Skip to main content
Advertisement

Main menu

  • Home
  • Articles
    • Accepted manuscripts
    • Latest complete issue
    • Issue archive
    • Archive by article type
    • Subject collections
    • Interviews
    • Sign up for alerts
  • About us
    • About DMM
    • Editors and Board
    • Editor biographies
    • Travelling Fellowships
    • Grants and funding
    • Journal Meetings
    • Workshops
    • The Company of Biologists
    • Journal news
  • For authors
    • Submit a manuscript
    • Aims and scope
    • Presubmission enquiries
    • Article types
    • Manuscript preparation
    • Cover suggestions
    • Editorial process
    • Promoting your paper
    • Open Access
    • Outstanding paper prize
    • Biology Open transfer
  • Journal info
    • Journal policies
    • Rights and permissions
    • Media policies
    • Reviewer guide
    • Sign up for alerts
  • Contact
    • Contact DMM
    • Advertising
    • Feedback
  • COB
    • About The Company of Biologists
    • Development
    • Journal of Cell Science
    • Journal of Experimental Biology
    • Disease Models & Mechanisms
    • Biology Open

User menu

  • Log in
  • Log out

Search

  • Advanced search
Disease Models & Mechanisms
  • COB
    • About The Company of Biologists
    • Development
    • Journal of Cell Science
    • Journal of Experimental Biology
    • Disease Models & Mechanisms
    • Biology Open

supporting biologistsinspiring biology

Disease Models & Mechanisms

Advanced search

RSS   Twitter   Facebook   YouTube

  • Home
  • Articles
    • Accepted manuscripts
    • Latest complete issue
    • Issue archive
    • Archive by article type
    • Subject collections
    • Interviews
    • Sign up for alerts
  • About us
    • About DMM
    • Editors and Board
    • Editor biographies
    • Travelling Fellowships
    • Grants and funding
    • Journal Meetings
    • Workshops
    • The Company of Biologists
    • Journal news
  • For authors
    • Submit a manuscript
    • Aims and scope
    • Presubmission enquiries
    • Article types
    • Manuscript preparation
    • Cover suggestions
    • Editorial process
    • Promoting your paper
    • Open Access
    • Outstanding paper prize
    • Biology Open transfer
  • Journal info
    • Journal policies
    • Rights and permissions
    • Media policies
    • Reviewer guide
    • Sign up for alerts
  • Contact
    • Contact DMM
    • Advertising
    • Feedback
REVIEW
Genetically engineered pigs as models for human disease
Carolin Perleberg, Alexander Kind, Angelika Schnieke
Disease Models & Mechanisms 2018 11: dmm030783 doi: 10.1242/dmm.030783 Published 22 January 2018
Carolin Perleberg
Chair of Livestock Biotechnology, School of Life Sciences, Technische Universität München, 85354 Freising, Germany
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Alexander Kind
Chair of Livestock Biotechnology, School of Life Sciences, Technische Universität München, 85354 Freising, Germany
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Angelika Schnieke
Chair of Livestock Biotechnology, School of Life Sciences, Technische Universität München, 85354 Freising, Germany
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Angelika Schnieke
  • For correspondence: schnieke@wzw.tum.de
  • Article
  • Figures & tables
  • Info & metrics
  • PDF
Loading

ABSTRACT

Genetically modified animals are vital for gaining a proper understanding of disease mechanisms. Mice have long been the mainstay of basic research into a wide variety of diseases but are not always the most suitable means of translating basic knowledge into clinical application. The shortcomings of rodent preclinical studies are widely recognised, and regulatory agencies around the world now require preclinical trial data from nonrodent species. Pigs are well suited to biomedical research, sharing many similarities with humans, including body size, anatomical features, physiology and pathophysiology, and they already play an important role in translational studies. This role is set to increase as advanced genetic techniques simplify the generation of pigs with precisely tailored modifications designed to replicate lesions responsible for human disease. This article provides an overview of the most promising and clinically relevant genetically modified porcine models of human disease for translational biomedical research, including cardiovascular diseases, cancers, diabetes mellitus, Alzheimer's disease, cystic fibrosis and Duchenne muscular dystrophy. We briefly summarise the technologies involved and consider the future impact of recent technical advances.

Introduction

New therapies and diagnostic methods are required for many human diseases. There are, however, no in vitro systems capable of modelling human whole-body pathophysiology; as such, disease research still relies on animals. Work with laboratory animals is carefully controlled, and workers in the field have a duty to ‘replace, reduce and refine’ their use whenever possible [see Article 47 of Directive 2010/63/EU (http://eur-lex.europa.eu/legal-content/EN/TXT/PDF/?uri=CELEX:32010L0063&from=EN) and www.nc3rs.org.uk]. It is thus important to ensure that all data gained are valuable and relevant to the disease studied. This is best achieved with well-defined animal models that replicate relevant aspects of human pathology as closely as possible.

Mice are now the most intensively studied and widely used mammalian species in biomedical research, mainly because they are convenient and cheap to house, and methods for their genetic modification are well advanced (Chu et al., 2016; Skarnes et al., 2011). Mouse studies have provided a wealth of information on the molecular basis of human disease and have enabled a host of proof-of-principle studies. Mice, however, do not always accurately model human disease pathology, reducing their predictive value for preclinical studies (Mak et al., 2014). Many new drugs fail in clinical trials because preclinical studies fail to predict safety and effectiveness in human patients (Justice and Dhillon, 2016; Ledford, 2011). Nonrodent species can provide additional information and improve the predictive value of preclinical studies (Bähr and Wolf, 2012).

Pigs share several key similarities with humans in terms of their body size, anatomical features, physiology, pathophysiological responses and diet, and are used to develop and refine biomedical procedures and medical equipment (Heinritz et al., 2013; Kararli, 1995; Schubert et al., 2016). Their use in biomedical research is aided by several practical factors, such as their favourable breeding characteristics. Pigs mature relatively quickly for a large species (6-7 months), have a short gestation period (∼114 days) and produce large litters (∼10 piglets per litter), depending on the breed (Sachs, 1994). Centuries of pig domestication have established suitable housing conditions, including specific pathogen-free conditions, which require only minor adaptation for research. Furthermore, as food animals, there is wide public acceptance of their humane use, which is not the case for other nonrodent species, such as primates.

The extension of genetic modification technology to pigs has greatly increased their value to biomedicine, motivating efforts to develop porcine models that replicate human disease, and so ‘bridge the gap between bench and bedside’. This review outlines the techniques used and describes the most promising and relevant porcine models of human disease (summarised in Table 1). This is, however, not a comprehensive account of all genetically modified pigs, and some reports of nonviable animals or animals with no relevant phenotype have been omitted. Our aim is to highlight the importance of porcine disease models for translational biomedical research, and to indicate those porcine models that recapitulate human disease pathology most accurately and those with the greatest potential.

View this table:
  • View inline
  • View popup
  • Download powerpoint
Table 1.

Summary of the genetically modified pig models for human diseases

Methods for generating porcine disease models

Genetic modification of livestock became a reality when transgenic rabbits, sheep and pigs were first produced by pronuclear deoxyribonucleic acid (DNA) microinjection (Fig. 1A) (Hammer et al., 1985). This method is straightforward but inefficient in terms of the proportion of transgenic animals produced, a significant problem with larger species (Hammer et al., 1985; Logan and Martin, 1994; Uchida et al., 2001). Porcine oocytes are also problematic for microinjection as their high lipid content makes them almost opaque and centrifugation is required to visualise the pronuclei (Fig. 2) (Kikuchi et al., 2002a). DNA microinjection in its basic form also results in random transgene integration, which lacks the precision and power of gene targeting. Nevertheless, this remained the only practical technique available to livestock biotechnologists for over a decade (Schnieke et al., 1997).

Fig. 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 1.

Methods used to generate genetically modified pigs. (A) Pronuclear microinjection of DNA results in random integration of transgenes into the genome, but does not enable gene targeted modifications. Viral vectors can also be microinjected to increase the frequency of transgenesis. (B) Somatic primary cells can be cultured and genetically modified by various methods to add random transgenes or for gene targeting. A genetically modified cell (shown in green) is introduced into the perivitelline space of an enucleated oocyte and an electrical pulse used to fuse the cell membranes and simultaneously activate the oocyte. (C) Endonuclease RNA or protein and guide RNA(s) are injected into the cytoplasm of the fertilised oocyte or zygote to directly modify the embryo genome.

Fig. 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 2.

Porcine oocytes. Note the opacity of the ooplasm caused by their high lipid content. Porcine oocytes require centrifugation to visualise their pronuclei for microinjection.

Meanwhile, the development of gene targeting via homologous recombination (HR) (see Glossary, Box 1) in mouse embryonic stem (ES) cells revolutionised the genetic modification of mice (Evans and Kaufman, 1981; Smithies et al., 1985; Thomas and Capecchi, 1987). The potential of this technology for livestock was recognised at an early stage, but the generation of fully functional ES, embryonic germ, or induced pluripotent stem cells from livestock species that were capable of germline transmission has been unsuccessful (Nowak-Imialek and Niemann, 2012). The search for a functional equivalent led to the development of nuclear transfer from primary somatic cells that could be transfected and analysed in culture. This was used first for random transgenesis (Schnieke et al., 1997) and, subsequently, for gene targeting (McCreath et al., 2000). The method was first demonstrated in sheep but soon extended to pigs (Dai et al., 2002; Lai et al., 2002), where it continues to be a mainstream approach.

Box 1. Glossary

Chylomicron – lipoprotein particles that are composed of triglycerides, cholesterol, phospholipids and apolipoproteins, including APOE and APOC.

CRISPR/Cas9 – the CRISPR/Cas9 system consists of the clustered regularly interspaced short palindromic repeat (CRISPR) locus, which contains sequences that guide the endonuclease Cas9 (CRISPR-associated) to foreign DNA via base complementary to cause targeted DNA cleavage.

Homologous recombination (HR) – nucleotide sequence exchange between two similar or identical sequences.

Maturity onset diabetes of the young (MODY) – a type of diabetes that is associated with monogenetic defects in β-cells and characterised by impaired insulin secretion (insulin function itself remains normal).

Meconium ileus – obstruction of the intestine by sticky meconium (the first stool of mammalian infants).

Nuclear transfer – transfer of a nucleus into an enucleated oocyte.

Recombinase-mediated cassette exchange (RMCE) – a recombination procedure that facilitates recombinases like Cre and Flp to replace, turn or remove gene cassettes.

ROSA26 locus – a genomic locus found in mice that yields ubiquitous and constitutive expression of any gene introduced into it via gene targeting. Homologues exist in pig and human and are called the same name.

Site-specific recombinase – enzymes that aid site-specific, not random, recombination processes in the cell.

Transcription activator-like effector nuclease (TALEN) – a complex consisting of the transcription activator-like effector (TALE), a DNA-binding transcription factor, and the nuclease domain of the FokI restriction enzyme, which creates targeted DNA breaks.

Zinc finger nuclease (ZFN) – an artificial construct composed of a DNA-binding zinc finger protein and the nuclease domain of the FokI restriction enzyme, which creates targeted DNA breaks.

Gene targeting in pigs is informed by mouse studies, and aided by bioinformatics and genome sequence data (Groenen et al., 2012). HR in primary porcine somatic cells is, however, much less efficient than in mouse ES cells, although some loci, such as porcine ROSA26, support efficient gene targeting (Li et al., 2014). The relatively short lifespan of cultured primary cells also strictly limits the time available for in vitro manipulation and cell expansion, while maintaining the ability to generate animals by nuclear transfer (Schnieke et al., 1997). Nuclear transfer is itself difficult and time intensive and, despite steady improvements, produces live viable healthy offspring with relatively low efficiency (Callesen et al., 2014; Kurome et al., 2013). Generating gene targeted pigs has thus been technically challenging, considerably more so than generating gene-targeted mice, as attested by the relatively few such pig lines available.

Nevertheless, other important genetic manipulation techniques developed in mouse ES cells have been extended to pigs (Fig. 1), such as site-specific recombination (see Glossary, Box 1) to control gene expression (Leuchs et al., 2012; Li et al., 2014) and the use of recombinase-mediated cassette exchange (RMCE; see Glossary, Box 1) to induce rearrangements (Clark et al., 2007; Jakobsen et al., 2013). Gene targeting using adeno-associated viral vectors (AAVs) has also been established in pigs (Luo et al., 2011). Improved methods of microinjection, including lentiviral vectors and transposon systems, have also increased rates of transgenesis in pigs (Clark et al., 2007; Garrels et al., 2011; Hofmann et al., 2003; Ivics et al., 2014; Whitelaw et al., 2004).

Gene editing

The development of synthetic, highly specific endonuclease technologies as tools for ‘gene editing’ has probably had the greatest impact on the genetic modification of pigs. The ability to introduce a single double-strand break (DSB) at a unique predetermined site enables genes to be inactivated by insertion or deletion mutations, introduced via nonhomologous end joining (NHEJ) repair, or by targeted sequence replacement via homology-directed repair with an exogenous homologous DNA fragment. The practicality and simplicity of gene editing has steadily improved in successive generations of endonuclease systems, beginning with zinc finger nucleases (ZFNs; see Glossary, Box 1) (Hauschild et al., 2011; Kwon et al., 2013), then transcription activator-like effector nucleases (TALENs; see Glossary, Box 1) (Carlson et al., 2012) and, most recently, through the use of the CRISPR/Cas9 system (see Glossary, Box 1) (Tan et al., 2013).

Highly efficient gene editing makes it possible to carry out genetic modification directly in zygotes and early-stage embryos and thus avoids nuclear transfer altogether. HR in mouse zygotes is normally very infrequent, <0.1% (Brinster et al., 1989), but the use of ZFNs raises this to 1.7-4.5% (Meyer et al., 2010). ZFN-mediated gene editing in embryos was first demonstrated in a nonrodent species, the rabbit, in 2011 (Flisikowska et al., 2011), and was subsequently extended to pigs (Lillico et al., 2013).

ZFNs and TALENs have now largely been superseded by CRISPR/Cas9, which is equally if not more efficient in inducing DSBs and in stimulating HR (Mali et al., 2013; Wu et al., 2016) (Fig. 3). The CRISPR/Cas9 system also offers improved target specificity, i.e. less off-target activity and better prediction of off-target effects (Cho et al., 2013; Fu et al., 2014; Mali et al., 2013). There have already been reports of potential porcine models of human disease based on gene knockouts generated by the injection of CRISPR/Cas9 components into zygotes (Hai et al., 2014; Wang et al., 2015; Whitworth et al., 2014; Yu et al., 2016). However, gene targeting by HR using CRISPR/Cas9 is more challenging because homology-directed repair is far less frequent than NHEJ, but this approach has been achieved in mice (Chu et al., 2016; Miyaoka et al., 2016). Genetically modified knock-in pigs have also been generated using CRISPR/Cas9 with single-stranded oligodeoxynucleotides as a template with an efficiency of 80% (Zhou et al., 2016).

Fig. 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 3.

Gene editing by CRISPR/Cas9 for gene inactivation and targeted sequence replacement. During gene editing by CRISPR/Cas 9, the endonuclease Cas9 (green) is led by the guide RNA (gRNA) to the genomic target site, where it cleaves the double-stranded DNA (dsDNA) at a point 3-5 bp upstream of the protospacer adjacent motif (PAM). The resulting double-strand break (DSB) can then be repaired by nonhomologous end joining (NHEJ, left) or by homology-directed repair (HDR, right). NHEJ is an error-prone mechanism that can lead to sequence deletion, insertion or both, which can disrupt gene function. The HDR pathway is more precise and uses template DNA to repair the DSB via homologous recombination. The introduction of an exogenous DNA template, as dsDNA or as single-stranded oligodeoxynucleotide (ssODN), allows desired sequence changes to be engineered.

Gene editing directly in zygotes is likely to replace nuclear transfer as the standard method of generating transgenic pigs because of its efficiency and simplicity.

Availability of porcine oocytes

An adequate supply of porcine oocytes is essential for nuclear transfer, as are fertilised oocytes or zygotes for newer methods of direct embryo manipulation. While unfertilised and fertilised oocytes can be collected by flushing the reproductive tract, this requires the use of many animals and is cost and labour intensive. Progress in this area came with the development of a robust method of in vitro oocyte maturation using ovaries taken from pigs at slaughter, and, more recently, from the production of early porcine embryos in vitro (Kikuchi et al., 2002a). This is a multistage process that involves in vitro oocyte maturation, in vitro sperm preparation, in vitro fertilisation (IVF) and embryo culture. Progress is being made in each of these areas, but IVF is at present the limiting step, because of the problem of fertilisation by multiple sperm (Romar et al., 2016). Different groups have reported that freezing semen (Suzuki et al., 2003) or performing IVF in rotating culture (Kitaji et al., 2015) improves monospermy. To date, there have been very few reports of pigs generated from entirely in vitro-produced pig embryos (Kikuchi et al., 2002b; Whitworth et al., 2014; Yoshioka et al., 2012).

