genome editing

 

  • [3] It has also enabled the editing of specific sequences within a genome as well as reduced off target effects.

  • [22] Zinc fingers have been more established in these terms and approaches such as modular assembly (where Zinc fingers correlated with a triplet sequence are attached in
    a row to cover the required sequence), OPEN (low-stringency selection of peptide domains vs. triplet nucleotides followed by high-stringency selections of peptide combination vs. the final target in bacterial systems), and bacterial one-hybrid
    screening of zinc finger libraries among other methods have been used to make site specific nucleases.

  • [31] It is also possible to fuse a protein constructed in this way with the catalytic domain of an endonuclease in order to induce a targeted DNA break, and therefore to use
    these proteins as genome engineering tools.

  • By crossing an organism containing the recombinase sites flanking the gene of interest with an organism that express the SSR under control of tissue specific promoters, it
    is possible to knock out or switch on genes only in certain cells.

  • [17] By creating DNA constructs that contain a template that matches the targeted genome sequence it is possible that the HR processes within the cell will insert the construct
    at the desired location.

  • [37] TALEN constructs are used in a similar way to designed zinc finger nucleases, and have three advantages in targeted mutagenesis: (1) DNA binding specificity is higher,
    (2) off-target effects are lower, and (3) construction of DNA-binding domains is easier.

  • Although, several methods have been discovered which target the inserted genes to specific sites within an organism genome.

  • [29] Meganucleases have the benefit of causing less toxicity in cells than methods such as Zinc finger nuclease (ZFN), likely because of more stringent DNA sequence recognition;[23]
    however, the construction of sequence-specific enzymes for all possible sequences is costly and time-consuming, as one is not benefiting from combinatorial possibilities that methods such as ZFNs and TALEN-based fusions utilize.

  • Meganucleases, found commonly in microbial species, have the unique property of having very long recognition sequences (>14bp) thus making them naturally very specific.

  • One drawback of this technology has been the random nature with which the DNA is inserted into the hosts genome, which can impair or alter other genes within the organism.

  • In order to study the function of these genes site specific recombinases were used.

  • Although the direct genome-wide characterization of zinc finger nuclease activity has not been reported, an assay that measures the total number of double-strand DNA breaks
    in cells found that only one to two such breaks occur above background in cells treated with zinc finger nucleases with a 24 bp composite recognition site and obligate heterodimer FokI nuclease domains.

  • [27] Another approach involves using computer models to try to predict as accurately as possible the activity of the modified meganucleases and the specificity of the recognized
    nucleic sequence.

  • Various selection techniques, using bacteria, yeast or mammal cells have been developed to identify the combinations that offer the best specificity and the best cell tolerance.

  • Further modifications of these systems allowed researchers to induce recombination only under certain conditions, allowing genes to be knocked out or expressed at desired
    times or stages of development.

  • [25][26] Yet others have attempted to alter the DNA interacting aminoacids of the meganuclease to design sequence specific meganucelases in a method named rationally designed
    meganuclease.

  • This could be used for research purposes, by targeting mutations to specific genes, and in gene therapy.

  • By inserting a functional gene into an organism and targeting it to replace the defective one it could be possible to cure certain genetic diseases.

  • Commonly used restriction enzymes are effective at cutting DNA, but generally recognize and cut at multiple sites.

  • Matching colors signify DNA recognition patterns The key to genome editing is creating a DSB at a specific point within the genome.

  • all three major classes of these enzymes—zinc finger nucleases, transcription activator-like effector nucleases and engineered meganucleases—were selected by Nature Methods
    as the 2011 Method of the Year.

  • Zinc finger nucleases[edit] As opposed to meganucleases, the concept behind ZFNs and TALEN technology is based on a non-specific DNA cutting catalytic domain, which can then
    be linked to specific DNA sequence recognizing peptides such as zinc fingers and transcription activator-like effectors (TALEs).

  • These fusion proteins serve as readily targetable “DNA scissors” for gene editing applications that enable to perform targeted genome modifications such as sequence insertion,
    deletion, repair and replacement in living cells.

  • NHEJ uses a variety of enzymes to directly join the DNA ends while the more accurate HDR uses a homologous sequence as a template for regeneration of missing DNA sequences
    at the break point.

  • [15][16] Background Genetic engineering as method of introducing new genetic elements into organisms has been around since the 1970s.

  • [32] The method generally adopted for this involves associating two DNA binding proteins – each containing 3 to 6 specifically chosen zinc fingers – with the catalytic domain
    of the FokI endonuclease which need to dimerize to cleave the double-strand DNA.

  • Meganucleases[edit] Meganucleases, discovered in the late 1980s, are enzymes in the endonuclease family which are characterized by their capacity to recognize and cut large
    DNA sequences (from 14 to 40 base pairs).

  • Gene targeting[edit] Homologous recombination[edit] Early methods to target genes to certain sites within a genome of an organism (called gene targeting) relied on homologous
    recombination.

  • This portion could then be linked to sequence recognizing peptides that could lead to very high specificity.

  • [18] Process Double strand break repair[edit] A common form of Genome editing relies on the concept of DNA double stranded break (DSB) repair mechanics.

