CRISPR-Cas and Its Wide-Ranging Applications: Comparison
Please note this is a comparison between Version 2 by Camila Xu and Version 1 by Simona Zaami.

The CRISPR-Cas system is a powerful tool for in vivo editing the genome of most organisms, including man.

  • CRISPR-Cas
  • germline genome editing
  • human embryo
  • bioethics
  • biosecurity
Please wait, diff process is still running!

References

  1. Ishino, Y.; Shinagawa, H.; Makino, K.; Amemura, M.; Nakatura, A. Nucleotide sequence of the iap gene, responsible for alkaline phosphatase isoenzyme conversion in Escherichia coli, and identification of the gene product. J. Bacteriol. 1987, 169, 5429–5433.
  2. Jinek, M.; Chylinski, K.; Fonfara, I.; Hauer, M.; Doudna, J.A.; Charpentier, E. A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity. Science 2012, 337, 816–821.
  3. Press Release: The Nobel Prize in Chemistry 2020. Available online: (accessed on 16 March 2021).
  4. Hynes, A.P.; Villion, M.; Moineau, S. Adaptation in bacterial CRISPR-Cas immunity can be driven by defective phages. Nat. Commun. 2014, 5, 1–6.
  5. Barrangou, R.; Marraffini, L.A. CRISPR-cas systems: Prokaryotes upgrade to adaptive immunity. Mol. Cell 2014, 54, 234–244.
  6. Westra, E.R.; Dowling, A.J.; Broniewski, J.M.; Van Houte, S. Evolution and Ecology of CRISPR. Annu. Rev. Ecol. Evol. Syst. 2016, 47, 307–331.
  7. Makarova, K.S.; Wolf, Y.I.; Iranzo, J.; Shmakov, S.A.; Alkhnbashi, O.S.; Brouns, S.J.J.; Charpentier, E.; Cheng, D.; Haft, D.H.; Horvath, P.; et al. Evolutionary classification of CRISPR–Cas systems: A burst of class 2 and derived variants. Nat. Rev. Microbiol. 2020, 18, 67–83.
  8. Cong, L.; Ran, F.A.; Cox, D.; Lin, S.; Barretto, R.; Habib, N.; Hsu, P.D.; Wu, X.; Jiang, W.; Marraffini, L.A.; et al. Multiplex Genome Engineering Using CRISPR/Cas Systems. Science 2013, 339, 819–823.
  9. Burgess, D.J. Technology: A CRISPR Genome-Editing Tool. Nat. Rev. Genet. 2013, 14, 80.
  10. Li, Q.; Sapkota, M.; van der Knaap, E. Perspectives of CRISPR/Cas-mediated cis-engineering in horticulture: Unlocking the neglected potential for crop improvement. Hortic. Res. 2020, 7, 36.
  11. Huang, X.; Hilscher, J.; Stoger, E.; Christou, P.; Zhu, C. Modification of cereal plant architecture by genome editing to improve yields. Plant Cell Rep. 2021, 1–26.
  12. Ku, H.-K.; Ha, S.-H. Improving Nutritional and Functional Quality by Genome Editing of Crops: Status and Perspectives. Front. Plant Sci. 2020, 11, 23.
  13. Arora, L.; Narula, A. Gene editing and crop improvement using CRISPR-cas9 system. Front. Plant Sci. 2017, 8, 1932.
  14. Martín-Pizarro, C.; Posé, D. Genome editing as a tool for fruit ripening manipulation. Front. Plant Sci. 2018, 9, 1415.
  15. Shipman, E.N.; Yu, J.; Zhou, J.; Albornoz, K.; Beckles, D.M. Can gene editing reduce postharvest waste and loss of fruit, vegetables, and ornamentals? Hortic. Res. 2021, 8, 1.
  16. Schenke, D.; Cai, D. Applications of CRISPR/Cas to Improve Crop Disease Resistance: Beyond Inactivation of Susceptibility Factors. iScience 2020, 23, 101478.
  17. Ahmad, S.; Wei, X.; Sheng, Z.; Hu, P.; Tang, S. CRISPR/Cas9 for development of disease resistance in plants: Recent progress, limitations and future prospects. Brief. Funct. Genom. 2020, 19, 26–39.
  18. Borrelli, V.M.G.; Brambilla, V.; Rogowsky, P.; Marocco, A.; Lanubile, A. The enhancement of plant disease resistance using crispr/cas9 technology. Front. Plant Sci. 2018, 9, 1245.
  19. Jaganathan, D.; Ramasamy, K.; Sellamuthu, G.; Jayabalan, S.; Venkataraman, G. CRISPR for crop improvement: An update review. Front. Plant Sci. 2018, 9, 985.
  20. Martignago, D.; Rico-Medina, A.; Blasco-Escámez, D.; Fontanet-Manzaneque, J.B.; Caño-Delgado, A.I. Drought Resistance by Engineering Plant Tissue-Specific Responses. Front. Plant Sci. 2020, 10, 1676.
  21. Zafar, S.A.; Zaidi, S.S.E.A.; Gaba, Y.; Singla-Pareek, S.L.; Dhankher, O.P.; Li, X.; Mansoor, S.; Pareek, A. Engineering abiotic stress tolerance via CRISPR/Cas-mediated genome editing. J. Exp. Bot. 2020, 71, 470–479.
  22. Wang, T.; Zhang, H.; Zhu, H. CRISPR technology is revolutionizing the improvement of tomato and other fruit crops. Hortic. Res. 2019, 6, 77.
  23. Corte, L.E.D.; Mahmoud, L.M.; Moraes, T.S.; Mou, Z.; Grosser, J.W.; Dutt, M. Development of improved fruit, vegetable, and ornamental crops using the CRISPR/cas9 genome editing technique. Plants 2019, 8, 601.
  24. Gao, Q.; Luo, H.; Li, Y.; Liu, Z.; Kang, C. Genetic modulation of RAP alters fruit coloration in both wild and cultivated strawberry. Plant Biotechnol. J. 2020, 18, 1550–1561.
  25. Naves, E.R.; de Ávila Silva, L.; Sulpice, R.; Araújo, W.L.; Nunes-Nesi, A.; Peres, L.E.P.; Zsögön, A. Capsaicinoids: Pungency beyond Capsicum. Trends Plant Sci. 2019, 24, 109–120.
  26. Sugano, S.; Hirose, A.; Kanazashi, Y.; Adachi, K.; Hibara, M.; Itoh, T.; Mikami, M.; Endo, M.; Hirose, S.; Maruyama, N.; et al. Simultaneous induction of mutant alleles of two allergenic genes in soybean by using site-directed mutagenesis. BMC Plant Biol. 2020, 20.
  27. Jouanin, A.; Gilissen, L.J.W.J.; Schaart, J.G.; Leigh, F.J.; Cockram, J.; Wallington, E.J.; Boyd, L.A.; van den Broeck, H.C.; van der Meer, I.M.; America, A.H.P.; et al. CRISPR/Cas9 Gene Editing of Gluten in Wheat to Reduce Gluten Content and Exposure—Reviewing Methods to Screen for Coeliac Safety. Front. Nutr. 2020, 7, 51.
  28. Sánchez-León, S.; Gil-Humanes, J.; Ozuna, C.V.; Giménez, M.J.; Sousa, C.; Voytas, D.F.; Barro, F. Low-gluten, nontransgenic wheat engineered with CRISPR/Cas9. Plant Biotechnol. J. 2018, 16, 902–910.
  29. Cowan, P.J.; Hawthorne, W.J.; Nottle, M.B. Xenogeneic transplantation and tolerance in the era of CRISPR-Cas9. Curr. Opin. Organ Transplant. 2019, 24, 5–11.
  30. Kararoudi, M.N.; Hejazi, S.S.; Elmas, E.; Hellström, M.; Kararoudi, M.N.; Padma, A.M.; Lee, D.; Dolatshad, H. Clustered regularly interspaced short palindromic repeats/Cas9 gene editing technique in xenotransplantation. Front. Immunol. 2018, 9, 1711.
  31. Menchaca, A.; dos Santos-Neto, P.C.; Mulet, A.P.; Crispo, M. CRISPR in livestock: From editing to printing. Theriogenology 2020, 150, 247–254.
  32. Khwatenge, C.N.; Nahashon, S.N. Recent Advances in the Application of CRISPR/Cas9 Gene Editing System in Poultry Species. Front. Genet. 2021, 12, 627714.
  33. Cyranoski, D. Gene-edited pigs to be sold as pets. Nature 2015, 526, 18.
  34. Zhou, W.; Wan, Y.; Guo, R.; Deng, M.; Deng, K.; Wang, Z.; Zhang, Y.; Wang, F. Generation of beta-lactoglobulin knock-out goats using CRISPR/Cas9. PLoS ONE 2017, 12, e0186056.
  35. Mukae, T.; Yoshii, K.; Watanobe, T.; Tagami, T.; Oishi, I. Production and characterization of eggs from hens with ovomucoid gene mutation. Poult. Sci. 2021, 100, 452–460.
  36. Torres-Ruiz, R.; Rodriguez-Perales, S. CRISPR-Cas9 technology: Applications and human disease modelling. Brief. Funct. Genom. 2017, 16, 4–12.
  37. Alagoz, M.; Kherad, N. Advance genome editing technologies in the treatment of human diseases: CRISPR therapy (Review). Int. J. Mol. Med. 2020, 46, 521–534.
  38. Van Giau, V.; Lee, H.; Shim, K.H.; Bagyinszky, E.; An, S.S.A. Genome-editing applications of CRISPR–Cas9 to promote in vitro studies of Alzheimer’s disease. Clin. Interv. Aging 2018, 13, 221–233.
  39. Safari, F.; Hatam, G.; Behbahani, A.B.; Rezaei, V.; Barekati-Mowahed, M.; Petramfar, P.; Khademi, F. CRISPR System: A High-throughput Toolbox for Research and Treatment of Parkinson’s Disease. Cell. Mol. Neurobiol. 2020, 40, 477–493.
  40. Vermersch, E.; Jouve, C.; Hulot, J.S. CRISPR/Cas9 gene-editing strategies in cardiovascular cells. Cardiovasc. Res. 2020, 116, 894–907.
  41. Sharma, G.; Sharma, A.R.; Bhattacharya, M.; Lee, S.S.; Chakraborty, C. CRISPR-Cas9: A Preclinical and Clinical Perspective for the Treatment of Human Diseases. Mol. Ther. 2021, 29, 571–586.
  42. Karimian, A.; Gorjizadeh, N.; Alemi, F.; Asemi, Z.; Azizian, K.; Soleimanpour, J.; Malakouti, F.; Targhazeh, N.; Majidinia, M.; Yousefi, B. CRISPR/Cas9 novel therapeutic road for the treatment of neurodegenerative diseases. Life Sci. 2020, 259.
  43. Chen, M.; Mao, A.; Xu, M.; Weng, Q.; Mao, J.; Ji, J. CRISPR-Cas9 for cancer therapy: Opportunities and challenges. Cancer Lett. 2019, 447, 48–55.
  44. Xing, H.; Meng, L. CRISPR-cas9: A powerful tool towards precision medicine in cancer treatment. Acta Pharmacol. Sin. 2020, 41, 583–587.
  45. Jiang, C.; Meng, L.; Yang, B.; Luo, X. Application of CRISPR/Cas9 gene editing technique in the study of cancer treatment. Clin. Genet. 2020, 97, 73–88.
  46. Mirza, Z.; Karim, S. Advancements in CRISPR/Cas9 technology—Focusing on cancer therapeutics and beyond. Semin. Cell Dev. Biol. 2019, 96, 13–21.
  47. Saber, A.; Liu, B.; Ebrahimi, P.; Haisma, H.J. CRISPR/Cas9 for overcoming drug resistance in solid tumors. DARU J. Pharm. Sci. 2020, 28, 295–304.
  48. Liu, B.; Saber, A.; Haisma, H.J. CRISPR/Cas9: A powerful tool for identification of new targets for cancer treatment. Drug Discov. Today 2019, 24, 955–970.
  49. Vallone, C.; Rigon, G.; Gulia, C.; Baffa, A.; Votino, R.; Morosetti, G.; Zaami, S.; Briganti, V.; Catania, F.; Gaffi, M.; et al. Non-Coding RNAs and Endometrial Cancer. Genes 2018, 9, 187.
  50. Esposito, R.; Bosch, N.; Lanzós, A.; Polidori, T.; Pulido-Quetglas, C.; Johnson, R. Hacking the Cancer Genome: Profiling Therapeutically Actionable Long Non-coding RNAs Using CRISPR-Cas9 Screening. Cancer Cell 2019, 35, 545–557.
  51. Piergentili, R.; Zaami, S.; Cavaliere, A.F.; Signore, F.; Scambia, G.; Mattei, A.; Marinelli, E.; Gulia, C.; Perelli, F. Non-Coding RNAs as Prognostic Markers for Endometrial Cancer. Int. J. Mol. Sci. 2021, 22, 3151.
  52. Pavani, G.; Fabiano, A.; Laurent, M.; Amor, F.; Cantelli, E.; Chalumeau, A.; Maule, G.; Tachtsidi, A.; Concordet, J.-P.; Cereseto, A.; et al. Correction of β-thalassemia by CRISPR/Cas9 editing of the α-globin locus in human hematopoietic stem cells. Blood Adv. 2021, 5, 1137–1153.
  53. Xiao, Q.; Guo, D.; Chen, S. Application of CRISPR/Cas9-based gene editing in HIV-1/AIDS therapy. Front. Cell. Infect. Microbiol. 2019, 9, 69.
  54. Xu, T.; Li, L.; Liu, Y.C.; Cao, W.; Chen, J.S.; Hu, S.; Liu, Y.; Li, L.Y.; Zhou, H.; Meng, X.M.; et al. Crispr/cas9-related technologies in liver diseases: From feasibility to future diversity. Int. J. Biol. Sci. 2020, 16, 2283–2295.
  55. Zuo, E.; Huo, X.; Yao, X.; Hu, X.; Sun, Y.; Yin, J.; He, B.; Wang, X.; Shi, L.; Ping, J.; et al. CRISPR/Cas9-mediated targeted chromosome elimination. Genome Biol. 2017, 18, 224.
  56. North, A.R.; Burt, A.; Godfray, H.C.J. Modelling the Suppression of a Malaria Vector Using a CRISPR-Cas9 Gene Drive to Reduce Female Fertility. BMC Biol. 2020, 18, 98.
  57. Annas, G.J.; Beisel, C.L.; Clement, K.; Crisanti, A.; Francis, S.; Galardini, M.; Galizi, R.; Grünewald, J.; Immobile, G.; Khalil, A.S.; et al. A Code of Ethics for Gene Drive Research. Cris. J. 2021, 4, 19–24.
  58. Callies, D.E. The ethical landscape of gene drive research. Bioethics 2019, 33, 1091–1097.
  59. Kang, X.; He, W.; Huang, Y.; Yu, Q.; Chen, Y.; Gao, X.; Sun, X.; Fan, Y. Introducing precise genetic modifications into human 3PN embryos by CRISPR/Cas-mediated genome editing. J. Assist. Reprod. Genet. 2016, 33, 581–588.
  60. Yang, Y.; Huang, Y. The CRIPSR /Cas gene-editing system—An immature but useful toolkit for experimental and clinical medicine. Anim. Model. Exp. Med. 2019, 2, 5–8.
  61. Ma, Y.; Zhang, L.; Qin, C. The first genetically gene-edited babies: It’s “irresponsible and too early”. Anim. Model. Exp. Med. 2019, 2, 1–4.
  62. Razzouk, S. CRISPR-Cas9: A cornerstone for the evolution of precision medicine. Ann. Hum. Genet. 2018, 82, 331–357.
  63. Ma, H.; Marti-Gutierrez, N.; Park, S.W.; Wu, J.; Lee, Y.; Suzuki, K.; Koski, A.; Ji, D.; Hayama, T.; Ahmed, R.; et al. Correction of a pathogenic gene mutation in human embryos. Nature 2017, 548, 413–419.
  64. Zaami, S.; Piergentili, R.; Marinelli, E.; Montanari Vergallo, G. Commentary—CRISPR-based techniques: Cas9, Cas13 and their applications in the era of COVID-19. Eur. Rev. Med. Pharmacol. Sci. 2021, 25, 1752–1761.
  65. Wang, H.; Yang, H. Gene-edited babies: What went wrong and what could go wrong. PLoS Biol. 2019, 17, e3000224.
  66. Krishan, K.; Kanchan, T.; Singh, B. Human Genome Editing and Ethical Considerations. Sci. Eng. Ethics 2016, 22, 597–599.
  67. Korablev, A.; Lukyanchikova, V.; Serova, I.; Battulin, N. On-target CRISPR/CAS9 activity can cause undesigned large deletion in mouse zygotes. Int. J. Mol. Sci. 2020, 21, 3604.
  68. Kosicki, M.; Tomberg, K.; Bradley, A. Repair of double-strand breaks induced by CRISPR–Cas9 leads to large deletions and complex rearrangements. Nat. Biotechnol. 2018, 36, 765.
  69. Przewrocka, J.; Rowan, A.; Rosenthal, R.; Kanu, N.; Swanton, C. Unintended on-target chromosomal instability following CRISPR/Cas9 single gene targeting. Ann. Oncol. 2020, 31, 1270–1273.
  70. Cullot, G.; Boutin, J.; Toutain, J.; Prat, F.; Pennamen, P.; Rooryck, C.; Teichmann, M.; Rousseau, E.; Lamrissi-Garcia, I.; Guyonnet-Duperat, V.; et al. CRISPR-Cas9 genome editing induces megabase-scale chromosomal truncations. Nat. Commun. 2019, 10, 1–14.
  71. Alanis-Lobato, G.; Zohren, J.; McCarthy, A.; Fogarty, N.M.E.; Kubikova, N.; Hardman, E.; Greco, M.; Wells, D.; Turner, J.M.A.; Niakan, K.K. Frequent loss-of-heterozygosity in CRISPR-Cas9-edited early human embryos. Proc. Natl. Acad. Sci. USA 2021.
  72. Zuccaro, M.V.; Xu, J.; Mitchell, C.; Marin, D.; Zimmerman, R.; Rana, B.; Weinstein, E.; King, R.T.; Palmerola, K.L.; Smith, M.E.; et al. Allele-Specific Chromosome Removal after Cas9 Cleavage in Human Embryos. Cell 2020, 183, 1650–1664.
  73. Abd El-Aziz, M.M.; Barragan, I.; O’Driscoll, C.A.; Goodstadt, L.; Prigmore, E.; Borrego, S.; Mena, M.; Pieras, J.I.; El-Ashry, M.F.; Safieh, L.A.; et al. EYS, encoding an ortholog of Drosophila spacemaker, is mutated in autosomal recessive retinitis pigmentosa. Nat. Genet. 2008, 40, 1285–1287.
  74. Sfeir, A.; Symington, L.S. Microhomology-Mediated End Joining: A Back-up Survival Mechanism or Dedicated Pathway? Trends Biochem. Sci. 2015, 40, 701–714.
  75. Liang, D.; Gutierrez, N.M.; Chen, T.; Lee, Y.; Park, S.W.; Ma, H.; Koski, A.; Ahmed, R.; Darby, H.; Li, Y.; et al. Frequent gene conversion in human embryos induced by double strand breaks. bioRxiv 2020.
  76. Anzalone, A.V.; Randolph, P.B.; Davis, J.R.; Sousa, A.A.; Koblan, L.W.; Levy, J.M.; Chen, P.J.; Wilson, C.; Newby, G.A.; Raguram, A.; et al. Search-and-replace genome editing without double-strand breaks or donor DNA. Nature 2019, 576, 149–157.
  77. Tu, Z.; Yang, W.; Yan, S.; Yin, A.; Gao, J.; Liu, X.; Zheng, Y.; Zheng, J.; Li, Z.; Yang, S.; et al. Promoting Cas9 degradation reduces mosaic mutations in non-human primate embryos. Sci. Rep. 2017, 7, 42081.
  78. Peng, R.; Lin, G.; Li, J. Potential pitfalls of CRISPR/Cas9-mediated genome editing. FEBS J. 2016, 283, 1218–1231.
  79. Brokowski, C.; Pollack, M.; Pollack, R. Cutting Eugenics Out of CRISPR-Cas9. Ethics Biol. Eng. Med. 2015, 6, 263–279.
  80. Brokowski, C.; Adli, M. CRISPR Ethics: Moral Considerations for Applications of a Powerful Tool. J. Mol. Biol. 2019, 431, 88–101.
  81. Wu, S.; Powers, S.; Zhu, W.; Hannun, Y.A. Substantial contribution of extrinsic risk factors to cancer development. Nature 2016, 529, 43–47.
  82. Napoletano, S.; Piersanti, V.; Rallo, G. CRISPR -Cas9: A groundbreaking new technique which ushers in new prospects and just as many doubts. Clin. Ter. 2021, 171, e52–e54.
  83. Marinelli, S.; Del Rio, A. Beginning of Life Ethics at the Dawn of a New Era of Genome Editing: Are Bioethical Precepts and Fast-Evolving Biotechnologies Irreconcilable? Clin. Ter. 2020, 171, e407–e411.
  84. Gilbert, S.F. When “Personhood” Begins in the Embryo: Avoiding a Syllabus of Errors. Birth Defects Res. C Embryo Today 2008, 84, 164–173.
  85. Charo, R.A. The hunting of the snark: The moral status of embryos, right-to-lifers, and Third World women. Stanf. Law Pol. Rev. 1995, 6, 11–37.
  86. Ledford, H. CRISPR gene editing in human embryos wreaks chromosomal mayhem. Nature 2020, 583, 17–18.
  87. Ricci, G.; Campanozzi, L.L.; Marinelli, S.; Midolo, E.; Ruggeri, L. The Human Embryo, Subjectivity and Legal Capacity. Notes in the Light of Art. 1 of the Italian Law on “Medically Assisted Procreation”. Clin. Ter. 2019, 170, e102–e107.
  88. Botkin, J.R. The Case for Banning Heritable Genome Editing. Genet. Med. 2020, 22, 487–489.
  89. Musunuru, K. Why Human Embryo Editing Should Be Banned. CRISPR J. 2019, 2, 356–358.
  90. Grant, E.V. FDA Regulation of Clinical Applications of CRISPR-CAS Gene-Editing Technology. Food Drug L. J. 2016, 71, 608–633.
  91. Public Law 114–113—18 December 2015. Consolidated Appropriations Act. 2016. Available online: (accessed on 15 February 2021).
  92. Williams, K.; Johnson, M.H. Adapting the 14-Day Rule for Embryo Research to Encompass Evolving Technologies. Reprod. Biomed. Soc. Online 2020, 10, 1–9.
  93. Law 40, 24 February 2004, Regulation of Medically Assisted Human Reproduction, Legge 24 Febbraio 2004, n. 40, Norme in Materia di Procreazione Medicalmente Assistita, G. U. N. 45 24-2-2004. Available online: (accessed on 15 February 2021).
  94. Montanari, V.G.; Zaami, S.; Bruti, V.; Signore, F. How the legislation in medically assisted procreation has evolved in Italy. Med. Law 2017, 36, 5.
  95. Negro, F.; Marinelli, S. Is There Anything Left of the Italian Law Governing Medically-Assisted Procreation? Clin. Ter. 2021, 171, e57–e59.
  96. Castelyn, G. Embryo Experimentation: Is There a Case for Moving beyond the “14-Day Rule”. Monash Bioeth. Rev. 2020, 38, 181–196.
  97. Dobler, R.; Dowling, D.K.; Morrow, E.H.; Reinhardt, K. A Systematic Review and Meta-Analysis Reveals Pervasive Effects of Germline Mitochondrial Replacement on Components of Health. Hum. Reprod. Update 2018, 24, 519–534.
  98. Cyranoski, D. The CRISPR-Baby Scandal: What’s next for Human Gene-Editing. Nature 2019, 566, 440–442.
  99. Miklavcic, J.J.; Flaman, P. Personhood Status of the Human Zygote, Embryo, Fetus. Linacre Q. 2017, 84, 130–144.
  100. Paul, P.J., II. You Shall Not Kill in Idem, The Gospel of Life (Evangelium Vitae); Times Books; Random House: New York, NY, USA, 1995; pp. 92–140.
  101. Nimbalkar, N. John Locke on Personal Identity. Mens. Sana Monogr. 2011, 9, 268–275.
  102. Manninen, B.A. Are Human Embryos Kantian Persons? Kantian Considerations in Favor of Embryonic Stem Cell Research. Philos. Ethics Humanit. Med. 2008, 3, 4.
  103. Donovan, P. When Does Personhood Begin? Fam. Plan. Perspect. 1983, 15, 40–44.
  104. Wagner, N.-F.; Northoff, G. Habits: Bridging the Gap between Personhood and Personal Identity. Front. Hum. Neurosci. 2014, 8, 330.