Modelling cardiovascular diseases

Cardiovascular diseases (CVDs) are the most common cause of morbidity and death worldwide [World Health Statistics 2017: Monitoring health for the SDGs (http://apps.who.int/iris/bitstream/10665/255336/1/9789241565486-eng.pdf?ua=1)]. Atherosclerosis, the primary culprit, is a chronic inflammatory condition characterised by lipid accumulation, thickening of arterial walls and plaque formation (Lusis, 2000). Chronic expansion of plaques reduces blood flow, and plaque rupture can precipitate acute thrombotic events, such as cardiac infarction and stroke (Falk, 2006).

Mutations of human genes involved in lipoprotein metabolism, including low-density lipoprotein receptor (LDLR) and proprotein convertase subtilisin/kexin type 9 (PCSK9), can cause familial hypercholesterolaemia (FH) and atherosclerosis or, as in the case of apolipoprotein E (APOE), increase disease risk. Several murine CVD models have been generated, including overexpression of wild-type Pcsk9, and knockout of Ldlr and Apoe (Maxwell and Breslow, 2004). Both Ldlr- and Apoe-deficient mice develop atherosclerotic lesions and hypercholesterolaemia and have provided valuable insights into atherogenic mechanisms (Ishibashi et al., 1993; Plump et al., 1999; Zhang et al., 1992). Importantly, however, these mouse models do not exhibit plaque rupture and thrombosis, a key feature of human atherosclerotic disease (Bentzon and Falk, 2010).

Pigs are well suited to model human CVDs because of similarities in their cardio- and cerebrovascular systems, blood parameters and vessel size, and have been used to develop and improve diagnostic tools and equipment, such as plaque localisation and imaging (Worthley et al., 2000). Also, unlike mice, pigs spontaneously develop atherosclerosis that can be accelerated by an atherogenic diet (Reiser et al., 1959; Skold et al., 1966).

FH has been modelled in Yucatan miniature pigs through AAV-mediated inactivation of LDLR (Davis et al., 2014). LDLR+/− heterozygous pigs developed hypercholesterolaemia, and LDLR−/− homozygotes developed more severe hypercholesterolaemia and atherosclerotic lesions in coronary arteries and abdominal aorta, disease locations common in humans with CVD. Disease severity was also increased by a diet rich in fat and cholesterol (Davis et al., 2014).

Another porcine FH model has been generated in Yucatan miniature pigs using DNA transposons to introduce a human PCSK9 transgene that carries the gain-of-function mutation D374Y controlled by a liver-specific human alpha1-antitrypsin promoter (Al-Mashhadi et al., 2013). These pigs showed increased degradation of LDLR, reduced hepatic LDLR, reduced plasma low-density lipoprotein uptake, hypercholesterolemia and atherosclerotic lesions, and have been used to test new imaging techniques to evaluate antiatherosclerotic drugs (Al-Mashhadi et al., 2013). The same model has also been used to investigate the influence of diabetes on atherosclerosis, revealing that poorly controlled blood glucose did not induce more advanced lesions nor increase plaque burden, findings consistent with human studies (Al-Mashhadi et al., 2015). However, none of the pig models described has been reported to show plaque rupture or thrombosis.

Lipid accumulation is an important risk factor in the development of atherosclerotic lesions. Apolipoprotein C3 (APOC3) is a major regulator of plasma triglyceride levels, and its overexpression is closely associated with hypertriglyceridaemia in patients with metabolic syndrome (Cohn et al., 2004). Minipigs that overexpress human APOC3 show increased plasma triglycerides owing to their delayed clearance, increased very low-density lipoprotein/chylomicron plasma fractions (see Glossary, Box 1) and reduced lipoprotein lipase activity, but not atherosclerotic lesions (Wei et al., 2012).

Thus, while the current generation of pig CVD models do not reproduce all aspects of human CVD pathology, they do recapitulate important early events, including dose-dependent hypercholesterolaemia, hypertriglyceridaemia and atherosclerotic lesions, and thus provide valuable tools to investigate early-stage FH and atherosclerosis, and for the development of new therapies.

Modelling cancer

Cancers are the second leading cause of death worldwide, and are set to increase as human populations age (World Health Statistics 2017: Monitoring health for the SDGs). In 2012, there were more than 14 million new cancer cases and 8 million cancer-associated deaths (Ferlay et al., 2015). It is thus concerning that only 5% of new anticancer agents are approved for patient use. Reasons for this have been ascribed to the complexity of cancers and the lack of good animal models for preclinical studies (Hutchinson and Kirk, 2011).

Genetically modified mouse strains have been vital tools for cancer research, but have also revealed that mouse and human cancers often differ. For example, murine cells are more easily transformed in vitro than human cells (Rangarajan et al., 2004), and different sets of genetic events are required for tumorigenesis (Kendall et al., 2005). Mouse cancer models might not, therefore, always provide the best representation of human disease.

Porcine oncology is a new field and the extent to which pigs replicate human cancers will become clearer as more models are characterised. Adam et al. were the first to address this question, using autologous transplantation of primary porcine cells transformed with viral oncogenic cDNAs (e.g. dominant-negative Tp53DD, c-MycT58A and H-RasG12V) to demonstrate that tumorigenesis in pigs resembles that in humans (Adam et al., 2007). Tumorigenesis has also been reported in pigs carrying randomly integrated transgenes that encode Cre-dependent KRASG12D and TP53R167H mutations (Box 2) (Schook et al., 2015). Primary porcine mesenchymal stem cells (MSCs) have also been transformed stepwise into sarcoma cells, showing that they resemble human MSCs in requiring the perturbation of TP53, RB1, KRAS and MYC signalling pathways to become fully transformed (Saalfrank et al., 2016).

Box 2. Generating inducible oncogenic mutations in pigs

The controlled activation of inducible and site-specific oncogenic mutations can mimic the spontaneous events that initiate and drive the progression of human cancers. This can be achieved by the introduction of oncogenic mutations silenced by an upstream transcriptional stop signal. Site-specific recombination, e.g. via Cre, excises the stop signal and allows the mutant gene to be expressed. Pigs that express the recombinase in a tissue-specific and/or drug-inducible manner can then be cross bred to activate mutant alleles in chosen tissues (Jin et al., 2014; Klymiuk et al., 2012a; Luo et al., 2014). Local administration of Cre in vivo via viral vectors is also possible (Schook et al., 2015).

Embedded Image

To monitor the pattern and extent of tissue-specific and/or drug-inducible Cre recombination in vivo, we have developed a ‘Cre-reporter’ pig that carries a dual fluorochrome cassette that switches expression from red to green after Cre recombination, enabling recombinase activity to be directly visualised (Li et al., 2014) [see figure above showing a piglet that carries a dual fluorochrome cassette in the ROSA26 locus (left) next to a wild-type piglet (right)]. Inducible porcine models have also been generated using the latent oncogenic mutations KRASG12D (Li et al., 2015; Schook et al., 2015) and TP53R167H (Leuchs et al., 2012; Schook et al., 2015), a mutation that is orthologous to the human R175H TP53 mutation; both of these represent mutations commonly found in human cancer (Levine and Oren, 2009; Pylayeva-Gupta et al., 2011).

The more recombinase driver lines that become available, the wider the range of tissues and cancers that can be investigated. This is an area where collaboration and synergy between livestock biotechnology researchers is clearly valuable.

Modelling colorectal cancer

Colorectal cancer (CRC) is the fourth most common cancer worldwide in both sexes (Ferlay et al., 2015). More than 80% of all sporadic cases are initiated by the functional disruption of the tumour suppressor gene adenomatous polyposis coli (APC) (Fearnhead et al., 2001; Morán et al., 2010). APC mutations are also responsible for a hereditary predisposition to CRC, familial adenomatous polyposis (FAP) (Kinzler et al., 1991). FAP varies considerably in severity, but patients typically develop adenomatous polyps in the colon and rectum in early life and have greatly increased risk of CRC (Croner et al., 2005).

Many murine models, most based on Apc mutations, have been generated to model human FAP (Karim and Huso, 2013). However, mutation of Apc alone does not replicate the pattern of human polyposis. For example, the widely used ApcMin mouse develops polyps mainly in the small intestine, and not in the colon, as in FAP patients (Karim and Huso, 2013). More complex mouse models based on additional mutant genes and models that use tissue-specific and locally activated oncogenes have been more successful at modelling FAP (Fearon, 2011; Hung et al., 2010; Tetteh et al., 2016).

Evidence is accumulating that pigs are perhaps more suitable than mice to model CRC. We have generated pigs that carry a translational stop signal at codon 1311 in porcine APC (APC1311), which is orthologous to the human APC1309 mutation responsible for a severe form of FAP. These pigs develop polyps in the colon and rectum as early as 4 months of age, which display epithelial features that are typical of the adenoma-carcinoma sequence, including aberrant crypt foci, and adenomatous polyps with low- and high-grade intraepithelial dysplasia (Flisikowska et al., 2012). Dysplastic adenomas also exhibit loss of APC heterozygosity; a hallmark of human FAP and of sporadic CRC (Albuquerque et al., 2002). Adenomas also showed marked accumulation of β-catenin and high expression of the β-catenin target, c-MYC, and frequent phosphorylation of ERK1/2 (MAPK3/1), a marker of MAPK pathway activation, and a known driver of intestinal tumorigenesis. Although invasive carcinoma has not yet been observed in this APC model, this is probably a function of time. Because mutation of porcine APC is sufficient to initiate polyposis without any further engineered mutation, spontaneous events that drive the transition from polyps to cancer can be investigated over time by colonoscopic monitoring and biopsy using standard human equipment. This is not possible in mice and would be very difficult in humans. Comparative transcriptome analysis of high- and low-grade porcine adenomas has also revealed the differential expression of gene sets similar to that in humans, as well as the upregulation of genes known to play a role in human CRC, and interesting new candidate genes involved in precancer development (Flisikowska et al., 2017). Heterozygous APC knockout pigs have also been generated by TALEN-mediated inactivation in cultured cells, but so far no phenotypic analysis has been reported (Tan et al., 2013).

The recapitulation of early human CRC pathogenesis was an important step in establishing pigs as a resource for studying human cancers. The FAP model is the first in a series of porcine cancer models to be generated, as discussed further below and in Box 2.

Modelling osteosarcoma

Osteosarcoma is relatively rare, but is the predominant form of primary bone cancer (Mirabello et al., 2009). It mainly affects young people and is highly malignant, requiring aggressive surgical resection and cytotoxic chemotherapy, the effects of which can be devastating (Durfee et al., 2016). There is thus a pressing need for animal models of this cancer to improve its surgical management, to develop new drugs and to better understand the molecular basis of its initiation and progression. Most human osteosarcomas are sporadic and of unknown cause, but can arise after radiation treatment. They frequently have TP53 mutations (Overholtzer et al., 2003) and/or alterations that affect cell cycle checkpoints, such as RB1 (Kansara et al., 2014). Patients with Li-Fraumeni syndrome, which is linked to TP53 mutations, are also predisposed to osteosarcoma (Ognjanovic et al., 2012).

Trp53 inactivation in mice results in diverse cancers, with ∼25% of heterozygotes and ∼4% of homozygotes developing osteosarcomas; homozygotes mainly develop lymphomas (Jacks et al., 1994). Improved mouse osteosarcoma models have been developed based on conditional Trp53 inactivation in the osteogenic lineage. These show highly penetrant osteosarcoma formation, but have been criticised because the primary tumours predominantly affect the axial skeleton, in contrast to human osteosarcomas, which tend to arise in the long bones of the limbs (Guijarro et al., 2014).

We have generated pigs that carry a latent TP53R167H mutation in exon 5 that can be activated by Cre-mediated excision of an upstream transcriptional stop signal (Box 2). In its uninduced form, transcription of the major TP53 transcript is blocked by a transcriptional stop signal (Leuchs et al., 2012; Saalfrank et al., 2016). Pigs heterozygous for the uninduced allele develop osteosarcomas after 16 months of age, while homozygotes show multiple osteosarcomas at 7-8 months (Leuchs et al., 2012; Saalfrank et al., 2016). The sarcomas primarily affect the long bones, skull and mandible, mirroring human pathology (Guijarro et al., 2014; Saalfrank et al., 2016). Porcine osteosarcoma cells show cytogenetic abnormalities characteristic of human TP53-mutant osteosarcoma (Boehm et al., 2000), including abnormal giant nuclei, micronuclei and multinuclear cells with fragmented nuclei and atypical mitotic figures (Saalfrank et al., 2016). Human osteosarcomas show genome-wide DNA instability (Overholtzer et al., 2003), and pig osteosarcoma cells are predominantly karyotypically abnormal (Saalfrank et al., 2016). Also, as in humans, pig osteosarcoma cells show increased resistance to radiation (Saalfrank et al., 2016). Data so far indicate that osteosarcoma is the predominant pathophenotype in these pigs. Little is known about the origin of human osteosarcoma, and the porcine model provides a valuable resource to study the genetic events involved.

Sieren et al. have generated Yucatan minipigs that carry an R176H mutation in the endogenous TP53 gene but, unlike the model above, this is expressed ubiquitously (Sieren et al., 2014). Heterozygous TP53R176H mutant pigs showed no tumour development even at 30 months of age, while those homozygotes that reached sexual maturity developed a variety of neoplastic lesions, including osteogenic tumours, lymphomas and renal tumours, recapitulating what has been observed in humans and mice with orthologous mutations.

The difference between the pathophenotypes exhibited by these two models is interesting and comparative analysis could shed light on the events that initiate this deadly disease.

Modelling diabetes mellitus

Diabetes mellitus is a diverse group of conditions characterised by loss of control of blood glucose levels. By far the most common form is type 2, which is mainly caused by insulin resistance combined with relative insulin deficiency and associated with excess body weight (American Diabetes, 2013). Many rodent models that target insulin signalling and action have provided valuable insights into diabetic disease mechanisms (Aigner et al., 2008; King, 2012).

Following food intake, the incretin hormones glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide-1 (GLP1; also known as GCG) are secreted to enhance glucose-induced insulin secretion (Baggio and Drucker, 2007). GIP function is impaired in type 2 diabetes, suggesting its involvement in early disease pathogenesis (Nauck et al., 2004). To model type 2 diabetes, Renner et al. generated pigs that express a human dominant-negative GIP receptor mutant (GIPRdn) in pancreatic islets controlled by the rat insulin promoter (Renner et al., 2010). At 11 weeks of age, these pigs showed reduced glucose tolerance due to delayed insulin secretion, and reduced insulin secretion and pancreatic β-cell mass with increasing age. The reduction in β-cell mass is caused by diminished cell proliferation and survival as a result of poor GIP signalling, consistent with findings in other species (Kim et al., 2008). Metabolic studies revealed changes in plasma amino acids and lipids that correlate significantly with β-cell mass (Renner et al., 2012). This pig model has been used to test whether the drug liraglutide, a GLP1 receptor agonist, can compensate for GIP deficiency by increasing GLP1 signalling (Streckel et al., 2015). The drug was found to improve insulin sensitivity, reduce weight gain and food uptake, but did not stop the loss of β-cell mass (Renner et al., 2016). These findings are similar to those from human diabetes patients, while studies in mouse models of diabetes show more disparate results (Shimoda et al., 2011; Tamura et al., 2015).

Mutations in the human insulin (INS) gene, e.g. INSC96Y, can cause permanent neonatal diabetes mellitus (Stoy et al., 2007). Pigs that express a porcine INSC94Y mutant insulin transgene (orthologous to human INSC96Y) in β-cells accumulated misfolded insulin in the endoplasmic reticulum and exhibited β-cell apoptosis. At 8 days after birth, INSC94Y animals developed cataracts and at 4.5 months they showed signs of permanent neonatal diabetes mellitus, including reduced body weight, decreased β-cell mass and reduced fasting insulin levels (Renner et al., 2013). No diabetes-associated pathological changes were detected in the kidney or nervous tissue during 1 year of observation.

