Granthaalayah
TRANSGENE MANUFACTURING: FROM TOOLS TO APPLICATIONS

Transgene Manufacturing: From Tools to Applications

 

Dr. Saeeda Wasim 1, Dr. Sharique Ahmad 2

 

1 Senior Consultant, Nova IVF Fertility, Hazratganj, Lucknow, Uttar Pradesh, India

2 Professor, Department of Pathology, Era's Lucknow Medical College and Hospital, Era University, Lucknow, Uttar Pradesh, India

 

A qr code with a tree and orange circles

Description automatically generated

A picture containing logo

Description automatically generated

ABSTRACT

Genetic modification generation is an important intersection of genetic engineering and biotechnology and involves the integration of foreign genes into the genome of organisms to produce specific proteins or repair genetic abnormalities. The machine has many applications in medicine, agriculture and biotechnology. The evolution of genetically modified production technologies from traditional methods to advanced genetic engineering tools demonstrates their evolution. This review discusses various methods incorporating modern and advanced technologies and explores their applications and future prospects. Early models of genetic modification generally involved organisms modified to produce human insulin, followed by the evolution of animals and plants. Traditional methods such as microinjection, retrovirus-mediated gene transfer and embryonic stem cell -mediated gene transfer are important for the production of transgenic animals. Similarly, Agrobacterium-mediated transformation and biolistic transformation methods are used to produce genetically modified plants. Although useful, these methods often result in synergistic and differential gene expression. Made with genetic modification. This technology allows modification of the target with minimal impact on the target, thus increasing the predictability and efficiency of genetic modification. Additionally, CRISPR/Cas9's ability to alter multiple genes simultaneously and adaptability to various organisms expands its applications in medicine and agriculture. Synthetic biology continues to advance genetic engineering by designing and building new biological materials and systems, enabling innovations such as genetic engineering, metabolic processes, and minimal genomes. Viral vectors such as adeno-associated virus (AAV), lentivirus, and adenovirus have unique advantages and clinical challenges. Nonviral vectors, including lipid nanoparticles, electroporated, and polymeric vectors, provide alternative delivery methods with varying efficacy and specificity. Painkiller. It can improve crop growth, livestock and disease resistance in agriculture. Applications of biotechnology include biofuels, biopharmaceuticals, and bioremediation. Future directions aim to improve delivery, expand applications, explore synthetic genomics, advance personalized medicine, and develop regenerative agriculture.

In summary, with the influence of advances in genetic engineering and technology, genetically modified production has become a complex field with many applications. Its future promises transformative solutions to global challenges in health, food security and environmental sustainability. To reach its full potential, continuous innovation and ethical thinking are essential.

 

Received 30 May 2024

Accepted 10 June 2024

Published 31 July 2024

Corresponding Author

Dr Sharique Ahmad, diagnopath@gmail.com  

DOI 10.29121/granthaalayah.v12.i7.2024.5709  

Funding: This research received no specific grant from any funding agency in the public, commercial, or not-for-profit sectors.

Copyright: © 2024 The Author(s). This work is licensed under a Creative Commons Attribution 4.0 International License.

With the license CC-BY, authors retain the copyright, allowing anyone to download, reuse, re-print, modify, distribute, and/or copy their contribution. The work must be properly attributed to its author.

 

Keywords: Transgene Manufacturing, Genetic Engineering, Gene Therapy Vectors, Synthetic Biology


1. INTRODUCTION

Transgene manufacturing is a sophisticated and multifaceted field that bridges the gap between genetic engineering and therapeutic applications. This process involves the integration of foreign genes into an organism’s genome to produce desired proteins or to correct genetic anomalies. Transgene manufacturing has applications in medicine, agriculture, and industrial biotechnology. This article delves into the methodologies of transgene manufacturing and explores the various techniques and innovations that have shaped this field over the years. The discussion will cover traditional methods like transgenic animal and plant models, advanced techniques such as CRISPR/Cas9, and emerging trends in synthetic biology and gene therapy vectors.

 

2. Historical Perspective

The journey of transgene manufacturing began in the 20th century with the advent of recombinant DNA technology. The first genetically modified organisms (GMOs) were bacteria engineered to produce human insulin. This breakthrough set the stage for more complex transgenic systems, including animals and plants Berg & Mertz (2010). The early methods relied heavily on random integration of transgenes, leading to variable expression and unintended effects. Over the decades, the field has evolved to adopt more precise and predictable techniques, paving the way for innovations in gene therapy, agricultural biotechnology, and synthetic biology.

