Please wait a minute...
 首页  期刊列表 期刊订阅 开放获取 关于我们
English
最新录用  |  在线预览  |  当期目录  |  过刊浏览  |  学科浏览  |  专题文章  |  热点文章  |  下载排行
Frontiers of Agricultural Science and Engineering    2020, Vol. 7 Issue (2) : 136-147     https://doi.org/10.15302/J-FASE-2019306
REVIEW
Developments in genetic modification of cattle and implications for regulation, safety and traceability
Jan Pieter VAN DER BERG1(), Gijs A. KLETER1, Evy BATTAGLIA1, Martien A. M. GROENEN2, Esther J. KOK1
1. Wageningen Food Safety Research, Wageningen University and Research, 6700 AE Wageningen, the Netherlands
2. Animal Breeding and Genomics, Wageningen University and Research, 6700 AH Wageningen, the Netherlands
全文: PDF(274 KB)   HTML
导出: BibTeX | EndNote | Reference Manager | ProCite | RefWorks
Abstract

Genetic modification techniques, in particular novel gene editing technologies, hold the yet unfulfilled promise of altering genetic traits in farm animals more efficiently than by crossbreeding, allowing for a more rapid development of new cattle breeds with distinct traits. Gene editing technologies allow for the directed alteration of specific traits and thereby have the potential to enhance, for instance, disease resilience, production yield and the production of desired substances in milk. The potential implications of these technological advancements, which are often combined with animal cloning methods, are discussed both for animal health and for consumer safety, also with consideration of available methods for the detection and identification of the related products in the food supply chain. Finally, an overview is provided of current regulatory approaches in the European Union (EU) and major countries exporting beef to the EU, for products from animals bred through established practices as well as modern biotechnologies.

Keywords cattle      food safety      gene editing      genetic modification      GMO detection      regulation     
最新录用日期:    在线预览日期:    发布日期: 2020-04-28
服务
推荐给朋友
免费邮件订阅
RSS订阅
作者相关文章
Jan Pieter VAN DER BERG
Gijs A. KLETER
Evy BATTAGLIA
Martien A. M. GROENEN
Esther J. KOK
引用本文:   
Jan Pieter VAN DER BERG,Gijs A. KLETER,Evy BATTAGLIA, et al. Developments in genetic modification of cattle and implications for regulation, safety and traceability[J]. Front. Agr. Sci. Eng. , 2020, 7(2): 136-147.
网址:  
https://journal.hep.com.cn/fase/EN/10.15302/J-FASE-2019306     OR     https://journal.hep.com.cn/fase/EN/Y2020/V7/I2/136
Trait Method Trait Reference
Milk composition and human Transgenesis using microinjection Introduction of gene encoding human lactoferrin [15]
Protein production Transgenesis using microinjection Introduction of gene encoding human α-lactalbumin [16,17]
Transgenesis in somatic cells, SCNT Introduction of gene encoding human bile salt-stimulated lipase [18]
Transgenesis in somatic cells, SCNT Introduction of gene encoding human immunoglobulin [19,20]
Transgenesis in somatic cells, SCNT Introduction of additional gene copies encoding bovine α- & k-casein [21]
Transgenesis in somatic cells, SCNT Introduction of gene encoding human lysozyme [22]
Gene editing using zinc finger nucleases, NHEJ repair, SCNT Disruption of β-lactoglobulin gene [12]
Transgenesis in somatic cells, SCNT Introduction of gene encoding humanized Caenorhabditis elegans n-3 fatty acid desaturase [11]
Transgenesis in somatic cells, SCNT Introduction of gene encoding human β-defensin-3 [23]
Transgenesis in somatic cells using TALENs & SCNT Introduction of gene encoding Sulfolobus solfataricus lactase [24]
Disease resilience
- Mastitis Transgenesis in somatic cells, SCNT Introduction of gene encoding Staphylococcus simulans lysostaphin [25]
- Bovine spongiform
encephalopathy
Transgenesis and embryonic cloning Disruption of prion protein via integration of knockout vectors [26]
