Hostname: page-component-8448b6f56d-dnltx Total loading time: 0 Render date: 2024-04-24T12:46:06.139Z Has data issue: false hasContentIssue false

Improved efficiencies in the generation of multigene-modified pigs by recloning and using sows as the recipient

Published online by Cambridge University Press:  28 June 2021

Jongki Cho
Affiliation:
College of Veterinary Medicine, Chungnam National University, Daejeon34134, Republic of Korea
Ghangyong Kim
Affiliation:
College of Veterinary Medicine, Chungnam National University, Daejeon34134, Republic of Korea
Ahmad Yar Qamar
Affiliation:
College of Veterinary Medicine, Chungnam National University, Daejeon34134, Republic of Korea
Xun Fang
Affiliation:
College of Veterinary Medicine, Chungnam National University, Daejeon34134, Republic of Korea
Pantu Kumar Roy
Affiliation:
College of Veterinary Medicine, Chungnam National University, Daejeon34134, Republic of Korea
Bereket Molla Tanga
Affiliation:
College of Veterinary Medicine, Chungnam National University, Daejeon34134, Republic of Korea
Seonggyu Bang
Affiliation:
College of Veterinary Medicine, Chungnam National University, Daejeon34134, Republic of Korea
Jong Keun Kim
Affiliation:
Department of Animal Resource and Science, Dankook University, Cheonan-si, Chungnam 31116, Republic of Korea
Cesare Galli
Affiliation:
Avantea, Laboratory of Reproductive Technologies, Via Porcellasco 7/f, Cremona26100, Italy
Andrea Perota
Affiliation:
Avantea, Laboratory of Reproductive Technologies, Via Porcellasco 7/f, Cremona26100, Italy
Young Tae Kim
Affiliation:
Department of Thoracic and Cardiovascular Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul03080, Republic of Korea
Jeong-Hwan Che
Affiliation:
Biomedical Center for Animal Resource and Development, Seoul National University College of Medicine, Seoul03080, Republic of Korea
Chung-Gyu Park*
Affiliation:
Department of Microbiology and Immunology; Xenotransplantation Research Center, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
*
Author for correspondence: Chung-Gyu Park. Department of Microbiology and Immunology; Xenotransplantation Research Center, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea. Email: chgpark@snu.ac.kr

Summary

This study was performed to improve production efficiency at the level of recipient pig and donor nuclei of transgenic cloned pigs used for xenotransplantation. To generate transgenic pigs, human endothelial protein C receptor (hEPCR) and human thrombomodulin (hTM) genes were introduced using the F2A expression vector into GalT–/–/hCD55+ porcine neonatal ear fibroblasts used as donor cells and cloned embryos were transferred to the sows and gilts. Cloned fetal kidney cells were also used as donor cells for recloning to increase production efficiency. Pregnancy and parturition rates after embryo transfer and preimplantation developmental competence were compared between cloned embryos derived from adult and fetal cells. Significantly higher parturition rates were shown in the group of sows (50.0 vs. 4.1%), natural oestrus (20.8 vs. 0%), and ovulated ovary (16.7 vs. 5.6%) compared with gilt, induced and non-ovulated, respectively (P < 0.05). When using gilts as recipients, final parturitions occurred in only the fetal cell groups and significantly higher blastocyst rates (15.1% vs. 21.3%) were seen (P < 0.05). Additionally, gene expression levels related to pluripotency were significantly higher in the fetal cell group (P < 0.05). In conclusion, sows can be recommended as recipients due to their higher efficiency in the generation of transgenic cloned pigs and cloned fetal cells also can be recommended as donor cells through correct nuclear reprogramming.

