<<

Recent Progress of Transgenic Models for Biomedicine and Pharmaceutical Research

Authors: Wiebke Garrels, Heiner Niemann. Corresponding author: Wilfried A. Kues

Abstract Whitelaw, 2003; Niemann and Kues, 2007; Robl et al., 2007). A The first transgenic were produced by the microinjection of bottleneck for porcine transgenesis is the lack of authentic pluri- foreign DNA into zygotic pronuclei in 1985. Since then, the me- potent stem cells that are suitable for blastocyst complementation thodological repertoire for porcine transgenesis was expanded to experiments (Brevini et al., 2008; Kues et al., 2010a). The semi- somatic nuclear transfer, lentiviral transgenesis and, recently, nal development of induced pluripotent stem cells (iPS) in mice cytoplasmic plasmid injection. The major impact of transgenic pigs and (Takahashi and Yamanaka, 2006) provides a new and minipigs took place in the fields of humanised pig models approach to this end. The results of the first attempts to generate and biomedical disease models, whereas agricultural applications porcine iPS cells were published recently (Esteban et al., 2009; did not find broad acceptance. The recent release of the porcine Wu et al., 2009; Ezashi et al., 2009), yet the potential of current whole genome sequence and parallel developments of highly spe- porcine iPS cells to contribute to chimera formation seems to be cific enzymes and RNAs now make it possible to perform precise limited (West et al., 2010). genetic modifications and fully exploit the advantages of this large This paper briefly discusses the current progress of transgenic animal model. We anticipate that genetically modified pigs and pig models for biomedical research. Comprehensive overviews minipigs will increasingly complement the commonly used small- about transgenic pigs and are available elsewhere (Clark animal models in biomedical research, since several aspects of and Whitelaw, 2003; Robl et al., 2007; Kues and Niemann, 2011; disease progression, physiology, metabolism and aging cannot Whyte and Prather, 2011). properly be mirrored in small-animal models. Basic and biomedical applications Introduction of transgenic pigs The production of transgenic pigs is labour-intensive and cost- In the last few years, an expanded methodological repertoire for intensive and depends on advanced techniques in molecular biolo- porcine gene transfer has been developed (Table 1), resulting in an gy and the micromanipulation of gametes and zygotes. At present, increasing number of transgenic approaches (Whyte and Prather, progress in reproductive techniques and gene-transfer methods 2011). At least 90% of genetically modified pigs are generated has allowed targeted modifications of the porcine genome (glos- for biomedical studies (Fig. 1A). Sequencing and annotation of sary box), albeit the overall success rates are still low (Clark and the porcine genome are important milestones for accelerating the

Fig.1. Increasing scientific interest in transgenic pig models A) Scientific interest in porcine transgenesis. Depicted are the numbers of total citations per year, as extracted from Thomson Reuters ISI Web of Knowledge for topic search terms “transgenic” and “pig model”. B) Transgenic boar exhibiting ubiquitous expression of the Venus fluorophor gene (Garrels et al., 2011). The boar is shown under specific excitation condi- tions of Venus, in front of the boar an autofluorescent toy is visible. Almost all somatic and germ cells are fluorescent.

Key words: Domestic animals, disease model, humanised, genome, large animal model Newsletter 36 Autumn 2011 17 ➤ Recent Progress of Transgenic Pig Models for Biomedicine and Pharmaceutical Research

Table 1. Progress of technologies for transgenesis in pigs and minipigs

Development Strategy Reference

First transgenic pigs PNI Hammer et al., 1985

Somatic cloning of transgenic pigs SCNT using transgenic donor cells Park et al., 2001

Sperm-mediated gene transfer SMGT Lavitrano et al., 2002; Chang et al., 2002

Knock-out in pigs Homologous recombination in somatic Dai et al., 2002; Lai et al., 2002 cells and SCNT Homozygous gene knockout Homozygous knockout Phelps et al., 2003

Lentiviral transgenesis Perivitelline injection of lentiviruses Hofmann et al., 2003; Whitelaw et al., 2004

SMGT / ICSI combination SMGT and ICSI Kurome et al, 2006

Conditional transgenesis PNI Kues et al., 2006

Episomal transgenesis SMGT and episomal plasmid Manzini et al., 2006; Giovannoni et al., 2010

