7
17 Newsletter 36 Autumn 2011 Abstract The first transgenic pigs were produced by the microinjection of foreign DNA into zygotic pronuclei in 1985. Since then, the me- thodological repertoire for porcine transgenesis was expanded to somatic cell nuclear transfer, lentiviral transgenesis and, recently, cytoplasmic plasmid injection. The major impact of transgenic pigs and minipigs took place in the fields of humanised pig models and biomedical disease models, whereas agricultural applications did not find broad acceptance. The recent release of the porcine whole genome sequence and parallel developments of highly spe- cific enzymes and RNAs now make it possible to perform precise genetic modifications and fully exploit the advantages of this large animal model. We anticipate that genetically modified pigs and minipigs will increasingly complement the commonly used small- animal models in biomedical research, since several aspects of disease progression, physiology, metabolism and aging cannot properly be mirrored in small-animal models. Introduction The production of transgenic pigs is labour-intensive and cost- intensive and depends on advanced techniques in molecular biolo- gy and the micromanipulation of gametes and zygotes. At present, progress in reproductive techniques and gene-transfer methods has allowed targeted modifications of the porcine genome (glos- sary box), albeit the overall success rates are still low (Clark and Whitelaw, 2003; Niemann and Kues, 2007; Robl et al., 2007). A bottleneck for porcine transgenesis is the lack of authentic pluri- potent stem cells that are suitable for blastocyst complementation experiments (Brevini et al., 2008; Kues et al., 2010a). The semi- nal development of induced pluripotent stem cells (iPS) in mice and humans (Takahashi and Yamanaka, 2006) provides a new approach to this end. The results of the first attempts to generate porcine iPS cells were published recently (Esteban et al., 2009; Wu et al., 2009; Ezashi et al., 2009), yet the potential of current porcine iPS cells to contribute to chimera formation seems to be limited (West et al., 2010). This paper briefly discusses the current progress of transgenic pig models for biomedical research. Comprehensive overviews about transgenic pigs and livestock are available elsewhere (Clark and Whitelaw, 2003; Robl et al., 2007; Kues and Niemann, 2011; Whyte and Prather, 2011). Basic and biomedical applications of transgenic pigs In the last few years, an expanded methodological repertoire for porcine gene transfer has been developed (Table 1), resulting in an increasing number of transgenic approaches (Whyte and Prather, 2011). At least 90% of genetically modified pigs are generated for biomedical studies (Fig. 1A). Sequencing and annotation of the porcine genome are important milestones for accelerating the Recent Progress of Transgenic Pig Models for Biomedicine and Pharmaceutical Research Authors: Wiebke Garrels, Heiner Niemann. Corresponding author: Wilfried A. Kues Key words: Domestic animals, disease model, humanised, genome, large animal model 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.

Ellegaard Göttingen Minipigs: Ellegaard Göttingen …...Xenotransplantation expression of human A20 (anti-apoptotic gene) Oropeza et al., 2009 Cystic fibrosis pig knockout of cystic

  • Upload
    others

  • View
    5

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Ellegaard Göttingen Minipigs: Ellegaard Göttingen …...Xenotransplantation expression of human A20 (anti-apoptotic gene) Oropeza et al., 2009 Cystic fibrosis pig knockout of cystic

17Newsletter 36 Autumn 2011

AbstractThe first transgenic pigs were produced by the microinjection of foreign DNA into zygotic pronuclei in 1985. Since then, the me-thodological repertoire for porcine transgenesis was expanded to somatic cell nuclear transfer, lentiviral transgenesis and, recently, cytoplasmic plasmid injection. The major impact of transgenic pigs and minipigs took place in the fields of humanised pig models and biomedical disease models, whereas agricultural applications did not find broad acceptance. The recent release of the porcine whole genome sequence and parallel developments of highly spe-cific enzymes and RNAs now make it possible to perform precise genetic modifications and fully exploit the advantages of this large animal model. We anticipate that genetically modified pigs and minipigs will increasingly complement the commonly used small-animal models in biomedical research, since several aspects of disease progression, physiology, metabolism and aging cannot properly be mirrored in small-animal models.

introductionThe production of transgenic pigs is labour-intensive and cost-intensive and depends on advanced techniques in molecular biolo-gy and the micromanipulation of gametes and zygotes. At present, progress in reproductive techniques and gene-transfer methods has allowed targeted modifications of the porcine genome (glos-sary box), albeit the overall success rates are still low (Clark and

Whitelaw, 2003; Niemann and Kues, 2007; Robl et al., 2007). A bottleneck for porcine transgenesis is the lack of authentic pluri-potent stem cells that are suitable for blastocyst complementation experiments (Brevini et al., 2008; Kues et al., 2010a). The semi-nal development of induced pluripotent stem cells (iPS) in mice and humans (Takahashi and Yamanaka, 2006) provides a new approach to this end. The results of the first attempts to generate porcine iPS cells were published recently (Esteban et al., 2009; Wu et al., 2009; Ezashi et al., 2009), yet the potential of current porcine iPS cells to contribute to chimera formation seems to be limited (West et al., 2010).

