). Thus, it was necessary to confirm that the porcine proteins can in fact reprogram somatic cells across species. As expected, the MEFs changed proliferation and morphology and upregulated endogenous OCT4 expression, as demonstrated by the presence of EGFP fluorescence and subsequently confirmed by RT-PCR. The picked colonies proliferated robustly and established cell lines with typical compact miPSC morphology and expression of key pluripotency markers such as AP, SSEA-1, Oct4, Sox2, Nanog, Klf4, Rex1, and Utf1. At the same time, the TetO-promoter-controlled transgenes were silenced, even in the presence of DOX. This result is in contrast with other reports, where transposon-delivered transgenes were not silenced by the reprogrammed cells if the culture medium was continuously supplemented with DOX[12, 17]. A possible explanation for our results is that the porcine transgenes are more efficiently recognized and silenced by the miPSC. Silencing of the reprogramming transcription factors has been described as an essential prerequisite for successful reprogramming[2, 7- 9]. Thus, porcine reprogramming factors may be useful for the production of miPSC, particularly where significant nucleotide sequence differences between the transgenes and the endogenous pluripotency genes are desired.
Another interesting question of this study was whether the mouse cells would balance the expression levels of key endogenous pluripotency genes with the non-silenced porcine transgenes. We obtained cell lines with silenced (TetO-regulated) and non-silenced (CAG-regulated) reprogramming cassettes, which enabled us to address this question by comparing the expression levels of endogenous pluripotency genes between the two groups. Surprisingly, we found no significant differences in the expression levels of endogenous mouse Oct4 and Sox2 between the cells with silenced vs. expressed transgenes. Additionally, the expression levels of these genes did not change even after the transgenes were removed by expression of CRE recombinase. These results suggest that the porcine transgenes did not participate in the regulation of endogenous Oct4 and Sox2 expression after the initial induction. Moreover, none of the cells from the CAG-group showed any signs of differentiation that would be expected from elevated total Oct4 and Sox2 expression levels. A possible explanation for this outcome could be the very low expression levels of the transgenes, as suggested by our real-time PCR analysis. Another possibility is that the specifying signals ensuing from elevated total Oct4 and Sox2 levels had balanced each other, preventing differentiation into any of the germ layers. On the other hand, the expression levels of the endogenous c-myc, Klf4, Rex1, and Utf1 were significantly lower in cell lines with non-silenced transgenes compared with the TetO group. Excision of the reprogramming cassette by CRE-recombinase resulted in significant upregulation of Utf1, but failed to upregulate the rest.
Despite the deviations in the expression of important pluripotency markers, the cell lines with residual expression of the four Yamanaka factors were capable of differentiation into derivatives of the three primary germ layers in vitro and in vivo (by forming fully differentiated teratomas) similar to the TetO- group. When we examined the expression levels of pluripotency and differentiation-related genes in teratomas and iPSC lines, we found that, as expected, the pluripotency markers were down-regulated while the differentiation markers were up-regulated, suggesting normal differentiation process in the CAG-iPSC.
Unlike the miPSC in an alternative state of pluripotency (F-state) described recently[12], our miPSC lines with non-silenced reprogramming factors have compact ES-like morphology and do not depend on the expression of the transgenes for their maintenance; therefore, they do not necessarily fall into an alternative pluripotency state category. Nevertheless, it has been shown that even subtle aberrations in miPSC with residual transgene expression could impair the full developmental competency of miPSC[11, 14]. The lower expression of important pluripotency markers such as Rex1, Utf1, and Klf4 in CAG-miPSC suggests that this may also apply for miPSC produced with non-silenced porcine transcription factors. In future work, the cells will be tested for chimera formation to ascertain whether they are fully competent iPSC.
Conclusions
In conclusion, we successfully established miPSC using four porcine reprogramming transcription factors and examined the effects of the non-silenced transgenes on the expression levels of key endogenous pluripotency markers. The residual expression of the transgenes affected expression levels of important pluripotency genes such as c-myc, Klf4, Rex1, and Utf1. These results suggest that porcine pluripotency transcription factors are suitable for reprogramming of mouse somatic cells to pluripotency, but epigenetic silencing of the xenogeneic transgenes may be necessary for complete acquisition of pluripotency.