- Mannheimia hemolytica
leukotoxin
Gene editing and homology-directed repair, SCNT Gene-edited CD18, substitution of a glutamine for a glycine codon in its signal peptide [27]
- Bovine tuberculosis Transgenesis in somatic cells using TALENs & SCNT Introduction of mouse nuclear body protein encoding gene SP110 [28]
Transgenesis in somatic cells using Cas9n & SCNT Introduction of additional genes encoding solute carrier family gene NRAMP1 [13]
Hornlessness Gene editing and homology-directed repair, SCNT Introduction of bovine Pc POLLED allele, resulting in hornless phenotype [29]
Thermotolerance Gene editing using TALENs, SCNT Introduction of the SLICK locus for improved thermotolerance [30,31]
Increased muscle growth Gene editing using TALEN mRNA Introduction of small deletions in the myostatin (GDF8) gene by use of gene editing [14]
Other fundamental research
-Reverse transcribed
gene transfer
Transgenesis via pronuclear injection of retroviral vector DNA Integration of hepatitis B surface antigen gene using a retroviral vector [32]
-Marker assisted
selection
Transgenesis via transfection of somatic cells with retroviral vector DNA, SCNT Integration of a β-galactosidase-neomycin fusion gene driven by cytomegalovirus promoter [33]
-Lentiviral infection Transgenesis in somatic cells using lentiviral vectors, SCNT Integration of eGFP using lentiviral vector [34]
-Transposon integration Transgenesis via microinjection of transposon DNA Sleeping Beauty and Piggybac transposons used to deliver sequences containing fluorescent protein genes [35]
-Targeted integration Transgenesis in somatic cells using TALENs & SCNT Integration of eGFP in Rosa26 “safe locus” [36]
Tab.1  Examples of transgenic and gene-edited cattle
Fig.1  Timeline of technological developments and milestones used to obtain GM cattle. MI, pronuclear microinjection; SCNT, somatic cell nuclear transfer; SDN, site-directed endonucleases. Important milestones are summarized on the right.
Country Animal cloning Transgenic livestock Gene-edited livestock Reference
EU member states Prohibited, until specific regulations on animal cloning are in place Requires approval according to EU Directive 2001/18/EC and Regulation (EC) No. 1829/2003, safety assessment performed by EFSA GMO Panel Requires approval according to EU Directive 2001/18/EC and Regulation (EC) No. 1829/2003, safety assessment performed by EFSA GMO Panel [52]
USA Allowed, a risk management plan and guidance for industry have been issued by the FDA Requires approval according to Federal FD&C Act, regulations for new animal drugs as stated in 2009 FDA Guidance for industry #187 (Draft guidance) and NEPA Requires approval according to Federal FD&C Act, regulations for new animal drugs as stated in 2009 FDA Guidance for industry #187 (Draft guidance) and NEPA [5355]
Canada Allowed, food products of cloned animals and clone progeny are considered “novel foods” and require pre-market safety assessments according to the regulations in Division 28, Part B, of the Food and Drug Regulations (Novel Foods) Requires approval according to the Canadian Environmental Protection Act, 1999, the New Substances Notification Regulations (Organisms) and Food and Drugs Act No specific policy on gene editing, may be considered “novel” and require case-by-case safety assessment by Health Canada [56,57]
Argentina Allowed Requires approval according to animal biotechnology regulation, case-by-case assessment by CONABIA Requires approval according to animal biotechnology regulation, case-by-case assessment by CONABIA [58]
Brazil Allowed, commercial animal cloning mostly in partnership with EMBRAPA, registration of cloned cattle at ABCZ Requires approval according to animal biotechnology regulation, case-by-case assessment by CTNBio Requires approval according to animal biotechnology regulation, case-by-case assessment by CTNBio, gene-edited animals lacking recombinant DNA are regarded non-GM according to Normative Resolution #16 [46]