 

3. Traditional Methods of Transgene Manufacturing

3.1. Transgenic Animals

Transgenic animals are those that have had a foreign gene deliberately inserted into their genome. The primary methods for creating transgenic animals include microinjection, retrovirus-mediated gene transfer, and embryonic stem cell-mediated gene transfer.

1)    Microinjection: This involves injecting a solution of DNA directly into the pronucleus of a fertilized egg. The egg is then implanted into a surrogate mother. This method has been widely used in mice, pigs, and other mammals. Although straightforward, the technique suffers from low efficiency and random integration of the transgene, often resulting in mosaic animals where only some cells carry the transgene Gordon et al. (1980).

2)    Retrovirus-Mediated Gene Transfer: Here, retroviruses are used to deliver the transgene into the host genome. This method allows for stable integration and is relatively efficient. However, it carries the risk of insertional mutagenesis, where the integration of the virus disrupts endogenous genes, and it is limited by the capacity of viral vectors to carry large genetic payloads Van der Putten et al. (1985).

3)    Embryonic Stem Cell-Mediated Gene Transfer: This involves manipulating embryonic stem cells to carry the transgene, which are then injected into blastocysts. This method allows for precise genetic modifications and has been pivotal in creating knockout and knock-in models. These models are essential for studying gene function and disease mechanisms Evans & Kaufman (1981).

 

3.2. Transgenic Plants

The production of transgenic plants primarily relies on two methods: Agrobacterium-mediated transformation and biolistic (gene gun) transformation.

1)    Agrobacterium-Mediated Transformation: Agrobacterium tumefaciens, a soil bacterium, naturally transfers DNA to plants. By modifying its Ti plasmid to carry desired genes, scientists can introduce new traits into plants. This method is highly efficient for dicotyledonous plants and involves fewer off-target effects compared to other methods Zambryski et al. (1989).

2)    Biolistic Transformation: Also known as the gene gun method, it involves shooting microscopic particles coated with DNA into plant cells. This method is versatile and can be used for both monocot and dicot plants, though it can cause damage to the plant tissue. It is particularly useful for plants that are resistant to Agrobacterium-mediated transformation Sanford et al. (1993).

 

4. Advanced Techniques in Transgene Manufacturing

4.1. CRISPR/Cas9 Technology

CRISPR/Cas9 has revolutionized genetic engineering by providing a precise, efficient, and versatile method for genome editing. This system uses a guide RNA (gRNA) to direct the Cas9 nuclease to a specific genomic location, where it creates double-strand breaks. The cell’s repair mechanisms then introduce the desired genetic modifications.

1)    High Precision: CRISPR/Cas9 allows for targeted modifications with minimal off-target effects compared to traditional methods. It offers a higher degree of control over the site of gene insertion, which is crucial for avoiding disruptions to other essential genes Jinek et al. (2012).

2)    Multiplexing: Multiple genes can be edited simultaneously, which is beneficial for studying complex traits and developing polygenic modifications. This capability is particularly valuable in plants and animals where traits often result from the interaction of multiple genes Cong et al. (2013).

3)    Versatility: CRISPR/Cas9 can be adapted for various organisms, from bacteria to humans, and can be used for both gene knockout and knock-in applications. Its applications range from creating disease models to developing crops with improved traits Hsu et al. (2014).

 

4.2. Synthetic Biology

Synthetic biology involves designing and constructing new biological parts, devices, and systems. It aims to create standardized genetic components that can be easily assembled to perform specific functions.

1)    Gene Circuits: Synthetic biology enables the creation of gene circuits that can control the timing and expression levels of transgenes, improving the predictability and functionality of genetic modifications. These circuits can mimic natural regulatory networks or implement entirely novel functionalities Elowitz & Leibler (2000).

2)    Metabolic Engineering: This approach allows optimize the metabolic pathways to get enhanced production of the desired compounds, such as pharmaceuticals and biofuels. By modifying the genes involved in metabolic pathways, scientists can increase the yield and efficiency of production Nielsen & Keasling (2016).

3)    Minimal Genomes: Researchers are developing minimal genomes, which are stripped-down versions of natural genomes, to serve as chassis for synthetic biology applications. These minimal genomes reduce complexity and increase stability in transgene expression, providing a more predictable platform for engineering new functions Hutchison et al. (2016).

