Journal of Biochemistry and Biotechnology

All submissions of the EM system will be redirected to Online Manuscript Submission System. Authors are requested to submit articles directly to Online Manuscript Submission System of respective journal.

Brief Report - Journal of Biochemistry and Biotechnology (2024) Volume 7, Issue 2

Gene Editing in Animals: Implications for Livestock Production and Conservation Efforts

Zhao Goodman*

Department of Synthetic Biology, Sorbonne University, France

*Corresponding Author:
Zhao Goodman
Department of Synthetic Biology
Sorbonne University, France
E-mail: goodmann@sorbonne.fr

Received: 02-Apr-2024, Manuscript No. AABB-24-134786; Editor assigned: 04-Apr-2024, Pre QC No. AABB-24-134786 (PQ); Reviewed: 16-Apr-2024, QC No. AABB-24-134786; Revised: 23-Apr-2024, Manuscript No. AABB-24-134786 (R); Published: 30-April-2024, DOI:10.35841/aabb-7.2.198

Citation: Goodman Z. Gene Editing in Animals: Implications for Livestock Production and Conservation Efforts. J Biochem Biotech 2024; 7(2):198

Visit for more related articles at Journal of Biochemistry and Biotechnology

Introduction

Gene editing technologies, particularly CRISPR-Cas9, have revolutionized the field of genetics, offering precise and efficient methods for modifying the genetic makeup of organisms. In the realm of animal agriculture and conservation, gene editing holds significant promise for enhancing livestock production, improving animal welfare, and facilitating conservation efforts. However, the widespread adoption of gene editing in animals also raises ethical, regulatory, and environmental considerations that warrant careful examination. In this article, we explore the implications of gene editing for livestock production and conservation efforts, considering both the opportunities and challenge it presents [1].

Disease resistance: Gene editing can be used to introduce genetic variations associated with disease resistance in livestock species, reducing the prevalence and impact of infectious diseases in agricultural settings. For example, researchers have successfully engineered pigs with resistance to Porcine Reproductive and Respiratory Syndrome (PRRS), a devastating viral disease that causes significant economic losses in the swine industry. Improved productivity: Gene editing can enhance traits related to animal productivity, such as growth rate, feed efficiency, and reproductive performance. By introducing genetic variations associated with desirable traits, researchers aim to develop livestock breeds with improved performance and profitability for farmers [2,3].

Animal welfare: Gene editing can be employed to improve animal welfare by reducing susceptibility to stress, disease, and adverse environmental conditions. For example, researchers have explored gene editing approaches to enhance heat tolerance in livestock species, mitigating the negative effects of climate change on animal health and productivity [4].

In addition to its applications in livestock production, gene editing holds promise for conservation efforts aimed at preserving endangered species, restoring ecosystems, and mitigating the impacts of biodiversity loss. Some potential applications include: Genetic rescue: Gene editing can be used to address genetic diversity loss and population decline in endangered species by introducing beneficial genetic variations, such as adaptive traits or disease resistance genes. This approach, known as genetic rescue, offers a promising strategy to enhance the resilience and viability of endangered populations facing extinction risk [5].

De-extinction: Gene editing technologies raise the possibility of de-extinction, whereby extinct species are brought back to life through genetic manipulation of closely related living species. While still in its early stages, de-extinction efforts hold potential for restoring ecosystems, revitalizing species interactions, and reversing the legacy of human-induced extinctions [6].

Ethical concerns: Gene editing raises ethical questions related to animal welfare, species integrity, and human-animal relationships. Critics argue that genetic manipulation of animals may compromise their welfare, autonomy, and intrinsic value, raising moral dilemmas about the ethical treatment of animals and the limits of human intervention in nature. Regulatory oversight: The regulation of gene editing in animals varies between countries, with some jurisdictions adopting strict regulations while others have more lenient policies. Harmonizing regulatory frameworks and ensuring transparent risk assessments are essential to address safety concerns, mitigate environmental risks, and safeguard public health and welfare [7,8].

Environmental impact: The release of genetically modified animals into the environment raises concerns about potential ecological consequences, including unintended effects on ecosystems, biodiversity, and species interactions. Long-term monitoring, ecological risk assessments, and adaptive management strategies are needed to assess and mitigate the environmental impacts of gene editing in animals [9,10].

Conclusion

Gene editing technologies offer unprecedented opportunities to enhance livestock production, conserve endangered species, and address pressing environmental challenges. By leveraging the power of genetic manipulation, researchers can develop innovative solutions to improve animal health, welfare, and productivity, while also contributing to biodiversity conservation and ecosystem restoration efforts.

References

  1. Lillico SG, Proudfoot C, Carlson DF, et al. Live pigs produced from genome edited zygotes. Sci Rep. 2013;3(1):2847.
  2. Indexed at, Google Scholar, Cross Ref

  3. O'Brien SJ, Menninger JC, Nash WG. Atlas of mammalian chromosomes. John Wiley Sons; 2006.
  4. Indexed at, Google Scholar, Cross Ref

  5. Church GM, Regis E. Regenesis: how synthetic biology will reinvent nature and us. Basic Books; 2014.
  6. Indexed at, Google Scholar, Cross Ref

  7. Ewbank AC, Duarte-Benvenuto A, Zamana-Ramblas R, et al. Herpesvirus and adenovirus surveillance in threatened wild West Indian (Trichechus manatus) and Amazonian manatees (Trichechus inunguis), Brazil. Acta Trop. 2023;237:106740.
  8. Indexed at, Google Scholar, Cross Ref

  9. Piergentili R, Del Rio A, Signore F. CRISPR-Cas and its wide-ranging applications: From human genome editing to environmental implications, technical limitations, hazards and bioethical issues. Cells. 2021;10(5):969.
  10. Indexed at, Google Scholar, Cross Ref

  11. National Academies of Sciences. Fostering integrity in research. National Academies Press; 2017.
  12. Indexed at, Google Scholar, Cross Ref

  13. Deagle BE, Jarman SN, Coissac E. DNA metabarcoding and the cytochrome c oxidase subunit I marker: not a perfect match. Bio letters. 2014;10(9):20140562.
  14. Indexed at, Google Scholar, Cross Ref

  15. Mascolino S, Mariani S, Benvenuto C. Behavioural responses in a congested sea: an observational study on a coastal nest-guarding fish. Eur Zool J. 2019;86(1):504-18.
  16. Indexed at, Google Scholar, Cross Ref

  17. Syková E, Rychmach P, Drahorádová I, et al. Transplantation of mesenchymal stromal cells in patients with amyotrophic lateral sclerosis: results of phase I/IIa clinical trial. Cell Transplant. 2017;26(4):647-58.
  18. Indexed at, Google Scholar, Cross Ref

  19. Garson J, Plutynski A, Sarkar S. The Routledge handbook of philosophy of biodiversity. Routledge; 2017.
  20. Indexed at, Google Scholar, Cross Ref

Get the App