Gene knock-down Knock-down of PERV genes with siRNA and Dieckhoff et al., 2008; Ramsoondar et al., SCNT 2009 Transposon transgenesis Sleeping Beauty transposition in zygotic genome Garrels et al., 2010; Kues et al., 2010b by CPI Transposon transgenesis Sleeping Beauty transposition in somatic cells Jacobsen et al., 2011; Carlson et al. 2011 and SCNT Targeted gene knockout Zinc finger nuclease-catalysed gene deletion in Whyte et al., 2011; Yang et al., 2011; primary cells and SCNT Hauschild et al., 2011 Targeted integration Recombination-mediated cassette exchange in Garrels et al., 2011 primary cells and SCNT

generation of transgenic models, even if the porcine genome with porcine alpha-galactosyltransferase knockout organs ( assembly still has gaps (annotated porcine genome data can be or heart) transplanted to (Kuwaki et al., 2005; Yamada found at: www.ensembl.org and www.pubmed.org). Since pig et al., 2005). and minipig physiology, anatomy, pathology, genome organisa- Extensive research has been conducted to reduce the risk of tion, body weight and life span are more similar to humans than porcine (PERV) transmission to are rodents, the domesticated pig represents a more appropriate patients (Switzer et al., 2001; Irgang et al., 2003). RNA interfe- biomedical model (Table 2). rence (RNAi) is a promising method for knocking down the PERV For certain biomedical therapies, such as xenotransplanta- expression. RNAi is based on small RNAs, either small interfering tion (transplantation of organs from one to another (e.g. RNA (siRNA) or short hairpin RNAs (shRNA). In the cytoplasm, porcine-to-human)), transgenic pigs are the only reasonable spe- small RNA molecules are incorporated into an RNA-induced cies (Niemann and Kues, 2003). seems to silencing complex (RISC) and targets binding to a complementary be one option for closing the widening gap between demand and transcript sequence, resulting in mRNA degradation (Plasterk, availability of appropriate human organs (Yang and Sykes, 2007). 2002; Dallas and Vlassow, 2006). The efficacy of RNAi for redu- The prerequisites for potential porcine–human xenotransplanta- cing PERV expression has been demonstrated in cloned piglets tion are: (i) overcoming immunological hurdles; (ii) preventing the (Dieckhoff et al., 2008; Ramsoondar et al., 2009). transmission of porcine pathogens to human recipients; and (iii) the For several approaches, a conditional gene expression is desi- compatibility of porcine organs with human physiology. rable over a constitutive transgenic expression. Initial animal mod- The suppression of hyperacute rejection of porcine xenografts els carrying the first generation of conditional promoter elements has been achieved by transgenic expression of human regula- suffered from high basal-expression levels and pleiotropic effects tors of complement activity (RCA) (Tucker et al., 2002) and a (Miller et al., 1989). Recent expression systems responsive to gene knockout of the porcine alpha, 1,3-galactosyltransferase exogenous tetracycline resulted in more tightly controlled expres- gene (Dai et al., 2002; Lai et al., 2002; Phelps et al., 2003). sion. In pigs, a tetracycline-controlled transgenic expression was Maximal survival rates of up to 3–6 months have been achieved achieved with a bicistronic expression cassette (Kues et al., 2006)

18 Newsletter 36 Autumn 2011 ➤

that was designed to give ubiquitous expression of human RCAs. tion. Recently, the first immunodeficient pigs were cloned by SCNT Crossbreeding of lines with two cassettes was necessary to over- (Mendicino et al., 2010; Ramsoondar et al., 2011), promising to come epigenetic silencing and to achieve tetracycline-sensitive serve as large-animal models for cell transplantation experiments. RCA expression. Conventional gain-of-function transgenesis is based on random Transgenic pigs have been shown to mimic human diseases integration of the transgene at sites of spontaneous double-strand such as atherosclerosis, non-insulin-dependent , cystic breaks of chromosomal DNA. The frequency of DNA double- , , ophthalmological and neurodegenerative disorders strand breaks at a defined locus can be considerably increased by (Kues and Niemann, 2004; Kragh et al., 2010; Rogers et al., introducing specifically designed endonuclease enzymes (Urnov 2008; Yang et al., 2010; Luo et al., 2011). An important exam- et al., 2005; Arnould et al., 2007). The artificial endonucleases ple is the minipig cystic fibrosis model, which develops disease are based on the DNA recognition sites of zinc finger transcription phenotypes that are highly similar to human patients (Rogers et factors, meganuclei or transcription factor like elements (TALE), al., 2008), whereas transgenic mouse models failed to exhibit and they can be designed to bind highly specifically to a single, lung, pancreatic and intestinal obstructions. Huntington’s disease predetermined sequence in the genome. Double-strand break- is a neurodegenerative disorder characterised by the expression repair pathways often create small deletions and, thus, designed of mutated huntingtin with expanded polyglutamine tracts. The endonucleases allow efficient gene knockouts. The proof-of-prin- misfolded protein accumulates in neurons and is suspected of trig- ciple to generate knockout pigs by synthetic zinc finger nucleases gering . Whereas genetic mouse models often failed to has been demonstrated by the inactivation of enhanced green replicate overt neurodegeneration and apoptosis, a minipig model fluorescent protein (EGFP), peroxisome proliferator-activated expressing the N-terminal huntingtin with a polyglutamine tract receptor (PPAR gamma) and alpha-galactosyltransferase (Whyte seems to do so (Yang et al., 2010). et al., 2011; Yang et al., 2011; Hauschild et al., 2011) in primary Truncation mutations in the elongation of a very long-chain fatty- somatic cells and the subsequent use of knockout cells for SCNT, acids-4 (ELOVL4) gene cause macular dystrophy. Photoreceptor respectively. Thus current lack of authentic porcine ES cells can be topography in the pig retina is more similar to that in humans as it circumvented for the purpose of generating knockout pigs. includes cone-rich, macula-like area centralis, whereas mice lack DNA-based transposons are mobile genetic elements that a macular. Transgenic pigs expressing disease-causing ELOVL4 move in the genome via a “cut-and-paste” mechanism. Most DNA mutations were generated by PNI and SCNT (Sommer et al., transposons are simply organised: they encode a transposase 2011). A detailed analysis showed photoreceptor loss, disorga- protein flanked by inverted terminal repeats (ITRs), which carry nised inner and outer segments, and diminished electroretinogra- transposase binding sites, and it has been possible to separate phy responses, suggesting that the transgenic pigs mirror macular the transposase coding sequence from ITR sequences. Any DNA degeneration and provide a unique model for therapeutic interven- flanked by ITRs will be recognised by the transposase and will