This paper briefly discusses the current progress of transgenic pig models for biomedical research. Comprehensive overviews about transgenic pigs and livestock are available elsewhere (Clark and Whitelaw, 2003; Robl et al., 2007; Kues and Niemann, 2011; Whyte and Prather, 2011).

basic and biomedical applications of transgenic pigsIn the last few years, an expanded methodological repertoire for porcine gene transfer has been developed (Table 1), resulting in an increasing number of transgenic approaches (Whyte and Prather, 2011). At least 90% of genetically modified pigs are generated for biomedical studies (Fig. 1A). Sequencing and annotation of the porcine genome are important milestones for accelerating the

Recent Progress of Transgenic Pig Models for Biomedicine and Pharmaceutical ResearchAuthors: Wiebke Garrels, Heiner Niemann. Corresponding author: Wilfried A. Kues

key words: Domestic animals, disease model, humanised, genome, large animal model

Fig.1. Increasing scientific interest in transgenic pig modelsA) 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.

Page 2: Ellegaard Göttingen Minipigs: Ellegaard Göttingen …...Xenotransplantation expression of human A20 (anti-apoptotic gene) Oropeza et al., 2009 Cystic fibrosis pig knockout of cystic

18 Newsletter 36 Autumn 2011

generation of transgenic models, even if the porcine genome assembly still has gaps (annotated porcine genome data can be found at: www.ensembl.org and www.pubmed.org). Since pig and minipig physiology, anatomy, pathology, genome organisa-tion, body weight and life span are more similar to humans than are rodents, the domesticated pig represents a more appropriate biomedical model (Table 2).

For certain biomedical therapies, such as xenotransplanta-tion (transplantation of organs from one species to another (e.g. porcine-to-human)), transgenic pigs are the only reasonable spe-cies (Niemann and Kues, 2003). Xenotransplantation seems to be one option for closing the widening gap between demand and availability of appropriate human organs (Yang and Sykes, 2007). The prerequisites for potential porcine–human xenotransplanta-tion are: (i) overcoming immunological hurdles; (ii) preventing the transmission of porcine pathogens to human recipients; and (iii) the compatibility of porcine organs with human physiology.

The suppression of hyperacute rejection of porcine xenografts has been achieved by transgenic expression of human regula-tors of complement activity (RCA) (Tucker et al., 2002) and a gene knockout of the porcine alpha, 1,3-galactosyltransferase gene (Dai et al., 2002; Lai et al., 2002; Phelps et al., 2003). Maximal survival rates of up to 3–6 months have been achieved

with porcine alpha-galactosyltransferase knockout organs (kidney or heart) transplanted to baboons (Kuwaki et al., 2005; Yamada et al., 2005).

Extensive research has been conducted to reduce the risk of porcine endogenous retrovirus (PERV) transmission to human patients (Switzer et al., 2001; Irgang et al., 2003). RNA interfe-rence (RNAi) is a promising method for knocking down the PERV expression. RNAi is based on small RNAs, either small interfering RNA (siRNA) or short hairpin RNAs (shRNA). In the cytoplasm, small RNA molecules are incorporated into an RNA-induced silencing complex (RISC) and targets binding to a complementary transcript sequence, resulting in mRNA degradation (Plasterk, 2002; Dallas and Vlassow, 2006). The efficacy of RNAi for redu-cing PERV expression has been demonstrated in cloned piglets (Dieckhoff et al., 2008; Ramsoondar et al., 2009).

For several approaches, a conditional gene expression is desi-rable over a constitutive transgenic expression. Initial animal mod-els carrying the first generation of conditional promoter elements suffered from high basal-expression levels and pleiotropic effects (Miller et al., 1989). Recent expression systems responsive to exogenous tetracycline resulted in more tightly controlled expres-sion. In pigs, a tetracycline-controlled transgenic expression was achieved with a bicistronic expression cassette (Kues et al., 2006)

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 cells and SCNT

Dai et al., 2002; Lai et al., 2002

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 SCNT

Dieckhoff et al., 2008; Ramsoondar et al., 2009

Transposon transgenesis Sleeping Beauty transposition in zygotic genome by CPI

Garrels et al., 2010; Kues et al., 2010b

Transposon transgenesis Sleeping Beauty transposition in somatic cells and SCNT

Jacobsen et al., 2011; Carlson et al. 2011

Targeted gene knockout Zinc finger nuclease-catalysed gene deletion in primary cells and SCNT