 

4.3. Gene Therapy Vectors

Gene therapy vectors are used to deliver therapeutic genes into patients’ cells to treat genetic disorders. The most commonly used vectors are viral and non-viral vectors.

 

4.3.1.  Viral Vectors

1)    Adeno-Associated Virus (AAV): AAV vectors are non-pathogenic and can transduce both dividing and non-dividing cells. They have been successfully used in clinical trials for diseases like hemophilia and retinal disorders. AAV vectors are known for their long-term expression and low immunogenicity, making them ideal for many therapeutic applications Kotin et al. (1990).

2)    Lentivirus: Lentiviral vectors can integrate into the host genome, providing long-term expression of the transgene. They are particularly useful for ex vivo gene therapy applications, such as modifying hematopoietic stem cells. Lentiviruses can carry larger genetic payloads compared to AAV, allowing for more complex genetic modifications Naldini et al. (1996).

3)    Adenovirus: Adenoviral vectors can accommodate large transgenes and have high transduction efficiency. However, they can induce strong immune responses, limiting their use in some applications. Despite this, they are useful for short-term gene expression and are often used in cancer gene therapy and vaccine development Shenk (1996).

 

4.3.2.  Non-Viral Vectors

1)    Lipid Nanoparticles: These are used to encapsulate and deliver nucleic acids into cells. They have gained prominence with the development of mRNA vaccines, such as for COVID-19. Lipid nanoparticles offer a non-immunogenic and versatile platform for gene delivery Pardi et al. (2018).

2)    Electroporation: This technique uses electric pulses to create pores in the cell membrane, allowing DNA to enter. It is widely used for gene delivery in plants and ex vivo cell therapy applications. Electroporation is highly efficient and can be used for a broad range of cell types Neumann et al. (1982).

3)    Polymeric Vectors: These are synthetic carriers that can protect and deliver nucleic acids into cells. They offer advantages in terms of biocompatibility and reduced immunogenicity. Polymeric vectors can be designed to target specific cell types, enhancing the precision of gene delivery Mintzer & Simanek (2009).

 

 

 

5. Applications of Transgene Manufacturing

5.1. Medicine

1)    Gene Therapy: Transgene manufacturing is at the heart of gene therapy, which aims to treat genetic disorders by introducing functional genes into patients' cells. Diseases like cystic fibrosis, muscular dystrophy, and certain cancers are being targeted using gene therapy approaches. Clinical trials have shown promising results, with some gene therapies receiving regulatory approval Ginn et al. (2018).

2)    Monoclonal Antibodies: Transgenic animals, such as genetically modified mice, are used to produce monoclonal antibodies. These antibodies are crucial for the treatment of various diseases, including cancer and autoimmune disorders. Transgene manufacturing allows for the production of humanized antibodies, reducing the risk of immune reactions in patients Scott et al. (2012).

3)    Regenerative Medicine: Transgenic techniques are used to create pluripotent stem cells with specific genetic modifications. These cells can differentiate into various cell types, offering potential treatments for conditions like Parkinson’s disease, spinal cord injuries, and heart disease Trounson & DeWitt (2016).

 

5.2. Agriculture

1)    Crop Improvement: Transgenic plants have been developed to exhibit traits like pest resistance, herbicide tolerance, and improved nutritional content. Examples include Bt cotton, which is resistant to bollworms, and Golden Rice, which is enriched with vitamin A. These innovations help increase crop yields, reduce reliance on chemical pesticides, and address nutritional deficiencies James (2018).

2)    Animal Husbandry: Transgenic animals have been created to improve agricultural productivity. For instance, transgenic cows produce milk with altered protein content, and pigs have been engineered to have leaner meat. These modifications can enhance food quality and reduce environmental impact Niemann & Kues (2007).

3)    Disease Resistance: Transgene manufacturing is used to develop plants and animals that are resistant to diseases. For example, transgenic bananas resistant to the banana wilt disease and transgenic salmon that are less susceptible to viral infections have been created. These developments help secure food supplies and reduce losses due to disease Rommens et al. (2007).

 

5.3. Industrial Biotechnology

1)    Biofuels: Transgenic microorganisms are engineered to produce biofuels from renewable resources. These organisms can convert biomass into ethanol, biodiesel, and other biofuels, providing a sustainable alternative to fossil fuels. Advances in metabolic engineering and synthetic biology are driving improvements in the efficiency and cost-effectiveness of biofuel production Steen et al. (2010).

