Non-viral and viral delivery systems for CRISPR-Cas9 technology in the biomedical field

logo

SCIENCE CHINA Life Sciences, Volume 60, Issue 5: 458-467(2017) https://doi.org/10.1007/s11427-017-9033-0

Non-viral and viral delivery systems for CRISPR-Cas9 technology in the biomedical field

More info
  • ReceivedJan 16, 2017
  • AcceptedMar 16, 2017
  • PublishedMay 2, 2017

Abstract

The clustered regularly interspaced short palindromic repeats (CRISPR)-associated protein 9 (CRISPR-Cas9) system provides a novel genome editing technology that can precisely target a genomic site to disrupt or repair a specific gene. Some CRISPR-Cas9 systems from different bacteria or artificial variants have been discovered or constructed by biologists, and Cas9 nucleases and single guide RNAs (sgRNA) are the major components of the CRISPR-Cas9 system. These Cas9 systems have been extensively applied for identifying therapeutic targets, identifying gene functions, generating animal models, and developing gene therapies. Moreover, CRISPR-Cas9 systems have been used to partially or completely alleviate disease symptoms by mutating or correcting related genes. However, the efficient transfer of CRISPR-Cas9 system into cells and target organs remains a challenge that affects the robust and precise genome editing activity. The current review focuses on delivery systems for Cas9 mRNA, Cas9 protein, or vectors encoding the Cas9 gene and corresponding sgRNA. Non-viral delivery of Cas9 appears to help Cas9 maintain its on-target effect and reduce off-target effects, and viral vectors for sgRNA and donor template can improve the efficacy of genome editing and homology-directed repair. Safe, efficient, and producible delivery systems will promote the application of CRISPR-Cas9 technology in human gene therapy.


Funded by

National High Technology Research and Development Program of China(2015AA020309 to Zhi-Yao He)

China Postdoctoral Science Foundation Funded Project(2015M570791 to Zhi-Yao He)

National Natural and Scientific Foundation of China(81602699 to Zhi-Yao He)


Acknowledgment

This work was supported by the National Natural and Scientific Foundation of China (81602699 to Zhi-Yao He, 81502677 to Ke Men, 81402302 to Yang Yang), the National High Technology Research and Development Program of China (2015AA020309 to Zhi-Yao He), and the China Postdoctoral Science Foundation Funded Project (2015M570791 to Zhi-Yao He).


Interest statement

The author(s) declare that they have no conflict of interest.


References

[1] Adler A.F., Grigsby C.L., Kulangara K., Wang H., Yasuda R., Leong K.W.. Nonviral direct conversion of primary mouse embryonic fibroblasts to neuronal cells. Mol Ther Nucleic Acids, 2012, 1: e32 CrossRef PubMed Google Scholar

[2] Aubrey B.J., Kelly G.L., Kueh A.J., Brennan M.S., O’Connor L., Milla L., Wilcox S., Tai L., Strasser A., Herold M.J.. An inducible lentiviral guide RNA platform enables the identification of tumor-essential genes and tumor-promoting mutations in vivo. Cell Rep, 2015, 10: 1422-1432 CrossRef PubMed Google Scholar

[3] Blinka S., Reimer Jr. M.H., Pulakanti K., Rao S.. Super-enhancers at the nanog locus differentially regulate neighboring pluripotency-associated genes. Cell Rep, 2016, 17: 19-28 CrossRef PubMed Google Scholar

[4] Chen Y., Wang Z., Ni H., Xu Y., Chen Q., Jiang L.. CRISPR/Cas9-mediated base-editing system efficiently generates gain-of-function mutations in Arabidopsis. Sci China Life Sci, 2017, : in press doi: 10.1007/s11427-017-9021-5 CrossRef PubMed Google Scholar

[5] Cheong T.C., Compagno M., Chiarle R.. Editing of mouse and human immunoglobulin genes by CRISPR-Cas9 system. Nat Commun, 2016, 7: 10934 CrossRef PubMed ADS Google Scholar

