Browse > Article
http://dx.doi.org/10.3345/cep.2020.02096

Application of CRISPR-Cas9 gene editing for congenital heart disease  

Seok, Heeyoung (Department of Life Sciences, Korea University)
Deng, Rui (Department of Cardiology, Boston Children's Hospital)
Cowan, Douglas B. (Department of Cardiology, Boston Children's Hospital)
Wang, Da-Zhi (Department of Cardiology, Boston Children's Hospital)
Publication Information
Clinical and Experimental Pediatrics / v.64, no.6, 2021 , pp. 269-279 More about this Journal
Abstract
Clustered regularly interspaced short palindromic repeats and CRISPR-associated protein 9 (CRISPR-Cas9) is an ancient prokaryotic defense system that precisely cuts foreign genomic DNA under the control of a small number of guide RNAs. The CRISPR-Cas9 system facilitates efficient double-stranded DNA cleavage that has been recently adopted for genome editing to create or correct inherited genetic mutations causing disease. Congenital heart disease (CHD) is generally caused by genetic mutations such as base substitutions, deletions, and insertions, which result in diverse developmental defects and remains a leading cause of birth defects. Pediatric CHD patients exhibit a spectrum of cardiac abnormalities such as septal defects, valvular defects, and abnormal chamber development. CHD onset occurs during the prenatal period and often results in early lethality during childhood. Because CRISPR-Cas9-based genome editing technology has gained considerable attention for its potential to prevent and treat diseases, we will review the CRISPR-Cas9 system as a genome editing tool and focus on its therapeutic application for CHD.
Keywords
CRISPR; Genome editing; Congenital heart diseases; Childhood onset;
Citations & Related Records
연도 인용수 순위
  • Reference
1 Fujiwara T, Takeda N, Hara H, Morita H, Kishihara J, Inuzuka R, et al. Distinct variants affecting differential splicing of TGFBR1 exon 5 cause either Loeys-Dietz syndrome or multiple self-healing squamous epithelioma. Eur J Hum Genet 2018;26:1151-8.   DOI
2 Thomas KR, Folger KR, Capecchi MR. High frequency targeting of genes to specific sites in the mammalian genome. Cell 1986;44:419-28.   DOI
3 Kelly TJ Jr, Smith HO. A restriction enzyme from Hemophilus influenzae. II. J Mol Biol 1970;51:393-409.   DOI
4 Basson CT, Bachinsky DR, Lin RC, Levi T, Elkins JA, Soults J, et al. Mutations in human TBX5 [corrected] cause limb and cardiac malformation in Holt-Oram syndrome. Nat Genet 1997;15:30-5.   DOI
5 Tartaglia M, Kalidas K, Shaw A, Song X, Musat DL, van der Burgt I, et al. PTPN11 mutations in Noonan syndrome: molecular spectrum, genotype-phenotype correlation, and phenotypic heterogeneity. Am J Hum Genet 2002;70:1555-63.   DOI
6 Ko JM, Kim JM, Kim GH, Yoo HW. PTPN11, SOS1, KRAS, and RAF1 gene analysis, and genotype-phenotype correlation in Korean patients with Noonan syndrome. J Hum Genet 2008;53:999-1006.   DOI
7 Mugikura SI, Katoh A, Watanabe S, Kimura M, Kajiwara K. Abnormal gait, reduced locomotor activity and impaired motor coordination in Dgcr2-deficient mice. Biochem Biophys Rep 2016;5:120-6.   DOI
8 Finsterer J, Stollberger C, Towbin JA. Left ventricular noncompaction cardiomyopathy: cardiac, neuromuscular, and genetic factors. Nat Rev Cardiol 2017;14:224-37.   DOI
9 Razmara E, Garshasbi M. Whole-exome sequencing identifies R1279X of MYH6 gene to be associated with congenital heart disease. BMC Cardiovasc Disord 2018;18:137.   DOI
10 Dietz HC, Cutting GR, Pyeritz RE, Maslen CL, Sakai LY, Corson GM, et al. Marfan syndrome caused by a recurrent de novo missense mutation in the fibrillin gene. Nature 1991;352:337-9.   DOI
11 Maeder ML, Stefanidakis M, Wilson CJ, Baral R, Barrera LA, Bounoutas GS, et al. Development of a gene-editing approach to restore vision loss in Leber congenital amaurosis type 10. Nat Med 2019;25:229-33.   DOI
12 Trembley MA, Velasquez LS, de Mesy Bentley KL, Small EM. Myocardin-related transcription factors control the motility of epicardium-derived cells and the maturation of coronary vessels. Development 2015; 142:21-30.   DOI
13 Basson CT, Huang T, Lin RC, Bachinsky DR, Weremowicz S, Vaglio A, et al. Different TBX5 interactions in heart and limb defined by Holt-Oram syndrome mutations. Proc Natl Acad Sci U S A 1999;96:2919-24.   DOI
14 Garrity DM, Childs S, Fishman MC. The heartstrings mutation in zebrafish causes heart/fin Tbx5 deficiency syndrome. Development 2002;129:4635-45.   DOI