Australia & New Zealand Allowed, generally in confined research environment Requires approval according to Gene Technology Act 2000, by OGTR Requires approval according to Gene Technology Act 2000, by OGTR, gene editing techniques that do not introduce new genetic material are not regulated as GMOs [59,60]
Uruguay No specific legislation on animal cloning, animal biotechnology performed in research institutes such as Institut Pasteur in Montevideo and the Animal Reproduction Institute of Uruguay No specific legislation on animal biotechnology, environmental release of GMOs and biosecurity is subject to prior authorization by competent authorities, as stated in article 23 of law No. 17283 on the protection of the environment No specific legislation on gene editing in animals, during a meeting of the CAS the minister of agriculture signed a declaration in favor of gene editing. Gene-edited animals may be subject to prior authorization according to law No. 17283 [61,62]
Tab.2  Regulation of animal cloning, transgenesis and gene editing
1 C Tait-Burkard, A Doeschl-Wilson, M J McGrew, A L Archibald, H M Sang, R D Houston, C B Whitelaw, M Watson. Livestock 2.0—genome editing for fitter, healthier, and more productive farmed animals. Genome Biology, 2018, 19(1): 204
https://doi.org/10.1186/s13059-018-1583-1 pmid: 30477539
2 U.S. Food and Drug Administration (FDA). AquAdvantage Salmon Approval Letter and Appendix (NADA 141–454). Available at FDA website on September 1, 2019
3 F Miglior, A Fleming, F Malchiodi, L F Brito, P Martin, C F A Baes. A 100-year review: identification and genetic selection of economically important traits in dairy cattle. Journal of Dairy Science, 2017, 100(12): 10251–10271
https://doi.org/10.3168/jds.2017-12968 pmid: 29153164
4 W Haires. What is an EBV and how can it help you? Available at The Cattle Site on September 1, 2019
5 R Mrode, J M K Ojango, A M Okeyo, J M Mwacharo. Genomic selection and use of molecular tools in breeding programs for indigenous and crossbred cattle in developing countries: current status and future prospects. Frontiers in Genetics, 2019, 9: 694
https://doi.org/10.3389/fgene.2018.00694 pmid: 30687382
6 B J Hayes, H A Lewin, M E Goddard. The future of livestock breeding: genomic selection for efficiency, reduced emissions intensity, and adaptation. Trends in Genetics, 2013, 29(4): 206–214
https://doi.org/10.1016/j.tig.2012.11.009 pmid: 23261029
7 S G Moore, J F A Hasler. A 100-year review: reproductive technologies in dairy science. Journal of Dairy Science, 2017, 100(12): 10314–10331
https://doi.org/10.3168/jds.2017-13138 pmid: 29153167
8 M Mikkola. Superovulation and embryo transfer in dairy cattle —effect of management factors with emphasis on sex-sorted semen (Academic Dissertation). Finland: University of Helsinki, 2017
9 G E Seidel, S Moore Seidel. Training Manual for Embryo Transfer in Cattle (FAO Animal Production and Health Paper 77). Rome: Food and Agriculture Organization of the United Nations, 1991
10 K C S Tavares, I S Carneiro, D B Rios, C Feltrin, A K C Ribeiro, S Gaudêncio-Neto, L T Martins, L H Aguiar, C R Lazzarotto, C E M Calderón, F E M Lopes, L P R Teixeira, M Bertolini, L R Bertolini. A fast and simple method for the polymerase chain reaction-based sexing of livestock embryos. Genetics and Molecular Research, 2016, 15(1): gmr7476
https://doi.org/10.4238/gmr.15017476 pmid: 27050974
11 X Wu, H Ouyang, B Duan, D Pang, L Zhang, T Yuan, L Xue, D Ni, L Cheng, S Dong, Z Wei, L Li, M Yu, Q Y Sun, D Y Chen, L Lai, Y Dai, G P Li. Production of cloned transgenic cow expressing omega-3 fatty acids. Transgenic Research, 2012, 21(3): 537–543