[6] Cho S.W., Kim S., Kim Y., Kweon J., Kim H.S., Bae S., Kim J.S.. Analysis of off-target effects of CRISPR/Cas-derived RNA-guided endonucleases and nickases. Genome Res, 2014, 24: 132-141 CrossRef PubMed Google Scholar

[7] Cong L., Ran F.A., Cox D., Lin S., Barretto R., Habib N., Hsu P.D., Wu X., Jiang W., Marraffini L.A., Zhang F.. Multiplex genome engineering using CRISPR/Cas systems. Science, 2013, 339: 819-823 CrossRef PubMed ADS Google Scholar

[8] Cox D.B.T., Platt R.J., Zhang F.. Therapeutic genome editing: prospects and challenges. Nat Med, 2015, 21: 121-131 CrossRef PubMed Google Scholar

[9] Croce C.M., Zhang K., Wei Y.. Announcing signal transduction and targeted therapy. Sig Transduct Target Ther, 2016, 1: 15006 CrossRef Google Scholar

[10] Cyranoski D.. Chinese scientists to pioneer first human CRISPR trial. Nature, 2016, 535: 476-477 CrossRef PubMed ADS Google Scholar

[11] Davis K.M., Pattanayak V., Thompson D.B., Zuris J.A., Liu D.R.. Small molecule-triggered Cas9 protein with improved genome-editing specificity. Nat Chem Biol, 2015, 11: 316-318 CrossRef PubMed Google Scholar

[12] Deng H., Li W., Wei Y.. Translational medicine center of West China Hospital. Sci China Life Sci, 2016, 59: 1055-1056 CrossRef PubMed Google Scholar

[13] Feng Y., Sassi S., Shen J.K., Yang X., Gao Y., Osaka E., Zhang J., Yang S., Yang C., Mankin H.J., Hornicek F.J., Duan Z.. Targeting Cdk11 in osteosarcoma cells using the CRISPR-cas9 system. J Orthop Res, 2015, 33: 199-207 CrossRef PubMed Google Scholar

[14] Fricano-Kugler C.J., Williams M.R., Salinaro J.R., Li M., Luikart B.. Designing, packaging, and delivery of high titer CRISPR retro and lentiviruses via stereotaxic injection. J Vis Exp, 2016, : in press doi: 10.3791/53783 CrossRef PubMed Google Scholar

[15] Fu Y., Sander J.D., Reyon D., Cascio V.M., Joung J.K.. Improving CRISPR-Cas nuclease specificity using truncated guide RNAs. Nat Biotechnol, 2014, 32: 279-284 CrossRef PubMed Google Scholar

[16] Gaj T., Gersbach C.A., Barbas Iii C.F.. ZFN, TALEN, and CRISPR/Cas-based methods for genome engineering. Trends Biotech, 2013, 31: 397-405 CrossRef PubMed Google Scholar

[17] Garneau J.E., Dupuis M.è., Villion M., Romero D.A., Barrangou R., Boyaval P., Fremaux C., Horvath P., Magadán A.H., Moineau S.. The CRISPR/Cas bacterial immune system cleaves bacteriophage and plasmid DNA. Nature, 2010, 468: 67-71 CrossRef PubMed ADS Google Scholar

[18] Harel I., Valenzano D.R., Brunet A.. Efficient genome engineering approaches for the short-lived African turquoise killifish. Nat Protoc, 2016, 11: 2010-2028 CrossRef PubMed Google Scholar

[19] He Z.Y., Deng F., Wei X.W., Ma C.C., Luo M., Zhang P., Sang Y.X., Liang X., Liu L., Qin H.X., Shen Y.L., Liu T., Liu Y.T., Wang W., Wen Y.J., Zhao X., Zhang X.N., Qian Z.Y., Wei Y.Q.. Ovarian cancer treatment with a tumor-targeting and gene expression-controllable lipoplex. Sci Rep, 2016, 6: 23764 CrossRef PubMed ADS Google Scholar