Table 2. Selected pig and minipig models for biomedicine and pharmaceutical research

Model Comment Reference Xenotransplantation knockout of alpha-galactosyltransferase Lai et al., 2002; Dai et al., 2002 Xenotransplantation expression of tumour factor ligand Klose et al., 2005 Xenotransplantation expression of human leukocyte Weiss et al., 2009 Xenotransplantation PERV-knock down Dieckhoff et al., 2008 Xenotransplantation expression of human thrombomodulin Petersen et al., 2009 Xenotransplantation expression of human A20 (anti-apoptotic gene) Oropeza et al., 2009 Cystic fibrosis pig knockout of cystic fibrosis transmembrane conductance receptor Rogers et al., 2008 Diabetes model expression of mutated hepatocyte nuclear factor-1 Umeyama et al., 2009 Diabetes model expression of mutated insulin 2 Renner et al., 2010 Immunodeficient pig knockout of light chain Ramsoondar et al., 2010 Immunodeficient pig knockout of joining gene cluster Mendicino et al., 2010 Huntington model expression of mutated huntingtin with polyglutamine tract Yang et al., 2010 Alzheimer model expression of mutated human amyloid precursor protein Kragh et al., 2010 Breast cancer knockout of BRCA1 gene Luo et al., 2011 Macular degeneration introduced deletion in ELOVL4 gene Sommer et al., 2011

Newsletter 36 Autumn 2011 19 ➤ Recent Progress of Transgenic Pig Models for Biomedicine and Pharmaceutical Research

Fig.2. Applications of transposon transgenesis Depicted is one integration site of a Venus transposon on chromosome X (red arrow). By means of targeted cassette exchange (via the Cre/loxP system), the Venus reporter gene can be replaced by a gene of choice (I), thus introducing a transgene in a pretested locus (Garrels et al., 2011) suitable for expression, and avoiding integration into heterochromatic regions or inser- tional mutagenesis. Alternatively, by supplying the SB transposase in trans, a remobilisation (II) of the transposon can be induced. The annotated pig genome sequence was extracted from www.ensembl.org.