  • The recognized sequences are short, made up of around 3 base pairs, but by combining 6 to 8 zinc fingers whose recognition sites have been characterized, it is possible to
    obtain specific proteins for sequences of around 20 base pairs.

  • While HDR based gene editing is similar to the homologous recombination based gene targeting, the rate of recombination is increased by at least three orders of magnitude.

  • It has also been possible to knock in genes or alter gene expression patterns.

  • [33] Several approaches are used to design specific zinc finger nucleases for the chosen sequences.

  • It is therefore possible to control the expression of a specific gene.

  • [30] The first step to this was to find an endonuclease whose DNA recognition site and cleaving site were separate from each other, a situation that is not the most common
    among restriction enzymes.

  • TALENs are artificial restriction enzymes designed by fusing the DNA cutting domain of a nuclease to TALE domains, which can be tailored to specifically recognize a unique
    DNA sequence.

  • One major advantage that CRISPR has over the ZFN and TALEN methods is that it can be directed to target different DNA sequences using its ~80nt CRISPR sgRNAs, while both ZFN
    and TALEN methods required construction and testing of the proteins created for targeting each DNA sequence.

  • For instance, the field of synthetic biology which aims to engineer cells and organisms to perform novel functions, is likely to benefit from the ability of engineered nuclease
    to add or remove genomic elements and therefore create complex systems.

  • Using global transcriptomics data to guide experimentation, the CRISPR based genome editing tool has made it feasible to disrupt or remove key genes in order to elucidate
    function in a human setting.

  • CRISPR[edit] Main article: CRISPR gene editing CRISPRs (Clustered Regularly Interspaced Short Palindromic Repeats) are genetic elements that bacteria use as a kind of acquired
    immunity to protect against viruses.

  • [56] A potentially successful example of the application of genome editing techniques in crop improvement can be found in banana, where scientists used CRISPR/Cas9 editing
    to inactivate the endogenous banana streak virus in the B genome of banana to overcome a major challenge in banana breeding.

  • Listed below are some specific tasks this method can carry out: • Targeted gene mutation • Gene therapy • Creating chromosome rearrangement • Study gene function with stem
    cells • Transgenic animals • Endogenous gene labeling • Targeted transgene addition Targeted gene modification in animals[edit] The combination of recent discoveries in genetic engineering, particularly gene editing and the latest improvement
    in bovine reproduction technologies (e.g.

  • The growth hormone-regulating gene in the Atlantic salmon is replaced with the growth hormone-regulating gene from the Pacific Chinook salmon and a promoter sequence from
    the ocean pout[50] Thanks to the parallel development of single-cell transcriptomics, genome editing and new stem cell models we are now entering a scientifically exciting period where functional genetics is no longer restricted to animal
    models but can be performed directly in human samples.

  • For instance, site-specific gene addition in major crop species can be used for ‘trait stacking’ whereby several desired traits are physically linked to ensure their co-segregation
    during the breeding processes.

  • [46] MAGE experiments can be divided into three classes, characterized by varying degrees of scale and complexity: (i) many target sites, single genetic mutations; (ii) single
    target site, many genetic mutations; and (iii) many target sites, many genetic mutations.

  • [51] Targeted gene modification in plants[edit] Overview of GEEN workflow and editing possibilities Genome editing using Meganuclease,[52] ZFNs, and TALEN provides a new strategy
    for genetic manipulation in plants and are likely to assist in the engineering of desired plant traits by modifying endogenous genes.

  • Genome editing with engineered nucleases will likely contribute to many fields of life sciences from studying gene functions in plants and animals to gene therapy in humans.

  • It achieves such efficiency because the DNA-binding element consists of an array of TALE subunits, each of them having the capability of recognizing a specific DNA nucleotide
    chain independent from others, resulting in a higher number of target sites with high precision.

  • [9] CRISPR also requires the least amount of expertise in molecular biology as the design lays in the guide RNA instead of the proteins.

  • It is only appropriate for precise editing requiring single nucleotide changes and has found to be highly efficient for this type of editing.

  • New TALE nucleases take about one week and a few hundred dollars to create, with specific expertise in molecular biology and protein engineering.

  • Applications As of 2012 efficient genome editing had been developed for a wide range of experimental systems ranging from plants to animals, often beyond clinical interest,
    and was becoming a standard experimental strategy in research labs.

  • [62] There is a need for reliable design and subsequent test of the nucleases, the absence of toxicity of the nucleases, the appropriate choice of the plant tissue for targeting,
    the routes of induction of enzyme activity, the lack of off-target mutagenesis, and a reliable detection of mutated cases.

  • [58] TALEN fusions have also been used by a U.S. food ingredient company, Calyxt,[59] to improve the quality of soybean oil products[60] and to increase the storage potential
    of potatoes[61] Several optimizations need to be made in order to improve editing plant genomes using ZFN-mediated targeting.

  • In particular CRISPR/Cas9 engineered endonucleases allows the use of multiple guide RNAs for simultaneous Knockouts (KO) in one step by cytoplasmic direct injection (CDI)
    on mammalian zygotes.

  • This is caused by the need of having a specific nucleotide at one end in order to produce the guide RNA that CRISPR uses to repair the double-strand break it induces.