Whyte et al., 2011; Yang et al., 2011; Hauschild et al., 2011

Targeted integration Recombination-mediated cassette exchange in primary cells and SCNT

Garrels et al., 2011

ReCent PRoGReSS of tRAnSGeniC PiG modelS foR biomediCine And PhARmACeutiCAl ReSeARCh➤

Page 3: Ellegaard Göttingen Minipigs: Ellegaard Göttingen …...Xenotransplantation expression of human A20 (anti-apoptotic gene) Oropeza et al., 2009 Cystic fibrosis pig knockout of cystic

19Newsletter 36 Autumn 2011

that was designed to give ubiquitous expression of human RCAs. Crossbreeding of lines with two cassettes was necessary to over-come epigenetic silencing and to achieve tetracycline-sensitive RCA expression.

Transgenic pigs have been shown to mimic human diseases such as atherosclerosis, non-insulin-dependent diabetes, cystic fibrosis, cancer, ophthalmological and neurodegenerative disorders (Kues and Niemann, 2004; Kragh et al., 2010; Rogers et al., 2008; Yang et al., 2010; Luo et al., 2011). An important exam-ple is the minipig cystic fibrosis model, which develops disease phenotypes that are highly similar to human patients (Rogers et al., 2008), whereas transgenic mouse models failed to exhibit lung, pancreatic and intestinal obstructions. Huntington’s disease is a neurodegenerative disorder characterised by the expression of mutated huntingtin with expanded polyglutamine tracts. The misfolded protein accumulates in neurons and is suspected of trig-gering apoptosis. Whereas genetic mouse models often failed to replicate overt neurodegeneration and apoptosis, a minipig model expressing the N-terminal huntingtin with a polyglutamine tract seems to do so (Yang et al., 2010).

Truncation mutations in the elongation of a very long-chain fatty-acids-4 (ELOVL4) gene cause macular dystrophy. Photoreceptor topography in the pig retina is more similar to that in humans as it includes cone-rich, macula-like area centralis, whereas mice lack a macular. Transgenic pigs expressing disease-causing ELOVL4 mutations were generated by PNI and SCNT (Sommer et al., 2011). A detailed analysis showed photoreceptor loss, disorga-nised inner and outer segments, and diminished electroretinogra-phy responses, suggesting that the transgenic pigs mirror macular degeneration and provide a unique model for therapeutic interven-

tion. Recently, the first immunodeficient pigs were cloned by SCNT (Mendicino et al., 2010; Ramsoondar et al., 2011), promising to serve as large-animal models for cell transplantation experiments.

Conventional gain-of-function transgenesis is based on random integration of the transgene at sites of spontaneous double-strand breaks of chromosomal DNA. The frequency of DNA double-strand breaks at a defined locus can be considerably increased by introducing specifically designed endonuclease enzymes (Urnov et al., 2005; Arnould et al., 2007). The artificial endonucleases are based on the DNA recognition sites of zinc finger transcription factors, meganuclei or transcription factor like elements (TALE), and they can be designed to bind highly specifically to a single, predetermined sequence in the genome. Double-strand break-repair pathways often create small deletions and, thus, designed endonucleases allow efficient gene knockouts. The proof-of-prin-ciple to generate knockout pigs by synthetic zinc finger nucleases has been demonstrated by the inactivation of enhanced green fluorescent protein (EGFP), peroxisome proliferator-activated receptor (PPAR gamma) and alpha-galactosyltransferase (Whyte et al., 2011; Yang et al., 2011; Hauschild et al., 2011) in primary somatic cells and the subsequent use of knockout cells for SCNT, respectively. Thus current lack of authentic porcine ES cells can be circumvented for the purpose of generating knockout pigs.

DNA-based transposons are mobile genetic elements that move in the genome via a “cut-and-paste” mechanism. Most DNA transposons are simply organised: they encode a transposase protein flanked by inverted terminal repeats (ITRs), which carry transposase binding sites, and it has been possible to separate the transposase coding sequence from ITR sequences. Any DNA 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 necrosis factor ligand Klose et al., 2005

Xenotransplantation expression of human leukocyte antigen 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

Page 4: Ellegaard Göttingen Minipigs: Ellegaard Göttingen …...Xenotransplantation expression of human A20 (anti-apoptotic gene) Oropeza et al., 2009 Cystic fibrosis pig knockout of cystic