Type
Research Article
Copyright
© The Author(s), 2021. Published by Cambridge University Press

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Ashworth, CJ, Sales, DI and Wilmut, I (1989). Evidence of an association between the survival of embryos and the periovulatory plasma progesterone concentration in the ewe. J Reprod Fertil 87, 2332.CrossRefGoogle ScholarPubMed
Bari, F, Khalid, M, Haresign, W, Murray, A and Merrell, B (2003). Factors affecting the survival of sheep embryos after transfer within a MOET program. Theriogenology 59, 1265–75.CrossRefGoogle ScholarPubMed
Betthauser, J, Forsberg, E, Augenstein, M, Childs, L, Eilertsen, K, Enos, J, Forsythe, T, Golueke, P, Jurgella, G, Koppang, R, Lesmeister, T, Mallon, K, Mell, G, Misica, P, Pace, M, Pfister-Genskow, M, Strelchenko, N, Voelker, G, Watt, S, Thompson, S and Bishop, M (2000). Production of cloned pigs from in vitro systems. Nat Biotechnol 18, 1055–9.CrossRefGoogle ScholarPubMed
Campbell, KH, Fisher, P, Chen, WC, Choi, I, Kelly, RD, Lee, JH and Xhu, J (2007). Somatic cell nuclear transfer: past, present and future perspectives. Theriogenology 68 Suppl 1, S214–31.CrossRefGoogle ScholarPubMed
Cao, Z, Li, Y, Wen, X, Li, Z, Mi, C, Zhang, Z, Li, N and Li, Q (2014). Recloned transgenic pigs possess normal reproductive performance and stable genetic transmission capacity. Zygote 22, 1824.CrossRefGoogle ScholarPubMed
Cho, B, Kim, SJ, Lee, EJ, Ahn, SM, Lee, JS, Ji, DY, Lee, SH and Kang, JT (2018). Production of cloned pigs derived from double gene knockout cells using CRISPR/Cas9 system and MACS-based enrichment system. J Emb Trans 33, 245–54.Google Scholar
Cho, J, Kim, H, Kang, DW, Yanagisawa, M and Ko, C (2012). Endothelin B receptor is not required but necessary for finite regulation of ovulation. Life Sci 91, 613–7.CrossRefGoogle Scholar
Clark, LK, Schinckel, AP, Singleton, WL, Einstein, ME and Teclaw, RF (1989). Use of farrowing rate as a measure of fertility of boars. J Am Vet Med Assoc 194, 239–43.Google ScholarPubMed
Fang, X, Qamar, AY, Shin, ST and Cho, J (2019). Improved preimplantation development of cloned porcine embryos through supplementation of histone deacetylase inhibitor MS-275. J Vet Clin 36, 253–8.CrossRefGoogle Scholar
Fujimura, T, Murakami, H, Kurome, M, Takahagi, Y, Shigehisa, T and Nagashima, H (2008). Effects of recloning on the efficiency of production of a1,3-galactosyltransferase knockout pigs. J Reprod Dev 54, 5862.CrossRefGoogle Scholar
Gutierrez, K, Dicks, N, Glanzner, WG, Agellon, LB and Bordignon, V (2015). Efficacy of the porcine species in biomedical research. Front Genet 6, 293.CrossRefGoogle ScholarPubMed
Han, NR, Baek, S, Lee, Y, Lee, J, Yun, JI, Lee, E and Lee, ST (2019). Establishment of in-vitro culture system for enhancing production of somatic cell nuclear transfer (SCNT) blastocysts with high performance in the colony formation and formation of colonies derived from SCNT blastocysts in pigs. J Anim Reprod Biotechnol 34, 130–8.CrossRefGoogle Scholar
Han, YM, Kang, YK, Koo, DB and Lee, KK (2003). Nuclear reprogramming of cloned embryos produced in vitro . Theriogenology 59, 3344.CrossRefGoogle ScholarPubMed
