Generic placeholder image

Recent Patents on Anti-Cancer Drug Discovery

Editor-in-Chief

ISSN (Print): 1574-8928
ISSN (Online): 2212-3970

Mini-Review Article

CRISPER/Cas in Plant Natural Product Research: Therapeutics as Anticancer and other Drug Candidates and Recent Patents

Author(s): Abhijit Dey* and Samapika Nandy

Volume 16, Issue 4, 2021

Published on: 06 July, 2021

Page: [460 - 468] Pages: 9

DOI: 10.2174/1574892816666210706155602

Price: $65

Abstract

Background: Clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR- associated9 (Cas9) endonuclease system is a facile, highly efficient and selective site-directed mutagenesis tool for RNA-guided genome-editing. CRISPR/Cas9 genome-editing strategy uses designed guide-RNAs that recognizes a 3 base-pair protospacer adjacent motif (PAM) sequence in the target-DNA. CRISPR/Cas-editing tools have mainly been employed in crop plants in relation to yield and stress tolerance. However, the immense potential of this technology has not yet been fully utilized in medicinal plants in deciphering or modulating secondary metabolic pathways producing therapeutically active phytochemicals against cancer and other diseases.

Objective: The present review elucidates the use of CRISPR-Cas9 as a promising genome-editing tool in plants and plant-derived natural products with anticancer and other therapeutic applications. It also includes recent patents on the therapeutic applications of CRISPR-CAS systems implicated to cancer and other human medical conditions.

Methods: Popular search engines, such as PubMed, Scopus, Google Scholar, Google Patents, Medline, ScienceDirect, SpringerLink, EMBASE, Mendeley, etc., were searched in order to retrieve literature using relevant keywords viz. CRISPER/Cas, plant natural product research, anticancer, therapeutics, etc., either singly or in various combinations.

Results: Retrieved citations and further cross-referencing among the literature have resulted in a total number of 71 publications and 3 patents are being cited in this work. Information presented in this review aims to support further biotechnological and clinical strategies to be carried using CRISPER/ Cas mediated optimization of plant natural products against cancer and an array of other human medical conditions.

Conclusion: Off late, knock-in and knock-out, point mutation, controlled tuning of gene-expression and targeted mutagenesis have enabled the versatile CRISPR/Cas-editing device to engineer medicinal plants’ genomes. In addition, by combining CRISPR/Cas-editing tool with next-generation sequencing (NGS) and various tools of system biology, many medicinal plants have been engineered genetically to optimize the production of valuable bioactive compounds of industrial significance.

Keywords: CRISPR/Cas, medicinal plants, natural products, next-generation sequencing, system biology, CRISPR-Cas9.