20 Newsletter 36 Autumn 2011

ReCent PRoGReSS of tRAnSGeniC PiG modelS foR biomediCine And PhARmACeutiCAl ReSeARCh

become enzymatically integrated into nuclear DNA. In a two-component system, the transposon is integrated solely by the trans-supplementation activity of transposase. The first transposon sufficiently active for use in vertebrates was the Sleeping Beauty (SB) transposon (Ivics et al., 1997; Clark et al., 2007). Many drawbacks of classical transgenic methods can be overcome by transposition-catalysed gene delivery, which increases the efficiency of chromosomal integration and facilitates single-copy (monomeric) insertion events. An additional advantage of trans-poson-catalysed transgenesis is that the integration of monomeric transgene units is directed to accessible euchromatic regions. Transposon transgenic pigs have been generated (Kues et al., 2010b; Garrels et al., 2011) by CPI (Iqbal et al., 2009), as well as by SCNT (Jakobsen et al., 2010; Carlson, 2011; Garrels, 2011). Ubiquitous expression of a fluorescent Venus protein, a derivative of the commonly used EGFP, was found in somatic and germ cells (differentiated spermatozoa) in own experiments (Fig. 1B, Garrels et al., 2011) for all integrations sites, strongly supporting the hypothesis that transposase preferentially integrates DNA into euchromatic regions. The robust transgenic expression of Venus is strictly copy-number dependent and facilitates cell-tracking experi-ments in cell-therapy approaches. The identification of integrations sites revealed that most transposon integration sites were found in intergenic regions of the porcine genome (Fig. 2). This approach made it possible to identify loci, which are suitable for transgenic

expression. Importantly, transposon-tagged loci can be read-dressed by recombination-mediated cassette exchange (RMCE) in cell culture. Via SCNT, the RMCE cells can be used to generate vital piglets carrying a targeted integration into a “safe harbour” locus (Garrels et al., 2011).

Since integrated transposons can be remobilised in the pre-sence of a transposase enzyme, these animals can provide the basis for performing whole genome mutagenesis screens in the pig. For the SB transposon, the phenomenon of local hopping after mobilisation has been described. The majority of secondary integrations take place at a distance of up to 5 megabases from the original integration. Figure 2 depicts one integration site on the gene-rich X chromosome. The neighbouring porcine genes are the von Hippel-Lindau binding gene (VBP1) and a novel gene, both about 10,000 base pairs away from the integration site. After mobilisation, the integration site can be screened for integration events in neighbouring genes, such as the VBP1. The VBP1 gene is of potential interest as an animal model, and the gene product is assumed to form a complex with the von Hippel-Lindau tumor sup-pressor (VHL). The von Hippel-Lindau syndrome is a dominantly inherited cancer syndrome predisposing carriers to several malig-nant and benign tumours. Thus, transposon transgenic pigs can be employed for performing unbiased and biased mutagenic events. It is anticipated that mutagenic screens with more advanced con-structs will be applied in the near future.

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.

Page 5: Ellegaard Göttingen Minipigs: Ellegaard Göttingen …...Xenotransplantation expression of human A20 (anti-apoptotic gene) Oropeza et al., 2009 Cystic fibrosis pig knockout of cystic

21Newsletter 36 Autumn 2011

ConclusionsMethodological improvements for gene transfer into the pig genome and a rapidly increasing list of biomedical pig models have been developed in recent years. Together with more accurate genome data and highly specific designed enzymes and RNAs, precise genetic modifications have become feasible. It is anticipa-ted that authentic pluripotent cells of the pig will be generated in the near future. Thus, porcine transgenesis will become a routine tool for generating relevant humanised porcine models. The most obvious application of transgenic pigs will be as disease models and biomedical therapies, which are not well-reflected in small rodent models. The progress expected in porcine transgenesis (increased success rates and decreasing costs), however, will make the pig an attractive complementary model for advanced approaches in biomedical research.

AcknowledgmentsThe expert technical support of Ms S. Holler, Ms Barg-Kues, Ms Herrmann and Ms Ziegler, and the financial support of the Deutsche Forschungsgemeinschaft (DFG) are gratefully acknow-ledged.

Conflicts of interestThe authors declare no conflicts of interest.

Wiebke Garrels, Heiner NiemannFriedrich-Loeffler-InstituteMariensee, DE-31535 Neustadt, Germany

Wilfried A. KuesFriedrich-Loeffler-InstituteInstitute of Farm Animal GeneticsMariensee, DE-31535 Neustadt, Germany0049 – (0)5034 871 1200049 – (0)5034 871 101

RefeRenCeS

Arnould S, Perez C, Cabaniols JP, Smith J, Gouble A, Grizot S, Epinat JC, Duclert A, Duchateau P, Pâques F (2007) Engineered I-CreI derivatives cleaving sequences from the human XPC gene can induce highly efficient gene correction in mammalian cells. J Mol Biol 371:49-65

Brevini, T.A., S. Antonini, G. Pennarossa, and F. Gandolfi. 2008. Recent progress in embryonic stem cell research and its application in domestic species. Reprod Domest Anim. 43 Suppl 2:193-199.