Maturity onset diabetes of the young (see Glossary, Box 1) is characterised by impaired insulin secretion with minimal impact on insulin action (American Diabetes, 2013), and is commonly caused by dominant-negative mutations in the gene encoding hepatocyte nuclear factor 1α (HNF-1α) (Yamagata et al., 1996). Piglets carrying a dominant-negative human HNF-1αP291fsinsC mutation developed hyperglycaemia at 2 weeks, and at 19 weeks showed distinct glomerular nodular lesions in the kidneys, a hallmark of diabetic nephropathy, that expanded over the 10-month observation period (Hara et al., 2014; Umeyama et al., 2009, 2013). However, this model lacks several diabetic renal features characteristic of human diabetic nephropathy (Hara et al., 2014).

Diabetes mellitus has a multitude of phenotypic manifestations that are unlikely to be recapitulated in a single animal model. The models mentioned above each show different features that can be investigated further and could lead to the identification of novel therapeutic targets.

Modelling Alzheimer's disease

Alzheimer's disease (AD) is a multifactorial progressive disease of the brain, characterised by memory loss and disorientation, and accounts for 50-80% of human dementia cases (Winblad et al., 2016). Although its aetiology is not fully understood, it features several key disease hallmarks, including the formation of extracellular beta amyloid protein (Aβ) senile plaques and of intraneuronal neurofibrillary tangles mainly composed of tau protein, as well as neuronal dysfunction and cell death (Ballard et al., 2011). Familial forms of AD are caused by missense gain-of-function mutations in the genes encoding amyloid-β precursor protein (APP), presenilin 1 (PSEN1) and presenilin 2 (PSEN2) (Wu et al., 2012). Abnormal processing and clearance of the transmembrane APP by secretase complexes, in part composed of PSEN1 and PSEN2, lead to Aβ senile plaque development (Citron et al., 1997; De Strooper et al., 1998; Wolfe et al., 1999). Although the aetiology of sporadic AD is more complex than the familial form, the clinical, neuropathological and biochemical similarities have led researchers to replicate the causative genetic lesions in animals.

Transgenic mice that express mutant human APP (e.g. double mutant K670N, M671L; V717F) develop senile plaques, but not neurofibrillary tangles or neuronal loss (Games et al., 1995; Hsiao et al., 1996; LaFerla and Green, 2012), and this was also the case when APP mutations were combined with PSEN1 mutations (Takeuchi et al., 2000), indicating that mice are not suitable to model AD.

Kragh et al. have generated Göttingen minipigs that carry a randomly integrated human APPsw transgene with two mutations (K670N and M671L) driven by the human platelet derived growth factor-beta promoter (Kragh et al., 2009). Despite high transgene expression in the brain, mutant pigs showed no phenotype. The same group also generated minipigs with a human mutant PSEN1M146I transgene driven by a cytomegalovirus (CMV)-enhanced human UbiC promoter, produced by RMCE and transposon delivery (Jakobsen et al., 2013). Although mutant PSEN1 protein was expressed, there was again no evidence of an AD phenotype. Jakobsen et al. have also generated double transgenic Göttingen minipigs that carry human PSEN1M146I and APPsw transgenes. This combination caused an increase in intraneuronal Aβ plaque formation between 10 and 18 months of age (Jakobsen et al., 2016). The authors hypothesised that this might be the first step in AD pathology and a precursor to extracellular senile plaque formation as similar developments have been observed in mice. Further analyses over time are clearly necessary.

These porcine models recapitulate early-stage human AD and confirm previous findings that porcine brain biology is similar to human (Dickerson and Dobbing, 1967; Glauser, 1966; Thibault and Margulies, 1998). As such, they will be useful in studying early-stage AD, but whether a representative model of the full disease can be produced in pigs or another animal remains to be seen.

Modelling cystic fibrosis

Cystic fibrosis (CF), one of the most common autosomal recessive genetic disorders in populations of northern European extraction, is caused by dysfunction of the CF transmembrane conductance regulator (CFTR), a chloride channel present in the epithelial lining of several tissues, including the airways, intestine, pancreatic ducts, testes and sweat glands (Gadsby et al., 2006). Many CFTR mutations have been identified, but 70% of cases are caused by deletion of a phenylalanine at position 508 (ΔF508) (Boyle and De Boeck, 2013; Rogers et al., 2008a). The most serious consequence of impaired chloride channel function caused by CFTR mutations is clogging of the airway with mucus, leaving it susceptible to bacterial infections, the main cause of morbidity and mortality (Stoltz et al., 2015). The intestine, pancreas, reproductive tract and biliary system are also affected by this disease.

Several Cftr mutant mouse models, including a ΔF508 model, have been generated, but none replicate obstructive lung disease (Wilke et al., 2011). Other species, including sheep and pigs, have been investigated in the search for more representative models. Pigs share similarities in lung function and anatomy with humans, and have been used to model pulmonary abnormalities that play key roles in CF, including infection and inflammation (Pabst, 1996; Pabst and Binns, 1994). CFTR mutant pigs have been generated in various ways, for example, by introducing a translational stop codon in exon 10 (Rogers et al., 2008b), the ΔF508 mutation in exon 10 (Ostedgaard et al., 2011), and a stop box in exon 1 (Klymiuk et al., 2012b). All three of these models replicate human pathology equally well. Piglets show features characteristic of CF, including exocrine pancreatic destruction, vas deferens abnormalities, focal biliary cirrhosis, and lung disease marked by inflammation and infection (Ostedgaard et al., 2011; Rogers et al., 2008b; Stoltz et al., 2013). Consistent with findings in humans, CF pigs also show abnormal tracheal structures, as well as axonal and demyelinating neuropathy (Klymiuk et al., 2012b; Meyerholz et al., 2010; Reznikov et al., 2013). However, all piglets also showed a severe form of meconium ileus (see Glossary, Box 1), which is lethal in early life. This occurs in humans but is much less common, affecting 15% of CF patients (Stoltz et al., 2013). Stoltz et al. have thus generated a ‘gut-corrected’ model that incorporates a normal CFTR transgene controlled by a Fabp2 promoter to restore CFTR expression in the intestine and thus viability (Stoltz et al., 2013). These animals have been used to test CFTR gene therapy using viral vectors, a treatment that successfully improved anion transport, tracheal surface lipid pH and bacterial killing (Cooney et al., 2016; Steines et al., 2016).

The three porcine models generated to model CF are all well characterised and show accurate replication of the human pathology. They are probably the most advanced of the porcine disease models and are already being used to test CFTR gene therapy.

Modelling Duchenne muscular dystrophy

Duchenne muscular dystrophy (DMD) is an X-linked lethal disorder characterised by progressive muscle weakness and wasting that affects approximately 1 in 3500-5000 human males (Mendell et al., 2012). It is caused by frameshift mutations in the DMD gene, which lead to loss of the muscle protein dystrophin. Mutation hotspots have been identified in exons 3-7 and 45-55 of the DMD gene (Koenig et al., 1989).

Several mouse models of DMD have been generated but, unlike humans, mice require more than one mutation to replicate the human disease phenotype (Araki et al., 1997; McGreevy et al., 2015). Spontaneous DMD mutations have also been identified in cats and dogs (McGreevy et al., 2015; Nakamura and Takeda, 2011). Dog models replicate the human phenotype better than do mouse models of this disease, but the causative mutation in dogs does not occur in human DMD patients (Yu et al., 2015).

Male pigs that carry a DMD gene lacking exon 52 replicate signs of human DMD pathology, including loss of dystrophin in skeletal muscle, progressive muscular dystrophy, increased serum creatine kinase levels and impaired mobility (Klymiuk et al., 2013). However, the animals’ lifespan is reduced to ≤3 months, precluding natural breeding. Transcriptome analysis of skeletal muscle at 3 months resembled that of human DMD patients (Klymiuk et al., 2013), as did proteome analysis (Frohlich et al., 2016). The mutation used occurs commonly in human DMD and can be treated by exon 51 skipping, as shown in human patients (Goemans et al., 2011; Heemskerk et al., 2010). This model could be useful in devising treatment strategies for DMD, but its practical value would be greatly enhanced by improved viability.

Another porcine DMD model has been generated by zygotic injection of Cas9 messenger ribonucleic acid (mRNA) and a single guide RNA (sgRNA) that targets DMD at exon 27 (Yu et al., 2016). Analysis of the mosaic founder showed 70% and 60% of dystrophin alleles to be mutated in skeletal and smooth muscle, respectively. Although mutations in exon 27 are not found in humans, piglets carrying this deletion replicate the degeneration and disorganisation of cardiac and skeletal muscle seen in human patients, and also reduced thickness of intestine and stomach smooth muscle. However both founder animals died of unreported causes. The group tested 14 likely CRISPR/Cas9 off-target sites, but could not detect such activity. Noteworthy is the extremely high efficiency (50%) of DMD targeting in zygotes, which if repeatable means that further animals and a wider range of mutations should be easy to generate.

Future perspectives

Niels Bohr reportedly joked that ‘Prediction is difficult, especially about the future’. This is a valuable caution given recent rapid changes. Nevertheless, here we indicate those techniques that are likely to help generate porcine models of human disease and the technical issues that need to be solved in the coming years.

Multiple recombinase systems using Flp, Cre and Dre recombinases or PhiC31 integrase are very powerful tools in mice that can be used to effect sequential genetic modifications and to control the expression and inactivation of multiple genes (Schönhuber et al., 2014). Extension of such methods to pigs would be very useful, enabling, for example, the roles of particular genes in cancer pathogenesis to be studied in different tissues and over time. These techniques also allow the ablation of specific cell types to investigate their role in a disease process (Schönhuber et al., 2014).

While our discussion of CRISPR/Cas9 has concentrated on germline alterations, this system also provides a means of inducing precise somatic mutations in vivo. Local gene editing within a tumour entity could, for example, be used to replicate the accumulation of somatic mutations responsible for cancer progression. The larger body size of pigs offers an advantage over rodents, allowing access and delivery of CRISPR/Cas9 using standard human surgical and endoscopic methods. Delivery of Cas9 via size-limited AAVs will be aided significantly by the recently developed intein-mediated split-Cas9, and by the discovery of a smaller Cas9 orthologue from Campylobacter jejuni (Kim et al., 2017; Truong et al., 2015). The repertoire of gene editing tools and variants suited to particular tasks will no doubt increase and improve the refinement with which diseases can be modelled and studied.

Nuclear transfer is still the standard method of producing gene-targeted pigs, but the difficulty and inefficiency of the process pose serious limitations. Direct modification of early embryos is currently the most promising alternative, and ever more sophisticated modifications are likely to be possible. This, however, places the focus on the need for reliable in vitro production of pig embryos, and in particular on solving the long-standing problem of polyspermy with porcine IVF. A quite different means of modifying the mammalian germline was first demonstrated in rats more than two decades ago. Spermatogonial stem cells (SSCs) can be transferred between animals and produce viable sperm in the recipient (Brinster and Avarbock, 1994; Brinster and Zimmermann, 1994), opening the possibility of modifying SSCs in vitro then transferring them to recipient ‘founder’ males that transmit the modified genotype via their sperm, with no need for embryo manipulation.

Excitingly, this technology has recently been extended to pigs by Park et al. (2017), who demonstrated transplantation of SSCs into boars rendered germline deficient by homozygous knockout of NANOS2. This method does, however, require the development of conditions for long-term SSC culture and transfection in vitro. These methods are not yet available for pig, but have been established in mice (Kubota et al., 2004).

Leaving technology aside, there are also whole new areas of biomedicine yet to explore. For example, human viral diseases, such as hepatitis B and human immunodeficiency virus (HIV), have been modelled in mice through the introduction of transgenic viral receptors (Chisari and Oldstone, 1996; Hatziioannou and Evans, 2012; Koike et al., 1994; Yang et al., 2014), but these diseases have yet to be investigated in pigs. Another area with great potential is the study of pigs with a humanised immune system. Mice have been humanised by genetic ablation of lymphocytes and re-colonisation with human immune cells (Ito et al., 2014; Suzuki et al., 2016). Pigs engineered in this way could provide information about the response of the human immune system to cancers, infections and grafts, and also provide an orthotopic tumour xenograft model to study the therapeutic response of a patient's tumour, as demonstrated in mice (Hidalgo et al., 2014). Finally, somewhat controversial, but of potential biomedical value, are chimeric pigs that carry human organs (Wu et al., 2017). With proper safeguards and ethical approval, pigs carrying human organs, such as the liver, could provide a powerful preclinical tool to study drug pharmacokinetics and toxicity and could also provide a possible source of organs for transplantation.

Acknowledgements

We thank Bernhard Klinger for porcine oocyte culture.

Footnotes

  • Competing interests

    The authors declare no competing or financial interests.

  • Funding

    Financial support was provided by Deutsche Forschungsgemeinschaft (German Research Foundation) (SCHN 971/3-2), Dr. Mildred Scheel Stiftung für Krebsforschung (Mildred Schiel Foundation for Cancer Research) (111902), Studienstiftung des Deutschen Volkes (German Academic Scholarship Foundation) and European Cooperation in Science and Technology (BM1308).

  • © 2018. Published by The Company of Biologists Ltd

This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/3.0), which permits unrestricted use, distribution and reproduction in any medium provided that the original work is properly attributed.