[20] Hille F., Charpentier E.. CRISPR-Cas: biology, mechanisms and relevance. Phil Trans R Soc B, 2016, 371: 20150496 CrossRef PubMed Google Scholar

[21] Jao L.E., Wente S.R., Chen W.. Efficient multiplex biallelic zebrafish genome editing using a CRISPR nuclease system. Proc Natl Acad Sci USA, 2013, 110: 13904-13909 CrossRef PubMed ADS Google Scholar

[22] Jiang C., Mei M., Li B., Zhu X., Zu W., Tian Y., Wang Q., Guo Y., Dong Y., Tan X.. A non-viral CRISPR/Cas9 delivery system for therapeutically targeting HBV DNA and pcsk9 in vivo. Cell Res, 2017, 27: 440-443 CrossRef PubMed Google Scholar

[23] Joung J.K., Sander J.D.. TALENs: a widely applicable technology for targeted genome editing. Nat Rev Mol Cell Biol, 2013, 14: 49-55 CrossRef PubMed Google Scholar

[24] Kennedy E.M., Bassit L.C., Mueller H., Kornepati A.V.R., Bogerd H.P., Nie T., Chatterjee P., Javanbakht H., Schinazi R.F., Cullen B.R.. Suppression of hepatitis B virus DNA accumulation in chronically infected cells using a bacterial CRISPR/Cas RNA-guided DNA endonuclease. Virology, 2015, 476: 196-205 CrossRef PubMed Google Scholar

[25] Kennedy E.M., Kornepati A.V.R., Goldstein M., Bogerd H.P., Poling B.C., Whisnant A.W., Kastan M.B., Cullen B.R.. Inactivation of the human papillomavirus E6 or E7 gene in cervical carcinoma cells by using a bacterial CRISPR/Cas RNA-guided endonuclease. J Virol, 2014, 88: 11965-11972 CrossRef PubMed Google Scholar

[26] Kim S., Kim D., Cho S.W., Kim J., Kim J.S.. Highly efficient RNA-guided genome editing in human cells via delivery of purified Cas9 ribonucleoproteins. Genome Res, 2014, 24: 1012-1019 CrossRef PubMed Google Scholar

[27] Kleinstiver B.P., Pattanayak V., Prew M.S., Tsai S.Q., Nguyen N.T., Zheng Z., Joung J.K.. High-fidelity CRISPR-Cas9 nucleases with no detectable genome-wide off-target effects. Nature, 2016, 529: 490-495 CrossRef PubMed ADS Google Scholar

[28] Li H., Eishingdrelo A., Kongsamut S., Eishingdrelo H.. G-protein-coupled receptors mediate 14-3-3 signal transduction. Sig Transduct Target Ther, 2016, 1: 16018 CrossRef Google Scholar

[29] Lin S.R., Yang H.C., Kuo Y.T., Liu C.J., Yang T.Y., Sung K.C., Lin Y.Y., Wang H.Y., Wang C.C., Shen Y.C., Wu F.Y., Kao J.H., Chen D.S., Chen P.J.. The CRISPR/Cas9 system facilitates clearance of the intrahepatic HBV templates in vivo. Mol Ther Nucleic Acids, 2014a, 3: e186 CrossRef PubMed Google Scholar

[30] Lin Y., Cradick T.J., Brown M.T., Deshmukh H., Ranjan P., Sarode N., Wile B.M., Vertino P.M., Stewart F.J., Bao G.. CRISPR/Cas9 systems have off-target activity with insertions or deletions between target DNA and guide RNA sequences. Nucleic Acids Res, 2014b, 42: 7473-7485 CrossRef PubMed Google Scholar

[31] Liu J., Shui S.L.. Delivery methods for site-specific nucleases: achieving the full potential of therapeutic gene editing. J Control Release, 2016, 244: 83-97 CrossRef PubMed Google Scholar

[32] Liu Y., Zeng Y., Liu L., Zhuang C., Fu X., Huang W., Cai Z.. Synthesizing AND gate genetic circuits based on CRISPR-Cas9 for identification of bladder cancer cells. Nat Commun, 2014, 5: 5393 CrossRef PubMed ADS Google Scholar