Chang K, Qian J, Jiang M, Liu YH, Wu MC, Chen CD, Lai CK, Lo HL, Hsiao CT, Brown L, Bolen J Jr, Huang HI, Ho PY, Shih PY, Yao CW, Lin WJ, Chen CH, Wu FY, Lin YJ, Xu J, Wang K (2002) Effective generation of transgenic pigs and mice by linker based sperm-mediated gene transfer. BMC Biotechnol 2:5

Carlson, DF, Garbe JR, Tan W, Martin MJ, Dobrinsky JR, Hackett PB, Clark KR, Fahrenkrug SC. 2011 Strategies for selec-tion marker-free swine transgenesis using the Sleeping Beauty

transposon system. Transgenic Res. 20, Jan. 9 (epub ahead of print).

Clark J, Whitelaw B (2003) A future for transgenic livestock. Nat Rev Genet 4:825-833

Clark, K.J:, Carlson, D.F., Fahrenkrug, S.C. 2007. Pigs taking wings with transposons and recombinases. Genome Biol. 8, Suppl 1: S13.

Dai Y, Vaught TD, Boone J, Chen SH, Phelps CJ, Ball S, Monahan JA, Jobst PM, McCreath KJ, Lamborn AE, Cowell-Lucero JL, Wells KD, Colman A, Polejaeva IA, Ayares DL (2002) Targeted disruption of the a1,3-galactosyltransferase gene in cloned pigs. Nat Biotechnol 20: 251-255

Dallas A, Vlassow A (2006) RNAi: A novel antisense technology and its therapeutic potential. Med Sci Monit 12 RA67-74

Dieckhoff B, Petersen B, Kues WA, Kurth R, Niemann H, Denner J (2008) Knockdown of porcine endogenous retrovirus (PERV) expression by PERV-specific shRNA in transgenic pigs. Xenotransplantation 15: 36-45

Esteban, M.A., J. Xu, J. Yang, M. Peng, D. Qin, W. Li, Z. Jiang, J. Chen, K. Deng, M. Zhong, J. Cai, L. Lai, and D. Pei. 2009. Generation of induced pluripotent stem cell lines from Tibetan miniature pig. J Biol Chem. 284:17634-17640.

Ezashi, T., B.P.V.L. Telugu, A.P. Alexenko, S. Sachdev, S. Sinha, and R.M. Roberts. 2009. Derivation of induced pluri-potent stem cells from pig somatic cells. Proceedings of the National Academy of Sciences of the United States of America. 106:10993-10998.

Garrels, W., Mates, L., Holler,S., Niemann, H., Izsvak, Z., Ivics, Z., Kues, W.A: 2010. Generation of transgenic pigs by the Sleeping Beauty transposition in zygotes. Reprod. Dom. Anim. 45, 65 (abstract).

Garrels, W., Mates, L., Holler, S., Dalda, A., Taylor, U., Petersen B, Niemann H, Izsvak Z, Ivics Z, Kues WA (2011). Germline transgenic pigs by Sleeping Beauty transposition in por-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 is progressively lost in vivo in multi-transgenic pigs produced by SMGT. Transgenic Research 19, 330 (abstract)

Hammer RE, Pursel VG, Rexroad CE Jr, Wall RJ, Bolt DJ, Ebert KM, Palmiter RD, Brinster RL (1985) Production of transgenic rabbits, sheep and pigs by microinjection. Nature 315: 680-683

Hauschild J, Petersen B, Santiago Y, Queisser AL, Carnwath JW, Lucas-Hahn A, Zhang L, Meng X, Gregory PD, Schwinzer R, Cost GJ, Niemann H. 2011. Efficient generation of a biallelic knockout in pigs using zinc-finger-nucleases. Proc Natl Acad Sci U S A 108, 12013-7. Epub 2011 Jul 5

Hofmann A, Kessler B, Ewerling S, Weppert M, Vogg B, Ludwig H, Stojkovic M, Boelhauve M, Brem G, Wolf E, Pfeifer A (2003) Efficient transgenesis in farm animals by lentiviral vectors. EMBO Rep 4:1054-1060

Iqbal, K., Barg-Kues, B., Broll, S., Bode, J., Niemann, H., Kues, W.A. 2009. Cytoplasmic injection of circular plasmids allows targeted expression in mammalian embryos. BioTechniques 47, 959-968.