Hua, Z, Xu, G, Liu, X, Bi, Y, Xiao, H, Hua, W, Li, L, Zhang, L, Ren, H and Zheng, X (2016). Impact of different sources of donor cells upon the nuclear transfer efficiency in Chinese indigenous Meishan pig. Pol J Vet Sci 19, 205–12.CrossRefGoogle ScholarPubMed
Huan, Y, Hu, K, Xie, B, Shi, Y, Wang, F, Zhou, Y, Liu, S, Huang, B, Zhu, J, Liu, Z, He, Y, Li, J, Kong, Q and Liu, Z (2015). Ovulation statuses of surrogate gilts are associated with the efficiency of excellent pig cloning. PLoS One 10, e0142549.CrossRefGoogle ScholarPubMed
Iwase, H, Hara, H, Ezzelarab, M, Li, T, Zhang, Z, Gao, B, Liu, H, Long, C, Wang, Y, Cassano, A, Klein, E, Phelps, C, Ayares, D, Humar, A, Wijkstrom, M and Cooper, DKC (2017). Immunological and physiological observations in baboons with life-supporting genetically engineered pig kidney grafts. Xenotransplantation 24, 10.1111/xen.12293.CrossRefGoogle Scholar
Jeon, RH, Kim, SJ and Lee, WJ (2017). The effect of preferable enrichments in the laboratory minipigs. J Emb Trans 32, 305–10.Google Scholar
Ji, SJ, Lee, G, Park, SH, Kim, KW, Byun, SJ, Ock, SA, Hwang, S, Woo, JS and Oh, KB (2017). Reproductive characteristic of transgenic Massachusetts general hospital miniature pigs for xenotransplantation. J Emb Trans 32, 165–70.Google Scholar
Kim, GA, Jin, JX, Lee, S, Taweechaipaisankul, A, Oh, HJ, Hwang, JI, Ahn, C, Saadeldin, IM and Lee, BC (2017). Postneonatal mortality and liver changes in cloned pigs associated with human tumor necrosis factor receptor I-FC and human heme oxygenase-1 overexpression. Biomed Res Int 2017, 5276576.CrossRefGoogle ScholarPubMed
Kim, G, Roy, PK, Fang, X, Hassan, BM and Cho, J (2019). Improved preimplantation development of porcine somatic cell nuclear transfer embryos by caffeine treatment. J Vet Sci 20, e31.CrossRefGoogle ScholarPubMed
Koo, OJ, Kang, JT, Kwon, DK, Park, HJ and Lee, BC (2010). Influence of ovulation status, seasonality and embryo transfer method on development of cloned porcine embryos. Reprod Domest Anim 45, 773–8.Google ScholarPubMed
Krupalnik, V and Hanna, JH (2014). Stem cells: the quest for the perfect reprogrammed cell. Nature 511, 160–2.CrossRefGoogle ScholarPubMed
Kurome, M, Ishikawa, T, Tomii, R, Ueno, S, Shimada, A, Yazawa, H and Nagashima, H (2008). Production of transgenic and non-transgenic clones in miniature pigs by somatic cell nuclear transfer. J Reprod Dev 54, 156–63.CrossRefGoogle ScholarPubMed
Kwon, DJ, Kim, DH, Hwang, IS, Kim, DE, Kim, HJ, Kim, JS, Lee, K, Im, GS, Lee, JW and Hwang, S (2017). Generation of α-1,3-galactosyltransferase knocked-out transgenic cloned pigs with knocked-in five human genes. Transgenic Res 26, 153–63.CrossRefGoogle ScholarPubMed
Lai, L and Prather, RS (2003). Production of cloned pigs by using somatic cells as donors. Cloning Stem Cells 5, 233–41.CrossRefGoogle ScholarPubMed
Lee, GS, Hyun, SH, Kim, HS, Kim, DY, Lee, SH, Lim, JM, Lee, ES, Kang, SK, Lee, BC and Hwang, WS (2003). Improvement of a porcine somatic cell nuclear transfer technique by optimizing donor cell and recipient oocyte preparations. Theriogenology 59, 1949–57.CrossRefGoogle ScholarPubMed