Potential Conflicts of Interests
None
Acknowledgements
Dr. Wilfried Kues (Institute of Farm Animal Genetics, Friedrich-Loeffler-Institute, Mariensee, Neustadt, Germany) for the injection of the immunodefficient mice. Dr. Hans Schoeler (Max-Planck-Institute for Molecular Biomedicine, Muenster, Germany) for providing the OG2 reporter mice. Dr. Zoltan Ivics (Paul-Ehrlich Institute, Langen, Germany) for providing the SB transposon backbone and the SB transposase expression plasmid. Dr. Poul Hyttel (University of Copenhagen, Copenhagen, Denmark) for allowing us to use and modify the transposon plasmid which was generated by Dr. Stoyan Petkov in his laboratory.

Lot 777 Lefroy Street Narrogin

Sponsor / Grants
German Research Foundation (Deutsche Forschungsgemeinschaft), Grant Ni 256/ 32-1.
Additional Information
Supplementary Information accompanies this article.
To view the Supplementary figures and table click here
To download the Supplementary Video click here
Abbreviations used in the article
Afp: Alpha-fetoprotein
AP: Alkaline Phosphatase
CAG: Synthetic Promoter containing the (C) cytomegalovirus early enhancer element, (A) the promoter, the first exon and the first intron of chicken beta-actin gene, and (G) the splice acceptor of the rabbit beta-globin gene
DMEM: Dulbecco’s Modified Eagle’s Medium
DOX: Doxycycline
EGFP: Enhanced Green Fluorescent Protein
FBS: Fetal Bovine Serum
iPSC: Induced Pluripotent Stem Cells
mESC: Mouse Embryonic Stem Cells
Klf4: Kruppel-Like Factor 4
MEFs: Mouse Embryonic Fibroblasts
MHC: Myosin Heavy Chain
Oct4: POU Domain, Class 5, Transcription Factor 1
Pax6: Paired Box Protein 6
PBS: Phosphate-Buffered Saline
PCR: Polymerase Chain Reaction
RT: Reverse Transcription
SB: Sleeping Beauty Transposon/Transposase
Sox2: SRY (sex determining region Y)-box 2
Sox17: SRY (sex determining region Y)-box 17
c-myc: V-Myc Avian Myelocytomatosis Viral Oncogene Homolog
UTF1: Undifferentiated Embryonic Cell Transcription Factor 1

Lot Airlines 777

References

Lot Boeing 777

  1. Slack JM. Origin of stem cells in organogenesis. Science. 2008;322(5907):1498-501.
  2. Jones DL, Wagers AJ. No place like home: anatomy and function of the stem cell niche. Nat Rev Mol Cell Biol. 2008;9(1):11-21.
  3. Spradling A, Drummond-Barbosa D, Kai T. Stem cells find their niche. Nature.2001;414(6859):98-104.
  4. Sell S. Stem cell origin of cancer and differentiation therapy. Crit Rev Oncol Hematol. 2004 ;51(1):1-28.
  5. Scadden DT. The stem-cell niche as an entity of action. Nature. 2006;441(7097):1075-9.
  6. Moore KA, Lemischka IR. Stem cells and their niches. Science. 2006;311(5769):1880-5.
  7. Tropepe V, Turksen K. The ontogeny of somatic stem cells. Stem Cell Rev. 2012;8(2):548-50.
  8. Cheung TH, Rando TA. Molecular regulation of stem cell quiescence. Nat Rev Mol Cell Biol. 2013;14(6):329-40.
  9. Clevers H, Loh KM, Nusse R. Stem cell signaling. An integral program for tissue renewal and regeneration: Wnt signaling and stem cell control. Science.2014;346(6205):1248012.
  10. Tang DG. Understanding cancer stem cell heterogeneity and plasticity. Cell Res. 2012;22(3):457-72.
  11. Li L, Clevers H. Coexistence of quiescent and active adult stem cells in mammals. Science. 2010;327(5965):542-5.
  12. Takeda N, Jain R, LeBoeuf MR, Wang Q, Lu MM, Epstein JA. Interconversion between intestinal stem cell populations in distinct niches. Science. 2011;334(6061):1420-4.