https://doi.org/10.1007/s11248-011-9554-2 pmid: 21918821
12 S Yu, J Luo, Z Song, F Ding, Y Dai, N Li. Highly efficient modification of beta-lactoglobulin (BLG) gene via zinc-finger nucleases in cattle. Cell Research, 2011, 21(11): 1638–1640
https://doi.org/10.1038/cr.2011.153 pmid: 21912434
13 Y Gao, H Wu, Y Wang, X Liu, L Chen, Q Li, C Cui, X Liu, J Zhang, Y Zhang. Single Cas9 nickase induced generation of NRAMP1 knockin cattle with reduced off-target effects. Genome Biology, 2017, 18(1): 13
https://doi.org/10.1186/s13059-016-1144-4 pmid: 28143571
14 C Proudfoot, D F Carlson, R Huddart, C R Long, J H Pryor, T J King, S G Lillico, A J Mileham, D G McLaren, C B A Whitelaw, S C Fahrenkrug. Genome edited sheep and cattle. Transgenic Research, 2015, 24(1): 147–153
https://doi.org/10.1007/s11248-014-9832-x pmid: 25204701
15 P Krimpenfort, A Rademakers, W Eyestone, A van der Schans, S van den Broek, P Kooiman, E Kootwijk, G Platenburg, F Pieper, R Strijker, H de Boer. Generation of transgenic dairy cattle using ‘in vitro’ embryo production. Nature Biotechnology, 1991, 9(9): 844–847
https://doi.org/10.1038/nbt0991-844 pmid: 1367358
16 J Wang, P Yang, B Tang, X Sun, R Zhang, C Guo, G Gong, Y Liu, R Li, L Zhang, Y Dai, N Li. Expression and characterization of bioactive recombinant human α-lactalbumin in the milk of transgenic cloned cows. Journal of Dairy Science, 2008, 91(12): 4466–4476
https://doi.org/10.3168/jds.2008-1189 pmid: 19038921
17 W H Eyestone. Production and breeding of transgenic cattle using in vitro embryo production technology. Theriogenology, 1999, 51(2): 509–517
https://doi.org/10.1016/S0093-691X(98)00244-1 pmid: 10729109
18 S H Chen, T D Vaught, J A Monahan, J Boone, E Emslie, P M Jobst, A E Lamborn, A Schnieke, L Robertson, A Colman, Y Dai, I A Polejaeva, D L Ayares. Efficient production of transgenic cloned calves using preimplantation screening. Biology of Reproduction, 2002, 67(5): 1488–1492
https://doi.org/10.1095/biolreprod.102.006981 pmid: 12390880
19 Y Kuroiwa, P Kasinathan, Y J Choi, R Naeem, K Tomizuka, E J Sullivan, J G Knott, A Duteau, R A Goldsby, B A Osborne, I Ishida, J M Robl. Cloned transchromosomic calves producing human immunoglobulin. Nature Biotechnology, 2002, 20(9): 889–894
https://doi.org/10.1038/nbt727 pmid: 12172556
20 L Grosse-Hovest, S Müller, R Minoia, E Wolf, V Zakhartchenko, H Wenigerkind, C Lassnig, U Besenfelder, M Müller, S D Lytton, G Jung, G Brem. Cloned transgenic farm animals produce a bispecific antibody for T cell-mediated tumor cell killing. Proceedings of the National Academy of Sciences of the United States of America, 2004, 101(18): 6858–6863
https://doi.org/10.1073/pnas.0308487101 pmid: 15105446
21 B Brophy, G Smolenski, T Wheeler, D Wells, P L’Huillier, G Laible. Cloned transgenic cattle produce milk with higher levels of β-casein and k-casein. Nature Biotechnology, 2003, 21(2): 157–162
https://doi.org/10.1038/nbt783 pmid: 12548290
22 B Yang, J Wang, B Tang, Y Liu, C Guo, P Yang, T Yu, R Li, J Zhao, L Zhang, Y Dai, N Li. Characterization of bioactive recombinant human lysozyme expressed in milk of cloned transgenic cattle. PLoS One, 2011, 6(3): e17593
https://doi.org/10.1371/journal.pone.0017593 pmid: 21436886
23 Y Yu, Y Wang, Q Tong, X Liu, F Su, F Quan, Z Guo, Y Zhang. A site-specific recombinase-based method to produce antibiotic selectable marker free transgenic cattle. PLoS One, 2013, 8(5): e62457
https://doi.org/10.1371/journal.pone.0062457 pmid: 23658729
24 X Su, S Wang, G Su, Z Zheng, J Zhang, Y Ma, Z Liu, H Zhou, Y Zhang, L Zhang. Production of microhomologous-mediated site-specific integrated LacS gene cow using TALENs. Theriogenology, 2018, 119: 282–288
https://doi.org/10.1016/j.theriogenology.2018.07.011 pmid: 30075414
