Transcription activator-like effector nuclease

Last updated
Spacefill drawing of dimeric TALE-FokI fusion (blue: TALE; green: FokI) bound to DNA (PDB: 1FOK, 3UGM ), by David Goodsell 180-TALEffectors TALEN.png
Spacefill drawing of dimeric TALE-FokI fusion (blue: TALE; green: FokI) bound to DNA ( PDB: 1FOK, 3UGM ), by David Goodsell

Transcription activator-like effector nucleases (TALEN) are restriction enzymes that can be engineered to cut specific sequences of DNA. They are made by fusing a TAL effector DNA-binding domain to a DNA cleavage domain (a nuclease which cuts DNA strands). Transcription activator-like effectors (TALEs) can be engineered to bind to practically any desired DNA sequence, so when combined with a nuclease, DNA can be cut at specific locations. [1] The restriction enzymes can be introduced into cells, for use in gene editing or for genome editing in situ , a technique known as genome editing with engineered nucleases. Alongside zinc finger nucleases and CRISPR/Cas9, TALEN is a prominent tool in the field of genome editing.

Contents

TALE DNA-binding domain

TAL effectors are proteins that are secreted by Xanthomonas bacteria via their type III secretion system when they infect plants. [2] The DNA binding domain contains a repeated highly conserved 33–34 amino acid sequence with divergent 12th and 13th amino acids. These two positions, referred to as the Repeat Variable Diresidue (RVD), are highly variable and show a strong correlation with specific nucleotide recognition. [3] [4] This straightforward relationship between amino acid sequence and DNA recognition has allowed for the engineering of specific DNA-binding domains by selecting a combination of repeat segments containing the appropriate RVDs. [1] Notably, slight changes in the RVD and the incorporation of "nonconventional" RVD sequences can improve targeting specificity. [5]

DNA cleavage domain

The non-specific DNA cleavage domain from the end of the FokI endonuclease can be used to construct hybrid nucleases that are active in a yeast assay. [6] [7] These reagents are also active in plant cells [8] [9] and in animal cells. [9] [10] [11] [12] Initial TALEN studies used the wild-type FokI cleavage domain, but some subsequent TALEN studies [11] [13] [14] also used FokI cleavage domain variants with mutations designed to improve cleavage specificity [15] [16] and cleavage activity. [17] The FokI domain functions as a dimer, requiring two constructs with unique DNA binding domains for sites in the target genome with proper orientation and spacing. Both the number of amino acid residues between the TALE DNA binding domain and the FokI cleavage domain and the number of bases between the two individual TALEN binding sites appear to be important parameters for achieving high levels of activity. [10] [18]

Engineering TALEN constructs

The simple relationship between amino acid sequence and DNA recognition of the TALE binding domain allows for the efficient engineering of proteins. In this case, artificial gene synthesis is problematic because of improper annealing of the repetitive sequence found in the TALE binding domain. [19] One solution to this is to use a publicly available software program (DNAWorks [20] ) to calculate oligonucleotides suitable for assembly in a two step PCR oligonucleotide assembly followed by whole gene amplification. A number of modular assembly schemes for generating engineered TALE constructs have also been reported. [9] [19] [21] [22] [23] [24] Both methods offer a systematic approach to engineering DNA binding domains that is conceptually similar to the modular assembly method for generating zinc finger DNA recognition domains.

Workflow of genome editing of Your Favorite Gene (YFG) using TALEN. The target sequence is identified, a corresponding TALEN sequence is engineered and inserted into a plasmid. The plasmid is inserted into the target cell where it is translated to produce the functional TALEN, which enters the nucleus and binds and cleaves the target sequence. Depending on the application, this can be used to introduce an error (to knock out a target gene) or to introduce a new DNA sequence into the target gene. TALEN copy.svg
Workflow of genome editing of Your Favorite Gene (YFG) using TALEN. The target sequence is identified, a corresponding TALEN sequence is engineered and inserted into a plasmid. The plasmid is inserted into the target cell where it is translated to produce the functional TALEN, which enters the nucleus and binds and cleaves the target sequence. Depending on the application, this can be used to introduce an error (to knock out a target gene) or to introduce a new DNA sequence into the target gene.