[1]
Cubbon A, Ivancic-Bace I, Bolt EL. CRISPR-CAS immunity, DNA repair and genome stability. Biosc Rep 2018; 38(5): BSR20180457.
[2]
Chen K, Wang Y, Zhang R, Zhang H, Gao C. CRISPR/Cas genome editing and precision plant breeding in agriculture. Annu Rev Plant Biol 2019; 70: 667-97.
[http://dx.doi.org/10.1146/annurev-arplant-050718-100049] [PMID: 30835493]
[3]
Gaj T, Gersbach CA, Barbas CF III. ZFN, TALEN, and CRISPR/Cas-based methods for genome engineering. Trends Biotechnol 2013; 31(7): 397-405.
[http://dx.doi.org/10.1016/j.tibtech.2013.04.004] [PMID: 23664777]
[4]
Wang Y, Li Z, Xu J, et al. The CRISPR/Cas system mediates efficient genome engineering in Bombyx mori. Cell Res 2013; 23(12): 1414-6.
[http://dx.doi.org/10.1038/cr.2013.146] [PMID: 24165890]
[5]
Leenay RT, Maksimchuk KR, Slotkowski RA, et al. Identifying and visualizing functional PAM diversity across CRISPR-CAS systems. Mol Cell 2016; 62(1): 137-47.
[http://dx.doi.org/10.1016/j.molcel.2016.02.031] [PMID: 27041224]
[6]
Dey A, De JN. Ethnobotanical survey of Purulia district, West Bengal, India for medicinal plants used against gastrointestinal disorders. J Ethnopharmacol 2012; 143(1): 68-80.
[http://dx.doi.org/10.1016/j.jep.2012.05.064] [PMID: 22721882]
[7]
Dey A, Bhattacharya R, Mukherjee A, Pandey DK. Natural products against Alzheimer’s disease: Pharmaco-therapeutics and biotechnological interventions. Biotechnol Adv 2017; 35(2): 178-216.
[http://dx.doi.org/10.1016/j.biotechadv.2016.12.005] [PMID: 28043897]
[8]
Anand U, Nandy S, Mundhra A, Das N, Pandey DK, Dey A. A review on antimicrobial botanicals, phytochemicals and natural resistance modifying agents from Apocynaceae family: Possible therapeutic approaches against multidrug resistance in pathogenic microorganisms. Drug Resist Updat 2020; 51: 100695-5.
[http://dx.doi.org/10.1016/j.drup.2020.100695] [PMID: 32442892]
[9]
Gupta AK, Gupta UD. Next generation sequencing and its applications. In: Animal Biotechnology Cambridge: Academic Press. 2014; pp. 345-67.
[http://dx.doi.org/10.1016/B978-0-12-416002-6.00019-5]
[10]
Niazian M. Application of genetics and biotechnology for improving medicinal plants. Planta 2019; 249(4): 953-73.
[http://dx.doi.org/10.1007/s00425-019-03099-1] [PMID: 30715560]
[11]
Belhaj K, Chaparro-Garcia A, Kamoun S, Patron NJ, Nekrasov V. Editing plant genomes with CRISPR/Cas9. Curr Opin Biotechnol 2015; 32: 76-84.
[http://dx.doi.org/10.1016/j.copbio.2014.11.007] [PMID: 25437637]
[12]
Feng Z, Zhang B, Ding W, et al. Efficient genome editing in plants using a CRISPR/Cas system. Cell Res 2013; 23(10): 1229-32.
[http://dx.doi.org/10.1038/cr.2013.114] [PMID: 23958582]
[13]
Jacobs TB, LaFayette PR, Schmitz RJ, Parrott WA. Targeted genome modifications in soybean with CRISPR/Cas9. BMC Biotechnol 2015; 15: 16.
[http://dx.doi.org/10.1186/s12896-015-0131-2] [PMID: 25879861]
[14]
Jiang W, Zhou H, Bi H, Fromm M, Yang B, Weeks DP. Demonstration of CRISPR/Cas9/sgRNA-mediated targeted gene modification in Arabidopsis, tobacco, sorghum and rice. Nucleic Acids Res 2013; 41(20): e188-8.
[http://dx.doi.org/10.1093/nar/gkt780] [PMID: 23999092]
[15]
Lawrenson T, Shorinola O, Stacey N, et al. Induction of targeted, heritable mutations in barley and Brassica oleracea using RNA-guided Cas9 nuclease. Genome Biol 2015; 16: 258.
[http://dx.doi.org/10.1186/s13059-015-0826-7] [PMID: 26616834]
[16]
Nekrasov V, Staskawicz B, Weigel D, Jones JD, Kamoun S. Targeted mutagenesis in the model plant Nicotiana benthamiana using Cas9 RNA-guided endonuclease. Nat Biotechnol 2013; 31(8): 691-3.
[http://dx.doi.org/10.1038/nbt.2655] [PMID: 23929340]
[17]