[33] Long C., Amoasii L., Mireault A.A., McAnally J.R., Li H., Sanchez-Ortiz E., Bhattacharyya S., Shelton J.M., Bassel-Duby R., Olson E.N.. Postnatal genome editing partially restores dystrophin expression in a mouse model of muscular dystrophy. Science, 2016, 351: 400-403 CrossRef PubMed ADS Google Scholar

[34] Maggio I., Holkers M., Liu J., Janssen J.M., Chen X., Gon?alves M.A.F.V.. Adenoviral vector delivery of RNA-guided CRISPR/Cas9 nuclease complexes induces targeted mutagenesis in a diverse array of human cells. Sci Rep, 2014, 4: 5105 CrossRef PubMed ADS Google Scholar

[35] Mandal P.K., Ferreira L.M.R., Collins R., Meissner T.B., Boutwell C.L., Friesen M., Vrbanac V., Garrison B.S., Stortchevoi A., Bryder D., Musunuru K., Brand H., Tager A.M., Allen T.M., Talkowski M.E., Rossi D.J., Cowan C.A.. Efficient ablation of genes in human hematopoietic stem and effector cells using CRISPR/Cas9. Cell Stem Cell, 2014, 15: 643-652 CrossRef PubMed Google Scholar

[36] Maruyama T., Dougan S.K., Truttmann M.C., Bilate A.M., Ingram J.R., Ploegh H.L.. Increasing the efficiency of precise genome editing with CRISPR-Cas9 by inhibition of nonhomologous end joining. Nat Biotechnol, 2015, 33: 538-542 CrossRef PubMed Google Scholar

[37] Mentis A.F.. Epigenomic engineering for Down syndrome. Neurosci Biobehav Rev, 2016, 71: 323-327 CrossRef PubMed Google Scholar

[38] Munshi N.V.. CRISPR (clustered regularly interspaced palindromic repeat)/Cas9 system. Circulation, 2016, 134: 777-779 CrossRef PubMed Google Scholar

[39] Nelson C.E., Hakim C.H., Ousterout D.G., Thakore P.I., Moreb E.A., Castellanos Rivera R.M., Madhavan S., Pan X., Ran F.A., Yan W.X., Asokan A., Zhang F., Duan D., Gersbach C.A.. In vivo genome editing improves muscle function in a mouse model of Duchenne muscular dystrophy. Science, 2016, 351: 403-407 CrossRef PubMed ADS Google Scholar

[40] Nishimasu H., Ran F.A., Hsu P.D., Konermann S., Shehata S.I., Dohmae N., Ishitani R., Zhang F., Nureki O.. Crystal structure of Cas9 in complex with guide RNA and target DNA. Cell, 2014, 156: 935-949 CrossRef PubMed Google Scholar

[41] Niu Y., Shen B., Cui Y., Chen Y., Wang J., Wang L., Kang Y., Zhao X., Si W., Li W., Xiang A.P., Zhou J., Guo X., Bi Y., Si C., Hu B., Dong G., Wang H., Zhou Z., Li T., Tan T., Pu X., Wang F., Ji S., Zhou Q., Huang X., Ji W., Sha J.. Generation of gene-modified cynomolgus monkey via Cas9/RNA-mediated gene targeting in one-cell embryos. Cell, 2014, 156: 836-843 CrossRef PubMed Google Scholar

[42] Osakabe Y., Watanabe T., Sugano S.S., Ueta R., Ishihara R., Shinozaki K., Osakabe K.. Optimization of CRISPR/Cas9 genome editing to modify abiotic stress responses in plants. Sci Rep, 2016, 6: 26685 CrossRef PubMed ADS Google Scholar

[43] Perez-Pinera P., Kocak D.D., Vockley C.M., Adler A.F., Kabadi A.M., Polstein L.R., Thakore P.I., Glass K.A., Ousterout D.G., Leong K.W., Guilak F., Crawford G.E., Reddy T.E., Gersbach C.A.. RNA-guided gene activation by CRISPR-Cas9-based transcription factors. Nat Meth, 2013, 10: 973-976 CrossRef PubMed Google Scholar