2)    Biopharmaceuticals: Transgenic systems are used to produce complex biopharmaceuticals, including hormones, enzymes, and vaccines. For instance, transgenic plants and animals have been developed to produce insulin, growth hormones, and antibodies. These systems offer scalable and cost-effective production platforms Fischer et al. (2012).

3)    Bioremediation: Transgenic microorganisms are employed to degrade environmental pollutants. These organisms can break down hazardous substances like oil spills, heavy metals, and plastic waste, contributing to environmental cleanup efforts. Advances in synthetic biology are enabling the development of organisms with enhanced capabilities for bioremediation Glick (2010).

 

6. Challenges and Future Directions

6.1. Challenges

1)    Efficiency and Precision: Despite advancements, achieving high efficiency and precision in transgene integration remains a challenge, especially in complex organisms. Random integration and off-target effects can lead to unintended consequences, affecting the safety and efficacy of transgenic products Kleinstiver et al. (2016).

2)    Regulatory and Ethical Issues: The development and use of transgenic organisms raise ethical concerns and face stringent regulatory scrutiny. Public acceptance and ethical considerations are critical factors influencing the progress of this field. Transparent risk assessments and ethical guidelines are necessary to address these concerns Kuzma (2016).

3)    Off-Target Effects: Techniques like CRISPR/Cas9, although precise, can still cause off-target mutations. Improving the specificity of these tools is an ongoing area of research. Efforts are being made to develop next-generation genome editing technologies with higher fidelity and reduced off-target effects Liang et al. (2015).

4)    Scalability: Scaling up transgene manufacturing processes for commercial production can be challenging. Factors such as production costs, yield consistency, and regulatory compliance must be addressed to ensure the viability of transgenic products Kwok (2019).

 

6.2. Future Directions

1)    Improving Delivery Systems: Enhancing the efficiency and specificity of delivery systems for gene therapy remains a priority. Innovations in nanotechnology and biomaterials hold promise in this regard. Advanced delivery systems, such as targeted nanoparticles and cell-penetrating peptides, are being developed to improve the precision and effectiveness of gene delivery Zhang & Satterlee (2016).

2)    Expanding Applications: The scope of transgene manufacturing is expanding beyond agriculture and medicine to areas like environmental remediation and industrial biotechnology. Emerging applications include the development of transgenic organisms for biofabrication, where biological systems are used to produce materials with unique properties Cameron et al. (2014).

3)    Synthetic Genomics: The creation of entirely synthetic genomes opens new possibilities for custom-designed organisms with novel functions. This could revolutionize fields like bioenergy, pharmaceuticals, and synthetic biology. Researchers are working on designing minimal genomes that serve as efficient platforms for synthetic biology applications Chan et al. (2005).

4)    Personalized Medicine: Advances in transgene manufacturing are paving the way for personalized medicine, where treatments are tailored to an individual's genetic profile. Gene editing technologies can be used to correct genetic mutations in patient-derived cells, offering customized therapeutic solutions Ylä-Herttuala (2012).

5)    Regenerative Agriculture: Transgenic techniques are being explored to develop crops that contribute to soil health and sustainability. For example, transgenic plants with enhanced nitrogen fixation capabilities can reduce the need for chemical fertilizers, promoting regenerative agricultural practices Vance (2001).

 

7. Conclusion

Transgene manufacturing has come a long way since its inception, evolving from rudimentary techniques to sophisticated methodologies that offer precision and versatility. The integration of advanced technologies like CRISPR/Cas9 and synthetic biology has opened new avenues for research and applications. Despite the challenges, the future of transgene manufacturing holds immense potential, promising to revolutionize medicine, agriculture, and beyond. As we continue to refine these techniques and address ethical and regulatory concerns, the impact of transgene manufacturing on society is bound to be profound and far-reaching. The ongoing advancements in this field are set to transform our approach to health, food security, and environmental sustainability, offering solutions to some of the most pressing challenges of our time.

 

CONFLICT OF INTERESTS

None. 

 

ACKNOWLEDGMENTS

None.