Transfection

Once the TALEN constructs have been assembled, they are inserted into plasmids; the target cells are then transfected with the plasmids, and the gene products are expressed and enter the nucleus to access the genome. Alternatively, TALEN constructs can be delivered to the cells as mRNAs, which removes the possibility of genomic integration of the TALEN-expressing protein. Using an mRNA vector can also dramatically increase the level of homology directed repair (HDR) and the success of introgression during gene editing.

Genome editing

Mechanisms

TALEN can be used to edit genomes by inducing double-strand breaks (DSB), which cells respond to with repair mechanisms.

Non-homologous end joining (NHEJ) directly ligates DNA from either side of a double-strand break where there is very little or no sequence overlap for annealing. This repair mechanism induces errors in the genome via indels (insertion or deletion), or chromosomal rearrangement; any such errors may render the gene products coded at that location non-functional. [10] Because this activity can vary depending on the species, cell type, target gene, and nuclease used, it should be monitored when designing new systems. A simple heteroduplex cleavage assay can be run which detects any difference between two alleles amplified by PCR. Cleavage products can be visualized on simple agarose gels or slab gel systems.

Alternatively, DNA can be introduced into a genome through NHEJ in the presence of exogenous double-stranded DNA fragments. [10]

Homology directed repair can also introduce foreign DNA at the DSB as the transfected double-stranded sequences are used as templates for the repair enzymes. [10]

Applications

TALEN has been used to efficiently modify plant genomes, [25] creating economically important food crops with favorable nutritional qualities. [26] They have also been harnessed to develop tools for the production of biofuels. [27] In addition, it has been used to engineer stably modified human embryonic stem cell and induced pluripotent stem cell (IPSCs) clones and human erythroid cell lines, [11] [28] to generate knockout C. elegans , [12] knockout rats, [13] knockout mice, [29] and knockout zebrafish. [14] [30] Moreover, the method can be used to generate knockin organisms. Wu et al.obtained a Sp110 knockin cattle using Talen nickases to induce increased resistance of tuberculosis. [31] This approach has also been used to generate knockin rats by TALEN mRNA microinjection in one-cell embryos. [32]

TALEN has also been utilized experimentally to correct the genetic errors that underlie disease. [33] For example, it has been used in vitro to correct the genetic defects that cause disorders such as sickle cell disease, [28] [34] xeroderma pigmentosum, [35] and epidermolysis bullosa. [36] Recently, it was shown that TALEN can be used as tools to harness the immune system to fight cancers; TALEN-mediated targeting can generate T cells that are resistant to chemotherapeutic drugs and show anti-tumor activity. [37] [38]

In theory, the genome-wide specificity of engineered TALEN fusions allows for correction of errors at individual genetic loci via homology-directed repair from a correct exogenous template. [33] In reality, however, the in situ application of TALEN is currently limited by the lack of an efficient delivery mechanism, unknown immunogenic factors, and uncertainty in the specificity of TALEN binding. [33]

Another emerging application of TALEN is its ability to combine with other genome engineering tools, such as meganucleases. The DNA binding region of a TAL effector can be combined with the cleavage domain of a meganuclease to create a hybrid architecture combining the ease of engineering and highly specific DNA binding activity of a TAL effector with the low site frequency and specificity of a meganuclease. [39]

In comparison to other genome editing techniques, TALEN falls in the middle in terms of difficulty and cost. Unlike ZFNs, TALEN recognizes single nucleotides. It's far more straightforward to engineer interactions between TALEN DNA binding domains and their target nucleotides than it is to create interactions with ZFNs and their target nucleotide triplets. [40] On the other hand, CRISPR relies on ribonucleotide complex formation instead of protein/DNA recognition. gRNAs[ definition needed ] have occasionally limitations regarding feasibility due to lack of PAM sites[ definition needed ] in the target sequence and even though they can be cheaply produced, the current development lead to a remarkable decrease of cost for TALENs, so that they are in a similar price and time range like CRISPR based genome editing[ clarification needed ].

TAL effector nuclease precision

The off-target activity of an active nuclease may lead to unwanted double-strand breaks and may consequently yield chromosomal rearrangements and/or cell death. Studies have been carried out to compare the relative nuclease-associated toxicity of available technologies. Based on these studies [18] and the maximal theoretical distance between DNA binding and nuclease activity, TALEN constructs are believed to have the greatest precision of the currently available technologies. [41]

See also

Related Research Articles

A restriction enzyme, restriction endonuclease, REase, ENase orrestrictase is an enzyme that cleaves DNA into fragments at or near specific recognition sites within molecules known as restriction sites. Restriction enzymes are one class of the broader endonuclease group of enzymes. Restriction enzymes are commonly classified into five types, which differ in their structure and whether they cut their DNA substrate at their recognition site, or if the recognition and cleavage sites are separate from one another. To cut DNA, all restriction enzymes make two incisions, once through each sugar-phosphate backbone of the DNA double helix.

