Gene editing (CRISPR-Cas) technology and fisheries sector

  1. A.D. Diwan1*,
  2. A.S. Ninawe2,
  3. S.N. Harke1

Authors Affiliation(s)

  • 1Mahatma Gandhi Missions Institute of Biosciences & Technology, MGM University of Health Sciences, Aurangabad 431003, Maharashtra, INDIA
  • 2Department of Biotechnology, Ministry of Science & Technology, Govt of India, CGO Complex, Lodhi Road, New Delhi 110003, INDIA

Can J Biotech, Volume 1, Issue 2, Pages 65-72, DOI: https://doi.org/10.24870/cjb.2017-000108

Received: Apr 8, 2017; Revised: Jun 20, 2017; Accepted: Jun 22, 2017

Abstract

Considering the advantages of gene editing technologies, in recent years, emphasis has been given on three main techniques of gene editing i.e. zinc finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs), and the CRISPR/Cas9 RNA-guided endonuclease system. In all of these three technologies one thing is common i.e. the technology utilizes restriction enzymes to break down the double stranded DNA molecule at a targeted location with the help of another molecule of homologous binding protein or RNA, which is also called as a guide RNA molecule. This targeted breaking and repair of the DNA molecule as per our requirement is generally viewed as a great breakthrough in gene therapy methods. In fisheries sector, in order to promote aquaculture/mariculture activities, aquaculture industries are facing number of challenges, particularly in area of quality and demand of seed production, control of health and disease management, production of quality traits with phenotypically improved varieties, and strengthening of immune system. Several efforts are being made both at public and private sectors to develop scientific technologies to meet these challenges and substantial achievements have also been made. But still these challenges remained as major constraints in hampering the growth of this industry as per the demand and expectations. However, with the advent of now recently developed gene editing techniques, we may have to explore and evaluate the possibilities of applications of this technology as a long lasting solution in addressing at least some of the vital issues of the aquaculture industry particularly in those related to altering targeted gene structure in the species for positive impact.