  13. Blau HM, Brazelton TR, Weimann JM. The evolving concept of a stem cell: entity or function? Cell. 2001;105(7):829-41.
  14. Clevers H. STEM CELLS. What is an adult stem cell? Science. 2015;350(6266):1319-20.
  15. Orkin SH, Zon LI. Hematopoiesis: an evolving paradigm for stem cell biology.Cell. 2008;132(4):631-44.
  16. Phillips RL, Ernst RE, Brunk B, Ivanova N, Mahan MA, Deanehan JK, Moore KA, Overton GC, Lemischka IR. The genetic program of hematopoietic stem cells. Science. 2000;288(5471):1635-40.
  17. Ding L, Morrison SJ. Haematopoietic stem cells and early lymphoid progenitors occupy distinct bone marrow niches. Nature. 2013;495(7440):231-5.
  18. Yokota T, Oritani K, Butz S, Kokame K, Kincade PW, Miyata T, Vestweber D, Kanakura Y. The endothelial antigen ESAM marks primitive hematopoietic progenitors throughout life in mice. Blood. 2009;113(13):2914-23.
  19. Morrison SJ, Spradling AC. Stem cells and niches: mechanisms that promote stem cell maintenance throughout life. Cell. 2008;132(4):598-611.
  20. Seita J, Weissman IL. Hematopoietic stem cell: self-renewal versus differentiation. Wiley Interdiscip Rev Syst Biol Med. 2010;2(6):640-53.
  21. Rieger MA, Hoppe PS, Smejkal BM, Eitelhuber AC, Schroeder T. Hematopoietic cytokines can instruct lineage choice. Science. 2009;325(5937):217-8.
  22. Ding L, Saunders TL, Enikolopov G, Morrison SJ. Endothelial and perivascular cells maintain haematopoietic stem cells. Nature. 2012;481(7382):457-62.
  23. Tweedell KS. The urodele limb regeneration blastema: the cell potential. ScientificWorldJournal. 2010;10:954-71.
  24. Cabezas-Wallscheid N, Trumpp A. STEM CELLS. Potency finds its niches. Science. 2016;351(6269):126-7.
  25. Khan JA, Mendelson A, Kunisaki Y, Birbrair A, Kou Y, Arnal-Estapé A, Pinho S, Ciero P, Nakahara F, Ma'ayan A, Bergman A, Merad M, Frenette PS. Fetal liver hematopoietic stem cell niches associate with portal vessels. Science. 2016;351(6269):176-80.
  26. Notta F, Zandi S, Takayama N, Dobson S, Gan OI, Wilson G, Kaufmann KB, McLeod J, Laurenti E, Dunant CF, McPherson JD, Stein LD, Dror Y, Dick JE. Distinct routes of lineage development reshape the human blood hierarchy across ontogeny. Science. 2016 Jan 8;351(6269):aab2116.
  27. Paul F, Arkin Y, Giladi A, Jaitin DA, Kenigsberg E, Keren-Shaul H, Winter D, Lara-Astiaso D, Gury M, Weiner A, David E, Cohen N, Lauridsen FK, Haas S, Schlitzer A, Mildner A, Ginhoux F, Jung S, Trumpp A, Porse BT, Tanay A, Amit I. Transcriptional Heterogeneity and Lineage Commitment in Myeloid Progenitors. Cell. 2015 ;163(7):1663-77.
  28. Gage FH. Mammalian neural stem cells. Science. 2000;287(5457):1433-8.
  29. Doetsch F, Caillé I, Lim DA, García-Verdugo JM, Alvarez-Buylla A.Subventricular zone astrocytes are neural stem cells in the adult mammalian brain. Cell. 1999;97(6):703-16.
  30. Götz M, Huttner WB. The cell biology of neurogenesis. Nat Rev Mol Cell Biol. 2005;6(10):777-88.
  31. Anthony TE, Klein C, Fishell G, Heintz N. Radial glia serve as neuronal progenitors in all regions of the central nervous system. Neuron. 2004;41(6):881-90.