D’Ambrosio C, Stigliani AL, Giorio G. CRISPR/Cas9 editing of carotenoid genes in tomato. Transgenic Res 2018; 27(4): 367-78.
[http://dx.doi.org/10.1007/s11248-018-0079-9] [PMID: 29797189]
[18]
Li R, Li R, Li X, et al. Multiplexed CRISPR/Cas9-mediated metabolic engineering of γ-aminobutyric acid levels in Solanum lycopersicum. Plant Biotechnol J 2018; 16(2): 415-27.
[http://dx.doi.org/10.1111/pbi.12781] [PMID: 28640983]
[19]
Zhang P, Du H, Wang J, et al. Multiplex CRISPR/Cas9-mediated metabolic engineering increases soya bean isoflavone content and resistance to soya bean mosaic virus. Plant Biotechnol J 2020; 18(6): 1384-95.
[http://dx.doi.org/10.1111/pbi.13302] [PMID: 31769589]
[20]
Berti M, Gesch R, Eynck C, Anderson J, Cermak S. Camelina uses, genetics, genomics, production, and management. Ind Crops Prod 2016; 94: 690-710.
[http://dx.doi.org/10.1016/j.indcrop.2016.09.034]
[21]
Dharavath RN, Singh S, Chaturvedi S, Luqman S. Camelina sativa (L.) Crantz A mercantile crop with speckled pharmacological activities. Ann Phytomed 2016; 5: 6-26.
[http://dx.doi.org/10.21276/ap.2016.5.2.2]
[22]
Sainger M, Jaiwal A, Sainger PA, Chaudhary D, Jaiwal R, Jaiwal PK. Advances in genetic improvement of Camelina sativa for biofuel and industrial bio-products. Renew Sustain Energy Rev 2017; 68: 623-37.
[http://dx.doi.org/10.1016/j.rser.2016.10.023]
[23]
Jiang WZ, Henry IM, Lynagh PG, Comai L, Cahoon EB, Weeks DP. Significant enhancement of fatty acid composition in seeds of the allohexaploid, Camelina sativa, using CRISPR/Cas9 gene editing. Plant Biotechnol J 2017; 15(5): 648-57.
[http://dx.doi.org/10.1111/pbi.12663] [PMID: 27862889]
[24]
Morineau C, Bellec Y, Tellier F, et al. Selective gene dosage by CRISPR-CAS9 genome editing in hexaploid Camelina sativa. Plant Biotechnol J 2017; 15(6): 729-39.
[http://dx.doi.org/10.1111/pbi.12671] [PMID: 27885771]
[25]
Aznar-Moreno JA, Durrett TP. Simultaneous targeting of multiple gene homeologs to alter seed oil production in Camelina sativa. Plant Cell Physiol 2017; 58(7): 1260-7.
[http://dx.doi.org/10.1093/pcp/pcx058] [PMID: 28444368]
[26]
Ozseyhan ME, Kang J, Mu X, Lu C. Mutagenesis of the FAE1 genes significantly changes fatty acid composition in seeds of Camelina sativa. Plant Physiol Biochem 2018; 123: 1-7.
[http://dx.doi.org/10.1016/j.plaphy.2017.11.021] [PMID: 29216494]
[27]
Waltz E. With a free pass, CRISPR-edited plants reach market in record time. Nat Biotechnol 2018; 36(1): 6-7.
[http://dx.doi.org/10.1038/nbt0118-6b] [PMID: 29319694]
[28]
Guo L, Qi J, Du D, Liu Y, Jiang X. Current advances of Dendrobium officinale polysaccharides in dermatology: A literature review. Pharm Biol 2020; 58(1): 664-73.
[http://dx.doi.org/10.1080/13880209.2020.1787470] [PMID: 32657196]
[29]
Kui L, Chen H, Zhang W, et al. Building a genetic manipulation tool box for orchid biology: Identification of constitutive promoters and application of CRISPR/Cas9 in the orchid, Dendrobium officinale. Front Plant Sci 2017; 7: 2036.
[http://dx.doi.org/10.3389/fpls.2016.02036] [PMID: 28127299]
[30]
Song W, Yan S, Li Y, Feng S, Zhang JJ, Li JR. Functional characterization of squalene epoxidase and NADPH-cytochrome P450 reductase in Dioscorea zingiberensis. Biochem Biophys Res Commun 2019; 509(3): 822-7.
[http://dx.doi.org/10.1016/j.bbrc.2019.01.010] [PMID: 30638657]
[31]
Zhou J, Xi Y, Zhang J, et al. Protective effect of Dioscorea zingiberensis ethanol extract on the disruption of blood-testes barrier in high-fat diet/streptozotocin-induced diabetic mice by upregulating ZO-1 and Nrf2. Andrologia 2020; 52(3): e13508.
[http://dx.doi.org/10.1111/and.13508] [PMID: 31957918]
[32]