25 R J Wall, A M Powell, M J Paape, D E Kerr, D D Bannerman, V G Pursel, K D Wells, N Talbot, H W Hawk. Genetically enhanced cows resist intramammary Staphylococcus aureus infection. Nature Biotechnology, 2005, 23(4): 445–451
https://doi.org/10.1038/nbt1078 pmid: 15806099
26 J A Richt, P Kasinathan, A N Hamir, J Castilla, T Sathiyaseelan, F Vargas, J Sathiyaseelan, H Wu, H Matsushita, J Koster, S Kato, I Ishida, C Soto, J M Robl, Y Kuroiwa. Production of cattle lacking prion protein. Nature Biotechnology, 2007, 25(1): 132–138
https://doi.org/10.1038/nbt1271 pmid: 17195841
27 S Shanthalingam, A Tibary, J E Beever, P Kasinathan, W C Brown, S Srikumaran. Precise gene editing paves the way for derivation of Mannheimia haemolytica leukotoxin-resistant cattle. Proceedings of the National Academy of Sciences of the United States of America, 2016, 113(46): 13186–13190
https://doi.org/10.1073/pnas.1613428113 pmid: 27799556
28 H Wu, Y Wang, Y Zhang, M Yang, J Lv, J Liu, Y Zhang. TALE nickase-mediated SP110 knockin endows cattle with increased resistance to tuberculosis. Proceedings of the National Academy of Sciences of the United States of America, 2015, 112(13): E1530–E1539
https://doi.org/10.1073/pnas.1421587112 pmid: 25733846
29 D F Carlson, C A Lancto, B Zang, E S Kim, M Walton, D Oldeschulte, C Seabury, T S Sonstegard, S C Fahrenkrug. Production of hornless dairy cattle from genome-edited cell lines. Nature Biotechnology, 2016, 34(5): 479–481
https://doi.org/10.1038/nbt.3560 pmid: 27153274
30 J Bellini. This gene-edited calf could transform Brazil’s beef industry. Available at The Wall Street Journal website on October 1, 2018
31 H J Huson, E S Kim, R W Godfrey, T A Olson, M C McClure, C C Chase, R Rizzi, A M P O’Brien, C P Van Tassell, J F Garcia, T S Sonstegard. Genome-wide association study and ancestral origins of the slick-hair coat in tropically adapted cattle. Frontiers in Genetics, 2014, 5: 101
https://doi.org/10.3389/fgene.2014.00101 pmid: 24808908
32 A W S Chan, E J Homan, L U Ballou, J C Burns, R D Bremel. Transgenic cattle produced by reverse-transcribed gene transfer in oocytes. Proceedings of the National Academy of Sciences of the United States of America, 1998, 95(24): 14028–14033
https://doi.org/10.1073/pnas.95.24.14028 pmid: 9826647
33 J B Cibelli, S L Stice, P J Golueke, J J Kane, J Jerry, C Blackwell, F A Ponce de León, J M Robl. Cloned transgenic calves produced from nonquiescent fetal fibroblasts. Science, 1998, 280(5367): 1256–1258
https://doi.org/10.1126/science.280.5367.1256 pmid: 9596577
34 A Hofmann, V Zakhartchenko, M Weppert, H Sebald, H Wenigerkind, G Brem, E Wolf, A Pfeifer. Generation of transgenic cattle by lentiviral gene transfer into oocytes. Biology of Reproduction, 2004, 71(2): 405–409
https://doi.org/10.1095/biolreprod.104.028472 pmid: 15044266
35 S Y Yum, S J Lee, H M Kim, W J Choi, J H Park, W W Lee, H S Kim, H J Kim, S H Bae, J H Lee, J Y Moon, J H Lee, C I Lee, B J Son, S H Song, S M Ji, S J Kim, G Jang. Efficient generation of transgenic cattle using the DNA transposon and their analysis by next-generation sequencing. Scientific Reports, 2016, 6(1): 27185
https://doi.org/10.1038/srep27185 pmid: 27324781
36 M Wang, Z Sun, Z Zou, F Ding, L Li, H Wang, C Zhao, N Li, Y Dai. Efficient targeted integration into the bovine Rosa26 locus using TALENs. Scientific Reports, 2018, 8(1): 10385
https://doi.org/10.1038/s41598-018-28502-x pmid: 29991797
37 W H Eyestone. Challenges and progress in the production of transgenic cattle. Reproduction, Fertility, and Development, 1994, 6(5): 647–652
https://doi.org/10.1071/RD9940647 pmid: 7569045
38 R J Wall, D E Kerr, K R Bondioli. Transgenic dairy cattle: genetic engineering on a large scale. Journal of Dairy Science, 1997, 80(9): 2213–2224
https://doi.org/10.3168/jds.S0022-0302(97)76170-8 pmid: 9313167