  32. Alvarez-Buylla A, García-Verdugo JM, Tramontin AD. A unified hypothesis on the lineage of neural stem cells. Nat Rev Neurosci. 2001;2(4):287-93.
  33. Ihrie RA, Alvarez-Buylla A. Cells in the astroglial lineage are neural stem cells. Cell Tissue Res. 2008;331(1):179-91.
  34. Kriegstein A, Alvarez-Buylla A. The glial nature of embryonic and adult neural stem cells. Annu Rev Neurosci. 2009;32:149-84.
  35. Grabel L. Developmental origin of neural stem cells: the glial cell that could. Stem Cell Rev. 2012;8(2):577-85.
  36. Lin R, Iacovitti L. Classic and novel stem cell niches in brain homeostasis and repair. Brain Res. 2015;1628(Pt B):327-42.
  37. Ottone C, Krusche B, Whitby A, Clements M, Quadrato G, Pitulescu ME, Adams RH, Parrinello S. Direct cell-cell contact with the vascular niche maintains quiescent neural stem cells. Nat Cell Biol. 2014;16(11):1045-56.
  38. Kawaguchi D, Furutachi S, Kawai H, Hozumi K, Gotoh Y. Dll1 maintains quiescence of adult neural stem cells and segregates asymmetrically during mitosis. Nat Commun. 2013;4:1880.
  39. 39. Jones KM, Sarić N, Russell JP, Andoniadou CL, Scambler PJ, Basson MA. CHD7 maintains neural stem cell quiescence and prevents premature stem cell depletion in the adult hippocampus. Stem Cells. 2015;33(1):196-210.
  40. Silva-Vargas V, Crouch EE, Doetsch F. Adult neural stem cells and their niche: a dynamic duo during homeostasis, regeneration, and aging. Curr Opin Neurobiol. 2013;23(6):935-42.
  41. Riquelme PA, Drapeau E, Doetsch F. Brain micro-ecologies: neural stem cell niches in the adult mammalian brain. Philos Trans R Soc Lond B Biol Sci. 2008 ;363(1489):123-37.
  42. Fuentealba LC, Obernier K, Alvarez-Buylla A. Adult neural stem cells bridge their niche. Cell Stem Cell. 2012;10(6):698-708.
  43. Tong CK, Chen J, Cebrián-Silla A, Mirzadeh Z, Obernier K, Guinto CD, Tecott LH, García-Verdugo JM, Kriegstein A, Alvarez-Buylla A. Axonal control of the adult neural stem cell niche. Cell Stem Cell. 2014;14(4):500-11.
  44. Dupin E, Real C, Glavieux-Pardanaud C, Vaigot P, Le Douarin NM. Reversal of developmental restrictions in neural crest lineages: transition from Schwann cells to glial-melanocytic precursors in vitro. Proc Natl Acad Sci U S A. 2003;100(9):5229-33.
  45. Jessen KR, Mirsky R. The origin and development of glial cells in peripheral nerves. Nat Rev Neurosci. 2005;6(9):671-82.
  46. Takahashi Y, Sipp D, Enomoto H. Tissue interactions in neural crest cell development and disease. Science. 2013;341(6148):860-3.
  47. Masaki T, Qu J, Cholewa-Waclaw J, Burr K, Raaum R, Rambukkana A. Reprogramming adult Schwann cells to stem cell-like cells by leprosy bacilli promotes dissemination of infection. Cell. 2013;152(1-2):51-67.
  48. Xin L, Ide H, Kim Y, Dubey P, Witte ON. In vivo regeneration of murine prostate from dissociated cellpopulations of postnatal epithelia and urogenital sinus mesenchyme . Proc Natl Acad Sci U S A. 2003; 100: (Suppl 1) 11896-903.
  49. Xin L, Lukacs RU, Lawson DA, Cheng D, Witte ON. Self-renewal and multilineage differentiation in vitro from murine prostate stem cells. Stem Cells. 2007;25(11):2760-9.
  50. Burger PE, Xiong X, Coetzee S, Salm SN, Moscatelli D, Goto K, Wilson EL. Sca-1 expression identifies stem cells in the proximal region of prostatic ducts with high capacity to reconstitute prostatic tissue. Proc Natl Acad Sci U S A. 2005;102(20):7180-5.