<span class="mw-page-title-main">Zinc finger</span> Small structural protein motif found mostly in transcriptional proteins

A zinc finger is a small protein structural motif that is characterized by the coordination of one or more zinc ions (Zn2+) which stabilizes the fold. It was originally coined to describe the finger-like appearance of a hypothesized structure from the African clawed frog (Xenopus laevis) transcription factor IIIA. However, it has been found to encompass a wide variety of differing protein structures in eukaryotic cells. Xenopus laevis TFIIIA was originally demonstrated to contain zinc and require the metal for function in 1983, the first such reported zinc requirement for a gene regulatory protein followed soon thereafter by the Krüppel factor in Drosophila. It often appears as a metal-binding domain in multi-domain proteins.

Gene knockdown is an experimental technique by which the expression of one or more of an organism's genes is reduced. The reduction can occur either through genetic modification or by treatment with a reagent such as a short DNA or RNA oligonucleotide that has a sequence complementary to either gene or an mRNA transcript.

<span class="mw-page-title-main">DNA-binding protein</span> Proteins that bind with DNA, such as transcription factors, polymerases, nucleases and histones

DNA-binding proteins are proteins that have DNA-binding domains and thus have a specific or general affinity for single- or double-stranded DNA. Sequence-specific DNA-binding proteins generally interact with the major groove of B-DNA, because it exposes more functional groups that identify a base pair.

<span class="mw-page-title-main">Germline mutation</span> Inherited genetic variation

A germline mutation, or germinal mutation, is any detectable variation within germ cells. Mutations in these cells are the only mutations that can be passed on to offspring, when either a mutated sperm or oocyte come together to form a zygote. After this fertilization event occurs, germ cells divide rapidly to produce all of the cells in the body, causing this mutation to be present in every somatic and germline cell in the offspring; this is also known as a constitutional mutation. Germline mutation is distinct from somatic mutation.

Zinc-finger nucleases (ZFNs) are artificial restriction enzymes generated by fusing a zinc finger DNA-binding domain to a DNA-cleavage domain. Zinc finger domains can be engineered to target specific desired DNA sequences and this enables zinc-finger nucleases to target unique sequences within complex genomes. By taking advantage of endogenous DNA repair machinery, these reagents can be used to precisely alter the genomes of higher organisms. Alongside CRISPR/Cas9 and TALEN, ZFN is a prominent tool in the field of genome editing.

<i>Fok</i>I Restriction enzyme

The restriction endonuclease Fok1, naturally found in Flavobacterium okeanokoites, is a bacterial type IIS restriction endonuclease consisting of an N-terminal DNA-binding domain and a non sequence-specific DNA cleavage domain at the C-terminal. Once the protein is bound to duplex DNA via its DNA-binding domain at the 5'-GGATG-3' recognition site, the DNA cleavage domain is activated and cleaves the DNA at two locations, regardless of the nucleotide sequence at the cut site. The DNA is cut 9 nucleotides downstream of the motif on the forward strand, and 13 nucleotides downstream of the motif on the reverse strand, producing two sticky ends with 4-bp overhangs.

Zinc finger protein chimera are chimeric proteins composed of a DNA-binding zinc finger protein domain and another domain through which the protein exerts its effect. The effector domain may be a transcriptional activator (A) or repressor (R), a methylation domain (M) or a nuclease (N).

Meganucleases are endodeoxyribonucleases characterized by a large recognition site ; as a result this site generally occurs only once in any given genome. For example, the 18-base pair sequence recognized by the I-SceI meganuclease would on average require a genome twenty times the size of the human genome to be found once by chance. Meganucleases are therefore considered to be the most specific naturally occurring restriction enzymes.