become enzymatically integrated into nuclear DNA. In a two- expression. Importantly, transposon-tagged loci can be read- component system, the transposon is integrated solely by the dressed by recombination-mediated cassette exchange (RMCE) in trans-supplementation activity of transposase. The first transposon cell culture. Via SCNT, the RMCE cells can be used to generate sufficiently active for use in vertebrates was the Sleeping Beauty vital piglets carrying a targeted integration into a “safe harbour” (SB) transposon (Ivics et al., 1997; Clark et al., 2007). Many locus (Garrels et al., 2011). drawbacks of classical transgenic methods can be overcome Since integrated transposons can be remobilised in the pre- by transposition-catalysed gene delivery, which increases the sence of a transposase enzyme, these animals can provide the efficiency of chromosomal integration and facilitates single-copy basis for performing whole genome mutagenesis screens in the (monomeric) insertion events. An additional advantage of trans- pig. For the SB transposon, the phenomenon of local hopping poson-catalysed transgenesis is that the integration of monomeric after mobilisation has been described. The majority of secondary transgene units is directed to accessible euchromatic regions. integrations take place at a distance of up to 5 megabases from Transposon transgenic pigs have been generated (Kues et al., the original integration. Figure 2 depicts one integration site on 2010b; Garrels et al., 2011) by CPI (Iqbal et al., 2009), as well as the gene-rich X chromosome. The neighbouring porcine genes by SCNT (Jakobsen et al., 2010; Carlson, 2011; Garrels, 2011). are the von Hippel-Lindau binding gene (VBP1) and a novel gene, Ubiquitous expression of a fluorescent Venus protein, a derivative both about 10,000 base pairs away from the integration site. After of the commonly used EGFP, was found in somatic and germ mobilisation, the integration site can be screened for integration cells (differentiated spermatozoa) in own experiments (Fig. 1B, events in neighbouring genes, such as the VBP1. The VBP1 gene Garrels et al., 2011) for all integrations sites, strongly supporting is of potential interest as an animal model, and the gene product is the hypothesis that transposase preferentially integrates DNA into assumed to form a complex with the von Hippel-Lindau tumor sup- euchromatic regions. The robust transgenic expression of Venus is pressor (VHL). The von Hippel-Lindau syndrome is a dominantly strictly copy-number dependent and facilitates cell-tracking experi- inherited cancer syndrome predisposing carriers to several malig- ments in cell-therapy approaches. The identification of integrations nant and benign tumours. Thus, transposon transgenic pigs can be sites revealed that most transposon integration sites were found in employed for performing unbiased and biased mutagenic events. intergenic regions of the porcine genome (Fig. 2). This approach It is anticipated that mutagenic screens with more advanced con- made it possible to identify loci, which are suitable for transgenic structs will be applied in the near future.