References

  1. ↵
    1. Adam, S. J.,
    2. Rund, L. A.,
    3. Kuzmuk, K. N.,
    4. Zachary, J. F.,
    5. Schook, L. B. and
    6. Counter, C. M.
    (2007). Genetic induction of tumorigenesis in swine. Oncogene 26, 1038-1045. doi:10.1038/sj.onc.1209892
    OpenUrlCrossRefPubMed
  2. ↵
    1. Aigner, B.,
    2. Rathkolb, B.,
    3. Herbach, N.,
    4. Hrabe de Angelis, M.,
    5. Wanke, R. and
    6. & Wolf, E.
    (2008). Diabetes models by screen for hyperglycemia in phenotype-driven ENU mouse mutagenesis projects. Am. J. Physiol. Endocrinol. Metab. 294, E232-E240. doi:10.1152/ajpendo.00592.2007
    OpenUrlCrossRefPubMed
  3. ↵
    1. Al-Mashhadi, R. H.,
    2. Sorensen, C. B.,
    3. Kragh, P. M.,
    4. Christoffersen, C.,
    5. Mortensen, M. B.,
    6. Tolbod, L. P.,
    7. Thim, T.,
    8. Du, Y.,
    9. Li, J.,
    10. Liu, Y. et al
    . (2013). Familial hypercholesterolemia and atherosclerosis in cloned minipigs created by DNA transposition of a human PCSK9 gain-of-function mutant. Sci. Transl. Med. 5, 166ra1. doi:10.1126/scitranslmed.3004853
    OpenUrlAbstract/FREE Full Text
  4. ↵
    1. Al-Mashhadi, R. H.,
    2. Bjorklund, M. M.,
    3. Mortensen, M. B.,
    4. Christoffersen, C.,
    5. Larsen, T.,
    6. Falk, E. &
    7. Bentzon, J. F.
    (2015). Diabetes with poor glycaemic control does not promote atherosclerosis in genetically modified hypercholesterolaemic minipigs. Diabetologia 58, 1926-1936. doi:10.1007/s00125-015-3637-1
    OpenUrlCrossRefPubMed
  5. ↵
    1. Albuquerque, C.,
    2. Breukel, C.,
    3. van der Luijt, R.,
    4. Fidalgo, P.,
    5. Lage, P.,
    6. Slors, F. J.,
    7. Leitao, C. N.,
    8. Fodde, R. and
    9. Smits, R.
    (2002). The ‘just-right’ signaling model: APC somatic mutations are selected based on a specific level of activation of the beta-catenin signaling cascade. Hum. Mol. Genet. 11, 1549-1560. doi:10.1093/hmg/11.13.1549
    OpenUrlCrossRefPubMedWeb of Science
  6. ↵
    American Diabetes, A. (2013). Diagnosis and classification of diabetes mellitus. Diabetes Care 36 Suppl 1, S67-S74. doi:10.2337/dc13-S067
    OpenUrlFREE Full Text
  7. ↵
    1. Araki, E.,
    2. Nakamura, K.,
    3. Nakao, K.,
    4. Kameya, S.,
    5. Kobayashi, O.,
    6. Nonaka, I.,
    7. Kobayashi, T. and
    8. Katsuki, M.
    (1997). Targeted disruption of exon 52 in the mouse dystrophin gene induced muscle degeneration similar to that observed in Duchenne muscular dystrophy. Biochem. Biophys. Res. Commun. 238, 492-497. doi:10.1006/bbrc.1997.7328
    OpenUrlCrossRefPubMed
  8. ↵
    1. Baggio, L. L. and
    2. Drucker, D. J.
    (2007). Biology of incretins: GLP-1 and GIP. Gastroenterology 132, 2131-2157. doi:10.1053/j.gastro.2007.03.054
    OpenUrlCrossRefPubMedWeb of Science
  9. ↵
    1. Bähr, A. &
    2. Wolf, E.
    (2012). Domestic animal models for biomedical research. Reprod. Domest. Anim. 47 Suppl. 4, 59-71. doi:10.1111/j.1439-0531.2012.02056.x
    OpenUrlCrossRefPubMed
  10. ↵
    1. Ballard, C.,
    2. Gauthier, S.,
    3. Corbett, A.,
    4. Brayne, C.,
    5. Aarsland, D. and
    6. Jones, E.
    (2011). Alzheimer's disease. Lancet 377, 1019-1031. doi:10.1016/S0140-6736(10)61349-9
    OpenUrlCrossRefPubMedWeb of Science
  11. ↵
    1. Bentzon, J. F. and
    2. Falk, E.
    (2010). Atherosclerotic lesions in mouse and man: is it the same disease? Curr. Opin. Lipidol. 21, 434-440. doi:10.1097/MOL.0b013e32833ded6a
    OpenUrlCrossRefPubMed
  12. ↵
    1. Boehm, A. K.,
    2. Neff, J. R.,
    3. Squire, J. A.,
    4. Bayani, J.,
    5. Nelson, M. and
    6. Bridge, J. A.
    (2000). Cytogenetic findings in 36 osteosarcoma specimens and a review of the literature. Pediatr. Pathol. Mol. Med. 19, 359-376. doi:10.1080/15513810009168645
    OpenUrlCrossRef
  13. ↵
    1. Boyle, M. P. and
    2. De Boeck, K.
    (2013). A new era in the treatment of cystic fibrosis: correction of the underlying CFTR defect. Lancet Respir. Med. 1, 158-163. doi:10.1016/S2213-2600(12)70057-7
    OpenUrlCrossRef
  14. ↵
    1. Brinster, R. L. and
    2. Avarbock, M. R.
    (1994). Germline transmission of donor haplotype following spermatogonial transplantation. Proc. Natl. Acad. Sci. USA 91, 11303-11307. doi:10.1073/pnas.91.24.11303
    OpenUrlAbstract/FREE Full Text
  15. ↵
    1. Brinster, R. L. and
    2. Zimmermann, J. W.
    (1994). Spermatogenesis following male germ-cell transplantation. Proc. Natl. Acad. Sci. USA 91, 11298-11302. doi:10.1073/pnas.91.24.11298
    OpenUrlAbstract/FREE Full Text
  16. ↵
    1. Brinster, R. L.,
    2. Braun, R. E.,
    3. Lo, D.,
    4. Avarbock, M. R.,
    5. Oram, F. and
    6. Palmiter, R. D.
    (1989). Targeted correction of a major histocompatibility class II E alpha gene by DNA microinjected into mouse eggs. Proc. Natl. Acad. Sci. USA 86, 7087-7091. doi:10.1073/pnas.86.18.7087
    OpenUrlAbstract/FREE Full Text
  17. ↵
    1. Callesen, H.,
    2. Liu, Y.,
    3. Pedersen, H. S.,
    4. Li, R. and
    5. Schmidt, M.
    (2014). Increasing efficiency in production of cloned piglets. Cell Reprogram 16, 407-410. doi:10.1089/cell.2014.0053
    OpenUrlCrossRefPubMed
  18. ↵
    1. Carlson, D. F.,
    2. Tan, W.,
    3. Lillico, S. G.,
    4. Stverakova, D.,
    5. Proudfoot, C.,
    6. Christian, M.,
    7. Voytas, D. F.,
    8. Long, C. R.,
    9. Whitelaw, C. B. and
    10. Fahrenkrug, S. C.
    (2012). Efficient TALEN-mediated gene knockout in livestock. Proc. Natl. Acad. Sci. USA 109, 17382-17387. doi:10.1073/pnas.1211446109
    OpenUrlAbstract/FREE Full Text
  19. ↵
    1. Chisari, F. V. and
    2. Oldstone, M. B. A.
    (1996). Transgenic Models of Human Viral and Immunological Disease. Berlin, New York: Springer-Verlag.
  20. ↵
    1. Cho, S. W.,
    2. Kim, S.,
    3. Kim, Y.,
    4. Kweon, J.,
    5. Kim, H. S.,
    6. Bae, S. and
    7. Kim, J. S.
    (2013). Analysis of off-target effects of CRISPR/Cas-derived RNA-guided endonucleases and nickases. Genome Res. 24, 132-141. doi:10.1101/gr.162339.113
    OpenUrlAbstract/FREE Full Text
  21. ↵
    1. Chu, V. T.,
    2. Weber, T.,
    3. Graf, R.,
    4. Sommermann, T.,
    5. Petsch, K.,
    6. Sack, U.,
    7. Volchkov, P.,
    8. Rajewsky, K. &
    9. Kuhn, R.
    (2016). Efficient generation of Rosa26 knock-in mice using CRISPR/Cas9 in C57BL/6 zygotes. BMC Biotechnol. 16, 4. doi:10.1186/s12896-016-0234-4
    OpenUrlCrossRefPubMed
  22. ↵
    1. Citron, M.,
    2. Westaway, D.,
    3. Xia, W.,
    4. Carlson, G.,
    5. Diehl, T.,
    6. Levesque, G.,
    7. Johnson-Wood, K.,
    8. Lee, M.,
    9. Seubert, P.,
    10. Davis, A. et al.
    (1997). Mutant presenilins of Alzheimer's disease increase production of 42-residue amyloid beta-protein in both transfected cells and transgenic mice. Nat. Med. 3, 67-72. doi:10.1038/nm0197-67
    OpenUrlCrossRefPubMedWeb of Science
  23. ↵
    1. Clark, K. J.,
    2. Carlson, D. F.,
    3. Foster, L. K.,
    4. Kong, B. W.,
    5. Foster, D. N. and
    6. Fahrenkrug, S. C.
    (2007). Enzymatic engineering of the porcine genome with transposons and recombinases. BMC Biotechnol. 7, 42. doi:10.1186/1472-6750-7-42
    OpenUrlCrossRefPubMed
  24. ↵
    1. Cohn, J. S.,
    2. Patterson, B. W.,
    3. Uffelman, K. D.,
    4. Davignon, J. and
    5. Steiner, G.
    (2004). Rate of production of plasma and very-low-density lipoprotein (VLDL) apolipoprotein C-III is strongly related to the concentration and level of production of VLDL triglyceride in male subjects with different body weights and levels of insulin sensitivity. J. Clin. Endocrinol. Metab. 89, 3949-3955. doi:10.1210/jc.2003-032056
    OpenUrlCrossRefPubMedWeb of Science
  25. ↵
    1. Cooney, A. L.,
    2. Abou Alaiwa, M. H.,
    3. Shah, V. S.,
    4. Bouzek, D. C.,
    5. Stroik, M. R.,
    6. Powers, L. S.,
    7. Gansemer, N. D.,
    8. Meyerholz, D. K.,
    9. Welsh, M. J.,
    10. Stoltz, D. A. et al
    . (2016). Lentiviral-mediated phenotypic correction of cystic fibrosis pigs. JCI Insight 1, e88730. doi:10.1172/jci.insight.88730
    OpenUrlCrossRef
  26. ↵
    1. Croner, R. S.,
    2. Brueckl, W. M.,
    3. Reingruber, B.,
    4. Hohenberger, W. and
    5. Guenther, K.
    (2005). Age and manifestation related symptoms in familial adenomatous polyposis. BMC Cancer 5, 24. doi:10.1186/1471-2407-5-24
    OpenUrlCrossRefPubMed
  27. ↵
    1. Dai, Y.,
    2. Vaught, T. D.,
    3. Boone, J.,
    4. Chen, S. H.,
    5. Phelps, C. J.,
    6. Ball, S.,
    7. Monahan, J. A.,
    8. Jobst, P. M.,
    9. McCreath, K. J.,
    10. Lamborn, A. E. et al
    . (2002). Targeted disruption of the alpha1,3-galactosyltransferase gene in cloned pigs. Nat. Biotechnol. 20, 251-255. doi:10.1038/nbt0302-251
    OpenUrlCrossRefPubMedWeb of Science
  28. ↵
    1. Davis, B. T.,
    2. Wang, X. J.,
    3. Rohret, J. A.,
    4. Struzynski, J. T.,
    5. Merricks, E. P.,
    6. Bellinger, D. A.,
    7. Rohret, F. A.,
    8. Nichols, T. C. &
    9. Rogers, C. S.
    (2014). Targeted disruption of LDLR causes hypercholesterolemia and atherosclerosis in Yucatan miniature pigs. PLoS ONE 9, e93457. doi:10.1371/journal.pone.0093457
    OpenUrlCrossRefPubMed
  29. ↵
    1. De Strooper, B.,
    2. Saftig, P.,
    3. Craessaerts, K.,
    4. Vanderstichele, H.,
    5. Guhde, G.,
    6. Annaert, W.,
    7. Von Figura, K. and
    8. Van Leuven, F
    . (1998). Deficiency of presenilin-1 inhibits the normal cleavage of amyloid precursor protein. Nature 391, 387-390. doi:10.1038/34910
    OpenUrlCrossRefPubMedWeb of Science
  30. ↵
    1. Dickerson, J. W. T. and
    2. Dobbing, J.
    (1967). Prenatal and postnatal growth and development of the central nervous system of the pig. Proc. R. Soc. Lond. B Biol. Sci. 166, 384-395. doi:10.1098/rspb.1967.0002
    OpenUrlAbstract/FREE Full Text
  31. ↵
    1. Durfee, R. A.,
    2. Mohammed, M. and
    3. Luu, H. H.
    (2016). Review of osteosarcoma and current management. Rheumatol. Ther. 3, 221-243. doi:10.1007/s40744-016-0046-y
    OpenUrlCrossRef
  32. ↵
    1. Evans, M. J. and
    2. Kaufman, M. H.
    (1981). Establishment in culture of pluripotential cells from mouse embryos. Nature 292, 154-156. doi:10.1038/292154a0
    OpenUrlCrossRefPubMedWeb of Science
  33. ↵
    1. Falk, E.
    (2006). Pathogenesis of atherosclerosis. J. Am. Coll. Cardiol. 47 Suppl. 8, C7-C12. doi:10.1016/j.jacc.2005.09.068
    OpenUrlFREE Full Text
  34. ↵
    1. Fearnhead, N. S.,
    2. Britton, M. P. and
    3. Bodmer, W. F.
    (2001). The ABC of APC. Hum. Mol. Genet. 10, 721-733. doi:10.1093/hmg/10.7.721
    OpenUrlCrossRefPubMedWeb of Science
  35. ↵
    1. Fearon, E. R.
    (2011). Molecular genetics of colorectal cancer. Annu. Rev. Pathol. 6, 479-507. doi:10.1146/annurev-pathol-011110-130235
    OpenUrlCrossRefPubMed
  36. ↵
    1. Ferlay, J.,
    2. Soerjomataram, I.,
    3. Dikshit, R.,
    4. Eser, S.,
    5. Mathers, C.,
    6. Rebelo, M.,
    7. Parkin, D. M.,
    8. Forman, D. and
    9. Bray, F.
    (2015). Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int. J. Cancer 136, E359-E386. doi:10.1002/ijc.29210
    OpenUrlCrossRefPubMed
  37. ↵
    1. Flisikowska, T.,
    2. Thorey, I. S.,
    3. Offner, S.,
    4. Ros, F.,
    5. Lifke, V.,
    6. Zeitler, B.,
    7. Rottmann, O.,
    8. Vincent, A.,
    9. Zhang, L.,
    10. Jenkins, S. et al
    . (2011). Efficient immunoglobulin gene disruption and targeted replacement in rabbit using zinc finger nucleases. PLoS ONE 6, e21045. doi:10.1371/journal.pone.0021045
    OpenUrlCrossRefPubMed
  38. ↵
    1. Flisikowska, T.,
    2. Merkl, C.,
    3. Landmann, M.,
    4. Eser, S.,
    5. Rezaei, N.,
    6. Cui, X.,
    7. Kurome, M.,
    8. Zakhartchenko, V.,
    9. Kessler, B.,
    10. Wieland, H. et al
    . (2012). A porcine model of familial adenomatous polyposis. Gastroenterology 143, 1173-1175.e1-7. doi:10.1053/j.gastro.2012.07.110
    OpenUrlCrossRefPubMed
  39. ↵
    1. Flisikowska, T.,
    2. Stachowiak, M.,
    3. Xu, H.,
    4. Wagner, A.,
    5. Hernandez-Caceres, A.,
    6. Wurmser, C.,
    7. Perleberg, C.,
    8. Pausch, H.,
    9. Perkowska, A.,
    10. Fischer, K. et al
    . (2017). Porcine familial adenomatous polyposis model enables systematic analysis of early events in adenoma progression. Sci. Rep. 7, 6613. doi:10.1038/s41598-017-06741-8
    OpenUrlCrossRef
  40. ↵
    1. Fröhlich, T.,
    2. Kemter, E.,
    3. Flenkenthaler, F.,
    4. Klymiuk, N.,
    5. Otte, K. A.,
    6. Blutke, A.,
    7. Krause, S.,
    8. Walter, M. C.,
    9. Wanke, R.,
    10. Wolf, E. et al
    . (2016). Progressive muscle proteome changes in a clinically relevant pig model of Duchenne muscular dystrophy. Sci. Rep. 6, 33362. doi:10.1038/srep33362
    OpenUrlCrossRef
  41. ↵
    1. Fu, Y.,
    2. Sander, J. D.,
    3. Reyon, D.,
    4. Cascio, V. M. and
    5. Joung, J. K.
    (2014). Improving CRISPR-Cas nuclease specificity using truncated guide RNAs. Nat. Biotechnol. 32, 279-284. doi:10.1038/nbt.2808
    OpenUrlCrossRefPubMed
  42. ↵
    1. Gadsby, D. C.,
    2. Vergani, P. &
    3. Csanády, L.
    (2006). The ABC protein turned chloride channel whose failure causes cystic fibrosis. Nature 440, 477-483. doi:10.1038/nature04712
    OpenUrlCrossRefPubMedWeb of Science
  43. ↵
    1. Games, D.,
    2. Adams, D.,
    3. Alessandrini, R.,
    4. Barbour, R.,
    5. Berthelette, P.,
    6. Blackwell, C.,
    7. Carr, T.,
    8. Clemens, J.,
    9. Donaldson, T.,
    10. Gillespie, F. et al.
    (1995). Alzheimer-type neuropathology in transgenic mice overexpressing V717F beta-amyloid precursor protein. Nature 373, 523-527. doi:10.1038/373523a0
    OpenUrlCrossRefPubMedWeb of Science
  44. ↵
    1. Garrels, W.,
    2. Mátés, L.,
    3. Holler, S.,
    4. Dalda, A.,
    5. Taylor, U.,
    6. Petersen, B.,
    7. Niemann, H.,
    8. Izsvak, Z.,
    9. Ivics, Z. &
    10. Kues, W. A.
    (2011). Germline transgenic pigs by Sleeping Beauty transposition in porcine zygotes and targeted integration in the pig genome. PLoS ONE 6, e23573. doi:10.1371/journal.pone.0023573
    OpenUrlCrossRefPubMed
  45. ↵
    1. Glauser, E. M.
    (1966). Advantages of piglets as experimental animals in pediatric research. Exp. Med. Surg. 24, 181-190.
    OpenUrlPubMedWeb of Science
  46. ↵
    1. Goemans, N. M.,
    2. Tulinius, M.,
    3. van den Akker, J. T.,
    4. Burm, B. E.,
    5. Ekhart, P. F.,
    6. Heuvelmans, N.,
    7. Holling, T.,
    8. Janson, A. A.,
    9. Platenburg, G. J.,
    10. Sipkens, J. A. et al.
    (2011). Systemic administration of PRO051 in Duchenne's muscular dystrophy. N. Engl. J. Med. 364, 1513-1522. doi:10.1056/NEJMoa1011367
    OpenUrlCrossRefPubMedWeb of Science
  47. ↵
    1. Groenen, M. A.,
    2. Archibald, A. L.,
    3. Uenishi, H.,
    4. Tuggle, C. K.,
    5. Takeuchi, Y.,
    6. Rothschild, M. F.,
    7. Rogel-Gaillard, C.,
    8. Park, C.,
    9. Milan, D.,
    10. Megens, H. J. et al.
    (2012). Analyses of pig genomes provide insight into porcine demography and evolution. Nature 491, 393-398. doi:10.1038/nature11622
    OpenUrlCrossRefPubMedWeb of Science
  48. ↵
    1. Guijarro, M. V.,
    2. Ghivizzani, S. C. and
    3. Gibbs, C. P.