Irgang M, Sauer IM, Karlas A, Zeilinger K, Gerlach J, Kurth R,

Page 6: Ellegaard Göttingen Minipigs: Ellegaard Göttingen …...Xenotransplantation expression of human A20 (anti-apoptotic gene) Oropeza et al., 2009 Cystic fibrosis pig knockout of cystic

22 Newsletter 36 Autumn 2011

ReCent PRoGReSS of tRAnSGeniC PiG modelS foR biomediCine And PhARmACeutiCAl ReSeARCh

Neuhaus P, Denner J (2003) Porcine endogenous retroviruses: no infection in patients treated with a bioreactor based on porcine liver cells. J Clin Virology 28:141-154

Ivics Z, Hackett PB, Plasterk RH, Izsvák Z (1997) Molecular reconstruction of Sleeping Beauty, a Tc1-like transposon from fish, and its transposition in human cells. Cell 91: 501-510

Jakobsen, J., Li, J., Kragh, P.M., Moldt, B., Lin, L., Liu, Y., Schmidt, M., Winther, K.D., Schyth, B.D., Holm, I.E., Vajta, G., Bolund, L., Callesen, H., Jorgensen, A.L., Nielsen, A.L., Mikkelsen, J.G., 2011. Pig transgenesis by Sleeping Beauty DNA transposition. Transgenic Research 20, 533-545.

Klose R, Kemter E, Bedke T, Bittmann I, Kelsser B, Endres R, Pfeffer K, Schwinzer R, Wolf E. 2005. Expression of biologically active human TRAIL in transgenic pigs. Transplantation. 2005 Jul 27;80(2):222-30

Kragh PM, Nielsen AL, Li J, Du Y, Lin L, Schmidt M, Bøgh IB, Holm IE, Jakobsen JE, Johansen MG, Purup S, Bolund L, Vajta G, Jørgensen AL. 2009. Hemizygous minipigs produced by random gene insertion and handmade cloning express the Alzheimer’s disease-causing dominant mutation APPsw. Transgenic Res. 2009 Aug;18(4):545-58.

Kues WA, Niemann H (2004) The contribution of farm animals to human health. Trends Biotechnol 22: 286-294

Kues WA, Schwinzer R, Wirth D, Verhoeyen E, Lemme E, Herrmann D, Barg-Kues B, Hauser H, Wonigeit H, Niemann H (2006) Epigenetic silencing and tissue independent expression of a novel tetracycline inducible system in double-transgenic pigs. FASEB Journal Express doi: 10.1096/fj.05-5415fje; printed short version: FASEB J 20: 1200-1202

Kues, W.A., Nowak-Imialek M, Haridoss S., Niemann H. 2010a. Strategies for the derivation of pluripotent cells from farm animals. Reprod. Dom Anim 45, Suppl 3, 25-31.

Kues, W.A., Garrels, W., Mates, L., Holler,S., Niemann, H., Izsvak, Z., Ivics, Z. 2010b. Production of transgenic pigs by the Sleeping Beauty transposon system. Transgenic Research 19, 336 (abstract).

Kues, W.A., Niemann H. 2011. Advances in transgenic farm animals. Prev. Vet. Med. (epub ahead of print), doi:10.1016/j.prevetmed.2011.04.009

Kurome M, Ueda H, Tomii R, Naruse K, Nagashima H. 2006. Production of transgenic-clone pigs by the combination of ICSI-mediated gene transfer with somatic cell nuclear transfer. Transgenic Res. 15, 229-40.

Kuwaki K, Tseng YL, Dor FJ, Shimizu A, House SL, Sanderson TM, Lanceros CJ, Rabharasuth DD, Cheng J, Moran K, Hisashi Y, Mueller N, Yamadoa K, Greenstein JL, Hawley RJ, Patience C, Awwad M, Fishman JA, Robson SC, Schuurman HJ, Sachs DH, Cooper DK (2005) Heart transplantation in baboons using 1, 3-glactosyltransferase knockout pigs as donors: initial experi-ments. Nat Med 11: 29-31

Lai L, Kolber-Simonds D, Park KW, Cheong HT, Greenstein JL, Im GS, Samuel M, Bonk A, Rieke A, Day BN, Murphy CN, Carter DB, Hawley RJ, Prather RS (2002) Production of a1, 3-galacto-syltransferase knockout pigs by nuclear transfer cloning. Science 295:1089-1092

Lavitrano M, Bacci ML, Forni M, Lazzereschi D, Di Stefano C, Fioretti D, Giancotti P, Marfé G, Pucci L, Renzi L, Wang H, Stoppacciaro A, Stassi G, Sargiacomo M, Sinibaldi P, Turchi V,

Giovannoni R, Della Casa G, Seren E, Rossi G. 2002. Efficient production by sperm-mediated gene transfer of human decay accelerating factor (hDAF) transgenic pigs for xenotransplantation. Proc Natl Acad Sci U S A. 2002 Oct 29;99(22):14230-5

Luo Y, Li J, Liu Y, Lin L, Du Y, Li S., Yang H, Vaitja G, Callesen H, Bolund L, Sorensen CB 2011. High efficiency of BRCA knock-out using rAAV-mediated gene targeting: developing a pig model for breast cancer. Transgenic Res.