Zhang X, Jin M, Tadesse N, et al. Dioscorea zingiberensis C. H. Wright: An overview on its traditional use, phytochemistry, pharmacology, clinical applications, quality control, and toxicity. J Ethnopharmacol 2018; 220: 283-93.
[http://dx.doi.org/10.1016/j.jep.2018.03.017] [PMID: 29602601]
[33]
Feng S, Song W, Fu R, Zhang H, Xu A, Li J. Application of the CRISPR/Cas9 system in Dioscorea zingiberensis. Plant Cell Tissue Organ Cult 2018; 135: 133-41.
[http://dx.doi.org/10.1007/s11240-018-1450-5]
[34]
Dewey RE, Xie J. Molecular genetics of alkaloid biosynthesis in Nicotiana tabacum. Phytochemistry 2013; 94: 10-27.
[http://dx.doi.org/10.1016/j.phytochem.2013.06.002] [PMID: 23953973]
[35]
Jutras PV, Dodds I, van der Hoorn RA. Proteases of Nicotiana benthamiana: An emerging battle for molecular farming. Curr Opin Biotechnol 2020; 61: 60-5.
[http://dx.doi.org/10.1016/j.copbio.2019.10.006] [PMID: 31765962]
[36]
Jansing J, Sack M, Augustine SM, Fischer R, Bortesi L. CRISPR/Cas9-mediated knockout of six glycosyltransferase genes in Nicotiana benthamiana for the production of recombinant proteins lacking β-1,2-xylose and core α-1,3-fucose. Plant Biotechnol J 2019; 17(2): 350-61.
[http://dx.doi.org/10.1111/pbi.12981] [PMID: 29969180]
[37]
Gao J, Wang G, Ma S, et al. CRISPR/Cas9-mediated targeted mutagenesis in Nicotiana tabacum. Plant Mol Biol 2015; 87(1-2): 99-110.
[http://dx.doi.org/10.1007/s11103-014-0263-0] [PMID: 25344637]
[38]
Schachtsiek J, Stehle F. Nicotine-free, nontransgenic tobacco (Nicotiana tabacum L.) edited by CRISPR-CAS9. Plant Biotechnol J 2019; 17: p. 2228.
[39]
Mercx S, Smargiasso N, Chaumont F, De Pauw E, Boutry M, Navarre C. Inactivation of the β(1,2)-xylosyltransferase and the α(1,3)-fucosyltransferase genes in Nicotiana tabacum BY-2 cells by a multiplex CRISPR/Cas9 strategy results in glycoproteins without plant-specific glycans. Front Plant Sci 2017; 8: 403.
[http://dx.doi.org/10.3389/fpls.2017.00403] [PMID: 28396675]
[40]
Mercx S, Tollet J, Magy B, Navarre C, Boutry M. Gene inactivation by CRISPR-CAS9 in Nicotiana tabacum BY-2 suspension cells. Front Plant Sci 2016; 7: 40.
[http://dx.doi.org/10.3389/fpls.2016.00040] [PMID: 26870061]
[41]
Raut RA, Ghotankar AM. Review of Khuskhus (Khaskhas) (Seeds of Papaver somniferum Linn.) with special reference of Ayurveda Medicine. J Ayurveda Integr Med Sci 2020; 4: 228-31.
[42]
Labanca F, Ovesna J, Milella L. Papaver somniferum L. taxonomy, uses and new insight in poppy alkaloid pathways. Phytochem Rev 2018; 17: 853-71.
[http://dx.doi.org/10.1007/s11101-018-9563-3]
[43]
Alagoz Y, Gurkok T, Zhang B, Unver T. Manipulating the biosynthesis of bioactive compound alkaloids for next-generation metabolic engineering in opium poppy using CRISPR-CAS 9 genome editing technology. Sci Rep 2016; 6: 30910.
[http://dx.doi.org/10.1038/srep30910] [PMID: 27483984]
[44]
Ren J, Fu L, Nile SH, Zhang J, Kai G. Salvia miltiorrhiza in treating cardiovascular diseases: A review on its pharmacological and clinical applications. Front Pharmacol 2019; 10: 753.
[http://dx.doi.org/10.3389/fphar.2019.00753] [PMID: 31338034]
[45]
Li B, Cui G, Shen G, et al. Targeted mutagenesis in the medicinal plant Salvia miltiorrhiza. Sci Rep 2017; 7: 43320.
[http://dx.doi.org/10.1038/srep43320] [PMID: 28256553]
[46]
Zhou Z, Tan H, Li Q, et al. CRISPR/Cas9-mediated efficient targeted mutagenesis of RAS in Salvia miltiorrhiza. Phytochemistry 2018; 148: 63-70.
[http://dx.doi.org/10.1016/j.phytochem.2018.01.015] [PMID: 29421512]
[47]
Kishi-Kaboshi M, Aida R, Sasaki K. Generation of gene-edited Chrysanthemum morifolium using multicopy transgenes as targets and markers. Plant Cell Physiol 2017; 58(2): 216-26.