[44] Ramakrishna S., Kwaku Dad A.B., Beloor J., Gopalappa R., Lee S.K., Kim H.. Gene disruption by cell-penetrating peptide-mediated delivery of Cas9 protein and guide RNA. Genome Res, 2014, 24: 1020-1027 CrossRef PubMed Google Scholar

[45] Ran F.A., Cong L., Yan W.X., Scott D.A., Gootenberg J.S., Kriz A.J., Zetsche B., Shalem O., Wu X., Makarova K.S., Koonin E.V., Sharp P.A., Zhang F.. In vivo genome editing using Staphylococcus aureus Cas9. Nature, 2015, 520: 186-191 CrossRef PubMed ADS Google Scholar

[46] Reardon S.. First CRISPR clinical trial gets green light from US panel. Nature, 2016, : in press doi: 10.1038/nature.2016.20137 CrossRef Google Scholar

[47] Sakuma, T., Masaki, K., Abe-Chayama, H., Mochida, K., Yamamoto, T., and Chayama, K. (2016). Highly multiplexed CRISPR-Cas9-nuclease and Cas9-nickase vectors for inactivation of hepatitis B virus. Genes Cells 21, 1253–1262. Google Scholar

[48] Savi? N., Schwank G.. Advances in therapeutic CRISPR/Cas9 genome editing. Transl Res, 2016, 168: 15-21 CrossRef PubMed Google Scholar

[49] Schumann K., Lin S., Boyer E., Simeonov D.R., Subramaniam M., Gate R.E., Haliburton G.E., Ye C.J., Bluestone J.A., Doudna J.A., Marson A.. Generation of knock-in primary human T cells using Cas9 ribonucleoproteins. Proc Natl Acad Sci USA, 2015, 112: 10437-10442 CrossRef PubMed ADS Google Scholar

[50] Sessions J.W., Skousen C.S., Price K.D., Hanks B.W., Hope S., Alder J.K., Jensen B.D.. CRISPR-Cas9 directed knock-out of a constitutively expressed gene using lance array nanoinjection. SpringerPlus, 2016, 5: 1521 CrossRef PubMed Google Scholar

[51] Shalem O., Sanjana N.E., Hartenian E., Shi X., Scott D.A., Mikkelsen T.S., Heckl D., Ebert B.L., Root D.E., Doench J.G., Zhang F.. Genome-scale CRISPR-Cas9 knockout screening in human cells. Science, 2014, 343: 84-87 CrossRef PubMed ADS Google Scholar

[52] Slaymaker I.M., Gao L., Zetsche B., Scott D.A., Yan W.X., Zhang F.. Rationally engineered Cas9 nucleases with improved specificity. Science, 2016, 351: 84-88 CrossRef PubMed ADS Google Scholar

[53] Su S., Hu B., Shao J., Shen B., Du J., Du Y., Zhou J., Yu L., Zhang L., Chen F., Sha H., Cheng L., Meng F., Zou Z., Huang X., Liu B.. CRISPR-Cas9 mediated efficient PD-1 disruption on human primary T cells from cancer patients. Sci Rep, 2016, 6: 20070 CrossRef PubMed ADS Google Scholar

[54] Suenaga T., Kohyama M., Hirayasu K., Arase H.. Engineering large viral DNA genomes using the CRISPR-Cas9 system. Microbiol Immunol, 2014, 58: 513-522 CrossRef PubMed Google Scholar

[55] Suresh, B., Ramakrishna, S., and Kim, H. (2017). Cell-penetrating peptide-mediated delivery of Cas9 protein and guide RNA for genome editing. Methods Mol Biol 1507, 81–94. Google Scholar

[56] Swiech, L., Heidenreich, M., Banerjee, A., Habib, N., Li, Y., Trombetta, J., Sur, M., and Zhang, F. (2015). In vivo interrogation of gene function in the mammalian brain using CRISPR-Cas9. Nat Biotechnol 33, 102–106. Google Scholar