 

REFERENCES

Berg, P., & Mertz, J. E. (2010). Personal Reflections on the Origins and Emergence of Recombinant DNA Technology. Genetics, 184(1), 9-17. https://doi.org/10.1534/genetics.109.112144

Cameron, D. E., Bashor, C. J., & Collins, J. J. (2014). A Brief History of Synthetic Biology. Nature Reviews Microbiology, 12(5), 381-390. https://doi.org/10.1038/nrmicro3239

Chan, L. Y., Kosuri, S., & Endy, D. (2005). Refactoring Bacteriophage T7. Molecular Systems Biology, 1(1), 0018. https://doi.org/10.1038/msb4100025

Cong, L., Ran, F. A., Cox, D., Lin, S., Barretto, R., Habib, N., & Zhang, F. (2013). Multiplex Genome Engineering Using CRISPR/Cas Systems. Science, 339(6121), 819-823. https://doi.org/10.1126/science.1231143

Elowitz, M. B., & Leibler, S. (2000). A Synthetic Oscillatory Network of Transcriptional Regulators. Nature, 403(6767), 335-338. https://doi.org/10.1038/35002125

Evans, M. J., & Kaufman, M. H. (1981). Establishment in Culture of Pluripotential Cells from Mouse Embryos. Nature, 292(5819), 154-156. https://doi.org/10.1038/292154a0

Fischer, R., Schillberg, S., Hellwig, S., Twyman, R. M., & Drossard, J. (2012). GMP Issues for Recombinant Plant-Derived Pharmaceutical Proteins. Biotechnology Advances, 30(2), 434-439. https://doi.org/10.1016/j.biotechadv.2011.08.007

Ginn, S. L., Amaya, A. K., Alexander, I. E., Edelstein, M., & Abedi, M. R. (2018). Gene Therapy Clinical Trials Worldwide to 2017: An Update. The Journal of Gene Medicine, 20(5). https://doi.org/10.1002/jgm.3015

Glick, B. R. (2010). Using Bacteria to Help Feed the World. Critical Reviews in Biotechnology, 30(3), 176-192.

Gordon, J. W., Scangos, G. A., Plotkin, D. J., Barbosa, J. A., & Ruddle, F. H. (1980). Genetic Transformation of Mouse Embryos by Microinjection of Purified DNA. Proceedings of the National Academy of Sciences, 77(12), 7380-7384. https://doi.org/10.1073/pnas.77.12.7380

Hsu, P. D., Lander, E. S., & Zhang, F. (2014). Development and Applications of CRISPR-Cas9 for Genome Engineering. Cell, 157(6), 1262-1278. https://doi.org/10.1016/j.cell.2014.05.010

Hutchison, C. A., Chuang, R. Y., Noskov, V. N., Assad-Garcia, N., Deerinck, T. J., Ellisman, M. H., & Venter, J. C. (2016). Design and Synthesis of a Minimal Bacterial Genome. Science, 351(6280). https://doi.org/10.1126/science.aad6253

James, C. (2018). Global Status of Commercialized Biotech/GM Crops: 2018. ISAAA Brief, 54.  

Jinek, M., Chylinski, K., Fonfara, I., Hauer, M., Doudna, J. A., & Charpentier, E. (2012). A Programmable Dual-RNA-Guided DNA Endonuclease in Adaptive Bacterial Immunity. Science, 337(6096), 816-821. https://doi.org/10.1126/science.1225829

Kleinstiver, B. P., Tsai, S. Q., Prew, M. S., Nguyen, N. T., Welch, M. M., Lopez, J. M., ... & Joung, J. K. (2016). Genome-Wide Specificities of CRISPR-Cas Cpf1 Nucleases in Human Cells. Nature Biotechnology, 34(8), 869-874. https://doi.org/10.1038/nbt.3620

Kotin, R. M., Siniscalco, M., Samulski, R. J., Zhu, X., Hunter, L., Laughlin, C. A., & Muzyczka, N. (1990). Site-Specific Integration by Adeno-Associated Virus. Proceedings of the National Academy of Sciences, 87(6), 2211-2215. https://doi.org/10.1073/pnas.87.6.2211

Kuzma, J. (2016). Rebooting the Debate Over Genetic Engineering. Issues in Science and Technology, 32(4), 80.

Kwok, P. S. (2019). Challenges in Scaling up Production of Gene Therapy Products. Molecular Therapy, 27(3), 543-544.