  • Chemically combined, synthetic single-stranded DNA (ssDNA) and a pool of oligionucleotides are introduced at targeted areas of the cell thereby creating genetic modifications.

  • [46] An example of class three was reflected in 2009, where Church and colleagues were able to program Escherichia coli to produce five times the normal amount of lycopene,
    an antioxidant normally found in tomato seeds and linked to anti-cancer properties.

  • Prior to this new revolution, researchers would have to do single-gene manipulations and tweak the genome one little section at a time, observe the phenotype, and start the
    process over with a different single-gene manipulation.

  • Gene editing in fish is currently experimental, but the possibilities include growth, disease resistance, sterility, controlled reproduction, and colour.

  • Cas (CRISPR associated proteins) process these sequences and cut matching viral DNA sequences.

  • [84][85] George Church has compiled a list of potential genetic modifications for possibly advantageous traits such as less need for sleep, cognition-related changes that
    protect against Alzheimer’s disease, disease resistances and enhanced learning abilities along with some of the associated studies and potential negative effects.

  • [84][85] In 2001 Australian researchers Ronald Jackson and Ian Ramshaw were criticized for publishing a paper in the Journal of Virology that explored the potential control
    of mice, a major pest in Australia, by infecting them with an altered mousepox virus that would cause infertility as the provided sensitive information could lead to the manufacture of biological weapons by potential bioterrorists who might
    use the knowledge to create vaccine resistant strains of other pox viruses, such as smallpox, that could affect humans.

  • [83] According to a September 2016 report by the Nuffield Council on Bioethics in the future it may be possible to enhance people with genes from other organisms or wholly
    synthetic genes to for example improve night vision and sense of smell.

  • Gene drive are a potential tool to alter the reproductive rate of invasive species, although there are significant associated risks.

  • The review also found that the risks and benefits of modifying a person’s genome – and having those changes pass on to future generations – are so complex that they demand
    urgent ethical scrutiny.

  • [88] They recommended that clinical trials for genome editing might one day be permitted once answers have been found to safety and efficiency problems “but only for serious
    conditions under stringent oversight.

  • [92] According to a September 2016 report by the Nuffield Council on Bioethics, the simplicity and low cost of tools to edit the genetic code will allow amateurs – or “biohackers”
    – to perform their own experiments, posing a potential risk from the release of genetically modified bugs.

  • Genome editing occurs also as a natural process without artificial genetic engineering.

  • [68] In February 2019, medical scientists working with Sangamo Therapeutics, headquartered in Richmond, California, announced the first ever “in body” human gene editing therapy
    to permanently alter DNA – in a patient with Hunter syndrome.

  • “[89] Risks In the 2016 Worldwide Threat Assessment of the US Intelligence Community statement United States Director of National Intelligence, James R. Clapper, named genome
    editing as a potential weapon of mass destruction, stating that genome editing conducted by countries with regulatory or ethical standards “different from Western countries” probably increases the risk of the creation of harmful biological
    agents or products.

  • CRISPR can help bridge the gap between this model and human clinical trials by creating transgenic disease models in larger animals such as pigs, dogs, and non-human primates.

  • This is advantageous over a virally delivered gene as there is no need to include the full coding sequences and regulatory sequences when only a small proportions of the gene
    needs to be altered as is often the case.

  • [64][65] The expression of the partially replaced genes is also more consistent with normal cell biology than full genes that are carried by viral vectors.

  • Although GEEN has higher efficiency than many other methods in reverse genetics, it is still not highly efficient; in many cases less than half of the treated populations
    obtain the desired changes.

  • [79] Human enhancement[edit] Many transhumanists see genome editing as a potential tool for human enhancement.

  • [44] In November 2018, He Jiankui announced that he had edited two human embryos, to attempt to disable the gene for CCR5, which codes for a receptor that HIV uses to enter
    cells.

  • [66][67] Extensive research has been done in cells and animals using CRISPR-Cas9 to attempt to correct genetic mutations which cause genetic diseases such as Down syndrome,
    spina bifida, anencephaly, and Turner and Klinefelter syndromes.

  • In the future, a possible method to identify secondary targets would be to capture broken ends from cells expressing the ZFNs and to sequence the flanking DNA using high-throughput
    sequencing.

  • [65] Because of the ease of use and cost-efficiency of CRISPR, extensive research is currently being done on it.

  • Such modifications might have unintended consequences which could harm not only the child, but also their future children, as the altered gene would be in their sperm or eggs.

  • [90][91][92] For instance technologies such as CRISPR could be used to make “killer mosquitoes” that cause plagues that wipe out staple crops.