    (2014). Animal models in osteosarcoma. Front Oncol. 4, 189. doi:10.3389/fonc.2014.00189
    OpenUrlCrossRef
  49. ↵
    1. Hai, T.,
    2. Teng, F.,
    3. Guo, R.,
    4. Li, W. and
    5. Zhou, Q.
    (2014). One-step generation of knockout pigs by zygote injection of CRISPR/Cas system. Cell Res. 24, 372-375. doi:10.1038/cr.2014.11
    OpenUrlCrossRefPubMedWeb of Science
  50. ↵
    1. Hammer, R. E.,
    2. Pursel, V. G.,
    3. Rexroad, C. E., Jr..,
    4. Wall, R. J.,
    5. Bolt, D. J.,
    6. Ebert, K. M.,
    7. Palmiter, R. D. and
    8. Brinster, R. L.
    (1985). Production of transgenic rabbits, sheep and pigs by microinjection. Nature 315, 680-683. doi:10.1038/315680a0
    OpenUrlCrossRefPubMed
  51. ↵
    1. Hara, S.,
    2. Umeyama, K.,
    3. Yokoo, T.,
    4. Nagashima, H. &
    5. Nagata, M.
    (2014). Diffuse glomerular nodular lesions in diabetic pigs carrying a dominant-negative mutant hepatocyte nuclear factor 1-alpha, an inheritant diabetic gene in humans. PLoS ONE 9, e92219. doi:10.1371/journal.pone.0092219
    OpenUrlCrossRef
  52. ↵
    1. Hatziioannou, T. and
    2. Evans, D. T.
    (2012). Animal models for HIV/AIDS research. Nat. Rev. Microbiol. 10, 852-867. doi:10.1038/nrmicro2911
    OpenUrlCrossRefPubMed
  53. ↵
    1. Hauschild, J.,
    2. Petersen, B.,
    3. Santiago, Y.,
    4. Queisser, A.-L.,
    5. Carnwath, J. W.,
    6. Lucas-Hahn, A.,
    7. Zhang, L.,
    8. Meng, X.,
    9. Gregory, P. D.,
    10. Schwinzer, R. et al.
    (2011). Efficient generation of a biallelic knockout in pigs using zinc-finger nucleases. Proc. Natl. Acad. Sci. USA 108, 12013-12017. doi:10.1073/pnas.1106422108
    OpenUrlAbstract/FREE Full Text
  54. ↵
    1. Heemskerk, H.,
    2. de Winter, C.,
    3. van Kuik, P.,
    4. Heuvelmans, N.,
    5. Sabatelli, P.,
    6. Rimessi, P.,
    7. Braghetta, P.,
    8. van Ommen, G. B.,
    9. de Kimpe, S.,
    10. Ferlini, A. et al.
    (2010). Preclinical PK and PD Studies on 2’-O-Methyl-phosphorothioate RNA Antisense Oligonucleotides in the mdx Mouse Model. Mol. Ther. 18, 1210-1217. doi:10.1038/mt.2010.72
    OpenUrlCrossRefPubMed
  55. ↵
    1. Heinritz, S. N.,
    2. Mosenthin, R. and
    3. Weiss, E.
    (2013). Use of pigs as a potential model for research into dietary modulation of the human gut microbiota. Nutr. Res. Rev. 26, 191-209. doi:10.1017/S0954422413000152
    OpenUrlCrossRefPubMed
  56. ↵
    1. Hidalgo, M.,
    2. Amant, F.,
    3. Biankin, A. V.,
    4. Budinska, E.,
    5. Byrne, A. T.,
    6. Caldas, C.,
    7. Clarke, R. B.,
    8. de Jong, S.,
    9. Jonkers, J.,
    10. Maelandsmo, G. M. et al.
    (2014). Patient-derived xenograft models: an emerging platform for translational cancer research. Cancer Discov. 4, 998-1013. doi:10.1158/2159-8290.CD-14-0001
    OpenUrlAbstract/FREE Full Text
  57. ↵
    1. Hofmann, A.,
    2. Kessler, B.,
    3. Ewerling, S.,
    4. Weppert, M.,
    5. Vogg, B.,
    6. Ludwig, H.,
    7. Stojkovic, M.,
    8. Boelhauve, M.,
    9. Brem, G.,
    10. Wolf, E. et al.
    (2003). Efficient transgenesis in farm animals by lentiviral vectors. EMBO Rep, 4, 1054-1060. doi:10.1038/sj.embor.7400007
    OpenUrlAbstract/FREE Full Text
  58. ↵
    1. Hsiao, K.,
    2. Chapman, P.,
    3. Nilsen, S.,
    4. Eckman, C.,
    5. Harigaya, Y.,
    6. Younkin, S.,
    7. Yang, F. and
    8. Cole, G.
    (1996). Correlative memory deficits, Abeta elevation, and amyloid plaques in transgenic mice. Science 274, 99-102. doi:10.1126/science.274.5284.99
    OpenUrlAbstract/FREE Full Text
  59. ↵
    1. Hung, K. E.,
    2. Maricevich, M. A.,
    3. Richard, L. G.,
    4. Chen, W. Y.,
    5. Richardson, M. P.,
    6. Kunin, A.,
    7. Bronson, R. T.,
    8. Mahmood, U. and
    9. Kucherlapati, R.
    (2010). Development of a mouse model for sporadic and metastatic colon tumors and its use in assessing drug treatment. Proc. Natl. Acad. Sci. USA 107, 1565-1570. doi:10.1073/pnas.0908682107
    OpenUrlAbstract/FREE Full Text
  60. ↵
    1. Hutchinson, L. and
    2. Kirk, R.
    (2011). High drug attrition rates--where are we going wrong? Nat. Rev. Clin. Oncol. 8, 189-190. doi:10.1038/nrclinonc.2011.34
    OpenUrlCrossRefPubMed
  61. ↵
    1. Ishibashi, S.,
    2. Brown, M. S.,
    3. Goldstein, J. L.,
    4. Gerard, R. D.,
    5. Hammer, R. E. and
    6. Herz, J.
    (1993). Hypercholesterolemia in low density lipoprotein receptor knockout mice and its reversal by adenovirus-mediated gene delivery. J. Clin. Invest. 92, 883-893. doi:10.1172/JCI116663
    OpenUrlCrossRefPubMedWeb of Science
  62. ↵
    1. Ito, T.,
    2. Sendai, Y.,
    3. Yamazaki, S.,
    4. Seki-Soma, M.,
    5. Hirose, K.,
    6. Watanabe, M.,
    7. Fukawa, K. and
    8. Nakauchi, H.
    (2014). Generation of recombination activating gene-1-deficient neonatal piglets: a model of T and B cell deficient severe combined immune deficiency. PLoS ONE 9, e113833. doi:10.1371/journal.pone.0113833
    OpenUrlCrossRef
  63. ↵
    1. Ivics, Z.,
    2. Garrels, W.,
    3. Mátés, L.,
    4. Yau, T. Y.,
    5. Bashir, S.,
    6. Zidek, V.,
    7. Landa, V.,
    8. Geurts, A.,
    9. Pravenec, M.,
    10. Rulicke, T. et al.
    (2014). Germline transgenesis in pigs by cytoplasmic microinjection of Sleeping Beauty transposons. Nat. Protoc. 9, 810-827. doi:10.1038/nprot.2014.010
    OpenUrlCrossRef
  64. ↵
    1. Jacks, T.,
    2. Remington, L.,
    3. Williams, B. O.,
    4. Schmitt, E. M.,
    5. Halachmi, S.,
    6. Bronson, R. T. and
    7. Weinberg, R. A.
    (1994). Tumor spectrum analysis in p53-mutant mice. Curr. Biol. 4, 1-7. doi:10.1016/S0960-9822(00)00002-6
    OpenUrlCrossRefPubMedWeb of Science
  65. ↵
    1. Jakobsen, J. E.,
    2. Johansen, M. G.,
    3. Schmidt, M.,
    4. Dagnaes-Hansen, F.,
    5. Dam, K.,
    6. Gunnarsson, A.,
    7. Liu, Y.,
    8. Kragh, P. M.,
    9. Li, R.,
    10. Holm, I. E. et al.
    (2013). Generation of minipigs with targeted transgene insertion by recombinase-mediated cassette exchange (RMCE) and somatic cell nuclear transfer (SCNT). Transgenic Res. 22, 709-723. doi:10.1007/s11248-012-9671-6
    OpenUrlCrossRef
  66. ↵
    1. Jakobsen, J. E.,
    2. Johansen, M. G.,
    3. Schmidt, M.,
    4. Liu, Y.,
    5. Li, R.,
    6. Callesen, H.,
    7. Melnikova, M.,
    8. Habekost, M.,
    9. Matrone, C.,
    10. Bouter, Y. et al.
    (2016). Expression of the Alzheimer's disease mutations AbetaPP695sw and PSEN1M146I in double-transgenic gottingen minipigs. J. Alzheimers Dis. 53, 1617-1630. doi:10.3233/JAD-160408
    OpenUrlCrossRef
  67. ↵
    1. Jin, Y.-X.,
    2. Jeon, Y.,
    3. Lee, S.-H.,
    4. Kwon, M.-S.,
    5. Kim, T.,
    6. Cui, X.-S.,
    7. Hyun, S.-H. &
    8. Kim, N.-H.
    (2014). Production of pigs expressing a transgene under the control of a tetracycline-inducible system. PLoS One 9, e86146. doi:10.1371/journal.pone.0086146
    OpenUrlCrossRef
  68. ↵
    1. Justice, M. J. and
    2. Dhillon, P.
    (2016). Using the mouse to model human disease: increasing validity and reproducibility. Dis. Model Mech. 9, 101-103. doi:10.1242/dmm.024547
    OpenUrlAbstract/FREE Full Text
  69. ↵
    1. Kansara, M.,
    2. Teng, M. W.,
    3. Smyth, M. J. and
    4. Thomas, D. M.
    (2014). Translational biology of osteosarcoma. Nat. Rev. Cancer 14, 722-735. doi:10.1038/nrc3838
    OpenUrlCrossRefPubMed
  70. ↵
    1. Kararli, T. T.
    (1995). Comparison of the gastrointestinal anatomy, physiology, and biochemistry of humans and commonly used laboratory animals. Biopharm. Drug. Dispos. 16, 351-380. doi:10.1002/bdd.2510160502
    OpenUrlCrossRefPubMedWeb of Science
  71. ↵
    1. Karim, B. O. and
    2. Huso, D. L.
    (2013). Mouse models for colorectal cancer. Am. J. Cancer Res. 3, 240-250.
    OpenUrlPubMedWeb of Science
  72. ↵
    1. Kendall, S. D.,
    2. Linardic, C. M.,
    3. Adam, S. J. and
    4. Counter, C. M.
    (2005). A network of genetic events sufficient to convert normal human cells to a tumorigenic state. Cancer Res. 65, 9824-9828. doi:10.1158/0008-5472.CAN-05-1543
    OpenUrlAbstract/FREE Full Text
  73. ↵
    1. Kikuchi, K.,
    2. Ekwall, H.,
    3. Tienthai, P.,
    4. Kawai, Y.,
    5. Noguchi, J.,
    6. Kaneko, H. and
    7. Rodriguez-Martinez, H.
    (2002a). Morphological features of lipid droplet transition during porcine oocyte fertilisation and early embryonic development to blastocyst in vivo and in vitro. Zygote 10, 355-366. doi:10.1017/S0967199402004100
    OpenUrlCrossRefPubMedWeb of Science
  74. ↵
    1. Kikuchi, K.,
    2. Onishi, A.,
    3. Kashiwazaki, N.,
    4. Iwamoto, M.,
    5. Noguchi, J.,
    6. Kaneko, H.,
    7. Akita, T. and
    8. Nagai, T.
    (2002b). Successful piglet production after transfer of blastocysts produced by a modified in vitro system. Biol. Reprod. 66, 1033-1041. doi:10.1095/biolreprod66.4.1033
    OpenUrlAbstract/FREE Full Text
  75. ↵
    1. Kim, S. J.,
    2. Nian, C.,
    3. Widenmaier, S. and
    4. McIntosh, C. H.
    (2008). Glucose-dependent insulinotropic polypeptide-mediated up-regulation of beta-cell antiapoptotic Bcl-2 gene expression is coordinated by cyclic AMP (cAMP) response element binding protein (CREB) and cAMP-responsive CREB coactivator 2. Mol. Cell Biol. 28, 1644-1656. doi:10.1128/MCB.00325-07
    OpenUrlAbstract/FREE Full Text
  76. ↵
    1. Kim, E.,
    2. Koo, T.,
    3. Park, S. W.,
    4. Kim, D.,
    5. Kim, K.,
    6. Cho, H. Y.,
    7. Song, D. W.,
    8. Lee, K. J.,
    9. Jung, M. H.,
    10. Kim, S. et al
    . (2017). In vivo genome editing with a small Cas9 orthologue derived from Campylobacter jejuni. Nat. Commun. 8, 14500. doi:10.1038/ncomms14500
    OpenUrlCrossRef
  77. ↵
    1. King, A. J.
    (2012). The use of animal models in diabetes research. Br. J. Pharmacol. 166, 877-894. doi:10.1111/j.1476-5381.2012.01911.x
    OpenUrlCrossRefPubMedWeb of Science
  78. ↵
    1. Kinzler, K. W.,
    2. Nilbert, M. C.,
    3. Su, L. K.,
    4. Vogelstein, B.,
    5. Bryan, T. M.,
    6. Levy, D. B.,
    7. Smith, K. J.,
    8. Preisinger, A. C.,
    9. Hedge, P.,
    10. McKechnie, D. et al.
    (1991). Identification of FAP locus genes from chromosome 5q21. Science. 253, 661-665. doi:10.1126/science.1651562
    OpenUrlAbstract/FREE Full Text
  79. ↵
    1. Kitaji, H.,
    2. Ookutsu, S.,
    3. Sato, M. and
    4. Miyoshi, K.
    (2015). A new rolling culture-based in vitro fertilization system capable of reducing polyspermy in porcine oocytes. Anim. Sci. J. 86, 494-498. doi:10.1111/asj.12327
    OpenUrlCrossRef
  80. ↵
    1. Klymiuk, N.,
    2. Bocker, W.,
    3. Schonitzer, V.,
    4. Bahr, A.,
    5. Radic, T.,
    6. Frohlich, T.,
    7. Wunsch, A.,
    8. Kessler, B.,
    9. Kurome, M.,
    10. Schilling, E. et al.
    (2012a). First inducible transgene expression in porcine large animal models. FASEB J. 26, 1086-1099. doi:10.1096/fj.11-185041
    OpenUrlAbstract/FREE Full Text
  81. ↵
    1. Klymiuk, N.,
    2. Mundhenk, L.,
    3. Kraehe, K.,
    4. Wuensch, A.,
    5. Plog, S.,
    6. Emrich, D.,
    7. Langenmayer, M. C.,
    8. Stehr, M.,
    9. Holzinger, A.,
    10. Kroner, C. et al.
    (2012b). Sequential targeting of CFTR by BAC vectors generates a novel pig model of cystic fibrosis. J. Mol. Med. (Berl). 90, 597-608. doi:10.1007/s00109-011-0839-y
    OpenUrlCrossRefPubMed
  82. ↵
    1. Klymiuk, N.,
    2. Blutke, A.,
    3. Graf, A.,
    4. Krause, S.,
    5. Burkhardt, K.,
    6. Wuensch, A.,
    7. Krebs, S.,
    8. Kessler, B.,
    9. Zakhartchenko, V.,
    10. Kurome, M. et al.
    (2013). Dystrophin-deficient pigs provide new insights into the hierarchy of physiological derangements of dystrophic muscle. Hum. Mol. Genet. 22, 4368-4382. doi:10.1093/hmg/ddt287
    OpenUrlCrossRefPubMedWeb of Science
  83. ↵
    1. Koenig, M.,
    2. Beggs, A. H.,
    3. Moyer, M.,
    4. Scherpf, S.,
    5. Heindrich, K.,
    6. Bettecken, T.,
    7. Meng, G.,
    8. Muller, C. R.,
    9. Lindlof, M.,
    10. Kaariainen, H. et al.
    (1989). The molecular basis for Duchenne versus Becker muscular dystrophy: correlation of severity with type of deletion. Am. J. Hum. Genet. 45, 498-506.
    OpenUrlPubMedWeb of Science
  84. ↵
    1. Koike, K.,
    2. Moriya, K.,
    3. Iino, S.,
    4. Yotsuyanagi, H.,
    5. Endo, Y.,
    6. Miyamura, T. and
    7. Kurokawa, K.
    (1994). High-level expression of hepatitis B virus HBx gene and hepatocarcinogenesis in transgenic mice. Hepatology 19, 810-819. doi:10.1002/hep.1840190403
    OpenUrlCrossRefPubMedWeb of Science
  85. ↵
    1. Kragh, P. M.,
    2. Nielsen, A. L.,
    3. Li, J.,
    4. Du, Y.,
    5. Lin, L.,
    6. Schmidt, M.,
    7. Bogh, I. B.,
    8. Holm, I. E.,
    9. Jakobsen, J. E.,
    10. Johansen, M. G. et al.
    (2009). Hemizygous minipigs produced by random gene insertion and handmade cloning express the Alzheimer's disease-causing dominant mutation APPsw. Transgenic Res. 18, 545-558. doi:10.1007/s11248-009-9245-4
    OpenUrlCrossRefPubMedWeb of Science
  86. ↵
    1. Kubota, H.,
    2. Avarbock, M. R. and
    3. Brinster, R. L.
    (2004). Growth factors essential for self-renewal and expansion of mouse spermatogonial stem cells. Proc. Natl. Acad. Sci. USA 101, 16489-16494. doi:10.1073/pnas.0407063101
    OpenUrlAbstract/FREE Full Text
  87. ↵
    1. Kurome, M.,
    2. Geistlinger, L.,
    3. Kessler, B.,
    4. Zakhartchenko, V.,
    5. Klymiuk, N.,
    6. Wuensch, A.,
    7. Richter, A.,
    8. Baehr, A.,
    9. Kraehe, K.,
    10. Burkhardt, K. et al
    . (2013). Factors influencing the efficiency of generating genetically engineered pigs by nuclear transfer: multi-factorial analysis of a large data set. BMC Biotechnol. 13, 43. doi:10.1186/1472-6750-13-43
    OpenUrlCrossRefPubMed
  88. ↵
    1. Kwon, D.-N.,
    2. Lee, K.,
    3. Kang, M.-J.,
    4. Choi, Y.-J.,
    5. Park, C.,
    6. Whyte, J. J.,
    7. Brown, A. N.,
    8. Kim, J. H.,
    9. Samuel, M.,
    10. Mao, J. et al
    . (2013). Production of biallelic CMP-Neu5Ac hydroxylase knock-out pigs. Sci. Rep. 3, 1981. doi:10.1038/srep01981
    OpenUrlCrossRefPubMed
  89. ↵
    1. LaFerla, F. M. and
    2. Green, K. N.
    (2012). Animal models of Alzheimer disease. Cold Spring Harb. Perspect. Med. 2, a006320. doi:10.1101/cshperspect.a006320
    OpenUrlAbstract/FREE Full Text
  90. ↵
    1. Lai, L.,
    2. Kolber-Simonds, D.,
    3. Park, K. W.,
    4. Cheong, H. T.,
    5. Greenstein, J. L.,
    6. Im, G. S.,
    7. Samuel, M.,
    8. Bonk, A.,
    9. Rieke, A.,
    10. Day, B. N. et al.
    (2002). Production of alpha-1,3-galactosyltransferase knockout pigs by nuclear transfer cloning. Science 295, 1089-1092. doi:10.1126/science.1068228
    OpenUrlAbstract/FREE Full Text
  91. ↵
    1. Ledford, H.
    (2011). Translational research: 4 ways to fix the clinical trial. Nature 477, 526-528. doi:10.1038/477526a
    OpenUrlCrossRefPubMedWeb of Science
  92. ↵
    1. Leuchs, S.,
    2. Saalfrank, A.,
    3. Merkl, C.,
    4. Flisikowska, T.,
    5. Edlinger, M.,
    6. Durkovic, M.,
    7. Rezaei, N.,
    8. Kurome, M.,
    9. Zakhartchenko, V.,
    10. Kessler, B. et al
    . (2012). Inactivation and inducible oncogenic mutation of p53 in gene targeted pigs. PLoS ONE 7, e43323. doi:10.1371/journal.pone.0043323