Manzini, S, Vargiolu A, Stehle IM, Bacci ML, Cerrito MG, Giovannoni R, Zannoni A, Bianco MR, Forni M, Donini P, Papa M, Lipps HJ, Lavitrano M, 2006. Genetically modified pigs produced with a nonviral episomal vector. Proc Natl Acad Sci USA 103, 17672-17677.

Mendicino, M., Ramsoondar, J., Phelps, C.,Vaught, T., Ball, S. LeLoith, T, Monahan, J., Chen, S., Dandro, A., Boone, J., Jobst, P., Vance, A., Wertz, N.,. Bergman, Z., Sun, X.-Z., Polejaeva, I., Butler, J., Dai, Y., Ayares, D., Wells, K. 2010. Generation of antibody- and B cell-deficient pigs by targeted disruption of the J-region gene segment of the heavy chain locus. Trans. Research. (epub ahead of print) DOI 10.1007/s11248-010-9444-z

Miller KF, Bolt DJ, Pursel VG, Hammer RE, Pinkert CA, Palmiter RD, Brinster RL (1989) Expression of human or bovine growth hormone gene with a mouse metallothionein-1 promoter in transgenic swine alters the secretion of porcine growth hormone and insulin-like growth factor-I. J Endocrinol 120: 481-488

Niemann H, Kues WA (2003) Application of transgenesis in livestock for agriculture and biomedicine. Anim Reprod Sci 79: 291-317

Niemann H, Kues, WA (2007) Transgenic farm animals: an update. Reprod Fertil Develop 19: 762-770

.Oropeza M, Petersen B, Carnwath JW, Lucas-Hahn A, Lemme E, Hassel P, Herrmann D, Barg-Kues B, Holler S, Queisser AL, Schwinzer R, Hinkel R, Kupatt C, Niemann H. 2009. Transgenic expression of the human A20 gene in cloned pigs provides protection against apoptotic and inflammatory stimuli. Xenotransplantation. 2009

Park KW, Cheong HT, Lai L, Im GS, Kühholzer B, Bonk A, Samuel M, Rieke A, Day BN, Murphy CN, Carter DB, Prather RS. 2001. Production of nuclear transfer-derived swine that express the enhanced green fluorescent protein. Anim Biotechnol 12, 173-181.

Petersen B, Ramackers W, Tiede A, Lucas-Hahn A, Herrmann D, Barg-Kues B, Schuettler W, Friedrich L, Schwinzer R, Winkler M, Niemann H. 2009. Pigs transgenic for human thrombomodulin have elevated production of activated protein C. Xenotransplantation. 2009 Nov-Dec;16(6):486-95.

Phelps CJ, Koike C, Vaught TD, Boone J, Wells KD, Chen SH, Ball S, Specht SM, Polejaeva IA, Monahan JA, Jobst PM, Sharma SB, Lamborn AE, Garst AS, Moore M, Demetris AJ, Rudert WA, Bottino R, Bertera S, Trucco M, Starzl TE, Dai Y, Ayares DL (2003) Production of alpha 1,3-galactosyltransferasedeficient pigs. Science 299: 411–414

Plasterk RH (2002) RNA silencing: the genomes immune sys-tem. Science 296:1263-1265

Ramsoondar J, Vaught T, Ball S, Mendicino M, Monahan J, Jobst P, Vance A, Duncan J, Wells K, Ayares D. Production of transgenic pigs that express porcine endogenous retrovirus small interfering RNAs. Xenotransplantation. 2009 May-Jun;16(3):164-80.

Page 7: Ellegaard Göttingen Minipigs: Ellegaard Göttingen …...Xenotransplantation expression of human A20 (anti-apoptotic gene) Oropeza et al., 2009 Cystic fibrosis pig knockout of cystic

23Newsletter 36 Autumn 2011

induced pluripotent stem cells produce chimeric offspring. Stem Cells Dev. 19:1211-1220.Whitelaw CB, Radcliffe PA, Ritchie WA, Carlisle A, Ellard FM, Pena RN, Rowe J, Clark AJ, King TJ, Mitrophanous KA (2004) Efficient generation of transgenic pigs using equine infectious anaemia virus (EIAV) derived vector. FEBS Lett 571: 233-236

Wu, Z., J. Chen, J. Ren, L. Bao, J. Liao, C. Chun, L. Rao, H. Li, Y. Gu, H. Dai, H. Zhu, X. Teng, L. Cheng, and L. Xiao. 2009. Generation of pig-induced pluripotent stem cells with a drug-inducible system. Journal of Molecular Cell Biology. 1:46-54

Whyte JJ, Prather RS. 2011. Genetic modifications of pigs for medicine and agriculture. Mol Reprod Dev. 2011 Jun 10. doi: 10.1002/mrd.21333. [Epub ahead of print]

Whyte JJ, Zhao J, Wells KD, Samuel MS, Whitworth KM, Walters EM, Laughlin MH, Prather RS. 2011. Gene targeting with zinc finger nucleases to produce cloned eGFP knockout pigs. Mol Reprod Dev. 78, 2.