 

Works Cited

[‘1. Saurabh S (March 2021). “Genome Editing: Revolutionizing the Crop Improvement”. Plant Molecular Biology Reporte. 39 (4): 752–772. doi:10.1007/s11105-021-01286-7. S2CID 233713026.
2. ^ Bak, Rasmus O.; Gomez-Ospina, Natalia; Porteus, Matthew H.
(August 2018). “Gene Editing on Center Stage”. Trends in Genetics. 34 (8): 600–611. doi:10.1016/j.tig.2018.05.004. ISSN 0168-9525. PMID 29908711. S2CID 49269023.
3. ^ Jump up to:a b c Woolf TM (April 1998). “Therapeutic repair of mutated nucleic
acid sequences”. Nature Biotechnology. 16 (4): 341–4. doi:10.1038/nbt0498-341. PMID 9555723. S2CID 9210810.
4. ^ “Method of the Year 2011”. Nature Methods. 9 (1): 1. January 2012. doi:10.1038/nmeth.1852. PMID 22312634.
5. ^ Science News Staff
(17 December 2015). “Breakthrough of the Year: CRISPR makes the cut”.
6. ^ Esvelt KM, Wang HH (2013). “Genome-scale engineering for systems and synthetic biology”. Molecular Systems Biology. 9 (1): 641. doi:10.1038/msb.2012.66. PMC 3564264. PMID
23340847.
7. ^ Tan WS, Carlson DF, Walton MW, Fahrenkrug SC, Hackett PB (2012). “Precision editing of large animal genomes”. Advances in Genetics Volume 80. Vol. 80. pp. 37–97. doi:10.1016/B978-0-12-404742-6.00002-8. ISBN 9780124047426. PMC 3683964.
PMID 23084873. {{cite book}}: |journal= ignored (help)
8. ^ Puchta H, Fauser F (2013). “Gene targeting in plants: 25 years later”. The International Journal of Developmental Biology. 57 (6–8): 629–37. doi:10.1387/ijdb.130194hp. PMID 24166445.
9. ^
Jump up to:a b c d e f Boglioli E, Richard M. “Rewriting the book of life: a new era in precision genome editing” (PDF). Boston Consulting Group. Retrieved November 30, 2015.
10. ^ Church G. “The future of genetic codes and BRAIN codes”. YouTube.
NIHvcast. Retrieved 10 February 2017.
11. ^ Cyranoski D (May 2019). “China set to introduce gene-editing regulation following CRISPR-baby furore”. Nature. doi:10.1038/d41586-019-01580-1. PMID 32424191. S2CID 182604140. Retrieved 20 May 2019.
12. ^
Cheong KH, Koh JM, Jones MC (July 2019). “Black Swans of CRISPR: Stochasticity and Complexity of Genetic Regulation”. BioEssays. 41 (7): e1900032. doi:10.1002/bies.201900032. PMID 31090950. S2CID 155086967.
13. ^ Jump up to:a b c Stokstad, Erik
(2021-05-26). “U.K. set to loosen rules for gene-edited crops and animals”. Science | AAAS. Retrieved 2021-05-27.
14. ^ “US Trial Shows 3 Cancer Patients Had Their Genomes Altered Safely by CRISPR”. ScienceAlert. 7 February 2020. Retrieved 2020-02-09.
15. ^
Altae-Tran, Han; Kannan, Soumya; Demircioglu, F. Esra; Oshiro, Rachel; Nety, Suchita P.; McKay, Luke J.; Dlakić, Mensur; Inskeep, William P.; Makarova, Kira S.; Macrae, Rhiannon K.; Koonin, Eugene V. (2021-10-01). “The widespread IS200/IS605 transposon
family encodes diverse programmable RNA-guided endonucleases”. Science. 374 (6563): 57–65. Bibcode:2021Sci…374…57A. doi:10.1126/science.abj6856. ISSN 0036-8075. PMC 8929163. PMID 34591643. S2CID 238237947.
16. ^ “New programmable gene editing
proteins found outside of CRISPR systems”. Broad Institute. 2021-09-09. Retrieved 2021-10-04.
17. ^ Lodish H, Berk A, Zipursky SL, Matsudaira P, Baltimore D, Darnell J (2000). “Chapter 8.5: Gene Replacement and Transgenic Animals: DNA Is Transferred
into Eukaryotic Cells in Various Ways”. Molecular Cell Biology (4th ed.). W. H. Freeman and Company. ISBN 978-0-7167-3136-8.
18. ^ Jump up to:a b Rocha-Martins M, Cavalheiro GR, Matos-Rodrigues GE, Martins RA (August 2015). “From Gene Targeting
to Genome Editing: Transgenic animals applications and beyond”. Anais da Academia Brasileira de Ciências. 87 (2 Suppl): 1323–48. doi:10.1590/0001-3765201520140710. PMID 26397828.
19. ^ Jump up to:a b “The Nobel Prize in Physiology or Medicine 2007”.
The Nobel Foundation. Retrieved December 15, 2008.
20. ^ Jasin M (June 1996). “Genetic manipulation of genomes with rare-cutting endonucleases”. Trends in Genetics. 12 (6): 224–8. doi:10.1016/0168-9525(96)10019-6. PMID 8928227.
21. ^ Stoddard
BL (February 2005). “Homing endonuclease structure and function”. Quarterly Reviews of Biophysics. 38 (1): 49–95. doi:10.1017/s0033583505004063. PMID 16336743. S2CID 27841011.