    OpenUrlCrossRefPubMed
  93. ↵
    1. Levine, A. J. and
    2. Oren, M.
    (2009). The first 30 years of p53: growing ever more complex. Nat. Rev. Cancer 9, 749-758. doi:10.1038/nrc2723
    OpenUrlCrossRefPubMedWeb of Science
  94. ↵
    1. Li, S.,
    2. Flisikowska, T.,
    3. Kurome, M.,
    4. Zakhartchenko, V.,
    5. Kessler, B.,
    6. Saur, D.,
    7. Kind, A.,
    8. Wolf, E.,
    9. Flisikowski, K. &
    10. Schnieke, A.
    (2014). Dual fluorescent reporter pig for Cre recombination: transgene placement at the ROSA26 locus. PLoS ONE 9, e102455. doi:10.1371/journal.pone.0102455
    OpenUrlCrossRef
  95. ↵
    1. Li, S.,
    2. Edlinger, M.,
    3. Saalfrank, A.,
    4. Flisikowski, K.,
    5. Tschukes, A.,
    6. Kurome, M.,
    7. Zakhartchenko, V.,
    8. Kessler, B.,
    9. Saur, D.,
    10. Kind, A. et al
    . (2015). Viable pigs with a conditionally-activated oncogenic KRAS mutation. Transgenic Res. 24, 509-517. doi:10.1007/s11248-015-9866-8
    OpenUrlCrossRef
  96. ↵
    1. Lillico, S. G.,
    2. Proudfoot, C.,
    3. Carlson, D. F.,
    4. Stverakova, D.,
    5. Neil, C.,
    6. Blain, C.,
    7. King, T. J.,
    8. Ritchie, W. A.,
    9. Tan, W.,
    10. Mileham, A. J. et al
    . (2013). Live pigs produced from genome edited zygotes. Sci. Rep. 3, 2847. doi:10.1038/srep02847
    OpenUrlCrossRefPubMed
  97. ↵
    1. Logan, J. S. and
    2. Martin, M. J.
    (1994). Transgenic swine as a recombinant production system for human hemoglobin. Methods Enzymol. 231, 435-445. doi:10.1016/0076-6879(94)31029-7
    OpenUrlCrossRefPubMed
  98. ↵
    1. Luo, Y.,
    2. Li, J.,
    3. Liu, Y.,
    4. Lin, L.,
    5. Du, Y.,
    6. Li, S.,
    7. Yang, H.,
    8. Vajta, G.,
    9. Callesen, H.,
    10. Bolund, L. et al.
    (2011). High efficiency of BRCA1 knockout using rAAV-mediated gene targeting: developing a pig model for breast cancer. Transgenic Res. 20, 975-988. doi:10.1007/s11248-010-9472-8
    OpenUrlCrossRefPubMed
  99. ↵
    1. Luo, W.,
    2. Li, Z.,
    3. Huang, Y.,
    4. Han, Y.,
    5. Yao, C.,
    6. Duan, X.,
    7. Ouyang, H. and
    8. Li, L.
    (2014). Generation of AQP2-Cre transgenic mini-pigs specifically expressing Cre recombinase in kidney collecting duct cells. Transgenic Res. 23, 365-375. doi:10.1007/s11248-013-9774-8
    OpenUrlCrossRef
  100. ↵
    1. Lusis, A. J.
    (2000). Atherosclerosis. Nature 407, 233-241. doi:10.1038/35025203
    OpenUrlCrossRefPubMedWeb of Science
  101. ↵
    1. Mak, I. W.,
    2. Evaniew, N. and
    3. Ghert, M.
    (2014). Lost in translation: animal models and clinical trials in cancer treatment. Am. J. Transl. Res. 6, 114-118.
    OpenUrlPubMed
  102. ↵
    1. Mali, P.,
    2. Yang, L.,
    3. Esvelt, K. M.,
    4. Aach, J.,
    5. Guell, M.,
    6. DiCarlo, J. E.,
    7. Norville, J. E. and
    8. Church, G. M.
    (2013). RNA-guided human genome engineering via Cas9. Science 339, 823-826. doi:10.1126/science.1232033
    OpenUrlAbstract/FREE Full Text
  103. ↵
    1. Maxwell, K. N. and
    2. Breslow, J. L.
    (2004). Adenoviral-mediated expression of Pcsk9 in mice results in a low-density lipoprotein receptor knockout phenotype. Proc. Natl. Acad. Sci. USA 101, 7100-7105. doi:10.1073/pnas.0402133101
    OpenUrlAbstract/FREE Full Text
  104. ↵
    1. McCreath, K. J.,
    2. Howcroft, J.,
    3. Campbell, K. H. S.,
    4. Colman, A.,
    5. Schnieke, A. E. and
    6. Kind, A. J.
    (2000). Production of gene-targeted sheep by nuclear transfer from cultured somatic cells. Nature 405, 1066-1069. doi:10.1038/35016604
    OpenUrlCrossRefPubMedWeb of Science
  105. ↵
    1. McGreevy, J. W.,
    2. Hakim, C. H.,
    3. McIntosh, M. A. and
    4. Duan, D.
    (2015). Animal models of Duchenne muscular dystrophy: from basic mechanisms to gene therapy. Dis. Model. Mech. 8, 195-213. doi:10.1242/dmm.018424
    OpenUrlAbstract/FREE Full Text
  106. ↵
    1. Mendell, J. R.,
    2. Shilling, C.,
    3. Leslie, N. D.,
    4. Flanigan, K. M.,
    5. al-Dahhak, R.,
    6. Gastier-Foster, J.,
    7. Kneile, K.,
    8. Dunn, D. M.,
    9. Duval, B.,
    10. Aoyagi, A. et al.
    (2012). Evidence-based path to newborn screening for Duchenne muscular dystrophy. Ann. Neurol. 71, 304-313. doi:10.1002/ana.23528
    OpenUrlCrossRefPubMed
  107. ↵
    1. Meyer, M.,
    2. de Angelis, M. H.,
    3. Wurst, W. and
    4. Kuhn, R.
    (2010). Gene targeting by homologous recombination in mouse zygotes mediated by zinc-finger nucleases. Proc. Natl. Acad. Sci. USA 107, 15022-15026. doi:10.1073/pnas.1009424107
    OpenUrlAbstract/FREE Full Text
  108. ↵
    1. Meyerholz, D. K.,
    2. Stoltz, D. A.,
    3. Namati, E.,
    4. Ramachandran, S.,
    5. Pezzulo, A. A.,
    6. Smith, A. R.,
    7. Rector, M. V.,
    8. Suter, M. J.,
    9. Kao, S.,
    10. McLennan, G. et al.
    (2010). Loss of cystic fibrosis transmembrane conductance regulator function produces abnormalities in tracheal development in neonatal pigs and young children. Am. J. Respir. Crit. Care Med. 182, 1251-1261. doi:10.1164/rccm.201004-0643OC
    OpenUrlCrossRefPubMedWeb of Science
  109. ↵
    1. Mirabello, L.,
    2. Troisi, R. J. and
    3. Savage, S. A.
    (2009). International osteosarcoma incidence patterns in children and adolescents, middle ages and elderly persons. Int. J. Cancer 125, 229-234. doi:10.1002/ijc.24320
    OpenUrlCrossRefPubMed
  110. ↵
    1. Miyaoka, Y.,
    2. Berman, J. R.,
    3. Cooper, S. B.,
    4. Mayerl, S. J.,
    5. Chan, A. H.,
    6. Zhang, B.,
    7. Karlin-Neumann, G. A. and
    8. Conklin, B. R.
    (2016). Systematic quantification of HDR and NHEJ reveals effects of locus, nuclease, and cell type on genome-editing. Sci. Rep. 6, 23549. doi:10.1038/srep23549
    OpenUrlCrossRefPubMed
  111. ↵
    1. Morán, A.,
    2. Ortega, P.,
    3. de Juan, C.,
    4. Fernandez-Marcelo, T.,
    5. Frias, C.,
    6. Sanchez-Pernaute, A.,
    7. Torres, A. J.,
    8. Diaz-Rubio, E.,
    9. Iniesta, P. &
    10. Benito, M.
    (2010). Differential colorectal carcinogenesis: molecular basis and clinical relevance. World J. Gastrointest. Oncol. 2, 151-158. doi:10.4251/wjgo.v2.i3.151
    OpenUrlCrossRefPubMed
  112. ↵
    1. Nakamura, A. and
    2. Takeda, S.
    (2011). Mammalian models of Duchenne muscular dystrophy: pathological characteristics and therapeutic applications. J. Biomed. Biotechnol. 2011, 184393. doi:10.1155/2011/184393
    OpenUrlCrossRefPubMed
  113. ↵
    1. Nauck, M. A.,
    2. El-Ouaghlidi, A.,
    3. Gabrys, B.,
    4. Hücking, K.,
    5. Holst, J. J.,
    6. Deacon, C. F.,
    7. Gallwitz, B.,
    8. Schmidt, W. E. &
    9. Meier, J. J.
    (2004). Secretion of incretin hormones (GIP and GLP-1) and incretin effect after oral glucose in first-degree relatives of patients with type 2 diabetes. Regul. Pept. 122, 209-217. doi:10.1016/j.regpep.2004.06.020
    OpenUrlCrossRefPubMedWeb of Science
  114. ↵
    1. Nowak-Imialek, M. and
    2. Niemann, H.
    (2012). Pluripotent cells in farm animals: state of the art and future perspectives. Reprod. Fertil. Dev. 25, 103-128. doi:10.1071/RD12265
    OpenUrlCrossRefWeb of Science
  115. ↵
    1. Ognjanovic, S.,
    2. Olivier, M.,
    3. Bergemann, T. L. and
    4. Hainaut, P.
    (2012). Sarcomas in TP53 germline mutation carriers: a review of the IARC TP53 database. Cancer 118, 1387-1396. doi:10.1002/cncr.26390
    OpenUrlCrossRefPubMed
  116. ↵
    1. Ostedgaard, L. S.,
    2. Meyerholz, D. K.,
    3. Chen, J.-H.,
    4. Pezzulo, A. A.,
    5. Karp, P. H.,
    6. Rokhlina, T.,
    7. Ernst, S. E.,
    8. Hanfland, R. A.,
    9. Reznikov, L. R.,
    10. Ludwig, P. S. et al
    . (2011). The DeltaF508 mutation causes CFTR misprocessing and cystic fibrosis-like disease in pigs. Sci. Transl. Med. 3, 74ra24. doi:10.1126/scitranslmed.3001868
    OpenUrlAbstract/FREE Full Text
  117. ↵
    1. Overholtzer, M.,
    2. Rao, P. H.,
    3. Favis, R.,
    4. Lu, X. Y.,
    5. Elowitz, M. B.,
    6. Barany, F.,
    7. Ladanyi, M.,
    8. Gorlick, R. and
    9. Levine, A. J.
    (2003). The presence of p53 mutations in human osteosarcomas correlates with high levels of genomic instability. Proc. Natl. Acad. Sci. USA 100, 11547-11552. doi:10.1073/pnas.1934852100
    OpenUrlAbstract/FREE Full Text
  118. ↵
    1. Pabst, R.
    (1996). The respiratory immune system of pigs. Vet. Immunol. Immunopathol. 54, 191-195. doi:10.1016/S0165-2427(96)05700-5
    OpenUrlCrossRefPubMedWeb of Science
  119. ↵
    1. Pabst, R. and
    2. Binns, R. M.
    (1994). The immune system of the respiratory tract in pigs. Vet. Immunol. Immunopathol. 43, 151-156. doi:10.1016/0165-2427(94)90131-7
    OpenUrlCrossRefPubMedWeb of Science
  120. ↵
    1. Park, K. E.,
    2. Kaucher, A. V.,
    3. Powell, A.,
    4. Waqas, M. S.,
    5. Sandmaier, S. E.,
    6. Oatley, M. J.,
    7. Park, C. H.,
    8. Tibary, A.,
    9. Donovan, D. M.,
    10. Blomberg, L. A. et al
    . (2017). Generation of germline ablated male pigs by CRISPR/Cas9 editing of the NANOS2 gene. Sci. Rep. 7, 40176. doi:10.1038/srep40176
    OpenUrlCrossRef
  121. ↵
    1. Plump, A. S.,
    2. Masucci-Magoulas, L.,
    3. Bruce, C.,
    4. Bisgaier, C. L.,
    5. Breslow, J. L. and
    6. Tall, A. R.
    (1999). Increased atherosclerosis in ApoE and LDL receptor gene knock-out mice as a result of human cholesteryl ester transfer protein transgene expression. Arterioscler. Thromb. Vasc. Biol. 19, 1105-1110. doi:10.1161/01.ATV.19.4.1105
    OpenUrlAbstract/FREE Full Text
  122. ↵
    1. Pylayeva-Gupta, Y.,
    2. Grabocka, E. and
    3. Bar-Sagi, D.
    (2011). RAS oncogenes: weaving a tumorigenic web. Nat. Rev. Cancer 11, 761-774. doi:10.1038/nrc3106
    OpenUrlCrossRefPubMedWeb of Science
  123. ↵
    1. Rangarajan, A.,
    2. Hong, S. J.,
    3. Gifford, A. and
    4. Weinberg, R. A.
    (2004). Species- and cell type-specific requirements for cellular transformation. Cancer Cell 6, 171-183. doi:10.1016/j.ccr.2004.07.009
    OpenUrlCrossRefPubMedWeb of Science
  124. ↵
    1. Reiser, R.,
    2. Sorrels, M. F. and
    3. Williams, M. C.
    (1959). Influence of high levels of dietary fats and cholesterol on atherosclerosis and lipid distribution in swine. Circ. Res. 7, 833-846. doi:10.1161/01.RES.7.6.833
    OpenUrlAbstract/FREE Full Text
  125. ↵
    1. Renner, S.,
    2. Fehlings, C.,
    3. Herbach, N.,
    4. Hofmann, A.,
    5. von Waldthausen, D. C.,
    6. Kessler, B.,
    7. Ulrichs, K.,
    8. Chodnevskaja, I.,
    9. Moskalenko, V.,
    10. Amselgruber, W. et al.
    (2010). Glucose intolerance and reduced proliferation of pancreatic beta-cells in transgenic pigs with impaired glucose-dependent insulinotropic polypeptide function. Diabetes 59, 1228-1238. doi:10.2337/db09-0519
    OpenUrlAbstract/FREE Full Text
  126. ↵
    1. Renner, S.,
    2. Romisch-Margl, W.,
    3. Prehn, C.,
    4. Krebs, S.,
    5. Adamski, J.,
    6. Goke, B.,
    7. Blum, H.,
    8. Suhre, K.,
    9. Roscher, A. A. and
    10. Wolf, E.
    (2012). Changing metabolic signatures of amino acids and lipids during the prediabetic period in a pig model with impaired incretin function and reduced beta-cell mass. Diabetes 61, 2166-2175. doi:10.2337/db11-1133
    OpenUrlAbstract/FREE Full Text
  127. ↵
    1. Renner, S.,
    2. Braun-Reichhart, C.,
    3. Blutke, A.,
    4. Herbach, N.,
    5. Emrich, D.,
    6. Streckel, E.,
    7. Wunsch, A.,
    8. Kessler, B.,
    9. Kurome, M.,
    10. Bahr, A. et al.
    (2013). Permanent neonatal diabetes in INS(C94Y) transgenic pigs. Diabetes 62, 1505-1511. doi:10.2337/db12-1065
    OpenUrlAbstract/FREE Full Text
  128. ↵
    1. Renner, S.,
    2. Blutke, A.,
    3. Streckel, E.,
    4. Wanke, R. and
    5. Wolf, E.
    (2016). Incretin actions and consequences of incretin-based therapies: lessons from complementary animal models. J. Pathol. 238, 345-358. doi:10.1002/path.4655
    OpenUrlCrossRef
  129. ↵
    1. Reznikov, L. R.,
    2. Dong, Q.,
    3. Chen, J.-H.,
    4. Moninger, T. O.,
    5. Park, J. M.,
    6. Zhang, Y.,
    7. Du, J.,
    8. Hildebrand, M. S.,
    9. Smith, R. J. H.,
    10. Randak, C. O. et al.
    (2013). CFTR-deficient pigs display peripheral nervous system defects at birth. Proc. Natl. Acad. Sci. USA 110, 3083-3088. doi:10.1073/pnas.1222729110
    OpenUrlAbstract/FREE Full Text
  130. ↵
    1. Rogers, C. S.,
    2. Hao, Y.,
    3. Rokhlina, T.,
    4. Samuel, M.,
    5. Stoltz, D. A.,
    6. Li, Y.,
    7. Petroff, E.,
    8. Vermeer, D. W.,
    9. Kabel, A. C.,
    10. Yan, Z. et al.
    (2008a). Production of CFTR-null and CFTR-DeltaF508 heterozygous pigs by adeno-associated virus-mediated gene targeting and somatic cell nuclear transfer. J. Clin. Invest. 118, 1571-1577. doi:10.1172/JCI34773
    OpenUrlCrossRefPubMedWeb of Science
  131. ↵
    1. Rogers, C. S.,
    2. Stoltz, D. A.,
    3. Meyerholz, D. K.,
    4. Ostedgaard, L. S.,
    5. Rokhlina, T.,
    6. Taft, P. J.,
    7. Rogan, M. P.,
    8. Pezzulo, A. A.,
    9. Karp, P. H.,
    10. Itani, O. A. et al.
    (2008b). Disruption of the CFTR gene produces a model of cystic fibrosis in newborn pigs. Science 321, 1837-1841. doi:10.1126/science.1163600
    OpenUrlAbstract/FREE Full Text
  132. ↵
    1. Romar, R.,
    2. Funahashi, H. and
    3. Coy, P.
    (2016). In vitro fertilization in pigs: new molecules and protocols to consider in the forthcoming years. Theriogenology 85, 125-134. doi:10.1016/j.theriogenology.2015.07.017
    OpenUrlCrossRef
  133. ↵
    1. Saalfrank, A.,
    2. Janssen, K. P.,
    3. Ravon, M.,
    4. Flisikowski, K.,
    5. Eser, S.,
    6. Steiger, K.,
    7. Flisikowska, T.,
    8. Muller-Fliedner, P.,
    9. Schulze, E.,
    10. Bronner, C. et al
    . (2016). A porcine model of osteosarcoma. Oncogenesis 5, e210. doi:10.1038/oncsis.2016.19
    OpenUrlCrossRef
  134. ↵
    1. Sachs, D. H.
    (1994). The pig as a potential xenograft donor. Vet. Immunol. Immunopathol. 43, 185-191. doi:10.1016/0165-2427(94)90135-X
    OpenUrlCrossRefPubMedWeb of Science
  135. ↵
    1. Schnieke, A. E.,
    2. Kind, A. J.,
    3. Ritchie, W. A.,
    4. Mycock, K.,
    5. Scott, A. R.,
    6. Ritchie, M.,
    7. Wilmut, I.,
    8. Colman, A. and
    9. Campbell, K. H.
    (1997). Human factor IX transgenic sheep produced by transfer of nuclei from transfected fetal fibroblasts. Science 278, 2130-2133. doi:10.1126/science.278.5346.2130
    OpenUrlAbstract/FREE Full Text
  136. ↵
    1. Schönhuber, N.,
    2. Seidler, B.,
    3. Schuck, K.,
    4. Veltkamp, C.,
    5. Schachtler, C.,
    6. Zukowska, M.,
    7. Eser, S.,
    8. Feyerabend, T. B.,
    9. Paul, M. C.,
    10. Eser, P. et al.
    (2014). A next-generation dual-recombinase system for time- and host-specific targeting of pancreatic cancer. Nat. Med. 20, 1340-1347. doi:10.1038/nm.3646
    OpenUrlCrossRefPubMed
  137. ↵
    1. Schook, L. B.,
    2. Collares, T. V.,
    3. Hu, W.,
    4. Liang, Y.,
    5. Rodrigues, F. M.,
    6. Rund, L. A.,
    7. Schachtschneider, K. M.,
    8. Seixas, F. K.,
    9. Singh, K.,
    10. Wells, K. D. et al
    . (2015). A Genetic Porcine Model of Cancer. PLoS ONE 10, e0128864. doi:10.1371/journal.pone.0128864