20 Newsletter 36 Autumn 2011 ➤

Conclusions transposon system. Transgenic Res. 20, Jan. 9 (epub ahead of Methodological improvements for gene transfer into the pig print). genome and a rapidly increasing list of biomedical pig models have Clark J, Whitelaw B (2003) A future for transgenic livestock. Nat been developed in recent years. Together with more accurate Rev Genet 4:825-833 genome data and highly specific designed enzymes and RNAs, Clark, K.J:, Carlson, D.F., Fahrenkrug, S.C. 2007. Pigs taking precise genetic modifications have become feasible. It is anticipa- wings with transposons and recombinases. Genome Biol. 8, Suppl ted that authentic pluripotent cells of the pig will be generated in 1: S13. the near future. Thus, porcine transgenesis will become a routine Dai Y, Vaught TD, Boone J, Chen SH, Phelps CJ, Ball S, tool for generating relevant humanised porcine models. The most Monahan JA, Jobst PM, McCreath KJ, Lamborn AE, Cowell- obvious application of transgenic pigs will be as disease models Lucero JL, Wells KD, Colman A, Polejaeva IA, Ayares DL (2002) and biomedical therapies, which are not well-reflected in small Targeted disruption of the a1,3-galactosyltransferase gene in rodent models. The progress expected in porcine transgenesis cloned pigs. Nat Biotechnol 20: 251-255 (increased success rates and decreasing costs), however, will Dallas A, Vlassow A (2006) RNAi: A novel antisense technology make the pig an attractive complementary model for advanced and its therapeutic potential. Med Sci Monit 12 RA67-74 approaches in biomedical research. Dieckhoff B, Petersen B, Kues WA, Kurth R, Niemann H, Denner J (2008) Knockdown of porcine endogenous retrovirus Acknowledgments (PERV) expression by PERV-specific shRNA in transgenic pigs. The expert technical support of Ms S. Holler, Ms Barg-Kues, Xenotransplantation 15: 36-45 Ms Herrmann and Ms Ziegler, and the financial support of the Esteban, M.A., J. Xu, J. Yang, M. Peng, D. Qin, W. Li, Z. Jiang, Deutsche Forschungsgemeinschaft (DFG) are gratefully acknow- J. Chen, K. Deng, M. Zhong, J. Cai, L. Lai, and D. Pei. 2009. ledged. Generation of induced pluripotent stem cell lines from Tibetan miniature pig. J Biol Chem. 284:17634-17640. Conflicts of interest Ezashi, T., B.P.V.L. Telugu, A.P. Alexenko, S. Sachdev, S. The authors declare no conflicts of interest. Sinha, and R.M. Roberts. 2009. Derivation of induced pluri- potent stem cells from pig somatic cells. Proceedings of the Wiebke Garrels, Heiner Niemann National Academy of Sciences of the United States of America. Friedrich-Loeffler-Institute 106:10993-10998. Mariensee, DE-31535 Neustadt, Germany Garrels, W., Mates, L., Holler,S., Niemann, H., Izsvak, Z., Ivics, Z., Kues, W.A: 2010. Generation of transgenic pigs by the Wilfried A. Kues Sleeping Beauty transposition in zygotes. Reprod. Dom. Anim. 45, Friedrich-Loeffler-Institute 65 (abstract). Institute of Farm Animal Genetics Garrels, W., Mates, L., Holler, S., Dalda, A., Taylor, U., Mariensee, DE-31535 Neustadt, Germany Petersen B, Niemann H, Izsvak Z, Ivics Z, Kues WA (2011). 0049 – (0)5034 871 120 Germline transgenic pigs by Sleeping Beauty transposition in por- 0049 – (0)5034 871 101 cine zygotes and targeted integration in the pig genome. PloS One 6, e23573, doi:10.1371/journal.pone.0023573. Giovannoni, R., Vargiolu, A., Manzini, S., De Cecco, M., Seruggia, D., Busnelli, M., Cerrito, M.G., Smolenski, R., Forni, M.., Bacci, M.L., Lavitrano, M. 2010. CMV-based episomal vector References is progressively lost in vivo in multi-transgenic pigs produced by SMGT. Transgenic Research 19, 330 (abstract) Arnould S, Perez C, Cabaniols JP, Smith J, Gouble A, Grizot S, Hammer RE, Pursel VG, Rexroad CE Jr, Wall RJ, Bolt DJ, Ebert Epinat JC, Duclert A, Duchateau P, Pâques F (2007) Engineered KM, Palmiter RD, Brinster RL (1985) Production of transgenic I-CreI derivatives cleaving sequences from the human XPC gene rabbits, sheep and pigs by microinjection. Nature 315: 680-683 can induce highly efficient gene correction in mammalian cells. J Hauschild J, Petersen B, Santiago Y, Queisser AL, Carnwath Mol Biol 371:49-65 JW, Lucas-Hahn A, Zhang L, Meng X, Gregory PD, Schwinzer Brevini, T.A., S. Antonini, G. Pennarossa, and F. Gandolfi. R, Cost GJ, Niemann H. 2011. Efficient generation of a biallelic 2008. Recent progress in embryonic stem cell research and its knockout in pigs using zinc-finger-nucleases. Proc Natl Acad Sci application in domestic species. Reprod Domest Anim. 43 Suppl U S A 108, 12013-7. Epub 2011 Jul 5 2:193-199. Hofmann A, Kessler B, Ewerling S, Weppert M, Vogg B, Ludwig Chang K, Qian J, Jiang M, Liu YH, Wu MC, Chen CD, Lai CK, H, Stojkovic M, Boelhauve M, Brem G, Wolf E, Pfeifer A (2003) Lo HL, Hsiao CT, Brown L, Bolen J Jr, Huang HI, Ho PY, Shih Efficient transgenesis in farm animals by lentiviral vectors. EMBO PY, Yao CW, Lin WJ, Chen CH, Wu FY, Lin YJ, Xu J, Wang K Rep 4:1054-1060 (2002) Effective generation of transgenic pigs and mice by linker Iqbal, K., Barg-Kues, B., Broll, S., Bode, J., Niemann, H., based sperm-mediated gene transfer. BMC Biotechnol 2:5 Kues, W.A. 2009. Cytoplasmic injection of circular plasmids allows Carlson, DF, Garbe JR, Tan W, Martin MJ, Dobrinsky JR, targeted expression in mammalian embryos. BioTechniques 47, Hackett PB, Clark KR, Fahrenkrug SC. 2011 Strategies for selec- 959-968. tion marker-free swine transgenesis using the Sleeping Beauty Irgang M, Sauer IM, Karlas A, Zeilinger K, Gerlach J, Kurth R,

Newsletter 36 Autumn 2011 21 ➤ Recent Progress of Transgenic Pig Models for Biomedicine and Pharmaceutical Research