[57] Tabebordbar M., Zhu K., Cheng J.K.W., Chew W.L., Widrick J.J., Yan W.X., Maesner C., Wu E.Y., Xiao R., Ran F.A., Cong L., Zhang F., Vandenberghe L.H., Church G.M., Wagers A.J.. In vivo gene editing in dystrophic mouse muscle and muscle stem cells. Science, 2016, 351: 407-411 CrossRef PubMed ADS Google Scholar

[58] Tanihara F., Takemoto T., Kitagawa E., Rao S., Do L.T.K., Onishi A., Yamashita Y., Kosugi C., Suzuki H., Sembon S., Suzuki S., Nakai M., Hashimoto M., Yasue A., Matsuhisa M., Noji S., Fujimura T., Fuchimoto D.I., Otoi T.. Somatic cell reprogramming-free generation of genetically modified pigs. Sci Adv, 2016, 2: e1600803-e1600803 CrossRef PubMed ADS Google Scholar

[59] Tao L., Zhang J., Meraner P., Tovaglieri A., Wu X., Gerhard R., Zhang X., Stallcup W.B., Miao J., He X., Hurdle J.G., Breault D.T., Brass A.L., Dong M.. Frizzled proteins are colonic epithelial receptors for C. difficile toxin B. Nature, 2016, 538: 350-355 CrossRef PubMed ADS Google Scholar

[60] Topalian S.L., Hodi F.S., Brahmer J.R., Gettinger S.N., Smith D.C., McDermott D.F., Powderly J.D., Carvajal R.D., Sosman J.A., Atkins M.B., Leming P.D., Spigel D.R., Antonia S.J., Horn L., Drake C.G., Pardoll D.M., Chen L., Sharfman W.H., Anders R.A., Taube J.M., McMiller T.L., Xu H., Korman A.J., Jure-Kunkel M., Agrawal S., McDonald D., Kollia G.D., Gupta A., Wigginton J.M., Sznol M.. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med, 2012, 366: 2443-2454 CrossRef PubMed Google Scholar

[61] Tóth E., Weinhardt N., Bencsura P., Huszár K., Kulcsár P.I., Tálas A., Fodor E., Welker E.. Cpf1 nucleases demonstrate robust activity to induce DNA modification by exploiting homology directed repair pathways in mammalian cells. Biol Direct, 2016, 11: 46 CrossRef PubMed Google Scholar

[62] Truong D.J.J., Kühner K., Kühn R., Werfel S., Engelhardt S., Wurst W., Ortiz O.. Development of an intein-mediated split-Cas9 system for gene therapy. Nucleic Acids Res, 2015, 43: 6450-6458 CrossRef PubMed Google Scholar

[63] Tsai S.Q., Wyvekens N., Khayter C., Foden J.A., Thapar V., Reyon D., Goodwin M.J., Aryee M.J., Joung J.K.. Dimeric CRISPR RNA-guided Fok I nucleases for highly specific genome editing. Nat Biotechnol, 2014, 32: 569-576 CrossRef PubMed Google Scholar

[64] Urnov F.D., Rebar E.J., Holmes M.C., Zhang H.S., Gregory P.D.. Genome editing with engineered zinc finger nucleases. Nat Rev Genet, 2010, 11: 636-646 CrossRef PubMed Google Scholar

[65] Valletta S., Dolatshad H., Bartenstein M., Yip B.H., Bello E., Gordon S., Yu Y., Shaw J., Roy S., Scifo L., Schuh A., Pellagatti A., Fulga T.A., Verma A., Boultwood J.. ASXL1 mutation correction by CRISPR/Cas9 restores gene function in leukemia cells and increases survival in mouse xenografts. Oncotarget, 2015, 6: 44061-44071 CrossRef PubMed Google Scholar