[PMID: 28049122]
[48]
Karkute SG, Singh AK, Gupta OP, Singh PM, Singh B. CRISPR/Cas9 mediated genome engineering for improvement of horticultural crops. Front Plant Sci 2017; 8: 1635.
[http://dx.doi.org/10.3389/fpls.2017.01635] [PMID: 28970844]
[49]
Iaffaldano B, Zhang Y, Cornish K. CRISPR/Cas9 genome editing of rubber producing dandelion Taraxacum kok-saghyz using Agrobacterium rhizogenes without selection. Ind Crops Prod 2016; 89: 356-62.
[http://dx.doi.org/10.1016/j.indcrop.2016.05.029]
[50]
Demirci Y, Zhang B, Unver T. CRISPR/Cas9: An RNA-guided highly precise synthetic tool for plant genome editing. J Cell Physiol 2018; 233(3): 1844-59.
[http://dx.doi.org/10.1002/jcp.25970] [PMID: 28430356]
[51]
Kimberland ML, Hou W, Alfonso-Pecchio A, et al. Strategies for controlling CRISPR/Cas9 off-target effects and biological variations in mammalian genome editing experiments. J Biotechnol 2018; 284: 91-101.
[http://dx.doi.org/10.1016/j.jbiotec.2018.08.007] [PMID: 30142414]
[52]
Shen B, Zhang W, Zhang J, et al. Efficient genome modification by CRISPR-CAS9 nickase with minimal off-target effects. Nat Methods 2014; 11(4): 399-402.
[http://dx.doi.org/10.1038/nmeth.2857] [PMID: 24584192]
[53]
Rees HA, Komor AC, Yeh WH, et al. Improving the DNA specificity and applicability of base editing through protein engineering and protein delivery. Nat Commun 2017; 8: 15790.
[http://dx.doi.org/10.1038/ncomms15790] [PMID: 28585549]
[54]
He Y, Su J, Lan B, Gao Y, Zhao J. Targeting off-target effects: Endoplasmic reticulum stress and autophagy as effective strategies to enhance temozolomide treatment. OncoTargets Ther 2019; 12: 1857-65.
[http://dx.doi.org/10.2147/OTT.S194770] [PMID: 30881038]
[55]
Morgens DW, Wainberg M, Boyle EA, et al. Genome-scale measurement of off-target activity using Cas9 toxicity in high-throughput screens. Nat Commun 2017; 8: 15178.
[http://dx.doi.org/10.1038/ncomms15178] [PMID: 28474669]
[56]
Yang S, Li S, Li XJ. Shortening the half-life of Cas9 maintains its gene editing ability and reduces neuronal toxicity. Cell Rep 2018; 25(10): 2653-2659.e3.
[http://dx.doi.org/10.1016/j.celrep.2018.11.019] [PMID: 30517854]
[57]
Markus BM, Bell GW, Lorenzi HA, Lourido S. Optimizing systems for Cas9 expression in Toxoplasma gondii. MSphere 2019; 4(3): e00386-19.
[http://dx.doi.org/10.1128/mSphere.00386-19] [PMID: 31243081]
[58]
Nagaraju S, Davies NK, Walker DJF, Köpke M, Simpson SD. Genome editing of Clostridium autoethanogenum using CRISPR/Cas9. Biotechnol Biofuels 2016; 9: 219.
[http://dx.doi.org/10.1186/s13068-016-0638-3] [PMID: 27777621]
[59]
Jiang W, Brueggeman AJ, Horken KM, Plucinak TM, Weeks DP. Successful transient expression of Cas9 and single guide RNA genes in Chlamydomonas reinhardtii. Eukaryot Cell 2014; 13(11): 1465-9.
[http://dx.doi.org/10.1128/EC.00213-14] [PMID: 25239977]
[60]
Staals RH, Jackson SA, Biswas A, Brouns SJ, Brown CM, Fineran PC. Interference-driven spacer acquisition is dominant over naive and primed adaptation in a native CRISPR-CAS system. Nat Commun 2016; 7: 12853.
[http://dx.doi.org/10.1038/ncomms12853] [PMID: 27694798]
[61]
Cañez C, Selle K, Goh YJ, Barrangou R. Outcomes and characterization of chromosomal self-targeting by native CRISPR-CAS systems in Streptococcus thermophilus. FEMS Microbiol Lett 2019; 366(9): fnz105.
[http://dx.doi.org/10.1093/femsle/fnz105] [PMID: 31077282]
[62]
Tong Y, Weber T, Lee SY. CRISPR/Cas-based genome engineering in natural product discovery. Nat Prod Rep 2019; 36(9): 1262-80.
[http://dx.doi.org/10.1039/C8NP00089A] [PMID: 30548045]
[63]