Neuhaus P, Denner J (2003) Porcine endogenous retroviruses: no Giovannoni R, Della Casa G, Seren E, Rossi G. 2002. Efficient infection in patients treated with a bioreactor based on porcine liver production by sperm-mediated gene transfer of human decay cells. J Clin Virology 28:141-154 accelerating factor (hDAF) transgenic pigs for xenotransplantation. Ivics Z, Hackett PB, Plasterk RH, Izsvák Z (1997) Molecular Proc Natl Acad Sci U S A. 2002 Oct 29;99(22):14230-5 reconstruction of Sleeping Beauty, a Tc1-like transposon from fish, Luo Y, Li J, Liu Y, Lin L, Du Y, Li S., Yang H, Vaitja G, Callesen and its transposition in human cells. Cell 91: 501-510 H, Bolund L, Sorensen CB 2011. High efficiency of BRCA knock- Jakobsen, J., Li, J., Kragh, P.M., Moldt, B., Lin, L., Liu, Y., out using rAAV-mediated gene targeting: developing a pig model Schmidt, M., Winther, K.D., Schyth, B.D., Holm, I.E., Vajta, for breast cancer. Transgenic Res. G., Bolund, L., Callesen, H., Jorgensen, A.L., Nielsen, A.L., Manzini, S, Vargiolu A, Stehle IM, Bacci ML, Cerrito MG, Mikkelsen, J.G., 2011. Pig transgenesis by Sleeping Beauty DNA Giovannoni R, Zannoni A, Bianco MR, Forni M, Donini P, Papa M, transposition. Transgenic Research 20, 533-545. Lipps HJ, Lavitrano M, 2006. Genetically modified pigs produced Klose R, Kemter E, Bedke T, Bittmann I, Kelsser B, Endres R, with a nonviral episomal vector. Proc Natl Acad Sci USA 103, Pfeffer K, Schwinzer R, Wolf E. 2005. Expression of biologically 17672-17677. active human TRAIL in transgenic pigs. Transplantation. 2005 Jul Mendicino, M., Ramsoondar, J., Phelps, C.,Vaught, T., Ball, S. 27;80(2):222-30 LeLoith, T, Monahan, J., Chen, S., Dandro, A., Boone, J., Jobst, Kragh PM, Nielsen AL, Li J, Du Y, Lin L, Schmidt M, Bøgh IB, P., Vance, A., Wertz, N.,. Bergman, Z., Sun, X.-Z., Polejaeva, Holm IE, Jakobsen JE, Johansen MG, Purup S, Bolund L, Vajta G, I., Butler, J., Dai, Y., Ayares, D., Wells, K. 2010. Generation of Jørgensen AL. 2009. Hemizygous minipigs produced by random - and B cell-deficient pigs by targeted disruption of the gene insertion and handmade cloning express the Alzheimer’s J-region gene segment of the heavy chain locus. Trans. Research. disease-causing dominant mutation APPsw. Transgenic Res. (epub ahead of print) DOI 10.1007/s11248-010-9444-z 2009 Aug;18(4):545-58. Miller KF, Bolt DJ, Pursel VG, Hammer RE, Pinkert CA, Kues WA, Niemann H (2004) The contribution of farm animals Palmiter RD, Brinster RL (1989) Expression of human or bovine to human health. Trends Biotechnol 22: 286-294 growth hormone gene with a mouse metallothionein-1 promoter in Kues WA, Schwinzer R, Wirth D, Verhoeyen E, Lemme E, transgenic swine alters the secretion of porcine growth hormone Herrmann D, Barg-Kues B, Hauser H, Wonigeit H, Niemann H and insulin-like growth factor-I. J Endocrinol 120: 481-488 (2006) Epigenetic silencing and tissue independent expression Niemann H, Kues WA (2003) Application of transgenesis in of a novel tetracycline inducible system in double-transgenic pigs. livestock for agriculture and biomedicine. Anim Reprod Sci 79: FASEB Journal Express doi: 10.1096/fj.05-5415fje; printed short 