[66] Wang D., Mou H., Li S., Li Y., Hough S., Tran K., Li J., Yin H., Anderson D.G., Sontheimer E.J., Weng Z., Gao G., Xue W.. Adenovirus-mediated somatic genome editing of Pten by CRISPR/Cas9 in mouse liver in spite of Cas9-specific immune responses. Hum Gene Therapy, 2015, 26: 432-442 CrossRef PubMed Google Scholar

[67] Wang L., Li F., Dang L., Liang C., Wang C., He B., Liu J., Li D., Wu X., Xu X., Lu A., Zhang G.. In vivo delivery systems for therapeutic genome editing. Int J Mol Sci, 2016, 17: 626 CrossRef PubMed Google Scholar

[68] Williams M.R., Fricano-Kugler C.J., Getz S.A., Skelton P.D., Lee J., Rizzuto C.P., Geller J.S., Li M., Luikart B.W.. A retroviral CRISPR-Cas9 system for cellular autism-associated phenotype discovery in developing neurons. Sci Rep, 2016, 6: 25611 CrossRef PubMed ADS Google Scholar

[69] Xie S.L., Bian W.P., Wang C., Junaid M., Zou J.X., Pei D.S.. A novel technique based on in vitro oocyte injection to improve CRISPR/Cas9 gene editing in zebrafish. Sci Rep, 2016, 6: 34555 CrossRef PubMed ADS Google Scholar

[70] Xu L., Park K.H., Zhao L., Xu J., El Refaey M., Gao Y., Zhu H., Ma J., Han R.. CRISPR-mediated genome editing restores dystrophin expression and function in mdx mice. Mol Ther, 2016, 24: 564-569 CrossRef PubMed Google Scholar

[71] Xue W., Chen S., Yin H., Tammela T., Papagiannakopoulos T., Joshi N.S., Cai W., Yang G., Bronson R., Crowley D.G., Zhang F., Anderson D.G., Sharp P.A., Jacks T.. CRISPR-mediated direct mutation of cancer genes in the mouse liver. Nature, 2014, 514: 380-384 CrossRef PubMed ADS Google Scholar

[72] Yang Y., Wang L., Bell P., McMenamin D., He Z., White J., Yu H., Xu C., Morizono H., Musunuru K., Batshaw M.L., Wilson J.M.. A dual AAV system enables the Cas9-mediated correction of a metabolic liver disease in newborn mice. Nat Biotechnol, 2016, 34: 334-338 CrossRef PubMed Google Scholar

[73] Yao S., He Z., Chen C.. CRISPR/Cas9-mediated genome editing of epigenetic factors for cancer therapy. Hum Gene Ther, 2015, 26: 463-471 CrossRef PubMed Google Scholar

[74] Yi L., Li J.. CRISPR-Cas9 therapeutics in cancer: promising strategies and present challenges. Biochim Biophys Acta, 2016, 1866: 197-207 CrossRef PubMed Google Scholar

[75] Yin H., Song C.Q., Dorkin J.R., Zhu L.J., Li Y., Wu Q., Park A., Yang J., Suresh S., Bizhanova A., Gupta A., Bolukbasi M.F., Walsh S., Bogorad R.L., Gao G., Weng Z., Dong Y., Koteliansky V., Wolfe S.A., Langer R., Xue W., Anderson D.G.. Therapeutic genome editing by combined viral and non-viral delivery of CRISPR system components in vivo. Nat Biotechnol, 2016, 34: 328-333 CrossRef PubMed Google Scholar

[76] Yin H., Xue W., Chen S., Bogorad R.L., Benedetti E., Grompe M., Koteliansky V., Sharp P.A., Jacks T., Anderson D.G.. Genome editing with Cas9 in adult mice corrects a disease mutation and phenotype. Nat Biotechnol, 2014, 32: 551-553 CrossRef PubMed Google Scholar

[77] Yuan M., Gao X., Chard L.S., Ali Z., Ahmed J., Li Y., Liu P., Lemoine N.R., Wang Y.. A marker-free system for highly efficient construction of vaccinia virus vectors using CRISPR Cas9. Mol Ther Methods Clin Dev, 2015a, 2: 15035 CrossRef PubMed Google Scholar