References

  1. DAHDF Annual Report (2015) Department of Animal Husbandry, Dairy and Fisheries, Government of India, New Delhi.
  2. Seruggia, D. and Montoliu, L. (2014) The new CRISPR-Cas system: RNA-guided genome engineering to efficiently produce any desired genetic alteration in animals. Transgenic Res 23:707716.Crossref
  3. FAO (2014) The State of World Fisheries & Aquaculture, Rome.
  4. Edun, O.M. and Uka, A. (2011) Biotechnology in Aquaculture: Prospects and Challenges. Nig J Biotech 22: 8- 12.
  5. Adelizi, P.D., Rosati, R.R., Warner, K., Wu, Y.V., Muench, T.R., White, M.R. and Brown, P.B. (1998). Evaluation of fish-meal free diets for rainbow trout, Oncorhynchus mykiss. Aquac Nutr 4: 255-262.Crossref
  6. Madany, I.M., Wahab, A.A.A. and Al-Alawi, Z. (1996) Trace metals concentrations in marine Organisms from the coastal areas of Bahrain, Arabian Gulf. Water Air Soil Pollut 91: 233-248.Crossref
  7. Verschuere, L., Rombaut, G., Sorgeloos, P. and Verstraete, W. (2000) Probiotic Bacteria as Biological Control Agents in Aquaculture. Microbiol Mol Biol Rev 64: 655-671.
  8. Beardmore, J.A., Mair, G.C. and Lewis, R.I. (2001) Monosex male production in finfish as exemplified by tilapia: applications, problems, and prospects. Aquaculture 197: 283-301.Crossref
  9. Mair, G.C. (2001) Genetics in Tilapia culture. In Proceedings of the Tilapia 2001 International Technical and Trade Conference (Subasinghe, S. and Singh, T. (eds)). 28-30 May 2001, Kuala Lumpur, Malaysia, INFOFISH, Kuala Lumpur, Malaysia: 136-148.
  10. Mair, G.C. (2001) Tilapia Genetics in Asia. Sixth Central American Aquaculture Symposium. Proceedings Tilapia session (Meyer, D.E. (ed.)) PD/A CRSP, Tegucigalpa , Honduras: 9-34.
  11. Mair, G.C. (2002). Tilapia Genetics and Breeding in Asia. In Tilapia farming in the 21st Century (Guerrero III, R.D. and Guerrero-del Custillo, M.R. (eds.)). Proceedings of the International Forum on Tilapia Farming in the 21st Century (Tilapia forum, 2002) pp184. Philippine Fisheries Association, Inc., Losbanos, Laguna, Philippines: 100-123.
  12. DBT Annual Report (2015) Department of Biotechnology, Government of India, New Delhi.
  13. Cheung, R.C., Ng, T.B., Wong, J.H. and Chan, W.Y. (2015) Chitosan: An Update on Potential Biomedical and Pharmaceutical Applications. Mar Drugs 13: 51565186. Crossref
  14. Elieh Ali Komi, D., Sharma, L. and Dela Cruz, C.S. (2017) Chitin and Its Effects on Inflammatory and Immune Responses. Clin Rev Allergy Immunol.Crossref
  15. Connelly, J.P., Barker, J.C., Pruett-Miller, S. and Porteus, M.H. (2010) Gene correction by homologous recombination with zinc finger nucleases in primary cells from a mouse model of a generic recessive genetic disease. Mol Ther 18: 1103-1110.Crossref
  16. Cong, L., Ran, F.A., Cox, D., Lin, S., Barretto, R., Habib, N., Hsu, P.D., Wu, X., Jiang, W., Marraffini, L.A. and Zhang, F. (2013) Multiplex genome engineering using CRISPR/Cas systems. Science 339: 819-823. Crossref
  17. Jinek, M., East, A., Cheng, A., Lin, S., Ma, E. and Doudna, J. (2013) RNA-programmed genome editing in human cells. Elife 2: e00471. Crossref
  18. Ishino, Y., Shinagawa, H., Makino, K., Amemura, M. and Nakata, A. (1987) Nucleotide sequence of the iap gene, responsible for alkaline phosphatase isozyme conversion in Escherichia coli, and identification of the gene product .J Bacteriol 169: 54295433.
  19. Jinek, M., Chylinski, K., Fonfara, I., Hauer, M., Doudna, J.A. and Charpentier, E. (2012) A Programmable Dual-RNAGuided DNA Endonuclease in Adaptive Bacterial Immunity. Science 337: 816821.Crossref
  20. Doudna, J.A. and Charpentier, E. (2014) The new frontier of genome engineering with CRISPR-Cas9. Science 346: 1258096. Crossref
  21. Makarova, K.S., Haft, D.H., Barrangou, R., Brouns, S.J., Charpentier, E., Horvath, P., Moineau, S., Mojica, F.J., Wolf, Y.I., Yakunin, A.F., van der Oost, J. and Koonin, E.V. (2011) Evolution and classification of the CRISPR-Cas systems. Nat Rev Microbiol 9: 467-477.Crossref
  22. Katayama, H., Nagata, K., Ohira, T., Yumoto, F., Tanokura, M. and Nagasawa, H. (2003) The solution structure of molt-inhibiting hormone from the kuruma prawn Marsupenaeus japonicus. J Biol Chem 278: 96209623. Crossref
  23. Katayama, H., Ohira, T., Nagata, S. and Nagasawa, H. (2004) Structure- Activity Relationship of Crustacean Molt-Inhibiting Hormone from the Kuruma Prawn Marsupenaeus japonicus. Biochemistry 43: 9629-9635.Crossref
  24. Nakatsuji, T., Lee, C.Y. and Watson, R.D. (2009) Crustacean molt-inhibiting hormone: structure, function, and cellular mode of action. Comp Biochem Physiol A Mol Integr Physiol 152: 139 148.Crossref
  25. Treerattrakool, S., Panyim, S. and Udomkit, A. (2011) Induction of ovarian maturation and spawning in Penaeus monodon broodstock by double-stranded RNA. Mar Biotechnol 13: 163 169.Crossref
  26. Das, R., Krishna, G., Priyadarshi, H., Gireesh-Babu, P., Pavan-Kumar, A., Rajendran, K.V., Reddy, A.K., Makesh, M. and Chaudhari, A. (2015) Captive maturation studies in Penaeus monodon by GIH silencing using constitutively expressed long hairpin RNA. Aquaculture 448: 512520.Crossref
  27. Blasky, A.J., Hass, A., Schiel, J.A., Prekeris, R. and Kelley, M.L.  Microinjection of zebra fish embryos using DharmaconTM Edit-RTM Cas9 Nuclease mRNA, synthetic crRNA, and tracrRNA for genome engineering. GE Healthcare.
  28. Hwang, W.Y., Fu, Y., Reyon, D., Maeder, M.L., Tsai, S.Q., Sander, J.D., Peterson, R.T., Yeh, J.R. and Joung, J.K. (2013) Efficient genome editing in zebrafish using a CRISPR-Cas system. Nat Biotechnol 31: 227229.Crossref
  29. Talbot, J.C. and Amacher, S.L. (2014) A streamlined CRISPR pipeline to reliably generate zebrafish frameshifting alleles. Zebrafish 11: 583585.Crossref
  30. Brown, A., Woods, W.S. and Perez-Pinera, P. (2016) Multiplexed Targeted Genome Engineering Using a Universal Nuclease-Assisted Vector Integration System. ACS Synth Biol 5: 582588.Crossref
  31. Carroll, D. (2014). Genome engineering with targetable nucleases. Annu Rev Biochem 83: 409-439.Crossref
  32. Sander, J.D. and Joung, J.K. (2014) CRISPR-Cas systems for editing, regulating and targeting genomes. Nat Biotechnol 32: 347-355. Crossref
  33. Li, H., Haurigot, V., Doyon, Y., Li, T., Wong, S.Y., Bhagwat, A.S., Malani, N., Anguela, X.M., Sharma, R., Ivanciu, L., Murphy, S.L., Finn, J.D., Khazi, F.R., Zhou, S., Paschon, D.E., Rebar, E.J., Bushman, F.D., Gregory, P.D., Holmes, M.C. and High, K.A. (2011) In vivo genome editing restores haemostasis in a mouse model of haemophilia . Nature 475: 217-221.Crossref
  34. Doyon, J.B., Zeitler, B., Cheng, J., Cheng, A.T., Cherone, J.M., Santiago, Y., Lee, A.H., Vo, T.D., Doyon, Y., Miller, J.C., Paschon, D.E., Zhang, L., Rebar, E.J., Gregory, P.D., Urnov, F.D. and Drubin, D.G. (2011) Rapid and efficient clathrin-mediated endocytosis revealed in genome-edited mammalian cells. Nat Cell Biol 13: 331-337. Crossref
  35. Savic, D., Partridge, E.C., Newberry, K.M., Smith, S.B., Meadows, S.K., Roberts, B.S., Mackiewicz, M., Mendenhall, E.M. and Myers, R.M (2015) CETCh-seq: CRISPR epitope tagging ChIP-seq of DNA-binding proteins. Genome Res 25: 1581-1589.Crossref
  36. Rath, D., Amlinger, L., Rath, A. and Lundgren, M. (2015) The CRISPR-Cas immune system: biology, mechanisms and applications. Biochimie 117: 119-128.Crossref