Generation and characterization of RAG2 knockout pigs as animal model for severe combined immunodeficiency

https://doi.org/10.1016/j.vetimm.2016.06.011Get rights and content

Highlights

  • We successfully generated RAG2 knockout pigs by somatic cell nuclear transfer.

  • RAG2 knockout pigs lacked T and B cells but not NK cells.

  • RAG2 knockout pigs had primordial lymph nodes and hypoplastic thymi.

  • RAG2 knockout pigs had cells resembling progenitors of T and B cells.

  • IL2RG/RAG2 knockout pigs lacked T, B, and NK cells altogether.

Abstract

Pigs with severe combined immunodeficiency (SCID) are versatile animal models for human medical research because of their biological similarities to humans, suitable body size, and longevity for practical research. SCID pigs with defined mutation(s) can be an invaluable tool for research on porcine immunity. In this study, we produced RAG2-knockout pigs via somatic cell nuclear transfer and analyzed their phenotype. The V(D)J recombination processes were confirmed as being inactivated. They consistently lacked mature T and B cells but had substantial numbers of cells considered to be T- or B-cell progenitors as well as NK cells. They also lacked thymic medulla and lymphoid aggregations in the spleen, mesenteric lymph nodes, and ileal Peyer’s patches. We showed more severe immunological defects in the RAG2 and IL2RG double-knockout pig through this study. Thus, SCID pigs could be promising animal models not only for translational medical research but also for immunological studies of pigs themselves.

Introduction

Immunodeficient animal models are versatile tools in biomedical research. They are naturally used as research models of immunity. In addition, they can be used as animal platforms for the generation of humanized animals that steadily maintain transplanted tissues and/or cells of humans and can be used for in vivo biological research of human. They are also useful in the preclinical trials of stem cell-based regenerative medicine to evaluate the effectiveness and safety of the transplantation of human stem cells, such as iPS cells. Severely immunodeficient mice models, such as NOG (Ito et al., 2002), NSG (Shultz et al., 2005), and BRG (Traggiai et al., 2004) mice have been intensively developed. The functional reconstitution of the human hematopoietic and immune systems has been achieved by transplanting human hematopoietic stem cells into these immunodeficient mice (Ito et al., 2002, Shultz et al., 2005, Traggiai et al., 2004), and research on human-specific infections has been performed using these models (Akkina, 2013, Bility et al., 2012, Bility et al., 2014). These models have also been used in human cancer and stem cell research (Cunningham et al., 2012, Shultz et al., 2014). However, mice models have considerable limitations for modeling human biological and clinical conditions. Their small size narrows the scope of surgical and clinical applications, and their short longevity precludes long-term evaluation of post-transplantation effects. Moreover, there are several genetic lines of difference in adaptive and innate immune systems (Mestas and Hughes, 2004) as well as inflammatory responses (Seok et al., 2013) between mice and humans.

Pigs are invaluable animal models in human medical research because of their similarities to humans in physiology, anatomy, nutrition, and genetics. In fact, several genetically modified pigs have been shown to recapitulate human disease with higher fidelity than mice models (Fan and Lai, 2013, Prather et al., 2013). Therefore, immunodeficient pigs can be excellent animal models for biomedical research, such as the development of humanized tissues and organs for transplantation and long-term evaluation of transplanted cancer or stem cells of human origin. Our group (Suzuki et al., 2012) and Watanabe et al. (2013) previously reported the development of severe combined immunodeficient (SCID) pigs by disrupting the interleukin 2 receptor gamma (IL2RG) gene. These pigs had extremely hypoplastic thymi and expressed a TB+NK phenotype. We also showed that these SCID pigs can be immune reconstituted by allogenic bone marrow (BM) transplantation (Suzuki et al., 2012). However, it is desirable that the SCID phenotype would be reinforced by additional gene modification(s) to achieve efficient engraftment of xenografts, including human stem cells.

Recombination activating gene (RAG) 1 and 2 are other genes where the inactivation of either gene induces a SCID phenotype in mice (Mombaerts et al., 1992, Shinkai et al., 1992), humans (Schwarz et al., 1996, Villa et al., 2001), and pigs (Huang et al., 2014, Ito et al., 2014, Lee et al., 2014). RAG1 and RAG2 collaborate in regulating the proper V(D)J rearrangements, which are indispensable for the generation of antigen receptor diversity in B cells and T cell receptor diversity in T cells (McBlane et al., 1995, Oettinger et al., 1990). The inactivation of RAG1 and/or RAG2 arrests lymphoid differentiation at an early stage and results in a lack of mature T and B cells. Therefore, we selected RAG2 as a target gene to be disrupted for the additional line in SCID pigs. Although the reports on RAG1- and/or RAG2-targeted pigs have already been published (Huang et al., 2014, Ito et al., 2014, Lee et al., 2014), they only showed the phenotypes of somatic-cell-cloned animals which can be accompanied with epigenetic abnormalities and they had not fully unraveled immunological traits of RAG-inactivated pigs. Therefore, additional research studies are still valuable in establishing the validated porcine SCID models. We report here the production of RAG2 knockout pigs via conventional gene targeting of somatic cells followed by somatic cell nuclear transfer and the characterization of immunological traits in their progenies which are free from epigenetic abnormalities. Moreover, we produced RAG2 and IL2RG double-knockout pigs toward the generation of more severe immunodeficient pigs which lacks not only T and B cells but also NK cells. The data presented here is useful for the development of an excellent humanized pig model as well as for veterinary research on porcine immunity.