291-317 version: FASEB J 20: 1200-1202 Niemann H, Kues, WA (2007) Transgenic farm animals: an Kues, W.A., Nowak-Imialek M, Haridoss S., Niemann H. update. Reprod Fertil Develop 19: 762-770 2010a. Strategies for the derivation of pluripotent cells from farm .Oropeza M, Petersen B, Carnwath JW, Lucas-Hahn A, Lemme animals. Reprod. Dom Anim 45, Suppl 3, 25-31. E, Hassel P, Herrmann D, Barg-Kues B, Holler S, Queisser Kues, W.A., Garrels, W., Mates, L., Holler,S., Niemann, H., AL, Schwinzer R, Hinkel R, Kupatt C, Niemann H. 2009. Izsvak, Z., Ivics, Z. 2010b. Production of transgenic pigs by the Transgenic expression of the human A20 gene in cloned pigs Sleeping Beauty transposon system. Transgenic Research 19, provides protection against apoptotic and inflammatory stimuli. 336 (abstract). Xenotransplantation. 2009 Kues, W.A., Niemann H. 2011. Advances in transgenic farm Park KW, Cheong HT, Lai L, Im GS, Kühholzer B, Bonk A, animals. Prev. Vet. Med. (epub ahead of print), doi:10.1016/j. Samuel M, Rieke A, Day BN, Murphy CN, Carter DB, Prather RS. prevetmed.2011.04.009 2001. Production of nuclear transfer-derived swine that express Kurome M, Ueda H, Tomii R, Naruse K, Nagashima H. the enhanced green fluorescent protein. Anim Biotechnol 12, 2006. Production of transgenic-clone pigs by the combination of 173-181. ICSI-mediated gene transfer with somatic cell nuclear transfer. Petersen B, Ramackers W, Tiede A, Lucas-Hahn A, Herrmann Transgenic Res. 15, 229-40. D, Barg-Kues B, Schuettler W, Friedrich L, Schwinzer R, Kuwaki K, Tseng YL, Dor FJ, Shimizu A, House SL, Sanderson Winkler M, Niemann H. 2009. Pigs transgenic for human TM, Lanceros CJ, Rabharasuth DD, Cheng J, Moran K, Hisashi thrombomodulin have elevated production of activated protein C. Y, Mueller N, Yamadoa K, Greenstein JL, Hawley RJ, Patience Xenotransplantation. 2009 Nov-Dec;16(6):486-95. C, Awwad M, Fishman JA, Robson SC, Schuurman HJ, Sachs Phelps CJ, Koike C, Vaught TD, Boone J, Wells KD, Chen SH, DH, Cooper DK (2005) in baboons using Ball S, Specht SM, Polejaeva IA, Monahan JA, Jobst PM, Sharma 1, 3-glactosyltransferase knockout pigs as donors: initial experi- SB, Lamborn AE, Garst AS, Moore M, Demetris AJ, Rudert WA, ments. Nat Med 11: 29-31 Bottino R, Bertera S, Trucco M, Starzl TE, Dai Y, Ayares DL Lai L, Kolber-Simonds D, Park KW, Cheong HT, Greenstein JL, (2003) Production of alpha 1,3-galactosyltransferasedeficient Im GS, Samuel M, Bonk A, Rieke A, Day BN, Murphy CN, Carter pigs. Science 299: 411–414 DB, Hawley RJ, Prather RS (2002) Production of a1, 3-galacto- Plasterk RH (2002) RNA silencing: the genomes immune sys- syltransferase knockout pigs by nuclear transfer cloning. Science tem. Science 296:1263-1265 295:1089-1092 Ramsoondar J, Vaught T, Ball S, Mendicino M, Monahan J, Jobst Lavitrano M, Bacci ML, Forni M, Lazzereschi D, Di Stefano P, Vance A, Duncan J, Wells K, Ayares D. Production of transgenic C, Fioretti D, Giancotti P, Marfé G, Pucci L, Renzi L, Wang H, pigs that express porcine endogenous retrovirus small interfering Stoppacciaro A, Stassi G, Sargiacomo M, Sinibaldi P, Turchi V, RNAs. Xenotransplantation. 2009 May-Jun;16(3):164-80.