Lee, G, Park, SH, Lee, H, Ji, SJ, Lee, JY, Byun, SJ, Hwang, S, Kim, KW, Ock, SA and Oh, KB (2017). Development of α1,3-galactosyltransferase inactivated and human membrane cofactor protein expressing homozygous transgenic pigs for xenotransplantation. J Emb Trans 32, 73–9.Google Scholar
Liu, T, Dou, H, Xiang, X, Li, L, Li, Y, Lin, L, Pang, X, Zhang, Y, Chen, Y, Luan, J, Xu, Y, Yang, Z, Yang, W, Liu, H, Li, F, Wang, H, Yang, H, Bolund, L, Vajta, G and Du, Y (2015). Factors determining the efficiency of porcine somatic cell nuclear transfer: data analysis with over 200,000 reconstructed embryos. Cell Reprogram 17, 463–71.CrossRefGoogle ScholarPubMed
Livak, KJ and Schmittgen, TD (2001). Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods 25, 402408.CrossRefGoogle Scholar
Luo, Y, Lin, L, Bolund, L, Jensen, TG and Sørensen, CB (2012). Genetically modified pigs for biomedical research. J Inherit Metab Dis 35, 695713.CrossRefGoogle ScholarPubMed
Macher, BA and Galili, U (2008). The Galα1,3Galβ1,4GlcNAc-R (α-Gal) epitope: a carbohydrate of unique evolution and clinical relevance. Biochim Biophys Acta 1780, 7588.CrossRefGoogle ScholarPubMed
Matoba, S and Zhang, Y (2018). Somatic cell nuclear transfer reprogramming: mechanisms and applications. Cell Stem Cell 23, 471–85.CrossRefGoogle ScholarPubMed
Nagaya, M, Watanabe, M, Kobayashi, M, Nakano, K, Arai, Y, Asano, Y, Takeishi, T, Umeki, I, Fukuda, T, Yashima, S, Takayanagi, S, Watanabe, N, Onodera, M, Matsunari, H, Umeyama, K and Nagashima, H (2016). A transgenic-cloned pig model expressing non-fluorescent modified plum. J Reprod Dev 62, 511–20.CrossRefGoogle ScholarPubMed
Niemann, H and Petersen, B (2016). The production of multi-transgenic pigs: update and perspectives for xenotransplantation. Transgenic Res 25, 361–74.CrossRefGoogle ScholarPubMed
Onishi, A, Iwamoto, M, Akita, T, Mikawa, S, Takeda, K, Awata, T, Hanada, H and Perry, AC (2000). Pig cloning by microinjection of fetal fibroblast nuclei. Science 289, 1188–90.CrossRefGoogle ScholarPubMed
Park, SJ, Cho, B, Koo, OJ, Kim, H, Kang, JT, Hurh, S, Kim, SJ, Yeom, HJ, Moon, J, Lee, EM, Choi, JY, Hong, JH, Jang, G, Hwang, JI, Yang, J, Lee, BC and Ahn, C (2014). Production and characterization of soluble human TNFRI-Fc and human HO-1(HMOX1) transgenic pigs by using the F2A peptide. Transgenic Res 23, 407–19.CrossRefGoogle ScholarPubMed
Perleberg, C, Kind, A and Schnieke, A (2018). Genetically engineered pigs as models for human disease. Dis Model Mech 11, dmm030783.CrossRefGoogle ScholarPubMed
Perota, A, Lagutina, I, Colleoni, S, Duchi, R, Lazzari, G, Cozzi, E, Lucchini, F and Galli, C (2011). Efficient expression of human endothelial protein C receptor and human thrombomodulin in transfected pig primary hCD55+-Gal–/– fibroblast using F2A expression vector. Reprod Fertil Dev 24, 229–30.CrossRefGoogle Scholar
Petersen, B, Lucas-Hahn, A, Oropeza, M, Hornen, N, Lemme, E, Hassel, P, Queisser, AL and Niemann, H (2008). Development and validation of a highly efficient protocol of porcine somatic cloning using preovulatory embryo transfer in peripubertal gilts. Cloning Stem Cells 10, 355–62.CrossRefGoogle ScholarPubMed