Section snippets

Experimental animals

Animals used in this study were cross-bred pigs (from Landrace, Large White and Duroc) conventionally reared in the NARO Institute of Livestock and Grassland Sciences. Their health was daily monitored by checking their appetites, gaits, feces, hair conditions and respiratory status. All animal experiments were approved by the Gene Recombination Experiment Safety Committee (#500035) and Animal Care Committee (#H18-038) of the National Institute of Agrobiological Sciences.

Targeting vector construction

A BAC clone containing

Generation of RAG2 knockout pigs by nuclear transfer of gene targeted fibroblasts

Following 7 rounds of fetal fibroblast transfection with a porcine RAG2 gene TV, subsequent selection, and PCR screening, four PCR-positive cell colonies were obtained out of 3453 puromycin-resistant colonies (0.11%). One of them was expanded and used as donor cells for nuclear transfer (Table 1). A total of 1431 Embryos which cleaved in 2 days after nuclear transfer (49.9%) were transferred to the oviducts of six recipient female pigs (Table 1).

Because PCR-positive colonies often contain a

Discussion

We successfully generated RAG2 knockout pigs via conventional gene targeting in somatic cells followed by somatic cell nuclear transfer. Inactivation of RAG1 and/or RAG2 in pigs has been reported in several studies (Huang et al., 2014, Ito et al., 2014, Lee et al., 2014). However, the previous studies reported each case of a specific strain and breeding condition respectively, the additional research studies are still necessary to unravel the overall effect of porcine RAG2 inactivation and

Acknowledgements

This work was supported in part by a Grant-in-Aid grant from the Ministry of Agriculture, Forestry and Fisheries of Japan. We are grateful to Dr. F. Ishikawa for his great contribution in generating IL2RG knockout pigs. We thank Dr. T. Yagi for providing PGK-Puro-p(A) and MC1-DTA-p(A) expression cassetes and Dr. T. Furusawa for his excellent technical assistance and valuable suggestions in FACS analysis. We also thank S. Iijima and staff in the Pig Management Section of NARO Institute of

References (43)

  • J.F. McBlane et al.

    Cleavage at a V(D)J recombination signal requires only RAG1 and RAG2 proteins and occurs in two steps

    Cell

    (1995)
  • P. Mombaerts et al.

    RAG-1-deficient mice have no mature B and T lymphocytes

    Cell

    (1992)
  • Y. Shinkai et al.

    RAG-2-deficient mice lack mature lymphocytes owing to inability to initiate V(D)J rearrangement

    Cell

    (1992)
  • S. Suzuki et al.

    Il2rg gene-targeted severe combined immunodeficiency pigs

    Cell Stem Cell

    (2012)
  • H. Uenishi et al.

    Genomic sequence encoding diversity segments of the pig TCR delta chain gene demonstrates productivity of highly diversified repertoire

    Mol. Immunol.

    (2009)
  • A. Villa et al.

    V(D)J recombination defects in lymphocytes due to RAG mutations: severe immunodeficiency with a spectrum of clinical presentations

    Blood

    (2001)
  • F.A. Zuckermann et al.

    Definition of the specificity of monoclonal antibodies against porcine CD45 and CD45R: report from the CD45/CD45R and CD44 subgroup of the Second International Swine CD Workshop

    Vet. Immunol. Immunopathol.

    (1998)
  • M.T. Bility et al.

    Generation of a humanized mouse model with both human immune system and liver cells to model hepatitis C virus infection and liver immunopathogenesis

    Nat. Protoc.

    (2012)
  • M.T. Bility et al.

    Hepatitis B virus infection and immunopathogenesis in a humanized mouse model: induction of human-specific liver fibrosis and M2-like macrophages

    PLoS Pathog.

    (2014)
  • J.J. Bleesing et al.

    Human B cells express a CD45 isoform that is similar to murine B220 and is downregulated with acquisition of the memory B-cell marker CD27

    Cytom. B. Clin. Cytom.

    (2003)
  • R.L. Coffman et al.

    B220: a B cell-specific member of th T200 glycoprotein family

    Nature

    (1981)
  • Cited by (33)

    • The RAG2 gene of yellow catfish (Tachysurus fulvidraco)and its immune response against Edwardsiella ictaluri infection

      2019, Developmental and Comparative Immunology
      Citation Excerpt :

      Initially, rag 2 was recognized as an initiator of the immunoglobulin and T-cell receptor genes rearrangement by the process of V(D)J recombination (Oettinger et al., 1990a; van Gent et al., 1995a), which is required for the early stages of B- and T-cell development during the adaptive immune response of vertebrates (Notarangelo et al., 1999). It was further reported that rag 2-deficient pigs with no mature B- and T-cells, lack medulla in the thymus, lymphoid accumulations in the spleen and aggregated lymphoid nodules in the ilea (Suzuki et al., 2016). Recently, rag 2 has been shown to have a close association with the production of pro-inflammatory cytokines (TNF-α and IL-1β) during bacterial infection (Mao et al., 2012).

    View all citing articles on Scopus
    View full text