22 Newsletter 36 Autumn 2011 ➤

Ramsoondar J, Mendicino M, Phelps C, Vaught T, Ball S, induced pluripotent stem cells produce chimeric offspring. Stem Monahan J, Chen S, Dandro A, Boone J, Jobst P, Vance A, Wertz Cells Dev. 19:1211-1220. N, Polejaeva I, Butler J, Dai Y, Ayares D, Wells K. 2011. Targeted Whitelaw CB, Radcliffe PA, Ritchie WA, Carlisle A, Ellard FM, disruption of the porcine immunoglobulin kappa light chain locus. Pena RN, Rowe J, Clark AJ, King TJ, Mitrophanous KA (2004) Transgenic Res. 2011 Jun;20(3):643-53. Efficient generation of transgenic pigs using equine infectious Renner S, Fehlings C, Herbach N, Hofmann A, von Waldthausen anaemia virus (EIAV) derived vector. FEBS Lett 571: 233-236 DC, Kessler B, Ulrichs K, Chodnevskaja I, Moskalenko V, Wu, Z., J. Chen, J. Ren, L. Bao, J. Liao, C. Chun, L. Rao, Amselgruber W, Göke B, Pfeifer A, Wanke R, Wolf E. 2010. H. Li, Y. Gu, H. Dai, H. Zhu, X. Teng, L. Cheng, and L. Xiao. Glucose intolerance and reduced proliferation of pancreatic beta- 2009. Generation of pig-induced pluripotent stem cells with a cells in transgenic pigs with impaired glucose-dependent insulino- drug-inducible system. Journal of Molecular Cell Biology. 1:46-54 tropic polypeptide function. Diabetes. 2010 May;59(5):1228-38. Whyte JJ, Prather RS. 2011. Genetic modifications of pigs for Robl JM, Wang Z, Kasinathan P, Kuroiwa Y (2007) Transgenic medicine and agriculture. Mol Reprod Dev. 2011 Jun 10. doi: animal production and animal biotechnology. Theriogenology 10.1002/mrd.21333. [Epub ahead of print] 67:127-133 (epub ahead of print, doi: 10.1016/j.theriogenol- Whyte JJ, Zhao J, Wells KD, Samuel MS, Whitworth KM, ogy.2006.09.034) Walters EM, Laughlin MH, Prather RS. 2011. Gene targeting with Rogers CS, Stoltz DA, Meyerholz DK, Ostedgaard LS, Rokhlina zinc finger nucleases to produce cloned eGFP knockout pigs. Mol T, Taft PJ, Rogan MP, Pezzulo AA, Karp PH, Itani OA, Kabel AC, Reprod Dev. 78, 2. Wohlford-Lenane CL, Davis GJ, Hanfland RA, Smith TL, Samuel Yamada K, Yazawa K, Shimizu A, Iwanaga T, Hisashi Y, Nuhn M, Wax D, Murphy CN, Rieke A, Whitworth K, Uc A, Starner TD, M, O’Malley P, Nobori S, Vagefi PA, Patience C, Fishman J, Brogden KA, Shilyansky J, McCray PB Jr, Zabner J, Prather RS, Cooper DK, Hawley RJ, Greenstein J, Schuurman HJ, Awwad M, Welsh MJ. (2008). Disruption of the CFTR gene produces a model Sykes M, Sachs DH (2005) Marked prolongation of porcine renal of cystic fibrosis in newborn pigs. Science. xenograft survival in baboons through the use of a1, 3-galacto- Sommer JR, Estrada JL, Collins EB, Bedell M, Alexander CA, syltransferase gene-knockout donors and the cotransplantation of Yang Z, Hughes G, Mir B, Gilger BC, Grob S, Wei X, Piedrahita vascularized thymic tissue. Nat Med 11: 32-34 JA, Shaw PX, Petters RM, Zhang K 2011 Production of ELOVL4 Yang YG, Sykes M (2007) Xenotransplantation: current status transgenic pigs: a large animal model for Stargardt-like macular and a perspective on the future. Nat Rev Immunol 7: 519-531 degeneration. Yang D, Wang CE, Zhao B, Li W, Ouyang Z, Liu Z, Yang H, Br J Ophthalmol. 2011 Aug 26. [Epub ahead of print] Fan P, O’Neill A, Gu W, Yi H, Li S, Lai L, Li XJ. 2010. Expression Switzer WM, Michler RE, Shangmugam V, Matthews A, of Huntington’s disease protein results in apoptotic neurons in Hussain AI, Wright A, Sandstrom P, Chapman L, Weber C, the brains of cloned transgenic pigs. Hum Mol Genet. 2010 Oct Safley S, Denny RD, Navarro A, Evans V, Norin AJ, Kwiatkowski 15;19(20):3983-94 P, Heneine W (2001) Lack of cross-species transmission of Yang D, Yang H, Li W, Zhao B, Ouyang Z, Liu Z, Zhao Y, porcine endogenous retrovirus infection to nonhuman primate Fan N, Song J, Tian J, Li F, Zhang J, Chang L, Pei D, Chen YE, recipients of porcine cells, tissues and organs. Transplantation Lai L. 2011. Generation of PPARγ mono-allelic knockout pigs via 71: 959-965 zinc-finger nucleases and nuclear transfer cloning. Cell Res. 2011 Takahashi K, Yamanaka S (2006) Induction of pluripotent stem Jun;21(6):979-82. Epub 2011 Apr 19 cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 126:663-676 Tucker A, Belcher C, Moloo B, Bell J, Mazzulli T, Humar Y, Hughes A, McArdle P, Talbot A (2002) The production of trans- genic pigs for potential use in clinical xenotransplantation: baseline clinical pathology and size studies. Xenotransplantation 9: 203-208 Umeyama K, Watanabe M, Saito H, Kurome M, Tohi S, Matsunari H, Miki K, Nagashima H. 2009. Dominant-negative mutant hepatocyte nuclear factor 1alpha induces diabetes in transgenic-cloned pigs. Transgenic Res. 2009 Oct;18(5):697-706 Urnov FD, Miller JC, Lee YL, Beausejour CM, Rock JM, Augustus S, Jamieson AC, Porteus MH, Gregory PD, Holmes MC (2005) Highly efficient endogenous human gene correction using designed zinc-finger nucleases. Nature 435: 646–651 Weiss EH, Lilienfeld BG, Müller S, Müller E, Herbach N, Kessler B, Wanke R, Schwinzer R, Seebach JD, Wolf E, Brem G. HLA-E/human beta2-microglobulin transgenic pigs: protection against xenogenic human anti-pig cytotoxicity. Transplantation. 2009 Jan 15;87(1):35-43. West, F.D., S.L. Terlouw, D.J. Kwon, J.L. Mumaw, S.K. Dhara, K. Hasneen, J.R. Dobrinsky, and S.L. Stice. 2010. Porcine

Newsletter 36 Autumn 2011 23