  51. Leong KG, Wang BE, Johnson L, Gao WQ. Generation of a prostate from a single adult stem cell. Nature. 2008;456(7223):804-8.
  52. Lawson DA, Xin L, Lukacs RU, Cheng D, Witte ON. Isolation and functional characterization of murine prostate stem cells. Proc Natl Acad Sci U S A. 2007 ;104(1):181-6.
  53. Richardson GD, Robson CN, Lang SH, Neal DE, Maitland NJ, Collins AT. CD133, a novel marker for human prostatic epithelial stem cells. J Cell Sci. 2004 ;117(Pt 16):3539-45.
  54. Garraway IP, Sun W, Tran CP, Perner S, Zhang B, Goldstein AS, Hahm SA, Haider M, Head CS, Reiter RE, Rubin MA, Witte ON. Human prostate sphere-forming cells represent a subset of basal epithelial cells capable of glandular regeneration in vivo. Prostate. 2010;70(5):491-501.
  55. Goldstein AS, Stoyanova T, Witte ON. Primitive origins of prostate cancer: in vivo evidence for prostate-regenerating cells and prostate cancer-initiating cells. Mol Oncol. 2010;4(5):385-96.
  56. Ousset M, Van Keymeulen A, Bouvencourt G, Sharma N, Achouri Y, Simons BD, Blanpain C. Multipotent and unipotent progenitors contribute to prostate postnatal development. Nat Cell Biol. 2012;14(11):1131-8.
  57. Shahi P, Seethammagari MR, Valdez JM, Xin L, Spencer DM. Wnt and Notch pathways have interrelated opposing roles on prostate progenitor cell proliferation and differentiation. Stem Cells. 2011;29(4):678-88.
  58. Luo W, Rodriguez M, Valdez JM, Zhu X, Tan K, Li D, Siwko S, Xin L, Liu M. Lgr4 is a key regulator of prostate development and prostate stem cell differentiation. Stem Cells. 2013;31(11):2492-505.
  59. Strand DW, Goldstein AS. The many ways to make a luminal cell and a prostate cancer cell. Endocr Relat Cancer. 2015;22(6):T187-97.
  60. Faraldo MM, Taddei-De La Hosseraye I, Teulière J, Deugnier MA, Moumen M, Thiery JP, Glukhova MA. [Mammary gland development: Role of basal myoepithelial cells]. J Soc Biol. 2006;200(2):193-8.
  61. Petersen OW, Polyak K. Stem cells in the human breast. Cold Spring Harb Perspect Biol. 2010;2(5):a003160.
  62. Shackleton M, Vaillant F, Simpson KJ, Stingl J, Smyth GK, Asselin-Labat ML, Wu L, Lindeman GJ, Visvader JE. Generation of a functional mammary gland from a single stem cell. Nature. 2006;439(7072):84-8.
  63. Wang D, Cai C, Dong X, Yu QC, Zhang XO, Yang L, Zeng YA. Identification of multipotent mammary stem cells by protein C receptor expression. Nature. 2015 ;517(7532):81-4.
  64. Van Keymeulen A, Rocha AS, Ousset M, Beck B, Bouvencourt G, Rock J, Sharma N, Dekoninck S, Blanpain C. Distinct stem cells contribute to mammary gland development and maintenance. Nature. 2011;479(7372):189-93.
  65. Hassiotou F, Beltran A, Chetwynd E, Stuebe AM, Twigger AJ, Metzger P, Trengove N, Lai CT, Filgueira L, Blancafort P, Hartmann PE. Breastmilk is a novel source of stem cells with multilineage differentiation potential. Stem Cells. 2012;30(10):2164-74.
  66. Rios AC, Fu NY, Lindeman GJ, Visvader JE. In situ identification of bipotent stem cells in the mammary gland. Nature. 2014;506(7488):322-7.
  67. LaBarge MA, Nelson CM, Villadsen R, Fridriksdottir A, Ruth JR, Stampfer MR, Petersen OW, Bissell MJ. Human mammary progenitor cell fate decisions are products of interactions with combinatorial microenvironments. Integr Biol (Camb). 2009;1(1):70-9.