Liang, P., Xu, Y., Zhang, X., Ding, C., Huang, R., Zhang, Z., ... & Huang, J. (2015). CRISPR/Cas9-Mediated Gene Editing in Human Tripronuclear Zygotes. Protein & Cell, 6(5), 363-372. https://doi.org/10.1007/s13238-015-0153-5

Mintzer, M. A., & Simanek, E. E. (2009). Nonviral Vectors for Gene Delivery. Chemical Reviews, 109(2), 259-302. https://doi.org/10.1021/cr800409e

Naldini, L., Blömer, U., Gallay, P., Ory, D., Mulligan, R., Gage, F. H., & Trono, D. (1996). In Vivo Gene Delivery and Stable Transduction of Nondividing Cells by a Lentiviral Vector. Science, 272(5259), 263-267. https://doi.org/10.1126/science.272.5259.263

Neumann, E., Schaefer-Ridder, M., Wang, Y., & Hofschneider, P. H. (1982). Gene Transfer into Mouse Lyoma Cells by Electroporation in High Electric Fields. The EMBO Journal, 1(7), 841-845. https://doi.org/10.1002/j.1460-2075.1982.tb01257.x

Nielsen, J., & Keasling, J. D. (2016). Engineering Cellular Metabolism. Cell, 164(6), 1185-1197. https://doi.org/10.1016/j.cell.2016.02.004

Niemann, H., & Kues, W. A. (2007). Transgenic Farm Animals: An Update. Reproduction, Fertility and Development, 19(6), 762-770. https://doi.org/10.1071/RD07040

Pardi, N., Hogan, M. J., Porter, F. W., & Weissman, D. (2018). mRNA Vaccines-a New Era in Vaccinology. Nature Reviews Drug Discovery, 17(4), 261-279. https://doi.org/10.1038/nrd.2017.243

Rommens, C. M., Haring, M. A., Swords, K., Davies, H. V., & Belknap, W. R. (2007). The Intragenic Approach as a New Extension to Traditional Plant Breeding. Trends in Plant Science, 12(9), 397-403. https://doi.org/10.1016/j.tplants.2007.08.001

Sanford, J. C., Smith, F. D., & Russell, J. A. (1993). Optimizing the Biolistic Process for Different Biological Applications. Methods in Enzymology, 217, 483-509. https://doi.org/10.1016/0076-6879(93)17086-K

Scott, A. M., Wolchok, J. D., & Old, L. J. (2012). Antibody Therapy of Cancer. Nature Reviews Cancer, 12(4), 278-287. https://doi.org/10.1038/nrc3236

Shenk, T. (1996). Adenoviruses. In Fields Virology. Lippincott-Raven, 2111-2148.

Steen, E. J., Kang, Y., Bokinsky, G., Hu, Z., Schirmer, A., McClure, A., & Keasling, J. D. (2010). Microbial Production of Fatty-Acid-Derived Fuels and Chemicals From Plant Biomass. Nature, 463(7280), 559-562. https://doi.org/10.1038/nature08721

Trounson, A., & DeWitt, N. D. (2016). Pluripotent Stem Cells Progressing to the Clinic. Nature Reviews Molecular Cell Biology, 17(3), 194-200. https://doi.org/10.1038/nrm.2016.10

Van der Putten, H., Botteri, F. M., Miller, A. D., Rosenfeld, M. G., & Verma, I. M. (1985). Efficient Insertion of Genes into the Mouse Germ Line Via Retroviral Vectors. Proceedings of the National Academy of Sciences, 82(18), 6148-6152. https://doi.org/10.1073/pnas.82.18.6148

Vance, C. P. (2001). Symbiotic Nitrogen Fixation and Phosphorus Acquisition. Plant Nutrition in a World of Declining Renewable Resources. Plant Physiology, 127(2), 390-397. https://doi.org/10.1104/pp.127.2.390

Ylä-Herttuala, S. (2012). Endgame: Glybera Finally Recommended for Approval as the First Gene Therapy Drug in the European Union. Molecular Therapy, 20(10), 1831-1832. https://doi.org/10.1038/mt.2012.194

Zambryski, P., Tempe, J., & Schell, J. (1989). Transfer and Function of T-DNA Genes from Agrobacterium Ti and Ri Plasmids in Plants. Cell, 56(2), 193-201. https://doi.org/10.1016/0092-8674(89)90892-1

Zhang, Y., & Satterlee, A. (2016). Nanoparticle Delivery of CRISPR/Cas9 for Genome Editing. In Applications of Nanomaterials in Human Health, 177-195. Springer.

     

 

 

 

Creative Commons Licence This work is licensed under a: Creative Commons Attribution 4.0 International License

© Granthaalayah 2014-2024. All Rights Reserved.