22. ^ Jump up to:a b de Souza N (January 2012). “Primer: genome editing
with engineered nucleases”. Nature Methods. 9 (1): 27. doi:10.1038/nmeth.1848. PMID 22312638. S2CID 26924628.
23. ^ Jump up to:a b c Smith J, Grizot S, Arnould S, Duclert A, Epinat JC, Chames P, et al. (2006). “A combinatorial approach to create
artificial homing endonucleases cleaving chosen sequences”. Nucleic Acids Research. 34 (22): e149. doi:10.1093/nar/gkl720. PMC 1702487. PMID 17130168.
24. ^ Seligman LM, Chisholm KM, Chevalier BS, Chadsey MS, Edwards ST, Savage JH, Veillet AL (September
2002). “Mutations altering the cleavage specificity of a homing endonuclease”. Nucleic Acids Research. 30 (17): 3870–9. doi:10.1093/nar/gkf495. PMC 137417. PMID 12202772.
25. ^ Chevalier BS, Kortemme T, Chadsey MS, Baker D, Monnat RJ, Stoddard BL
(October 2002). “Design, activity, and structure of a highly specific artificial endonuclease”. Molecular Cell. 10 (4): 895–905. doi:10.1016/S1097-2765(02)00690-1. PMID 12419232.
26. ^ Arnould S, Chames P, Perez C, Lacroix E, Duclert A, Epinat JC,
et al. (January 2006). “Engineering of large numbers of highly specific homing endonucleases that induce recombination on novel DNA targets”. Journal of Molecular Biology. 355 (3): 443–58. doi:10.1016/j.jmb.2005.10.065. PMID 16310802.
27. ^ Rationally-designed
meganucleases with altered sequence specificity and DNA-binding affinity, 2006-10-18, retrieved 2018-08-11
28. ^ Ashworth J, Taylor GK, Havranek JJ, Quadri SA, Stoddard BL, Baker D (September 2010). “Computational reprogramming of homing endonuclease
specificity at multiple adjacent base pairs”. Nucleic Acids Research. 38 (16): 5601–8. doi:10.1093/nar/gkq283. PMC 2938204. PMID 20435674.
29. ^ Redondo P, Prieto J, Muñoz IG, Alibés A, Stricher F, Serrano L, et al. (November 2008). “Molecular basis
of xeroderma pigmentosum group C DNA recognition by engineered meganucleases”. Nature. 456 (7218): 107–11. Bibcode:2008Natur.456..107R. doi:10.1038/nature07343. PMID 18987743. S2CID 4300643.
30. ^ Jump up to:a b Baker M (January 2012). “Gene-editing
nucleases”. Nature Methods. 9 (1): 23–6. doi:10.1038/nmeth.1807. PMID 22312637. S2CID 37050234.
31. ^ Rebar EJ, Huang Y, Hickey R, Nath AK, Meoli D, Nath S, et al. (December 2002). “Induction of angiogenesis in a mouse model using engineered transcription
factors”. Nature Medicine. 8 (12): 1427–32. doi:10.1038/nm1202-795. PMID 12415262. S2CID 23318821.
32. ^ Kim YG, Cha J, Chandrasegaran S (February 1996). “Hybrid restriction enzymes: zinc finger fusions to Fok I cleavage domain”. Proceedings of
the National Academy of Sciences of the United States of America. 93 (3): 1156–60. Bibcode:1996PNAS…93.1156K. doi:10.1073/pnas.93.3.1156. PMC 40048. PMID 8577732.
33. ^ Jump up to:a b c d Urnov FD, Rebar EJ, Holmes MC, Zhang HS, Gregory PD (September
2010). “Genome editing with engineered zinc finger nucleases”. Nature Reviews. Genetics. 11 (9): 636–46. doi:10.1038/nrg2842. PMID 20717154. S2CID 205484701.
34. ^ Reik A, et al. (2008). “Zinc finger nucleases targeting the glucocorticoid receptor
allow IL-13 zetakine transgenic CTLs to kill glioblastoma cells in vivo in the presence of immunosuppressing glucocorticoids”. Mol. Ther. 16 (Supplement 1): S13–S14. doi:10.1016/S1525-0016(16)39437-0.
35. ^ Holt N, Wang J, Kim K, Friedman G, Wang
X, Taupin V, et al. (August 2010). “Human hematopoietic stem/progenitor cells modified by zinc-finger nucleases targeted to CCR5 control HIV-1 in vivo”. Nature Biotechnology. 28 (8): 839–47. doi:10.1038/nbt.1663. PMC 3080757. PMID 20601939.
36. ^
Gaj T, Gersbach CA, Barbas CF (July 2013). “ZFN, TALEN, and CRISPR/Cas-based methods for genome engineering”. Trends in Biotechnology. 31 (7): 397–405. doi:10.1016/j.tibtech.2013.04.004. PMC 3694601. PMID 23664777.
37. ^ Pérez-Quintero AL, Rodriguez-R
LM, Dereeper A, López C, Koebnik R, Szurek B, Cunnac S (2013-07-15). “An improved method for TAL effectors DNA-binding sites prediction reveals functional convergence in TAL repertoires of Xanthomonas oryzae strains”. PLOS ONE. 8 (7): e68464. Bibcode:2013PLoSO…868464P.
doi:10.1371/journal.pone.0068464. PMC 3711819. PMID 23869221.
38. ^ Young S (11 February 2014). “Genome Surgery”. MIT Technology Review.
39. ^ Woolf TM, Chase JM, Stinchcomb DT (August 1995). “Toward the therapeutic editing of mutated RNA sequences”.
Proceedings of the National Academy of Sciences of the United States of America. 92 (18): 8298–302. Bibcode:1995PNAS…92.8298W. doi:10.1073/pnas.92.18.8298. PMC 41144. PMID 7545300.
40. ^ Woolf TM, Gurumurthy CB, Boyce F, Kmiec EB (April 2017).
“To cleave or not to cleave: therapeutic gene editing with and without programmable nucleases”. Nature Reviews. Drug Discovery. 16 (4): 296. doi:10.1038/nrd.2017.42. PMID 28303022.
41. ^ Jump up to:a b Komor AC, Kim YB, Packer MS, Zuris JA, Liu
DR (May 2016). “Programmable editing of a target base in genomic DNA without double-stranded DNA cleavage”. Nature. 533 (7603): 420–4. Bibcode:2016Natur.533..420K. doi:10.1038/nature17946. PMC 4873371. PMID 27096365.SharedIt
42. ^ Cervantes-Gracia
K, Gramalla-Schmitz A, Weischedel J, Chahwan R (2021). “APOBECs orchestrate genomic and epigenomic editing across health and disease”. Trends Genet. 37 (11): 1028–1043. doi:10.1016/j.tig.2021.07.003. ISSN 0168-9525. PMID 34353635. S2CID 236934922.
43. ^
Khan, Sikandar Hayat (2019-06-07). “Genome-Editing Technologies: Concept, Pros, and Cons of Various Genome-Editing Techniques and Bioethical Concerns for Clinical Application”. Molecular Therapy. Nucleic Acids. 16. Elsevier BV: 326–334. doi:10.1016/j.omtn.2019.02.027.
ISSN 2162-2531. PMC 6454098. PMID 30965277.
44. ^ Jump up to:a b c d Barrangou R, Doudna JA (September 2016). “Applications of CRISPR technologies in research and beyond”. Nature Biotechnology. 34 (9): 933–941. doi:10.1038/nbt.3659. PMID 27606440.
S2CID 21543486.
45. ^ Kim H, Kim JS (May 2014). “A guide to genome engineering with programmable nucleases”. Nature Reviews. Genetics. 15 (5): 321–34. doi:10.1038/nrg3686. PMID 24690881. S2CID 9373606.
46. ^ Jump up to:a b c d Gallagher RR, Li
Z, Lewis AO, Isaacs FJ (October 2014). “Rapid editing and evolution of bacterial genomes using libraries of synthetic DNA”. Nature Protocols. 9 (10): 2301–16. doi:10.1038/nprot.2014.082. PMID 25188632. S2CID 16447825.
47. ^ Jump up to:a b McMahon
MA, Rahdar M, Porteus M (December 2011). “Gene editing: not just for translation anymore”. Nature Methods. 9 (1): 28–31. doi:10.1038/nmeth.1811. PMID 22205513. S2CID 2144013.
48. ^ Daigneault BW, Rajput S, Smith GW, Ross PJ (May 2018). “Embryonic
POU5F1 is Required for Expanded Bovine Blastocyst Formation”. Scientific Reports. 8 (1): 7753. Bibcode:2018NatSR…8.7753D. doi:10.1038/s41598-018-25964-x. PMC 5958112. PMID 29773834.
49. ^ Wargelius, Anna (2019). “Application of genome editing
in aquatic farm animals”. Transgenic Research. 28 (Suppl 2): 101–105. doi:10.1007/s11248-019-00163-0. PMID 31321691. S2CID 197665984. Retrieved 29 April 2021.
50. ^ Yaskowiak, Edward S.; Shears, Margaret A.; Agarwal-Mawal, Alka; Fletcher, Garth
L. (2006). “Characterization and multi-generational stability of the growth hormone transgene (EO-1α) responsible for enhanced growth rates in Atlantic Salmon”. Transgenic Research. 15 (4): 465–480. doi:10.1007/s11248-006-0020-5. PMID 16906447. S2CID
22247493.
51. ^ Ortega NM, Winblad N, Plaza Reyes A, Lanner F (October 2018). “Functional genetics of early human development”. Current Opinion in Genetics & Development. 52: 1–6. doi:10.1016/j.gde.2018.04.005. PMID 29729430.
52. ^ Arnould S,
Delenda C, Grizot S, Desseaux C, Pâques F, Silva GH, Smith J (January 2011). “The I-CreI meganuclease and its engineered derivatives: applications from cell modification to gene therapy”. Protein Engineering, Design & Selection. 24 (1–2): 27–31. doi:10.1093/protein/gzq083.
PMID 21047873.
53. ^ Jump up to:a b c Townsend JA, Wright DA, Winfrey RJ, Fu F, Maeder ML, Joung JK, Voytas DF (May 2009). “High-frequency modification of plant genes using engineered zinc-finger nucleases”. Nature. 459 (7245): 442–5. Bibcode:2009Natur.459..442T.
doi:10.1038/nature07845. PMC 2743854. PMID 19404258.
54. ^ Zhang F, Maeder ML, Unger-Wallace E, Hoshaw JP, Reyon D, Christian M, et al. (June 2010). “High frequency targeted mutagenesis in Arabidopsis thaliana using zinc finger nucleases”. Proceedings
of the National Academy of Sciences of the United States of America. 107 (26): 12028–33. Bibcode:2010PNAS..10712028Z. doi:10.1073/pnas.0914991107. PMC 2900673. PMID 20508152.
55. ^ Osakabe K, Osakabe Y, Toki S (June 2010). “Site-directed mutagenesis
in Arabidopsis using custom-designed zinc finger nucleases”. Proceedings of the National Academy of Sciences of the United States of America. 107 (26): 12034–9. Bibcode:2010PNAS..10712034O. doi:10.1073/pnas.1000234107. PMC 2900650. PMID 20508151.
56. ^
Jump up to:a b Shukla VK, Doyon Y, Miller JC, DeKelver RC, Moehle EA, Worden SE, et al. (May 2009). “Precise genome modification in the crop species Zea mays using zinc-finger nucleases”. Nature. 459 (7245): 437–41. Bibcode:2009Natur.459..437S. doi:10.1038/nature07992.
PMID 19404259. S2CID 4323298.
57. ^ Tripathi JN, Ntui VO, Ron M, Muiruri SK, Britt A, Tripathi L (2019-01-31). “CRISPR/Cas9 editing of endogenous banana streak virus in the B genome of Musa spp. overcomes a major challenge in banana breeding”. Communications
Biology. 2 (1): 46. doi:10.1038/s42003-019-0288-7. PMC 6355771. PMID 30729184.
58. ^ Townson J (2017-01-01). “Recent developments in genome editing for potential use in plants”. Bioscience Horizons. 10. doi:10.1093/biohorizons/hzx016.
59. ^ Regalado
A (19 December 2017). “These Are Not Your Father’s GMOs”. MIT Technology Review. Retrieved 16 April 2018.
60. ^ Haun W, Coffman A, Clasen BM, Demorest ZL, Lowy A, Ray E, et al. (September 2014). “Improved soybean oil quality by targeted mutagenesis
of the fatty acid desaturase 2 gene family”. Plant Biotechnology Journal. 12 (7): 934–40. doi:10.1111/pbi.12201. PMID 24851712.
61. ^ Clasen BM, Stoddard TJ, Luo S, Demorest ZL, Li J, Cedrone F, et al. (January 2016). “Improving cold storage and
processing traits in potato through targeted gene knockout”. Plant Biotechnology Journal. 14 (1): 169–76. doi:10.1111/pbi.12370. PMID 25846201.
62. ^ Jump up to:a b Puchta H, Hohn B (June 2010). “Breaking news: plants mutate right on target”. Proceedings
of the National Academy of Sciences of the United States of America. 107 (26): 11657–8. Bibcode:2010PNAS..10711657P. doi:10.1073/pnas.1006364107. PMC 2900667. PMID 20554917.
63. ^ Jump up to:a b c Paul JW, Qi Y (July 2016). “CRISPR/Cas9 for plant
genome editing: accomplishments, problems and prospects”. Plant Cell Reports. 35 (7): 1417–27. doi:10.1007/s00299-016-1985-z. PMID 27114166. S2CID 8035222.
64. ^ Monga, Isha; Qureshi, Abid; Thakur, Nishant; Gupta, Amit Kumar; Kumar, Manoj (September
2017). “ASPsiRNA: A Resource of ASP-siRNAs Having Therapeutic Potential for Human Genetic Disorders and Algorithm for Prediction of Their Inhibitory Efficacy”. G3. 7 (9): 2931–2943. doi:10.1534/g3.117.044024. PMC 5592921. PMID 28696921.
65. ^ Jump
up to:a b c Carroll D (November 2008). “Progress and prospects: zinc-finger nucleases as gene therapy agents”. Gene Therapy. 15 (22): 1463–8. doi:10.1038/gt.2008.145. PMC 2747807. PMID 18784746.
66. ^ Pollack A (2015-11-05). “A Cell Therapy Untested
in Humans Saves a Baby With Cancer”. The New York Times. ISSN 0362-4331. Retrieved 2015-11-30.
67. ^ Couzin-Frankel J (November 2015). “CANCER IMMUNOTHERAPY. Baby’s leukemia recedes after novel cell therapy”. Science. 350 (6262): 731. doi:10.1126/science.350.6262.731.
PMID 26564829.
68. ^ Mentis AF (December 2016). “Epigenomic engineering for Down syndrome”. Neuroscience and Biobehavioral Reviews. 71: 323–327. doi:10.1016/j.neubiorev.2016.09.012. PMID 27646312. S2CID 24192441.
69. ^ Marchione M (7 February
2019). “Tests suggest scientists achieved 1st ‘in body’ gene editing”. AP News. Retrieved 7 February 2019.
70. ^ Staff (2 February 2019). “Ascending Dose Study of Genome Editing by the Zinc Finger Nuclease (ZFN) Therapeutic SB-913 in Subjects
With MPS II”. ClinicalTrials.gov. U.S. National Library of Medicine. Retrieved 7 February 2019.
71. ^ Hammond A, Galizi R, Kyrou K, Simoni A, Siniscalchi C, Katsanos D, et al. (January 2016). “A CRISPR-Cas9 gene drive system targeting female reproduction
in the malaria mosquito vector Anopheles gambiae”. Nature Biotechnology. 34 (1): 78–83. doi:10.1038/nbt.3439. PMC 4913862. PMID 26641531.
72. ^ Fletcher M (2018-08-11). “Mutant mosquitoes: Can gene editing kill off malaria?”. The Telegraph. ISSN
0307-1235. Retrieved 2018-08-12.
73. ^ Begley S (28 November 2018). “Amid uproar, Chinese scientist defends creating gene-edited babies – STAT”. STAT.
74. ^ Science China Press (23 January 2019). “Gene-edited disease monkeys cloned in China”.
EurekAlert!. Retrieved 24 January 2019.
75. ^ Mandelbaum RF (23 January 2019). “China’s Latest Cloned-Monkey Experiment Is an Ethical Mess”. Gizmodo. Retrieved 24 January 2019.
76. ^ Teboul L, Herault Y, Wells S, Qasim W, Pavlovic G (June 2020).
“Variability in Genome Editing Outcomes: Challenges for Research Reproducibility and Clinical Safety”. Molecular Therapy. 28 (6): 1422–1431. doi:10.1016/j.ymthe.2020.03.015. PMC 7264426. PMID 32243835.
77. ^ Im W, Moon J, Kim M (September 2016).
“Applications of CRISPR/Cas9 for Gene Editing in Hereditary Movement Disorders”. Journal of Movement Disorders. 9 (3): 136–43. doi:10.14802/jmd.16029. PMC 5035944. PMID 27667185.
78. ^ Jump up to:a b Hsu PD, Lander ES, Zhang F (June 2014). “Development
and applications of CRISPR-Cas9 for genome engineering”. Cell. 157 (6): 1262–1278. doi:10.1016/j.cell.2014.05.010. PMC 4343198. PMID 24906146.
79. ^ Johnson JA, Altwegg R, Evans DM, Ewen JG, Gordon IJ, Pettorelli N, Young JK (2016-04-01). “Is there
a future for genome-editing technologies in conservation?”. Animal Conservation. 19 (2): 97–101. doi:10.1111/acv.12273. ISSN 1469-1795. S2CID 27633404.
80. ^ Pearlman A (2015-12-03). “Geneticists Are Concerned Transhumanists Will Use CRISPR on Themselves”.
Vice Motherboard. Retrieved 26 December 2016.
81. ^ Jorgensen E. “How DIY bio-hackers are changing the conversation around genetic engineering”. The Washington Post. Retrieved 26 December 2016.
82. ^ “Human Enhancement”. Pew Research Center. 2016-07-26.
Retrieved 26 December 2016.
83. ^ Regalado A. “Engineering the Perfect Baby”. MIT Technology Review. Retrieved 26 December 2016.
84. ^ Jump up to:a b Sample I (30 September 2016). “Experts warn home ‘gene editing’ kits pose risk to society”.
The Guardian. Retrieved 26 December 2016.
85. ^ Jump up to:a b c d “Genome editing: an ethical review” (PDF). Nuffield Council on Bioethics. September 2016. Retrieved 27 December 2016.
86. ^ “George Church told us why he’s listing “superhuman”
gene hacks”. Futurism. Retrieved 25 July 2021.
87. ^ “Protective alleles”. arep.med.harvard.edu. Retrieved 25 July 2021.
88. ^ Harmon A (2017-02-14). “Human Gene Editing Receives Science Panel’s Support”. The New York Times. ISSN 0362-4331. Retrieved
2017-02-17.
89. ^ “Scientists OK genetically engineering babies”. New York Post. Reuters. 2017-02-14. Retrieved 2017-02-17.
90. ^ Clapper JR (9 February 2016). “Worldwide Threat Assessment of the US Intelligence Community” (PDF). Retrieved 26
December 2016.
91. ^ Warmflash D (2016-09-06). “Genome editing: Is it a national security threat?”. Retrieved 26 December 2016.
92. ^ Jump up to:a b Regalado A. “Top U.S. Intelligence Official Calls Gene Editing a WMD Threat”. MIT Technology Review.
Retrieved 26 December 2016.
93. ^ Jackson R, Ramshaw I (January 2010). “The mousepox experience. An interview with Ronald Jackson and Ian Ramshaw on dual-use research. Interview by Michael J. Selgelid and Lorna Weir”. EMBO Reports. 11 (1): 18–24.
doi:10.1038/embor.2009.270. PMC 2816623. PMID 20010799.
94. ^ Broad WJ (23 January 2001). “Australians Create a Deadly Mouse Virus”. The New York Times. Retrieved 27 December 2016.
95. ^ Radford T (10 January 2001). “Lab creates killer virus by
accident”. The Guardian. Retrieved 27 December 2016.
96. ^ “The Nobel Prize in Chemistry 2020”. Nobel Foundation. Archived from the original on 7 October 2020. Retrieved 7 October 2020.
“WHO launches global registry on human genome editing.” PharmaBiz,
31 Aug. 2019. Gale General OneFile, Accessed 27 Apr. 2020.
Photo credit: https://www.flickr.com/photos/meteomarco/5955475577/’]