    OpenUrlCrossRefPubMed
  138. ↵
    1. Schubert, R.,
    2. Frank, F.,
    3. Nagelmann, N.,
    4. Liebsch, L.,
    5. Schuldenzucker, V.,
    6. Schramke, S.,
    7. Wirsig, M.,
    8. Johnson, H.,
    9. Kim, E. Y.,
    10. Ott, S. et al
    . (2016). Neuroimaging of a minipig model of Huntington's disease: feasibility of volumetric, diffusion-weighted and spectroscopic assessments. J. Neurosci. Methods, 265, 46-55. doi:10.1016/j.jneumeth.2015.11.017
    OpenUrlCrossRef
  139. ↵
    1. Shimoda, M.,
    2. Kanda, Y.,
    3. Hamamoto, S.,
    4. Tawaramoto, K.,
    5. Hashiramoto, M.,
    6. Matsuki, M. and
    7. Kaku, K.
    (2011). The human glucagon-like peptide-1 analogue liraglutide preserves pancreatic beta cells via regulation of cell kinetics and suppression of oxidative and endoplasmic reticulum stress in a mouse model of diabetes. Diabetologia 54, 1098-1108. doi:10.1007/s00125-011-2069-9
    OpenUrlCrossRefPubMedWeb of Science
  140. ↵
    1. Sieren, J. C.,
    2. Meyerholz, D. K.,
    3. Wang, X.-J.,
    4. Davis, B. T.,
    5. Newell, J. D., Jr..,
    6. Hammond, E.,
    7. Rohret, J. A.,
    8. Rohret, F. A.,
    9. Struzynski, J. T.,
    10. Goeken, J. A. et al.
    (2014). Development and translational imaging of a TP53 porcine tumorigenesis model. J. Clin. Invest. 124, 4052-4066. doi:10.1172/JCI75447
    OpenUrlCrossRefPubMed
  141. ↵
    1. Skarnes, W. C.,
    2. Rosen, B.,
    3. West, A. P.,
    4. Koutsourakis, M.,
    5. Bushell, W.,
    6. Iyer, V.,
    7. Mujica, A. O.,
    8. Thomas, M.,
    9. Harrow, J.,
    10. Cox, T. et al.
    (2011). A conditional knockout resource for the genome-wide study of mouse gene function. Nature 474, 337-342. doi:10.1038/nature10163
    OpenUrlCrossRefPubMedWeb of Science
  142. ↵
    1. Skold, B. H.,
    2. Getty, R. and
    3. Ramsey, F. K.
    (1966). Spontaneous atherosclerosis in the arterial system of aging swine. Am. J. Vet. Res. 27, 257-273.
    OpenUrlPubMedWeb of Science
  143. ↵
    1. Smithies, O.,
    2. Gregg, R. G.,
    3. Boggs, S. S.,
    4. Koralewski, M. A. and
    5. Kucherlapati, R. S.
    (1985). Insertion of DNA sequences into the human chromosomal beta-globin locus by homologous recombination. Nature 317, 230-234. doi:10.1038/317230a0
    OpenUrlCrossRefPubMedWeb of Science
  144. ↵
    1. Steines, B.,
    2. Dickey, D. D.,
    3. Bergen, J.,
    4. Excoffon, K. J.,
    5. Weinstein, J. R.,
    6. Li, X.,
    7. Yan, Z.,
    8. Abou Alaiwa, M. H.,
    9. Shah, V. S.,
    10. Bouzek, D. C. et al
    . (2016). CFTR gene transfer with AAV improves early cystic fibrosis pig phenotypes. JCI Insight 1, e88728. doi:10.1172/jci.insight.88728
    OpenUrlCrossRef
  145. ↵
    1. Stoltz, D. A.,
    2. Rokhlina, T.,
    3. Ernst, S. E.,
    4. Pezzulo, A. A.,
    5. Ostedgaard, L. S.,
    6. Karp, P. H.,
    7. Samuel, M. S.,
    8. Reznikov, L. R.,
    9. Rector, M. V.,
    10. Gansemer, N. D. et al.
    (2013). Intestinal CFTR expression alleviates meconium ileus in cystic fibrosis pigs. J. Clin. Invest. 123, 2685-2693. doi:10.1172/JCI68867
    OpenUrlCrossRefPubMedWeb of Science
  146. ↵
    1. Stoltz, D. A.,
    2. Meyerholz, D. K. and
    3. Welsh, M. J.
    (2015). Origins of cystic fibrosis lung disease. N. Engl. J. Med. 372, 351-362. doi:10.1056/NEJMra1300109
    OpenUrlCrossRefPubMedWeb of Science
  147. ↵
    1. Stoy, J.,
    2. Edghill, E. L.,
    3. Flanagan, S. E.,
    4. Ye, H.,
    5. Paz, V. P.,
    6. Pluzhnikov, A.,
    7. Below, J. E.,
    8. Hayes, M. G.,
    9. Cox, N. J.,
    10. Lipkind, G. M. et al.
    (2007). Insulin gene mutations as a cause of permanent neonatal diabetes. Proc. Natl. Acad. Sci. USA 104, 15040-15044. doi:10.1073/pnas.0707291104
    OpenUrlAbstract/FREE Full Text
  148. ↵
    1. Streckel, E.,
    2. Braun-Reichhart, C.,
    3. Herbach, N.,
    4. Dahlhoff, M.,
    5. Kessler, B.,
    6. Blutke, A.,
    7. Bahr, A.,
    8. Ubel, N.,
    9. Eddicks, M.,
    10. Ritzmann, M. et al
    . (2015). Effects of the glucagon-like peptide-1 receptor agonist liraglutide in juvenile transgenic pigs modeling a pre-diabetic condition. J. Transl. Med. 13, 73. doi:10.1186/s12967-015-0431-2
    OpenUrlCrossRefPubMed
  149. ↵
    1. Suzuki, H.,
    2. Saito, Y.,
    3. Kagawa, N. and
    4. Yang, X.
    (2003). In vitro fertilization and polyspermy in the pig: factors affecting fertilization rates and cytoskeletal reorganization of the oocyte. Microsc. Res. Tech. 61, 327-334. doi:10.1002/jemt.10345
    OpenUrlCrossRefPubMed
  150. ↵
    1. Suzuki, S.,
    2. Iwamoto, M.,
    3. Hashimoto, M.,
    4. Suzuki, M.,
    5. Nakai, M.,
    6. Fuchimoto, D.,
    7. Sembon, S.,
    8. Eguchi-Ogawa, T.,
    9. Uenishi, H. and
    10. Onishi, A.
    (2016). Generation and characterization of RAG2 knockout pigs as animal model for severe combined immunodeficiency. Vet. Immunol. Immunopathol. 178, 37-49. doi:10.1016/j.vetimm.2016.06.011
    OpenUrlCrossRef
  151. ↵
    1. Takeuchi, A.,
    2. Irizarry, M. C.,
    3. Duff, K.,
    4. Saido, T. C.,
    5. Hsiao Ashe, K.,
    6. Hasegawa, M.,
    7. Mann, D. M.,
    8. Hyman, B. T. and
    9. Iwatsubo, T.
    (2000). Age-related amyloid beta deposition in transgenic mice overexpressing both Alzheimer mutant presenilin 1 and amyloid beta precursor protein Swedish mutant is not associated with global neuronal loss. Am. J. Pathol. 157, 331-339. doi:10.1016/S0002-9440(10)64544-0
    OpenUrlCrossRefPubMedWeb of Science
  152. ↵
    1. Tamura, K.,
    2. Minami, K.,
    3. Kudo, M.,
    4. Iemoto, K.,
    5. Takahashi, H. and
    6. Seino, S.
    (2015). Liraglutide improves pancreatic Beta cell mass and function in alloxan-induced diabetic mice. PLoS ONE 10, e0126003. doi:10.1371/journal.pone.0126003
    OpenUrlCrossRef
  153. ↵
    1. Tan, W.,
    2. Carlson, D. F.,
    3. Lancto, C. A.,
    4. Garbe, J. R.,
    5. Webster, D. A.,
    6. Hackett, P. B. and
    7. Fahrenkrug, S. C.
    (2013). Efficient nonmeiotic allele introgression in livestock using custom endonucleases. Proc. Natl. Acad. Sci. USA 110, 16526-16531. doi:10.1073/pnas.1310478110
    OpenUrlAbstract/FREE Full Text
  154. ↵
    1. Tetteh, P. W.,
    2. Kretzschmar, K.,
    3. Begthel, H.,
    4. van den Born, M.,
    5. Korving, J.,
    6. Morsink, F.,
    7. Farin, H.,
    8. van Es, J. H.,
    9. Offerhaus, G. J. and
    10. Clevers, H.
    (2016). Generation of an inducible colon-specific Cre enzyme mouse line for colon cancer research. Proc. Natl. Acad. Sci. USA 113, 11859-11864. doi:10.1073/pnas.1614057113
    OpenUrlAbstract/FREE Full Text
  155. ↵
    1. Thibault, K. L. and
    2. Margulies, S. S.
    (1998). Age-dependent material properties of the porcine cerebrum: effect on pediatric inertial head injury criteria. J. Biomech. 31, 1119-1126. doi:10.1016/S0021-9290(98)00122-5
    OpenUrlCrossRefPubMedWeb of Science
  156. ↵
    1. Thomas, K. R. and
    2. Capecchi, M. R.
    (1987). Site-directed mutagenesis by gene targeting in mouse embryo-derived stem cells. Cell 51, 503-512. doi:10.1016/0092-8674(87)90646-5
    OpenUrlCrossRefPubMedWeb of Science
  157. ↵
    1. Truong, D.-J.,
    2. Kuhner, K.,
    3. Kuhn, R.,
    4. Werfel, S.,
    5. Engelhardt, S.,
    6. Wurst, W. &
    7. Ortiz, O.
    (2015). Development of an intein-mediated split-Cas9 system for gene therapy. Nucleic. Acids Res. 43, 6450-6458. doi:10.1093/nar/gkv601
    OpenUrlCrossRefPubMed
  158. ↵
    1. Uchida, M.,
    2. Shimatsu, Y.,
    3. Onoe, K.,
    4. Matsuyama, N.,
    5. Niki, R.,
    6. Ikeda, J. E. and
    7. Imai, H.
    (2001). Production of transgenic miniature pigs by pronuclear microinjection. Transgenic Res. 10, 577-582. doi:10.1023/A:1013059917280
    OpenUrlCrossRefPubMedWeb of Science
  159. ↵
    1. Umeyama, K.,
    2. Watanabe, M.,
    3. Saito, H.,
    4. Kurome, M.,
    5. Tohi, S.,
    6. Matsunari, H.,
    7. Miki, K. and
    8. Nagashima, H.
    (2009). Dominant-negative mutant hepatocyte nuclear factor 1alpha induces diabetes in transgenic-cloned pigs. Transgenic Res. 18, 697-706. doi:10.1007/s11248-009-9262-3
    OpenUrlCrossRefPubMedWeb of Science
  160. ↵
    1. Umeyama, K.,
    2. Honda, K.,
    3. Matsunari, H.,
    4. Nakano, K.,
    5. Hidaka, T.,
    6. Sekiguchi, K.,
    7. Mochizuki, H.,
    8. Takeuchi, Y.,
    9. Fujiwara, T.,
    10. Watanabe, M. et al.
    (2013). Production of diabetic offspring using cryopreserved epididymal sperm by in vitro fertilization and intrafallopian insemination techniques in transgenic pigs. J. Reprod. Dev. 59, 599-603. doi:10.1262/jrd.2013-069
    OpenUrlCrossRef
  161. ↵
    1. Wang, Y.,
    2. Du, Y.,
    3. Shen, B.,
    4. Zhou, X.,
    5. Li, J.,
    6. Liu, Y.,
    7. Wang, J.,
    8. Zhou, J.,
    9. Hu, B.,
    10. Kang, N., et al
    . (2015). Efficient generation of gene-modified pigs via injection of zygote with Cas9/sgRNA. Sci. Rep. 5, 8256. doi:10.1038/srep08256
    OpenUrlCrossRefPubMed
  162. ↵
    1. Wei, J.,
    2. Ouyang, H.,
    3. Wang, Y.,
    4. Pang, D.,
    5. Cong, N. X.,
    6. Wang, T.,
    7. Leng, B.,
    8. Li, D.,
    9. Li, X.,
    10. Wu, R. et al.
    (2012). Characterization of a hypertriglyceridemic transgenic miniature pig model expressing human apolipoprotein CIII. FEBS J. 279, 91-99. doi:10.1111/j.1742-4658.2011.08401.x
    OpenUrlCrossRefPubMed
  163. ↵
    1. Whitelaw, C. B.,
    2. Radcliffe, P. A.,
    3. Ritchie, W. A.,
    4. Carlisle, A.,
    5. Ellard, F. M.,
    6. Pena, R. N.,
    7. Rowe, J.,
    8. Clark, A. J.,
    9. King, T. J. and
    10. Mitrophanous, K. A.
    (2004). Efficient generation of transgenic pigs using equine infectious anaemia virus (EIAV) derived vector. FEBS Lett. 571, 233-236. doi:10.1016/j.febslet.2004.06.076
    OpenUrlCrossRefPubMedWeb of Science
  164. ↵
    1. Whitworth, K. M.,
    2. Lee, K.,
    3. Benne, J. A.,
    4. Beaton, B. P.,
    5. Spate, L. D.,
    6. Murphy, S. L.,
    7. Samuel, M. S.,
    8. Mao, J.,
    9. O'Gorman, C.,
    10. Walters, E. M. et al
    . (2014). Use of the CRISPR/Cas9 system to produce genetically engineered pigs from in vitro-derived oocytes and embryos. Biol. Reprod. 91, 78. doi:10.1095/biolreprod.114.121723
    OpenUrlAbstract/FREE Full Text
  165. ↵
    1. Wilke, M.,
    2. Buijs-Offerman, R. M.,
    3. Aarbiou, J.,
    4. Colledge, W. H.,
    5. Sheppard, D. N.,
    6. Touqui, L.,
    7. Bot, A.,
    8. Jorna, H.,
    9. de Jonge, H. R. and
    10. Scholte, B. J.
    (2011). Mouse models of cystic fibrosis: phenotypic analysis and research applications. J. Cyst. Fibros. 10 Suppl 2, S152-S171. doi:10.1016/S1569-1993(11)60020-9
    OpenUrlCrossRefPubMed
  166. ↵
    1. Winblad, B.,
    2. Amouyel, P.,
    3. Andrieu, S.,
    4. Ballard, C.,
    5. Brayne, C.,
    6. Brodaty, H.,
    7. Cedazo-Minguez, A.,
    8. Dubois, B.,
    9. Edvardsson, D.,
    10. Feldman, H. et al.
    (2016). Defeating Alzheimer's disease and other dementias: a priority for European science and society. Lancet Neurol. 15, 455-532. doi:10.1016/S1474-4422(16)00062-4
    OpenUrlCrossRefPubMed
  167. ↵
    1. Wolfe, M. S.,
    2. Xia, W.,
    3. Ostaszewski, B. L.,
    4. Diehl, T. S.,
    5. Kimberly, W. T. and
    6. Selkoe, D. J.
    (1999). Two transmembrane aspartates in presenilin-1 required for presenilin endoproteolysis and gamma-secretase activity. Nature 398, 513-517. doi:10.1038/19077
    OpenUrlCrossRefPubMedWeb of Science
  168. ↵
    1. Worthley, S. G.,
    2. Helft, G.,
    3. Fuster, V.,
    4. Fayad, Z. A.,
    5. Rodriguez, O. J.,
    6. Zaman, A. G.,
    7. Fallon, J. T. and
    8. Badimon, J. J.
    (2000). Noninvasive in vivo magnetic resonance imaging of experimental coronary artery lesions in a porcine model. Circulation 101, 2956-2961. doi:10.1161/01.CIR.101.25.2956
    OpenUrlAbstract/FREE Full Text
  169. ↵
    1. Wu, L.,
    2. Rosa-Neto, P.,
    3. Hsiung, G. Y.,
    4. Sadovnick, A. D.,
    5. Masellis, M.,
    6. Black, S. E.,
    7. Jia, J. and
    8. Gauthier, S.
    (2012). Early-onset familial Alzheimer's disease (EOFAD). Can. J. Neurol. Sci. 39, 436-445. doi:10.1017/S0317167100013949
    OpenUrlCrossRefPubMed
  170. ↵
    1. Wu, M.,
    2. Wei, C.,
    3. Lian, Z.,
    4. Liu, R.,
    5. Zhu, C.,
    6. Wang, H.,
    7. Cao, J.,
    8. Shen, Y.,
    9. Zhao, F.,
    10. Zhang, L. et al
    . (2016). Rosa26-targeted sheep gene knock-in via CRISPR-Cas9 system. Sci. Rep. 6, 24360. doi:10.1038/srep24360
    OpenUrlCrossRef
  171. ↵
    1. Wu, J.,
    2. Platero-Luengo, A.,
    3. Sakurai, M.,
    4. Sugawara, A.,
    5. Gil, M. A.,
    6. Yamauchi, T.,
    7. Suzuki, K.,
    8. Bogliotti, Y. S.,
    9. Cuello, C.,
    10. Morales Valencia, M. et al
    . (2017). Interspecies Chimerism with Mammalian Pluripotent Stem Cells. Cell 168, 473-486 e15. doi:10.1016/j.cell.2016.12.036
    OpenUrlCrossRef
  172. ↵
    1. Yamagata, K.,
    2. Oda, N.,
    3. Kaisaki, P. J.,
    4. Menzel, S.,
    5. Furuta, H.,
    6. Vaxillaire, M.,
    7. Southam, L.,
    8. Cox, R. D.,
    9. Lathrop, G. M.,
    10. Boriraj, V. V. et al.
    (1996). Mutations in the hepatocyte nuclear factor-1alpha gene in maturity-onset diabetes of the young (MODY3). Nature 384, 455-458. doi:10.1038/384455a0
    OpenUrlCrossRefPubMedWeb of Science
  173. ↵
    1. Yang, D.,
    2. Liu, L.,
    3. Zhu, D.,
    4. Peng, H.,
    5. Su, L.,
    6. Fu, Y. X. and
    7. Zhang, L.
    (2014). A mouse model for HBV immunotolerance and immunotherapy. Cell Mol. Immunol. 11, 71-78. doi:10.1038/cmi.2013.43
    OpenUrlCrossRefPubMed
  174. ↵
    1. Yoshioka, K.,
    2. Noguchi, M. and
    3. Suzuki, C.
    (2012). Production of piglets from in vitro-produced embryos following non-surgical transfer. Anim. Reprod. Sci. 131, 23-29. doi:10.1016/j.anireprosci.2012.01.018
    OpenUrlCrossRefPubMed
  175. ↵
    1. Yu, X.,
    2. Bao, B.,
    3. Echigoya, Y. and
    4. Yokota, T.
    (2015). Dystrophin-deficient large animal models: translational research and exon skipping. Am. J. Transl. Res. 7, 1314-1331.
    OpenUrl
  176. ↵
    1. Yu, H. H.,
    2. Zhao, H.,
    3. Qing, Y. B.,
    4. Pan, W. R.,
    5. Jia, B. Y.,
    6. Zhao, H. Y.,
    7. Huang, X. X. and
    8. Wei, H. J.
    (2016). Porcine zygote injection with Cas9/sgRNA results in DMD-modified pig with muscle dystrophy. Int. J. Mol. Sci. 17, E1668. doi:10.3390/ijms17101668
    OpenUrlCrossRef
  177. ↵
    1. Zhang, S. H.,
    2. Reddick, R. L.,
    3. Piedrahita, J. A. and
    4. Maeda, N.
    (1992). Spontaneous hypercholesterolemia and arterial lesions in mice lacking apolipoprotein E. Science 258, 468-471. doi:10.1126/science.1411543
    OpenUrlAbstract/FREE Full Text
  178. ↵
    1. Zhou, X.,
    2. Wang, L.,
    3. Du, Y.,
    4. Xie, F.,
    5. Li, L.,
    6. Liu, Y.,
    7. Liu, C.,
    8. Wang, S.,
    9. Zhang, S.,
    10. Huang, X. et al.
    (2016). Efficient generation of gene-modified pigs harboring precise orthologous human mutation via crispr/cas9-induced homology-directed repair in zygotes. Hum. Mutat. 37, 110-118. doi:10.1002/humu.22913
    OpenUrlCrossRefPubMed
Previous ArticleNext Article
Back to top
Previous ArticleNext Article