[78] Yuan M., Zhang W., Wang J., Al Yaghchi C., Ahmed J., Chard L., Lemoine N.R., Wang Y.. Efficiently editing the vaccinia virus genome by using the CRISPR-Cas9 system. J Virol, 2015b, 89: 5176-5179 CrossRef PubMed Google Scholar

[79] Zhang D., Li J.F.. DNA-guided genome editing tool. Sci China Life Sci, 2016, 59: 740-741 CrossRef PubMed Google Scholar

[80] Zhang T., Yin Y., Liu H., Du W., Ren C., Wang L., Lu H., Zhang Z.. Generation of VDR knock-out mice via zygote injection of CRISPR/Cas9 system. PLoS ONE, 2016, 11: e0163551 CrossRef PubMed ADS Google Scholar

[81] Zhang X., Wang S.. From the first human gene-editing to the birth of three-parent baby. Sci China Life Sci, 2016, 59: 1341-1342 CrossRef PubMed Google Scholar

[82] Zuris J.A., Thompson D.B., Shu Y., Guilinger J.P., Bessen J.L., Hu J.H., Maeder M.L., Joung J.K., Chen Z.Y., Liu D.R.. Cationic lipid-mediated delivery of proteins enables efficient protein-based genome editing in vitro and in vivo. Nat Biotechnol, 2015, 33: 73-80 CrossRef PubMed Google Scholar

  • Figure 1

    Schematic diagram of CRISPR-Cas9-mediated genome editing. Cas9 is guided by an sgRNA to induce a double-strand DNA break (DSB) at a desired genomic locus. The DSB can be repaired by NHEJ causing random insertion or deletion (indel) mutations or by HDR using a donor DNA template, enabling the introduction of desired sequence changes for precise genome editing purposes.

  • Figure 2

    Delivery vectors for CRISPR-Cas9 systems. Human codon-optimized Cas9 and sgRNA sequences were packaged into a viral vector (e.g., adenovirus, rAAV, lentivirus) for genome editing. Cas9 protein, mRNA of Cas9 and sgRNA, or a plasmid encoding Cas9 and sgRNA was incorporated into a nanoparticle to formulate a nano-Cas9 complex for non-viral delivery.

  • Table 1   Non-viral and viral vectors for CRISPR-Cas9 system and their applications in the biomedical field

    Delivery methods

    Advantages

    Disadvantages

    Applications

    Microinjection

    High efficiency in vitro

    Low-throughput

    Genome editing for oocytes or embryos; generation of model animals

    Electroporation

    High transfection efficiency in vitro

    Cytotoxicity, difficult for in vivo use

    Genome editing for various cell types in vitro

    Hydrodynamic injection

    Feasible for in vivo gene editing in small animals

    Low efficiency, difficult for clinical use

    Gene function study in vivo

    CPP

    Low off-target effects

    Low efficiency, immunogenicity, difficult for in vivo use

    Genome editing for cells in vitro

    Cationic vectors

    Easy to produce, large packaging capacity

    Low efficiency

    Genome editing for various cell types in vitro; gene therapy for cancer, HBV, genetic diseases, etc.

    Retrovirus

    High efficiency in vivo, integrating target gene into host cell genome

    Insertional mutagenesis, oncogene activation

    Gene therapy for cancer, genetic diseases, etc.

    Lentivirus

    High efficiency, high throughput in vitro and in vivo

    Prone to rearrangements of cargo genes, liable to transgene silencing

    Genomic screen and gene function study in vitro and in vivo

    Adenovirus

    High efficiency in vivo, high packaging capacity

    Immunoreactivity, difficult to produce in large scale

    Gene therapy for genetic diseases

    AAV

    High efficiency in vivo, non-pathogenic

    Limited packaging capacity, high cost

    Gene therapy for various genetic diseases

Copyright 2019 Science China Press Co., Ltd. 科学大众杂志社有限责任公司 版权所有

京ICP备18024590号-1