Polejaeva, IA, Chen, SH, Vaught, TD, Page, RL, Mullins, J, Ball, S, Dai, Y, Boone, J, Walker, S, Ayares, DL, Colman, A and Campbell, KH (2000). Cloned pigs produced by nuclear transfer from adult somatic cells. Nature 407, 8690.CrossRefGoogle ScholarPubMed
Richter, A, Kurome, M, Kessler, B, Zakhartchenko, V, Klymiuk, N, Nagashima, H, Wolf, E and Wuensch, A (2012). Potential of primary kidney cells for somatic cell nuclear transfer mediated transgenesis in pig. BMC Biotechnol 12, 84.CrossRefGoogle ScholarPubMed
Roy, PK, Qamar, AY, Fang, X, Hassan, BMS and Cho, J (2020). Effects of cobalamin on meiotic resumption and developmental competence of growing porcine oocytes. Theriogenology 154, 2430.CrossRefGoogle ScholarPubMed
Roy, PK, Qamar, AY, Fang, X, Kim, G, Bang, S, De Zoysa, M, Shin, ST and Cho, J (2021). Chitosan nanoparticles enhance developmental competence of in vitro-matured porcine oocytes. Reprod Domest Anim 56, 342–50.CrossRefGoogle Scholar
Shimizu, I, Smith, NR, Zhao, G, Medof, E and Sykes, M (2006). Decay-accelerating factor prevents acute humoral rejection induced by low levels of anti-aGal natural antibodies. Transplantation 81, 95100.CrossRefGoogle Scholar
Wei, H, Qing, Y, Pan, W, Zhao, H, Li, H, Cheng, W, Zhao, L, Xu, C, Li, H, Li, S, Ye, L, Wei, T, Li, X, Fu, G, Li, W, Xin, J and Zeng, Y (2013). Comparison of the efficiency of Banna miniature inbred pig somatic cell nuclear transfer among different donor cells. PLoS One 8, e57728.CrossRefGoogle ScholarPubMed
Weng, XG, Cai, MM, Zhang, YT, Liu, Y, Liu, C and Liu, ZH (2020). Improvement in the in vitro development of cloned pig embryos after kdm4a overexpression and an H3K9me3 methyltransferase inhibitor treatment. Theriogenology 146, 162–70.CrossRefGoogle Scholar
Yazaki, S, Iwamoto, M, Onishi, A, Miwa, Y, Hashimoto, M, Oishi, T, Suzuki, S, Fuchimoto, D, Sembon, S, Furusawa, T, Liu, D, Nagasaka, T, Kuzuya, T, Ogawa, H, Yamamoto, K, Iwasaki, K, Haneda, M, Maruyama, S and Kobayashi, T (2012). Production of cloned pigs expressing human thrombomodulin in endothelial cells. Xenotransplantation 19, 8291.CrossRefGoogle ScholarPubMed
Zhang, X, Li, X, Yang, Z, Tao, K, Wang, Q, Dai, B, Qu, S, Peng, W, Zhang, H, Cooper, DKC and Dou, K (2019). A review of pig liver xenotransplantation: current problems and recent progress. Xenotransplantation 26, e12497.CrossRefGoogle ScholarPubMed
Zhang, Z, Zhai, Y, Ma, X, Zhang, S, An, X, Yu, H and Li, Z (2018). Down-regulation of H3K4me3 by MM-102 facilitates epigenetic reprogramming of porcine somatic cell nuclear transfer embryos. Cell Physiol Biochem 45, 1529–40.CrossRefGoogle ScholarPubMed
Zhao, H, Li, Y, Wiriyahdamrong, T, Yuan, Z, Qing, Y, Li, H, Xu, K, Guo, J, Jia, B, Zhang, X, Cheng, W, Su, Y, Long, W, Wang, J, Zou, D, Kinoshita, K, Zhao, HY and Wei, HJ (2020). Improved production of GTKO/hCD55/hCD59 triple-gene-modified Diannan miniature pigs for xenotransplantation by recloning. Transgenic Res 29, 369–79.CrossRefGoogle ScholarPubMed