  68. LaBarge MA. On stem cells in the human breast. Cold Spring Harb Perspect Biol. 2012;4(5). pii: a013441.
  69. Mishra L, Shetty K, Tang Y, Stuart A, Byers SW. The role of TGF-beta and Wnt signaling in gastrointestinal stem cells and cancer. Oncogene. 2005;24(37):5775-89.
  70. Medema JP, Vermeulen L. Microenvironmental regulation of stem cells in intestinal homeostasis and cancer. Nature. 2011;474(7351):318-26.
  71. Cheng H, Leblond CP. Origin, differentiation and renewal of the four main epithelial cell types in the mouse small intestine. V. Unitarian Theory of the origin of the four epithelial cell types. Am J Anat. 1974;141(4):537-61.
  72. Clevers H. The intestinal crypt, a prototype stem cell compartment. Cell. 2013;154(2):274-84.
  73. Clevers H, Batlle E. SnapShot: the intestinal crypt. Cell. 2013;152(5):1198-1198.e2.
  74. Clevers H. Stem Cells: A unifying theory for the crypt. Nature. 2013;495(7439):53-4.
  75. Barker N, van de Wetering M, Clevers H. The intestinal stem cell. Genes Dev. 2008;22(14):1856-64.
  76. Potten CS. Extreme sensitivity of some intestinal crypt cells to X and gamma irradiation. Nature. 1977;269(5628):518-21.
  77. Li VS, Clevers H. In vitro expansion and transplantation of intestinal crypt stem cells. Gastroenterology. 2012;143(1):30-4.
  78. Sangiorgi E, Capecchi MR. Bmi1 is expressed in vivo in intestinal stem cells. Nat Genet. 2008;40(7):915-20.
  79. Buczacki SJ, Zecchini HI, Nicholson AM, Russell R, Vermeulen L, Kemp R, Winton DJ. Intestinal label-retaining cells are secretory precursors expressing Lgr5. Nature. 2013;495(7439):65-9.
  80. van Es JH, Sato T, van de Wetering M, Lyubimova A, Nee AN, Gregorieff A, Sasaki N, Zeinstra L, van den Born M, Korving J, Martens AC, Barker N, van Oudenaarden A, Clevers H. Dll1+ secretory progenitor cells revert to stem cells upon crypt damage. Nat Cell Biol. 2012;14(10):1099-104.
  81. Barker N. Adult intestinal stem cells: critical drivers of epithelial homeostasis and regeneration. Nat Rev Mol Cell Biol. 2014;15(1):19-33.
  82. Carulli AJ, Samuelson LC, Schnell S. Unraveling intestinal stem cell behavior with models of crypt dynamics. Integr Biol (Camb). 2014;6(3):243-57.
  83. Yan KS, Chia LA, Li X, Ootani A, Su J, Lee JY, Su N, Luo Y, Heilshorn SC, Amieva MR, Sangiorgi E, Capecchi MR, Kuo CJ. The intestinal stem cell markers Bmi1 and Lgr5 identify two functionally distinct populations. Proc Natl Acad Sci U S A. 2012;109(2):466-71.
  84. Visvader JE, Clevers H. Tissue-specific designs of stem cell hierarchies. Nat Cell Biol. 2016;18(4):349-55.
  85. Booth BW, Mack DL, Androutsellis-Theotokis A, McKay RD, Boulanger CA, Smith GH. The mammary microenvironment alters the differentiation repertoire of neural stem cells. Proc Natl Acad Sci U S A. 2008;105(39):14891-6.
  86. Boulanger CA, Mack DL, Booth BW, Smith GH. Interaction with the mammary microenvironment redirects spermatogenic cell fate in vivo. Proc Natl Acad Sci U S A. 2007;104(10):3871-6.

Wordalot 777

Corresponding Author

Lot 777 Sierra Verde Ranch

  1. Stoyan Petkov, German Primate Center, Kellnerweg 4,37077 Göttingen, Germany; Email: [email protected]
  2. Heiner Niemann, Institute for Farm Animal Genetics, Friedrich-Loeffler-Institute, Hoeltystrasse 10, 31535 Neustadt am Ruebenberge, Germany; Email: [email protected]