39 R L Krisher, J R Gibbons, R S Canseco, J L Johnson, C G Russell, D R Notter, W H Velander, F C Gwazdauskas. Influence of time of gene microinjection on development and DNA detection frequency in bovine embryos. Transgenic Research, 1994, 3(4): 226–231
https://doi.org/10.1007/BF02336775 pmid: 7920738
40 M Gagné, F Pothier, M A Sirard. Effect of microinjection time during postfertilization S-phase on bovine embryonic development. Molecular Reproduction and Development, 1995, 41(2): 184–194
https://doi.org/10.1002/mrd.1080410209 pmid: 7654372
41 C Galli, I Lagutina, A Perota, S Colleoni, R Duchi, F Lucchini, G Lazzari. Somatic cell nuclear transfer and transgenesis in large animals: current and future insights. Reproduction in Domestic Animals, 2012, 47(Suppl 3): 2–11
https://doi.org/10.1111/j.1439-0531.2012.02045.x pmid: 22681293
42 C A Hodges, S L Stice. Generation of bovine transgenics using somatic cell nuclear transfer. Reproductive Biology and Endocrinology, 2003, 1(1): 81
https://doi.org/10.1186/1477-7827-1-81 pmid: 14613543
43 S Dikmen, F A Khan, H J Huson, T S Sonstegard, J I Moss, G E Dahl, P J Hansen. The SLICK hair locus derived from Senepol cattle confers thermotolerance to intensively managed lactating Holstein cows. Journal of Dairy Science, 2014, 97(9): 5508–5520
https://doi.org/10.3168/jds.2014-8087 pmid: 24996281
44 A L Norris, S S Lee, K J Greenlees, D A Tadesse, M F Miller, H Lombardi. Template plasmid integration in germline genome-edited cattle. bioRxiv 2019 [Preprint]
https://doi.org/10.1101/715482
45 A E Young, T A Mansour, B R McNabb, J R Owen, J F Trott, C T Brown, A L Van Eenennaam. Genomic and phenotypic analyses of six offspring of a genome-edited hornless bull. Nature Biotechnology, 2019 [Published Online]
https://doi.org/10.1038/s41587-019-0266-0 pmid: 31591551
46 United States Department of Agriculture, Foreign Agricultural Service (USDA FAS). Brazil—Agricultural Biotechnology Annual, BR 1818. Washington DC: USDA FAS, 2018
47 M Drost. Complications during gestation in the cow. Theriogenology, 2007, 68(3): 487–491
https://doi.org/10.1016/j.theriogenology.2007.04.023 pmid: 17531308
48 A Schurmann, D N Wells, B Oback. Early zygotes are suitable recipients for bovine somatic nuclear transfer and result in cloned offspring. Reproduction, 2006, 132(6): 839–848
https://doi.org/10.1530/REP-06-0054 pmid: 17127744
49 D Miao, M I Giassetti, M Ciccarelli, B Lopez-Biladeau, J M Oatley. Simplified pipelines for genetic engineering of mammalian embryos by CRISPR-Cas9 electroporation. Biology of Reproduction, 2019, 101(1): 177–187
https://doi.org/10.1093/biolre/ioz075 pmid: 31095680
50 J Ryu, R S Prather, K Lee. Use of gene-editing technology to introduce targeted modifications in pigs. Journal of Animal Science and Biotechnology, 2018, 9(1): 5
https://doi.org/10.1186/s40104-017-0228-7 pmid: 29423214
51 J P van der Berg, G A Kleter, E J Kok. Regulation and safety considerations of somatic cell nuclear transfer-cloned farm animals and their offspring used for food production. Theriogenology, 2019, 135: 85–93
https://doi.org/10.1016/j.theriogenology.2019.06.001 pmid: 31203092
52 United States Department of Agriculture, Foreign Agricultural Service (USDA FAS). EU-28—Agricultural Biotechnology Annual FR1827. Washington DC: USDA FAS, 2018
53 U.S. Food and Drug Administration (FDA). Animal Cloning—Risk Management Plan for Clones and Their Progeny. Available at FDA website on August 10, 2019
54 U.S. Food and Drug Administration (FDA). Guidance for Industry—Use of Animal Clones and Clone Progeny for Human Food and Animal Feed. Available at FDA website on August 10, 2019
55 U.S. Food and Drug Administration (FDA). Guidance for Industry #187 Regulation of Intentionally Altered Genomic DNA in Animals—Draft Guidance. Available at FDA website on August 10, 2019