Eid A, Alshareef S, Mahfouz MM. CRISPR base editors: Genome editing without double-stranded breaks. Biochem J 2018; 475: 1955-64.
[64]
Tong Y, Whitford CM, Blin K, Jørgensen TS, Weber T, Lee SY. CRISPR-CAS9, CRISPRi and CRISPR-BEST-mediated genetic manipulation in streptomycetes. Nat Protoc 2020; 15(8): 2470-502.
[http://dx.doi.org/10.1038/s41596-020-0339-z] [PMID: 32651565]
[65]
Gupta S, Khanna VK, Singh R, Garg GK. Strategies for overcoming genotypic limitations of in vitro regeneration and determination of genetic components of variability of plant regeneration traits in sorghum. Plant Cell Tissue Organ Cult 2006; 86: 379-88.
[http://dx.doi.org/10.1007/s11240-006-9140-0]
[66]
Zhang Y, Zhang F, Li X, et al. Transcription activator-like effector nucleases enable efficient plant genome engineering. Plant Physiol 2013; 161(1): 20-7.
[http://dx.doi.org/10.1104/pp.112.205179] [PMID: 23124327]
[67]
Andersson M, Turesson H, Nicolia A, Fält AS, Samuelsson M, Hofvander P. Efficient targeted multiallelic mutagenesis in tetraploid potato (Solanum tuberosum) by transient CRISPR-CAS9 expression in protoplasts. Plant Cell Rep 2017; 36(1): 117-28.
[http://dx.doi.org/10.1007/s00299-016-2062-3] [PMID: 27699473]
[68]
Mout R, Ray M, Yesilbag Tonga G, et al. Direct cytosolic delivery of CRISPR/Cas9-ribonucleoprotein for efficient gene editing. ACS Nano 2017; 11(3): 2452-8.
[http://dx.doi.org/10.1021/acsnano.6b07600] [PMID: 28129503]
[69]
Zhang L, Wang P, Feng Q, et al. Lipid nanoparticle-mediated efficient delivery of CRISPR/Cas9 for tumor therapy. NPG Asia Mater 2017; 9: e441-1.
[http://dx.doi.org/10.1038/am.2017.185]
[70]
Lian J, Schultz C, Cao M, HamediRad M, Zhao H. Multi-functional genome-wide CRISPR system for high throughput genotype-phenotype mapping. Nat Commun 2019; 10(1): 5794.
[http://dx.doi.org/10.1038/s41467-019-13621-4] [PMID: 31857575]
[71]
Voytas DF. Conferring resistance to geminiviruses in plants using CRISPR/CAS systems. US20160237451, 2018.
[72]
Sharp PA. Delivery, use and therapeutic applications of the CRISPR-CAS systems and compositions for modeling competition of multiple cancer mutations in vivo. WO2016049024, 2016.
[73]
Tang H. CRISPR/Cas-mediated genome editing to treat EGFR-mutant lung cancer. US10240145, 2019.
[74]
Gelvin SB. Plant proteins involved in Agrobacterium-mediated genetic transformation. Annu Rev Phytopathol 2010; 48: 45-68.
[http://dx.doi.org/10.1146/annurev-phyto-080508-081852] [PMID: 20337518]
[75]
Kalbande BB, Patil AS. Plant tissue culture independent Agrobacterium tumefaciens mediated In-planta transformation strategy for upland cotton (Gossypium hirsutum). J Genet Eng Biotechnol 2016; 14(1): 9-18.
[http://dx.doi.org/10.1016/j.jgeb.2016.05.003] [PMID: 30647592]
[76]
Jiang Y, Chen B, Duan C, Sun B, Yang J, Yang S. Multigene editing in the Escherichia coli genome via the CRISPR-CAS9 system. Appl Environ Microbiol 2015; 81(7): 2506-14.
[http://dx.doi.org/10.1128/AEM.04023-14] [PMID: 25636838]
[77]
Bennett-Baker PE, Mueller JL. CRISPR-mediated isolation of specific megabase segments of genomic DNA. Nucleic Acids Res 2017; 45(19): e165-5.
[http://dx.doi.org/10.1093/nar/gkx749] [PMID: 28977642]
[78]
McInally SG, Hagen KD, Nosala C, et al. Robust and stable transcriptional repression in Giardia using CRISPRi. Mol Biol Cell 2019; 30(1): 119-30.
[http://dx.doi.org/10.1091/mbc.E18-09-0605] [PMID: 30379614]
[79]
Bester AC, Lee JD, Chavez A, et al. An integrated genome-wide CRISPRa approach to functionalize lncRNAs in drug resistance. Cell 2018; 173(3): 649-664.e20.
[http://dx.doi.org/10.1016/j.cell.2018.03.052] [PMID: 29677511]

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy