Skip to main content
Animal Models and Experimental Medicine logoLink to Animal Models and Experimental Medicine
. 2019 Mar 13;2(1):5–8. doi: 10.1002/ame2.12061

The CRIPSR/Cas gene‐editing system—an immature but useful toolkit for experimental and clinical medicine

Yuyan Yang 1,2, Yue Huang 1,2,
PMCID: PMC6431121  PMID: 31016281

Abstract

A Chinese scientist, Jiankui He, and his creation of the world's first genetically altered baby made headlines recently. As a newly developed gene‐editing technique, the CRISPR/Cas system should not be applied to human beings for reproductive purposes until it has been extensively tested. However, numerous experimental research studies in human somatic, germline cells, and even in embryos, have been conducted, which have shown CRISPR/Cas to be a useful tool for human genome editing and a potential therapeutic method for future clinical use.

Keywords: CRISPR‐Cas, gene editing, gene therapy, human embryos, recombinant DNA


With the development of recombinant DNA technology, programmable nuclease‐based genome‐editing tools are gradually being created. Zinc finger nucleases (ZNFs)1 and transcriptional activator‐like effector nucleases (TALENs)2 have been well studied and are already being applied in several gene therapy clinical trials for serious inherited genetic diseases, and the newly developed CRISPR/Cas9 system3 is turning out to be a more promising tool. In nature, CRISPR/Cas systems provide bacteria with RNA‐guided adaptive immunity against foreign genetic elements by creating doubled‐stranded breaks (DSBs) with their directing nuclease activity.4, 5 Among them, the designated class 2 CRISPR/Cas9 systems with single guide RNA (sgRNA) are being applied to mammalian genome editing, including in human cells.6 Besides gene editing, the derived epigenetic editing tools CRISPR interference (CRISPRi), CRISPR activation (CRISPRa), and catalytically deficient Cas9 (dCas9) have also been extensively used and modified.7, 8, 9 Since the first application in mammalian cells in 2013, CRISPR/Cas9 systems have been researched in human somatic cells, embryonic or germline cells, and even in embryos prior to germ‐layer formation. Despite its practical immaturity and drawbacks, the CRISPR/Cas system still holds great promise for disease treatment and prevention.10, 11, 12

1. GENETIC SCREENING AND PROGRAMMABLE NUCLEIC ACID IMAGING

The CRISPR/Cas9‐based gene‐editing system has prompted functional genome‐wide genetic screening, which enables the study of gene functions and genetic interactions in complex heritable diseases. Up to now, a number of cell types have been studied.13, 14 Diseases such fragile X syndrome, type I diabetes, acute kidney injury, and murine muscular dystrophy have all been researched.15, 16 Furthermore, spatiotemporal localization of specific genomic loci can also be detected by using dCas9. Dysregulated molecular localization can result in or aggravate diseases.17 Since this tool for rapid detection of specific loci (DNA) or transcripts (RNA) is partially adaptable for clinical use, related point‐of‐care diagnostics should be available in the near future. The CRISPR/Cas9 system is therefore a powerful screening tool for systematically elucidating gene function in health and disease states, which is crucial for understanding the pathogenic mechanism.

2. GENE THERAPY WITH SOMATIC CELL GENE EDITING

The first clinical trial of conventional somatic gene therapy was allowed to proceed in the 1990s, but the ensuing issues of safety and efficacy, and several tragic medical accidents, largely delayed its translation to the clinic. Recently developed nuclease‐based gene‐editing technologies have already breathed new life into somatic gene therapy. CRISPR/Cas9‐based gene editing, with its high efficiency, flexibility, and accuracy, can be applied to correct mutations or inactivate defective exons in animal models or human cells with pathologies such as Duchenne muscular dystrophy (DMD), amyotrophic lateral sclerosis and Huntington's disease.18, 19, 20, 21 Recently, researchers succeeded in eliminating an entire chromosome in different cell types, including human induced pluripotent stem (iPS) cells with trisomy 21.22 Furthermore, on November 30, 2018, Editas Medicine announced FDA acceptance of their Investigative New Drug (IND) application for EDIT‐101, a CRISPR genome‐editing tool that reverses the IVS26 mutation using the CRISPR/Cas9 system to restore the function of photoreceptor cells.23

Cancer is characterized by multiple genetic alterations leading to malignant cell proliferation. Newly developed cancer immunotherapies, such as chimeric antigen receptor T‐cells (CAR‐T), have shown striking efficacy against multiple cancers in clinical trials. Currently, both CRISPR‐Cas9 and TALENs gene‐editing systems are being applied in T‐cell engineering.24 In 2015, oncologists at Sichuan University injected CRISPR‐Cas9‐modified T cells into patients with aggressive lung cancer at the West China Hospital, in the first CRISPR‐Cas9 application in a clinical trial.25

3. EPIGENETIC‐EDITING AND RNA‐TARGETING CRISPR SYSTEMS FOR CLINICAL USE

Epigenetic editing does not induce DSBs, so it turns out to be safer than direct gene editing. dCas9 is the most useful scaffold for incorporating multiple modulators to perturb transcription without permanent DNA alternation. Chromatin and histone modifications are the most direct way to regulate inherited gene expression.26, 27 Moreover, based on the dCas9 platform, targeted DNA methylation and demethylation, and deployment of long non‐coding RNA (ncRNA) to ectopic genomic sites can be used in both the study of gene expression and regulation, and also potential therapeutic strategies.28, 29

Posttranscriptional engineering mainly focuses on RNA. It can be used for eliminating pathogenic RNA molecules, adjusting aberrant mRNA expression and splicing. Nowadays, artificial RNA‐targeting Cas9s have programmable RNA modulating activity independent of Protospacer Adjacent Motif (PAM)‐presenting oligonucleotides (PAMmers).30 CRISPR‐Cas13, another class 2 type VI CRISPR‐Cas system, specially targets RNA molecules and can be used for targeted gene knockdown in human cells.31, 32 Although the CRISPR‐Cas13a system is currently only in its infancy, we still expect it to be applicable in anti‐virus prophylaxis and other disease treatments.

4. GENE EDITING IN HUMAN GERMLINE CELLS AND EMBRYOS, AND RELATED ETHICAL ISSUES

Apart from gene editing in somatic cells, the CRISPR/Cas9 system has been applied in human germline cells and embryos. Recent research has proved that this system is effective in HBB and G6PD point‐mutation correction in human zygotes.33 In 2017, Dr Shoukhrat Mitalipov and his team claimed success in correction of the heterozygous MYBPC3 mutation in human preimplantation embryos using a CRISPR/Cas9 system.34 However, the relevant safety and efficacy issues of gene editing in human embryos are still being debated.35, 36, 37

Surprisingly, and unfortunately, Dr Jiankui He from the Southern University of Science and Technology of China recently announced that he has created the world's first gene‐edited babies using CRISPR‐Cas9 technology. This breaking news incurred world‐wide criticism and controversy. In the current circumstances, and in agreement with the Chinese Academy of Medical Sciences response,38 we are opposed to any clinical application of human embryo genome editing for reproductive purposes, which, in the absence of full scientific evaluation, is in violation of laws, regulations, and ethical norms.

5. SAFETY

Safety is the prerequisite for every therapeutic technique. Cas9‐mediated single‐base editors provide the potential to correct point mutations of disease‐related genes without inducing DSBs, and thus without small insertions or deletions.39, 40 Off‐target effects of the Cas endonucleases are also an important concern of this newly developed gene‐editing tool. Recent research has revealed that, with appropriately designed sgRNA, and using a highly sensitive strategy for identifying such effects, the CRISPR/Cas9 system can achieve in vivo editing without detectable genome‐wide off‐target mutations.41 These studies all indicate that the CRISPR/Cas9 system can be further improved and finally become clinically applicable.

In conclusion, research into the CRISPR/Cas9 system has shown that, although the system is still not mature enough for clinical application in humans, steady progress has been made. While we should undoubtedly condemn Jiankui He's irresponsible actions, at the same time we should not deny that the CRISPR/Cas gene‐editing technique has a bright future.

CONFLICT OF INTEREST

None.

ACKNOWLEDGEMENTS

The present work was supported in part by the National Key Research and Development Program of China (2016YFA0100103 to YH), and the CAMS Innovation Fund for Medical Sciences (2016‐I2M‐3‐002 to YH).

Yang Y, Huang Y. The CRIPSR/Cas gene‐editing system—an immature but useful toolkit for experimental and clinical medicine. Anim Models Exp Med. 2019;2:5–8. 10.1002/ame2.12061

REFERENCES

  • 1. Urnov FD, Rebar EJ, Holmes MC, Zhang HS, Gregory PD. Genome editing with engineered zinc finger nucleases. Nat Rev Genet. 2010;11:636‐646. [DOI] [PubMed] [Google Scholar]
  • 2. Joung JK, Sander JD. TALENs: a widely applicable technology for targeted genome editing. Nat Rev Mol Cell Biol. 2012;14:49‐55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Jinek M, Chylinski K, Fonfara I, Hauer M, Doudna JA, Charpentier E. A programmable dual‐RNA‐guided DNA endonuclease in adaptive bacterial immunity. Science. 2012;337:816‐821. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Wagner R, Pul Ü. CRISPR: a Bacterial Immunity System Based on Small RNAs In: Erdmann V, Barciszewski J, eds. From Nucleic Acids Sequences to Molecular Medicine. Berlin, Heidelberg: Springer Press; 2012:121‐143. [Google Scholar]
  • 5. Wiedenheft B, Sternberg SH, Doudna JA. RNA‐guided genetic silencing systems in bacteria and archaea. Nature. 2012;482:331‐338. [DOI] [PubMed] [Google Scholar]
  • 6. Koonin EV, Makarova KS, Zhang F. Diversity, classification and evolution of CRISPR‐Cas systems. Curr Opin Microbiol. 2017;37:67‐78. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Larson MH, Gilbert LA, Wang X, Lim WA, Weissman JS, Qi LS. CRISPR interference (CRISPRi) for sequence‐specific control of gene expression. Nat Protoc. 2013;8:2180‐2196. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Gilbert LA, Horlbeck MA, Adamson B, et al. Genome‐scale CRISPR‐mediated control of gene repression and activation. Cell. 2014;159:647‐661. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Pulecio J, Verma N, Mejía‐Ramírez E, Huangfu D, Raya A. CRISPR/Cas9‐Based engineering of the epigenome. Cell Stem Cell. 2017;21:431. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Hsu P, Lander E, Zhang F. Development and applications of CRISPR‐Cas9 for genome engineering. Cell. 2014;157:1262‐1278. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Cox DB, Platt RJ, Zhang F. Therapeutic genome editing: prospects and challenges. Nat Med. 2015;21:121‐131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Knott GJ, Doudna JA. CRISPR‐Cas guides the future of genetic engineering. Science. 2018;361:866‐869. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Bak RO, Dever DP, Reinisch A, Cruz Hernandez D, Majeti R, Porteus MH. Multiplexed genetic engineering of human hematopoietic stem and progenitor cells using CRISPR/Cas9 and AAV6. ELife. 2017;6:e27873. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Zheng Y, Shen W, Zhang J, et al. CRISPR interference‐based specific and efficient gene inactivation in the brain. Nat Neurosci. 2018;21:447‐454. [DOI] [PubMed] [Google Scholar]
  • 15. Liu XS, Wu H, Krzisch M, et al. Rescue of fragile X syndrome neurons by DNA methylation editing of the FMR1 gene. Cell. 2018;172:979‐992. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Liao HK, Hatanaka F, Araoka T, et al. In Vivo target gene activation via CRISPR/Cas9‐mediated trans‐epigenetic modulation. Cell. 2017;171:1495‐1507. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Knight S, Tjian R, Doudna J. Genomes in focus: development and applications of CRISPR‐Cas9 imaging technologies. Angew Chem Int Ed Engl. 2019;57:4329‐4337. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Zhang Y, Long C, Li H, et al. CRISPR‐Cpf1 correction of muscular dystrophy mutations in human cardiomyocytes and mice. Sci Advs. 2017;3:e1602814. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Jon C. In dogs, CRISPR fixes a muscular dystrophy. Science. 2018;361:835. [DOI] [PubMed] [Google Scholar]
  • 20. Gaj T, Ojala DS, Ekman FK, Byrne LC, Limsirichai P, Schaffer DV. In vivo genome editing improves motor function and extends survival in a mouse model of ALS. Sci Adv. 2017;3:eaar3952. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Staahl BT, Benekareddy M, Coulon‐Bainier C, et al. Efficient genome editing in the mouse brain by local delivery of engineered Cas9 ribonucleoprotein complexes. Nat Biotechnol. 2017;35:431‐434. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Zuo E, Huo X, Yao X, et al. CRISPR/Cas9‐mediated targeted chromosome elimination. Genome Biol. 2017;18:224. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Maeder ML, Stefanidakis M, Wilson CJ, et al. Development of a gene‐editing approach to restore vision loss in Leber congenital amaurosis type 10. Nat Med. 2019;25:229‐233. [DOI] [PubMed] [Google Scholar]
  • 24. Rupp LJ, Schumann K, Roybal KT, et al. CRISPR/Cas9‐mediated PD‐1 disruption enhances anti‐tumor efficacy of human chimeric antigen receptor T cells. Sci Rep. 2017;7(1):737. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. News N. CRISPR gene‐editing tested in a person for the first time. Nature. 2016;539:479. [DOI] [PubMed] [Google Scholar]
  • 26. Keung AJ, Joung JK, Khalil AS, Collins JJ. Chromatin regulation at the frontier of synthetic biology. Nat Rev Genet. 2015;16(3):159‐171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Polstein LR, Perez‐Pinera P, Kocak DD, et al. Genome‐wide specificity of DNA binding, gene regulation, and chromatin remodeling by TALE‐ and CRISPR/Cas9‐based transcriptional activators. Genome Res. 2015;25:1158‐1169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Morita S, Noguchi H, Horii T, et al. Targeted DNA demethylation in vivo using dCas9‐peptide repeat and scFv‐TET1 catalytic domain fusions. Nat Biotechnol. 2016;34:1060‐1065. [DOI] [PubMed] [Google Scholar]
  • 29. Shechner DM, Hacisuleyman E, Younger ST, Rinn JL. Multiplexable, locus‐specific targeting of long RNAs with CRISPR‐Display. Nat Methods. 2015;12:664‐670. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Dugar G, Leenay RT, Eisenbart SK, et al. CRISPR RNA‐dependent binding and cleavage of endogenous RNAs by the Campylobacter jejuni Cas9. Mol Cell. 2018;69:893‐905. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Abudayyeh OO, Gootenberg JS, Essletzbichler P, et al. RNA targeting with CRISPR–Cas13. Nature. 2017;550:280‐284. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Cox DBT, Gootenberg JS, Abudayyeh OO, et al. RNA editing with CRISPR–Cas13. Science. 2017;358:1019‐1027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Tang L, Zeng Y, Du H, et al. CRISPR/Cas9‐mediated gene editing in human zygotes using Cas9 protein. Mol Genet Genomics. 2017;292:525‐533. [DOI] [PubMed] [Google Scholar]
  • 34. Ma H, Marti‐Gutierrez N, Park SW, et al. Correction of a pathogenic gene mutation in human embryos. Nature. 2017;548:413‐419. [DOI] [PubMed] [Google Scholar]
  • 35. Egli D, Zuccaro MV, Kosicki M, Church GM, Bradley A, Jasin M. Inter‐homologue repair in fertilized human eggs? Nature. 2018;560:E5‐E7. [DOI] [PubMed] [Google Scholar]
  • 36. Adikusuma F, Piltz S, Corbett MA, et al. Large deletions induced by Cas9 cleavage. Nature. 2018;560:E8‐E9. [DOI] [PubMed] [Google Scholar]
  • 37. Ma H, Martigutierrez N, Park SW, et al. Ma et al. reply. Nature. 2018;560:E10‐E23. [DOI] [PubMed] [Google Scholar]
  • 38. Wang C, Zhai X, Zhang X, et al. Gene‐edited babies: Chinese Academy of Medical Sciences’ response and action. Lancet. 2019;393:25‐26. [DOI] [PubMed] [Google Scholar]
  • 39. Komor AC, Kim YB, Packer MS, Zuris JA, Liu DR. Programmable editing of a target base in genomic DNA without double‐stranded DNA cleavage. Nature. 2016;533:420‐424. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Gaudelli NM, Komor AC, Rees HA, et al. Programmable base editing of A•T to G•C in genomic DNA without DNA cleavage. Nature. 2017;551:464‐471. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Akcakaya P, Bobbin ML, Guo JA, et al. In vivo CRISPR editing with no detectable genome‐wide off‐target mutations. Nature. 2018;561:416‐419. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Animal Models and Experimental Medicine are provided here courtesy of Wiley

RESOURCES