This Issue

RSSRSS

Keywords

  • Disease models
  • Genetic modification
  • Pig
  • Swine

 Download PDF

Email

Thank you for your interest in spreading the word on Disease Models & Mechanisms.

NOTE: We only request your email address so that the person you are recommending the page to knows that you wanted them to see it, and that it is not junk mail. We do not capture any email address.

Enter multiple addresses on separate lines or separate them with commas.
Genetically engineered pigs as models for human disease
(Your Name) has sent you a message from Disease Models & Mechanisms
(Your Name) thought you would like to see the Disease Models & Mechanisms web site.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Share
REVIEW
Genetically engineered pigs as models for human disease
Carolin Perleberg, Alexander Kind, Angelika Schnieke
Disease Models & Mechanisms 2018 11: dmm030783 doi: 10.1242/dmm.030783 Published 22 January 2018
del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo
Citation Tools
REVIEW
Genetically engineered pigs as models for human disease
Carolin Perleberg, Alexander Kind, Angelika Schnieke
Disease Models & Mechanisms 2018 11: dmm030783 doi: 10.1242/dmm.030783 Published 22 January 2018

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Alerts

Please log in to add an alert for this article.

Sign in to email alerts with your email address

Article navigation

  • Top
  • Article
    • ABSTRACT
    • Introduction
    • Methods for generating porcine disease models
    • Modelling cardiovascular diseases
    • Modelling cancer
    • Modelling diabetes mellitus
    • Modelling Alzheimer's disease
    • Modelling cystic fibrosis
    • Modelling Duchenne muscular dystrophy
    • Future perspectives
    • Acknowledgements
    • Footnotes
    • References
  • Figures & tables
  • Info & metrics
  • PDF

Related articles

Cited by...

More in this TOC section

  • Hyperoxia-induced bronchopulmonary dysplasia: better models for better therapies
  • Modelling epilepsy in the mouse: challenges and solutions
  • AIRE deficiency, from preclinical models to human APECED disease
Show more REVIEW

Similar articles

Other journals from The Company of Biologists

Development

Journal of Cell Science

Journal of Experimental Biology

Biology Open

Advertisement

DMM and COVID-19

We are aware that the COVID-19 pandemic is having an unprecedented impact on researchers worldwide. The Editors of all The Company of Biologists’ journals have been considering ways in which we can alleviate concerns that members of our community may have around publishing activities during this time. Read about the actions we are taking at this time.

Please don’t hesitate to contact the Editorial Office if you have any questions or concerns.


Monica Justice bids farewell to DMM

In her farewell Editorial, outgoing Editor-in-Chief Monica Justice reminds us of the past half-decade of growth and of DMM's commitment to support the disease modelling community, concluding, “The knowledge and experience I gained during my time as Senior Editor and EiC at DMM is invaluable: working within a not-for-profit community publishing environment is a joy.”


3D imaging of beta cell mass in diabetic mouse models

In their inducible mouse model of diabetes, Roostalu et al. demonstrate how quantitative light-sheet imaging can capture changes in individual islets to help pharmacological research in diabetes.

Visit our YouTube channel to watch more videos from DMM, our sister journals and the Company.


Modelling Joubert syndrome patient-derived mutations in C. elegans

In this issue’s Editor’s choice, Karen Lange and colleagues used C. elegans to model and characterise two patient-derived mutations that cause the ciliopathy Joubert syndrome.


Interview – Karen Lange

First author of our current Editor’s choice, Karen Lange takes us behind the scenes of the paper, and shares her thoughts on how the lack of both time and job security will impact her research.

Articles

  • Accepted manuscripts
  • Latest complete issue
  • Issue archive
  • Archive by article type
  • Subject collections
  • Interviews
  • Sign up for alerts

About us

  • About DMM
  • Editors and Board
  • Editor biographies
  • Travelling Fellowships
  • Grants and funding
  • Journal Meetings
  • Workshops
  • The Company of Biologists

For Authors

  • Submit a manuscript
  • Aims and scope
  • Presubmission enquiries
  • Article types
  • Manuscript preparation
  • Cover suggestions
  • Editorial process
  • Promoting your paper
  • Open Access
  • Biology Open transfer

Journal Info

  • Journal policies
  • Rights and permissions
  • Media policies
  • Reviewer guide
  • Sign up for alerts

Contact

  • Contact DMM
  • Advertising
  • Feedback

Twitter   YouTube   LinkedIn

© 2021   The Company of Biologists Ltd   Registered Charity 277992