56 Health Canada. Food Directorate Interim Policy on Foods from Cloned Animals. Ontario: Health Canada, 2003
57 Canada Food Inspection Agency (CFIA). Animals and Animal Products Derived Through Modern Biotechnology. Available at CFIA website on August 10, 2019
58 United States Department of Agriculture, Foreign Agricultural Service (USDA FAS). Argentina—Agricultural Biotechnology Annual. Washington DC: USDA FAS, 2019
59 United States Department of Agriculture, Foreign Agricultural Service (USDA FAS). Australia—Agricultural Biotechnology Annual AS1826. Washington DC: USDA FAS, 2018
60 Office of the Gene Technology Regulator (OGTR). Technical Review of the Gene Technology Regulations 2001—Decision Regulation Impact Statement. Available at the OGTR website on August 10, 2019
61 United States Department of Agriculture, Foreign Agricultural Service (USDA FAS). Uruguay Annual Biotechnology Report 2012. Washington DC: USDA FAS, 2012
62 Government of Argentina. Argentina presented to the WTO the Declaration on Gene Edition Techniques of the Southern Agricultural Council. Available at Government of Argentina website on August 9, 2019 (in Spanish)
63 European Commission (EC). EU Meat Market Observatory—Beef and veal. Available at EC website (Beef Statistics) on August 9, 2019
64 Enropean Group on Ethics in Science and New Technologies (EGE). Ethical aspects of animal cloning for food supply. Luxembourg: Office for Official Publications of the European Communities, 2008
65 European Parliament (EP). Cloning of animals kept and reproduced for farming purposes. Brussels, Belgium: European Parliament, 2015
66 European Food Safety Authority (EFSA). Food safety, animal health and welfare and environmental impact of animals derived from cloning by somatic cell nucleus transfer (SCNT) and their offspring and products obtained from those animals. EFSA Journal, 2008, 767: 1–49
67 International Embryo Transfer Society (IETS). Health assessment and care for animals involved in the cloning process. Champaign: International Embryo Technology Society, 2008
68 S Mallapaty. Australian gene-editing rules adopt ‘middle ground’. Nature, 2019 [Published Online] doi: 10.1038/d41586-019-01282-8
69 Ministry of Livestock, Agriculture and Fisheries (MGAP), Government of Uruguay. Regulations and Documents—Legislation. Available at MGAP website on August 6, 2019 (in Spanish)
70 World Trade Organization (WTO). Ministerial declaration of the southern agricultural council (CAS) on gene editing techniques—Communication from Argentina. Geneva, Switzerland: WTO, 2019
71 C Alimentarius. Guideline for the Conduct of Food Safety Assessment of Foods Produced using Recombinant-DNA Animals (CAC/GL 68–2008). Rome: FAO/WHO Joint Publications, 2008
72 European Food Safety Authority (EFSA). Guidance on the risk assessment of food and feed from genetically modified animals and on animal health and welfare aspects. EFSA Journal, 2012, 10(1): 2501
https://doi.org/10.2903/j.efsa.2012.2501
73 Food and Agriculture Organization of the United Nations (FAO). Development of integrated multipurpose animal recording systems. Rome: FAO, 2016
74 European Commission (EC). Study on the labelling of products from cloned animals and their offspring. Luxembourg: European Commission, 2015
75 J Huffman. Special Skretting feed key to RAS salmon grower’s ‘non-GMO’ label. Available at Undercurrent News website on December 19, 2019
76 Eurofins Gene Scan Technologies GmbH. A New Kit for the Detection of Genetically Modified Salmon in Food and Feed. Available at the Eurofins website on December 19, 2019
No related articles found!
Viewed
Full text


Abstract

Cited

  Shared   
  Discussed   
版权所有 © 2015 高等教育出版社.
电话: 010-58556848 (技术); 010-58556485 (订阅) E-mail: subscribe@hep.com.cn