Yamada K, Yazawa K, Shimizu A, Iwanaga T, Hisashi Y, Nuhn M, O’Malley P, Nobori S, Vagefi PA, Patience C, Fishman J, Cooper DK, Hawley RJ, Greenstein J, Schuurman HJ, Awwad M, Sykes M, Sachs DH (2005) Marked prolongation of porcine renal xenograft survival in baboons through the use of a1, 3-galacto-syltransferase gene-knockout donors and the cotransplantation of vascularized thymic tissue. Nat Med 11: 32-34

Yang YG, Sykes M (2007) Xenotransplantation: current status and a perspective on the future. Nat Rev Immunol 7: 519-531

Yang D, Wang CE, Zhao B, Li W, Ouyang Z, Liu Z, Yang H, Fan P, O’Neill A, Gu W, Yi H, Li S, Lai L, Li XJ. 2010. Expression of Huntington’s disease protein results in apoptotic neurons in the brains of cloned transgenic pigs. Hum Mol Genet. 2010 Oct 15;19(20):3983-94

Yang D, Yang H, Li W, Zhao B, Ouyang Z, Liu Z, Zhao Y, Fan N, Song J, Tian J, Li F, Zhang J, Chang L, Pei D, Chen YE, Lai L. 2011. Generation of PPAR mono-allelic knockout pigs via zinc-finger nucleases and nuclear transfer cloning. Cell Res. 2011 Jun;21(6):979-82. Epub 2011 Apr 19

Ramsoondar J, Mendicino M, Phelps C, Vaught T, Ball S, Monahan J, Chen S, Dandro A, Boone J, Jobst P, Vance A, Wertz N, Polejaeva I, Butler J, Dai Y, Ayares D, Wells K. 2011. Targeted disruption of the porcine immunoglobulin kappa light chain locus. Transgenic Res. 2011 Jun;20(3):643-53.

Renner S, Fehlings C, Herbach N, Hofmann A, von Waldthausen DC, Kessler B, Ulrichs K, Chodnevskaja I, Moskalenko V, Amselgruber W, Göke B, Pfeifer A, Wanke R, Wolf E. 2010. Glucose intolerance and reduced proliferation of pancreatic beta-cells in transgenic pigs with impaired glucose-dependent insulino-tropic polypeptide function. Diabetes. 2010 May;59(5):1228-38.

Robl JM, Wang Z, Kasinathan P, Kuroiwa Y (2007) Transgenic animal production and animal biotechnology. Theriogenology 67:127-133 (epub ahead of print, doi: 10.1016/j.theriogenol-ogy.2006.09.034)

Rogers CS, Stoltz DA, Meyerholz DK, Ostedgaard LS, Rokhlina T, Taft PJ, Rogan MP, Pezzulo AA, Karp PH, Itani OA, Kabel AC, Wohlford-Lenane CL, Davis GJ, Hanfland RA, Smith TL, Samuel M, Wax D, Murphy CN, Rieke A, Whitworth K, Uc A, Starner TD, Brogden KA, Shilyansky J, McCray PB Jr, Zabner J, Prather RS, Welsh MJ. (2008). Disruption of the CFTR gene produces a model of cystic fibrosis in newborn pigs. Science.

Sommer JR, Estrada JL, Collins EB, Bedell M, Alexander CA, Yang Z, Hughes G, Mir B, Gilger BC, Grob S, Wei X, Piedrahita JA, Shaw PX, Petters RM, Zhang K 2011 Production of ELOVL4 transgenic pigs: a large animal model for Stargardt-like macular degeneration.

Br J Ophthalmol. 2011 Aug 26. [Epub ahead of print]Switzer WM, Michler RE, Shangmugam V, Matthews A,

Hussain AI, Wright A, Sandstrom P, Chapman L, Weber C, Safley S, Denny RD, Navarro A, Evans V, Norin AJ, Kwiatkowski P, Heneine W (2001) Lack of cross-species transmission of porcine endogenous retrovirus infection to nonhuman primate recipients of porcine cells, tissues and organs. Transplantation 71: 959-965

Takahashi K, Yamanaka S (2006) Induction of pluripotent stem 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 organ 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 natural killer cell 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