<span class="mw-page-title-main">Transcription activator-like effector</span>

TALeffectors are proteins secreted by some β- and γ-proteobacteria. Most of these are Xanthomonads. Plant pathogenic Xanthomonas bacteria are especially known for TALEs, produced via their type III secretion system. These proteins can bind promoter sequences in the host plant and activate the expression of plant genes that aid bacterial infection. The TALE domain responsible for binding to DNA is known to have 1.5 to 33.5 short sequences that are repeated multiple times. Each of these repeats was found to be specific for a certain base pair of the DNA. These repeats also have repeat variable residues (RVD) that can detect specific DNA base pairs. They recognize plant DNA sequences through a central repeat domain consisting of a variable number of ~34 amino acid repeats. There appears to be a one-to-one correspondence between the identity of two critical amino acids in each repeat and each DNA base in the target sequence. These proteins are interesting to researchers both for their role in disease of important crop species and the relative ease of retargeting them to bind new DNA sequences. Similar proteins can be found in the pathogenic bacterium Ralstonia solanacearum and Burkholderia rhizoxinica, as well as yet unidentified marine microorganisms. The term TALE-likes is used to refer to the putative protein family encompassing the TALEs and these related proteins.

Recombinant adeno-associated virus (rAAV) based genome engineering is a genome editing platform centered on the use of recombinant AAV vectors that enables insertion, deletion or substitution of DNA sequences into the genomes of live mammalian cells. The technique builds on Mario Capecchi and Oliver Smithies' Nobel Prize–winning discovery that homologous recombination (HR), a natural hi-fidelity DNA repair mechanism, can be harnessed to perform precise genome alterations in mice. rAAV mediated genome-editing improves the efficiency of this technique to permit genome engineering in any pre-established and differentiated human cell line, which, in contrast to mouse ES cells, have low rates of HR.

<span class="mw-page-title-main">Genome editing</span> Type of genetic engineering

Genome editing, or genome engineering, or gene editing, is a type of genetic engineering in which DNA is inserted, deleted, modified or replaced in the genome of a living organism. Unlike early genetic engineering techniques that randomly inserts genetic material into a host genome, genome editing targets the insertions to site-specific locations. The basic mechanism involved in genetic manipulations through programmable nucleases is the recognition of target genomic loci and binding of effector DNA-binding domain (DBD), double-strand breaks (DSBs) in target DNA by the restriction endonucleases, and the repair of DSBs through homology-directed recombination (HDR) or non-homologous end joining (NHEJ).

<span class="mw-page-title-main">Genetic engineering techniques</span> Methods used to change the DNA of organisms

Genetic engineering techniques allow the modification of animal and plant genomes. Techniques have been devised to insert, delete, and modify DNA at multiple levels, ranging from a specific base pair in a specific gene to entire genes. There are a number of steps that are followed before a genetically modified organism (GMO) is created. Genetic engineers must first choose what gene they wish to insert, modify, or delete. The gene must then be isolated and incorporated, along with other genetic elements, into a suitable vector. This vector is then used to insert the gene into the host genome, creating a transgenic or edited organism.

<span class="mw-page-title-main">Cas9</span> Microbial protein found in Streptococcus pyogenes M1 GAS

Cas9 is a 160 kilodalton protein which plays a vital role in the immunological defense of certain bacteria against DNA viruses and plasmids, and is heavily utilized in genetic engineering applications. Its main function is to cut DNA and thereby alter a cell's genome. The CRISPR-Cas9 genome editing technique was a significant contributor to the Nobel Prize in Chemistry in 2020 being awarded to Emmanuelle Charpentier and Jennifer Doudna.

A protospacer adjacent motif (PAM) is a 2–6-base pair DNA sequence immediately following the DNA sequence targeted by the Cas9 nuclease in the CRISPR bacterial adaptive immune system. The PAM is a component of the invading virus or plasmid, but is not found in the bacterial host genome and hence is not a component of the bacterial CRISPR locus. Cas9 will not successfully bind to or cleave the target DNA sequence if it is not followed by the PAM sequence. PAM is an essential targeting component which distinguishes bacterial self from non-self DNA, thereby preventing the CRISPR locus from being targeted and destroyed by the CRISPR-associated nuclease.

Transcription Activator-Like Effector-Likes (TALE-likes) are a group of bacterial DNA binding proteins named for the first and still best-studied group, the TALEs of Xanthomonas bacteria. TALEs are important factors in the plant diseases caused by Xanthomonas bacteria, but are known primarily for their role in biotechnology as programmable DNA binding proteins, particularly in the context of TALE nucleases. TALE-likes have additionally been found in many strains of the Ralstonia solanacearum bacterial species complex, in Paraburkholderia rhizoxinica strain HKI 454, and in two unknown marine bacteria. Whether or not all these proteins form a single phylogenetic grouping is as yet unclear.