  105. Fan, H.-C.; Chi, C.-S.; Lee, Y.-J.; Tsai, J.-D.; Lin, S.-Z.; Harn, H.-J. The Role of Gene Editing in Neurodegenerative Diseases. Cell Transplant. 2018, 27, 364–378.
  106. Gallego, C.; Gonçalves, M.A.F.V.; Wijnholds, J. Novel Therapeutic Approaches for the Treatment of Retinal Degenerative Diseases: Focus on CRISPR/Cas-Based Gene Editing. Front. Neurosci. 2020, 14, 838.
  107. Gaj, T.; Perez-Pinera, P. The Continuously Evolving CRISPR Barcoding Toolbox. Genome Biol. 2018, 19, 143.
  108. Wang, H.; La Russa, M.; Qi, L.S. CRISPR/Cas9 in Genome Editing and Beyond. Annu. Rev. Biochem. 2016, 85, 227–264.
  109. Zhang, J.-H.; Adikaram, P.; Pandey, M.; Genis, A.; Simonds, W.F. Optimization of Genome Editing through CRISPR-Cas9 Engineering. Bioengineered 2016, 7, 166–174.
  110. Ben Jehuda, R.; Shemer, Y.; Binah, O. Genome Editing in Induced Pluripotent Stem Cells Using CRISPR/Cas9. Stem Cell Rev. Rep. 2018, 14, 323–336.
  111. Geng, B.; Choi, K.-H.; Wang, S.; Chen, P.; Pan, X.; Dong, N.; Ko, J.-K.; Zhu, H. A Simple, Quick, and Efficient CRISPR/Cas9 Genome Editing Method for Human Induced Pluripotent Stem Cells. Acta Pharmacol. Sin. 2020, 41, 1427–1432.
  112. Lu, F.; Zhang, Y. Cell Totipotency: Molecular Features, Induction, and Maintenance. Natl. Sci. Rev. 2015, 2, 217–225.
  113. Le, R.; Huang, Y.; Zhao, A.; Gao, S. Lessons from Expanded Potential of Embryonic Stem Cells: Moving toward Totipotency. J. Genet. Genom. 2020, 47, 123–130.
  114. De Masi, C.; Spitalieri, P.; Murdocca, M.; Novelli, G.; Sangiuolo, F. Application of CRISPR/Cas9 to Human-Induced Pluripotent Stem Cells: From Gene Editing to Drug Discovery. Hum. Genom. 2020, 14, 25.
  115. Spitalieri, P.; Quitadamo, M.C.; Orlandi, A.; Guerra, L.; Giardina, E.; Casavola, V.; Novelli, G.; Saltini, C.; Sangiuolo, F. Rescue of Murine Silica-Induced Lung Injury and Fibrosis by Human Embryonic Stem Cells. Eur. Respir. J. 2012, 39, 446–457.
  116. Nie, J.; Li, Q.; Wu, J.; Zhao, C.; Hao, H.; Liu, H.; Zhang, L.; Nie, L.; Qin, H.; Wang, M.; et al. Establishment and Validation of a Pseudovirus Neutralization Assay for SARS-CoV-2. Emerg. Microbes Infect. 2020, 9, 680–686.
  117. Uddin, F.; Rudin, C.M.; Sen, T. CRISPR Gene Therapy: Applications, Limitations, and Implications for the Future. Front. Oncol. 2020, 10, 1387.
  118. Nguyen, T.M.; Zhang, Y.; Pandolfi, P.P. Virus against Virus: A Potential Treatment for 2019-NCov (SARS-CoV-2) and Other RNA Viruses. Cell. Res. 2020, 30, 189–190.
  119. Straiton, J. CRISPR vs. COVID-19: How Can Gene Editing Help Beat a Virus? BioTechniques 2020, 69, 327–329.
  120. Abbott, T.R.; Dhamdhere, G.; Liu, Y.; Lin, X.; Goudy, L.; Zeng, L.; Chemparathy, A.; Chmura, S.; Heaton, N.S.; Debs, R.; et al. Development of CRISPR as an Antiviral Strategy to Combat SARS-CoV-2 and Influenza. Cell 2020, 181, 865–876.
More