<span class="mw-page-title-main">Daniel Voytas</span> American geneticist

Daniel Voytas is an American geneticist who is Professor of Genetics, Cell Biology and Development at the University of Minnesota and Director of the Center for Precision Plant Genomics. He is also the Chief Scientific Officer of Calyxt, an agricultural biotechnology company focused on developing crops that provide consumer benefit.

J. Keith Joung is an American pathologist and molecular biologist who holds the Robert B. Colvin Endowed Chair in Pathology at Massachusetts General Hospital and is Professor of Pathology at Harvard Medical School. He is a leading figure in the field of genome editing and has pioneered the development of designer nucleases and sensitive off-target detection methods.

Off-target genome editing refers to nonspecific and unintended genetic modifications that can arise through the use of engineered nuclease technologies such as: clustered, regularly interspaced, short palindromic repeats (CRISPR)-Cas9, transcription activator-like effector nucleases (TALEN), meganucleases, and zinc finger nucleases (ZFN). These tools use different mechanisms to bind a predetermined sequence of DNA (“target”), which they cleave, creating a double-stranded chromosomal break (DSB) that summons the cell's DNA repair mechanisms and leads to site-specific modifications. If these complexes do not bind at the target, often a result of homologous sequences and/or mismatch tolerance, they will cleave off-target DSB and cause non-specific genetic modifications. Specifically, off-target effects consist of unintended point mutations, deletions, insertions inversions, and translocations.

Since antiretroviral therapy requires a lifelong treatment regimen, research to find more permanent cures for HIV infection is currently underway. It is possible to synthesize zinc finger nucleotides with zinc finger components that selectively bind to specific portions of DNA. Conceptually, targeting and editing could focus on host cellular co-receptors for HIV or on proviral HIV DNA.

References

  1. 1 2 Boch J (February 2011). "TALEs of genome targeting". Nature Biotechnology. 29 (2): 135–6. doi:10.1038/nbt.1767. PMID   21301438. S2CID   304571.
  2. Boch J, Bonas U (September 2010). "Xanthomonas AvrBs3 family-type III effectors: discovery and function". Annual Review of Phytopathology. 48: 419–36. doi:10.1146/annurev-phyto-080508-081936. PMID   19400638.
  3. Boch J, Scholze H, Schornack S, Landgraf A, Hahn S, Kay S, Lahaye T, Nickstadt A, Bonas U (December 2009). "Breaking the code of DNA binding specificity of TAL-type III effectors". Science. 326 (5959): 1509–12. Bibcode:2009Sci...326.1509B. doi:10.1126/science.1178811. PMID   19933107. S2CID   206522347.
  4. Moscou MJ, Bogdanove AJ (December 2009). "A simple cipher governs DNA recognition by TAL effectors". Science. 326 (5959): 1501. Bibcode:2009Sci...326.1501M. doi:10.1126/science.1178817. PMID   19933106. S2CID   6648530.
  5. Juillerat A, Pessereau C, Dubois G, Guyot V, Maréchal A, Valton J, Daboussi F, Poirot L, Duclert A, Duchateau P (January 2015). "Optimized tuning of TALEN specificity using non-conventional RVDs". Scientific Reports. 5: 8150. Bibcode:2015NatSR...5E8150J. doi:10.1038/srep08150. PMC   4311247 . PMID   25632877.
  6. Christian M, Cermak T, Doyle EL, Schmidt C, Zhang F, Hummel A, Bogdanove AJ, Voytas DF (October 2010). "Targeting DNA double-strand breaks with TAL effector nucleases". Genetics. 186 (2): 757–61. doi:10.1534/genetics.110.120717. PMC   2942870 . PMID   20660643.
  7. Li T, Huang S, Jiang WZ, Wright D, Spalding MH, Weeks DP, Yang B (January 2011). "TAL nucleases (TALNs): hybrid proteins composed of TAL effectors and FokI DNA-cleavage domain". Nucleic Acids Research. 39 (1): 359–72. doi:10.1093/nar/gkq704. PMC   3017587 . PMID   20699274.
  8. Mahfouz MM, Li L, Shamimuzzaman M, Wibowo A, Fang X, Zhu JK (February 2011). "De novo-engineered transcription activator-like effector (TALE) hybrid nuclease with novel DNA binding specificity creates double-strand breaks". Proceedings of the National Academy of Sciences of the United States of America. 108 (6): 2623–8. Bibcode:2011PNAS..108.2623M. doi: 10.1073/pnas.1019533108 . PMC   3038751 . PMID   21262818.
  9. 1 2 3 Cermak T, Doyle EL, Christian M, Wang L, Zhang Y, Schmidt C, Baller JA, Somia NV, Bogdanove AJ, Voytas DF (July 2011). "Efficient design and assembly of custom TALEN and other TAL effector-based constructs for DNA targeting". Nucleic Acids Research. 39 (12): e82. doi:10.1093/nar/gkr218. PMC   3130291 . PMID   21493687.
  10. 1 2 3 4 5 Miller JC, Tan S, Qiao G, Barlow KA, Wang J, Xia DF, Meng X, Paschon DE, Leung E, Hinkley SJ, Dulay GP, Hua KL, Ankoudinova I, Cost GJ, Urnov FD, Zhang HS, Holmes MC, Zhang L, Gregory PD, Rebar EJ (February 2011). "A TALE nuclease architecture for efficient genome editing". Nature Biotechnology. 29 (2): 143–8. doi:10.1038/nbt.1755. PMID   21179091. S2CID   53549397.
  11. 1 2 3 Hockemeyer D, Wang H, Kiani S, Lai CS, Gao Q, Cassady JP, Cost GJ, Zhang L, Santiago Y, Miller JC, Zeitler B, Cherone JM, Meng X, Hinkley SJ, Rebar EJ, Gregory PD, Urnov FD, Jaenisch R (July 2011). "Genetic engineering of human pluripotent cells using TALE nucleases". Nature Biotechnology. 29 (8): 731–4. doi:10.1038/nbt.1927. PMC   3152587 . PMID   21738127.
  12. 1 2 Wood AJ, Lo TW, Zeitler B, Pickle CS, Ralston EJ, Lee AH, Amora R, Miller JC, Leung E, Meng X, Zhang L, Rebar EJ, Gregory PD, Urnov FD, Meyer BJ (July 2011). "Targeted genome editing across species using ZFNs and TALENs". Science. 333 (6040): 307. Bibcode:2011Sci...333..307W. doi:10.1126/science.1207773. PMC   3489282 . PMID   21700836.
  13. 1 2 Tesson L, Usal C, Ménoret S, Leung E, Niles BJ, Remy S, Santiago Y, Vincent AI, Meng X, Zhang L, Gregory PD, Anegon I, Cost GJ (August 2011). "Knockout rats generated by embryo microinjection of TALENs". Nature Biotechnology. 29 (8): 695–6. doi: 10.1038/nbt.1940 . PMID   21822240.
  14. 1 2 Huang P, Xiao A, Zhou M, Zhu Z, Lin S, Zhang B (August 2011). "Heritable gene targeting in zebrafish using customized TALENs". Nature Biotechnology. 29 (8): 699–700. doi:10.1038/nbt.1939. PMID   21822242. S2CID   28802632.
  15. Doyon Y, Vo TD, Mendel MC, Greenberg SG, Wang J, Xia DF, Miller JC, Urnov FD, Gregory PD, Holmes MC (January 2011). "Enhancing zinc-finger-nuclease activity with improved obligate heterodimeric architectures". Nature Methods. 8 (1): 74–9. doi:10.1038/nmeth.1539. PMID   21131970. S2CID   14334237.
  16. Szczepek M, Brondani V, Büchel J, Serrano L, Segal DJ, Cathomen T (July 2007). "Structure-based redesign of the dimerization interface reduces the toxicity of zinc-finger nucleases" (PDF). Nature Biotechnology. 25 (7): 786–93. doi:10.1038/nbt1317. PMID   17603476. S2CID   22079561.
  17. Guo J, Gaj T, Barbas CF (July 2010). "Directed evolution of an enhanced and highly efficient FokI cleavage domain for zinc finger nucleases". Journal of Molecular Biology. 400 (1): 96–107. doi:10.1016/j.jmb.2010.04.060. PMC   2885538 . PMID   20447404.
  18. 1 2 Mussolino C, Morbitzer R, Lütge F, Dannemann N, Lahaye T, Cathomen T (November 2011). "A novel TALE nuclease scaffold enables high genome editing activity in combination with low toxicity". Nucleic Acids Research. 39 (21): 9283–93. doi:10.1093/nar/gkr597. PMC   3241638 . PMID   21813459.
  19. 1 2 Zhang F, Cong L, Lodato S, Kosuri S, Church GM, Arlotta P (February 2011). "Efficient construction of sequence-specific TAL effectors for modulating mammalian transcription". Nature Biotechnology. 29 (2): 149–53. doi:10.1038/nbt.1775. PMC   3084533 . PMID   21248753.
  20. Hoover D (2012). "Using DNAWorks in Designing Oligonucleotides for PCR-Based Gene Synthesis". Gene Synthesis. Methods in Molecular Biology. Vol. 852. pp. 215–23. doi:10.1007/978-1-61779-564-0_16. ISBN   978-1-61779-563-3. PMID   22328436.
  21. Morbitzer R, Elsaesser J, Hausner J, Lahaye T (July 2011). "Assembly of custom TALE-type DNA binding domains by modular cloning". Nucleic Acids Research. 39 (13): 5790–9. doi:10.1093/nar/gkr151. PMC   3141260 . PMID   21421566.
  22. Li T, Huang S, Zhao X, Wright DA, Carpenter S, Spalding MH, Weeks DP, Yang B (August 2011). "Modularly assembled designer TAL effector nucleases for targeted gene knockout and gene replacement in eukaryotes". Nucleic Acids Research. 39 (14): 6315–25. doi:10.1093/nar/gkr188. PMC   3152341 . PMID   21459844.
  23. Geissler R, Scholze H, Hahn S, Streubel J, Bonas U, Behrens SE, Boch J (2011). Shiu S (ed.). "Transcriptional activators of human genes with programmable DNA-specificity". PLOS ONE. 6 (5): e19509. Bibcode:2011PLoSO...619509G. doi: 10.1371/journal.pone.0019509 . PMC   3098229 . PMID   21625585.
  24. Weber E, Gruetzner R, Werner S, Engler C, Marillonnet S (2011). Bendahmane M (ed.). "Assembly of designer TAL effectors by Golden Gate cloning". PLOS ONE. 6 (5): e19722. Bibcode:2011PLoSO...619722W. doi: 10.1371/journal.pone.0019722 . PMC   3098256 . PMID   21625552.
  25. Zhang Y, Zhang F, Li X, Baller JA, Qi Y, Starker CG, Bogdanove AJ, Voytas DF (January 2013). "Transcription activator-like effector nucleases enable efficient plant genome engineering". Plant Physiology. 161 (1): 20–7. doi:10.1104/pp.112.205179. PMC   3532252 . PMID   23124327.
  26. Haun W, Coffman A, Clasen BM, Demorest ZL, Lowy A, Ray E, Retterath A, Stoddard T, Juillerat A, Cedrone F, Mathis L, Voytas DF, Zhang F (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.
  27. Daboussi F, Leduc S, Maréchal A, Dubois G, Guyot V, Perez-Michaut C, Amato A, Falciatore A, Juillerat A, Beurdeley M, Voytas DF, Cavarec L, Duchateau P (May 2014). "Genome engineering empowers the diatom Phaeodactylum tricornutum for biotechnology". Nature Communications. 5: 3831. Bibcode:2014NatCo...5.3831D. doi: 10.1038/ncomms4831 . PMID   24871200.
  28. 1 2 Wienert B, Funnell AP, Norton LJ, Pearson RC, Wilkinson-White LE, Lester K, Vadolas J, Porteus MH, Matthews JM, Quinlan KG, Crossley M (2015). "Editing the genome to introduce a beneficial naturally occurring mutation associated with increased fetal globin". Nature Communications. 6: 7085. Bibcode:2015NatCo...6.7085W. doi: 10.1038/ncomms8085 . PMID   25971621.
  29. Davies B, Davies G, Preece C, Puliyadi R, Szumska D, Bhattacharya S (2013). "Site specific mutation of the Zic2 locus by microinjection of TALEN mRNA in mouse CD1, C3H and C57BL/6J oocytes". PLOS ONE. 8 (3): e60216. Bibcode:2013PLoSO...860216D. doi: 10.1371/journal.pone.0060216 . PMC   3610929 . PMID   23555929.
  30. Sander JD, Cade L, Khayter C, Reyon D, Peterson RT, Joung JK, Yeh JR (August 2011). "Targeted gene disruption in somatic zebrafish cells using engineered TALENs". Nature Biotechnology. 29 (8): 697–8. doi:10.1038/nbt.1934. PMC   3154023 . PMID   21822241.
  31. Wu H, Wang Y, Zhang Y, Yang M, Lv J, Liu J, Zhang Y (March 2015). "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. 112 (13): E1530-9. Bibcode:2015PNAS..112E1530W. doi: 10.1073/pnas.1421587112 . PMC   4386332 . PMID   25733846.
  32. Ponce de León V, Mérillat AM, Tesson L, Anegón I, Hummler E (2014). "Generation of TALEN-mediated GRdim knock-in rats by homologous recombination". PLOS ONE. 9 (2): e88146. Bibcode:2014PLoSO...988146P. doi: 10.1371/journal.pone.0088146 . PMC   3921256 . PMID   24523878.
  33. 1 2 3 Carlson DF, Fahrenkrug SC, Hackett PB (January 2012). "Targeting DNA With Fingers and TALENs". Molecular Therapy: Nucleic Acids. 1 (3): e3. doi:10.1038/mtna.2011.5. PMC   3381595 . PMID   23344620.
  34. Ramalingam S, Annaluru N, Kandavelou K, Chandrasegaran S (2014). "TALEN-mediated generation and genetic correction of disease-specific human induced pluripotent stem cells". Current Gene Therapy. 14 (6): 461–72. doi:10.2174/1566523214666140918101725. PMID   25245091.
  35. Dupuy A, Valton J, Leduc S, Armier J, Galetto R, Gouble A, Lebuhotel C, Stary A, Pâques F, Duchateau P, Sarasin A, Daboussi F (2013). "Targeted gene therapy of xeroderma pigmentosum cells using meganuclease and TALEN™". PLOS ONE. 8 (11): e78678. Bibcode:2013PLoSO...878678D. doi: 10.1371/journal.pone.0078678 . PMC   3827243 . PMID   24236034.
  36. Osborn MJ, Starker CG, McElroy AN, Webber BR, Riddle MJ, Xia L, DeFeo AP, Gabriel R, Schmidt M, von Kalle C, Carlson DF, Maeder ML, Joung JK, Wagner JE, Voytas DF, Blazar BR, Tolar J (June 2013). "TALEN-based gene correction for epidermolysis bullosa". Molecular Therapy. 21 (6): 1151–9. doi:10.1038/mt.2013.56. PMC   3677309 . PMID   23546300.
  37. Valton J, Guyot V, Marechal A, Filhol JM, Juillerat A, Duclert A, Duchateau P, Poirot L (September 2015). "A Multidrug-resistant Engineered CAR T Cell for Allogeneic Combination Immunotherapy". Molecular Therapy. 23 (9): 1507–18. doi:10.1038/mt.2015.104. PMC   4817890 . PMID   26061646.
  38. Poirot L, Philip B, Schiffer-Mannioui C, Le Clerre D, Chion-Sotinel I, Derniame S, Potrel P, Bas C, Lemaire L, Galetto R, Lebuhotel C, Eyquem J, Cheung GW, Duclert A, Gouble A, Arnould S, Peggs K, Pule M, Scharenberg AM, Smith J (September 2015). "Multiplex Genome-Edited T-cell Manufacturing Platform for "Off-the-Shelf" Adoptive T-cell Immunotherapies". Cancer Research. 75 (18): 3853–64. doi: 10.1158/0008-5472.CAN-14-3321 . PMID   26183927.
  39. Boissel S, Jarjour J, Astrakhan A, Adey A, Gouble A, Duchateau P, Shendure J, Stoddard BL, Certo MT, Baker D, Scharenberg AM (February 2014). "megaTALs: a rare-cleaving nuclease architecture for therapeutic genome engineering". Nucleic Acids Research. 42 (4): 2591–601. doi:10.1093/nar/gkt1224. PMC   3936731 . PMID   24285304.
  40. "Pros and Cons Of ZFNS, TALENS, AND CRISPR/CAS". The Jackson Laboratory. March 2014.
  41. Boglioli, Elsy; Richard, Magali. "Boston Consulting Group - Report on Gene Editing Precision" (PDF).