15 Fire A, Xu S, Montgomery MK, Kostas SA, Driver SE, Mello CC. Potent and specific genetic interference by double-stranded RNA in Caenorhabditis elegans. Nature 1998;391:806-11.   DOI
16 Lyons I, Parsons LM, Hartley L, Li R, Andrews JE, Robb L, et al. Myogenic and morphogenetic defects in the heart tubes of murine embryos lacking the homeo box gene Nkx2-5. Genes Dev 1995;9:1654-66.   DOI
17 Carniel E, Taylor MR, Sinagra G, Di Lenarda A, Ku L, Fain PR, et al. Alpha-myosin heavy chain: a sarcomeric gene associated with dilated and hypertrophic phenotypes of cardiomyopathy. Circulation 2005;112:54-9.   DOI
18 Posch MG, Waldmuller S, Muller M, Scheffold T, Fournier D, Andrade-Navarro MA, et al. Cardiac alpha-myosin (MYH6) is the predominant sarcomeric disease gene for familial atrial septal defects. PLoS One 2011;6:e28872.   DOI
19 Hanses U, Kleinsorge M, Roos L, Yigit G, Li Y, Barbarics B, et al. Intronic CRISPR repair in a preclinical model of noonan syndrome-associated cardiomyopathy. Circulation 2020;142:1059-76.   DOI
20 Rand TA, Petersen S, Du F, Wang X. Argonaute2 cleaves the anti-guide strand of siRNA during RISC activation. Cell 2005;123:621-9.   DOI
21 Pierpont ME, Digilio MC. Cardiovascular disease in Noonan syndrome. Curr Opin Pediatr 2018;30:601-8.   DOI
22 Cirino A, Aurigemma I, Franzese M, Lania G, Righelli D, Ferrentino R, et al. Chromatin and transcriptional response to loss of TBX1 in early differentiation of mouse cells. Front Cell Dev Biol 2020;8:571501.   DOI
23 Watanabe S, Sakurai T, Nakamura S, Miyoshi K, Sato M. The combinational use of CRISPR/Cas9 and targeted toxin technology enables efficient isolation of bi-allelic knockout non-human mammalian clones. Int J Mol Sci 2018;19:1075.   DOI
24 Molinard-Chenu A, Dayer A. The candidate schizophrenia risk gene DGCR2 regulates early steps of corticogenesis. Biol Psychiatry 2018; 83:692-706.   DOI
25 Garg V, Kathiriya IS, Barnes R, Schluterman MK, King IN, Butler CA, et al. GATA4 mutations cause human congenital heart defects and reveal an interaction with TBX5. Nature 2003;424:443-7.   DOI
26 Hirayama-Yamada K, Kamisago M, Akimoto K, Aotsuka H, Nakamura Y, Tomita H, et al. Phenotypes with GATA4 or NKX2.5 mutations in familial atrial septal defect. Am J Med Genet A 2005;135:47-52.
27 Gifford CA, Ranade SS, Samarakoon R, Salunga HT, de Soysa TY, Huang Y, et al. Oligogenic inheritance of a human heart disease involving a genetic modifier. Science 2019;364:865-70.   DOI
28 Stainier DYR, Raz E, Lawson ND, Ekker SC, Burdine RD, Eisen JS, et al. Guidelines for morpholino use in zebrafish. PLoS Genet 2017;13:e1007000.   DOI
29 Nyboe C, Olsen MS, Nielsen-Kudsk JE, Hjortdal VE. Atrial fibrillation and stroke in adult patients with atrial septal defect and the long-term effect of closure. Heart 2015;101:706-11.   DOI
30 Ching YH, Ghosh TK, Cross SJ, Packham EA, Honeyman L, Loughna S, et al. Mutation in myosin heavy chain 6 causes atrial septal defect. Nat Genet 2005;37:423-8.   DOI
31 Boyle Anderson EAT, Ho RK. A transcriptomics analysis of the Tbx5 paralogues in zebrafish. PLoS One 2018;13:e0208766.   DOI
32 Zhu L, Belmont JW, Ware SM. Genetics of human heterotaxias. Eur J Hum Genet 2006;14:17-25.   DOI
33 Li S, Liu S, Chen W, Yuan Y, Gu R, Song Y, et al. A novel ZIC3 gene mutation identified in patients with heterotaxy and congenital heart disease. Sci Rep 2018;8:12386.   DOI
34 Macrae IJ, Zhou K, Li F, Repic A, Brooks AN, Cande WZ, et al. Structural basis for double-stranded RNA processing by Dicer. Science 2006;311:195-8.   DOI
35 Wang S, Li Y, Xu Y, Ma Q, Lin Z, Schlame M, et al. AAV gene therapy prevents and reverses heart failure in a murine knockout model of barth syndrome. Circ Res 2020;126:1024-39.   DOI
36 Schott JJ, Benson DW, Basson CT, Pease W, Silberbach GM, Moak JP, et al. Congenital heart disease caused by mutations in the transcription factor NKX2-5. Science 1998;281:108-11.   DOI
37 Haaning AM, Quinn ME, Ware SM. Heterotaxy-spectrum heart defects in Zic3 hypomorphic mice. Pediatr Res 2013;74:494-502.   DOI
38 Houtmeyers R, Souopgui J, Tejpar S, Arkell R. The ZIC gene family encodes multi-functional proteins essential for patterning and morphogenesis. Cell Mol Life Sci 2013;70:3791-811.   DOI
39 Paulussen AD, Steyls A, Vanoevelen J, van Tienen FH, Krapels IP, Claes GR, et al. Rare novel variants in the ZIC3 gene cause X-linked heterotaxy. Eur J Hum Genet 2016;24:1783-91.   DOI
40 Gasiunas G, Barrangou R, Horvath P, Siksnys V. Cas9-crRNA ribonucleoprotein complex mediates specific DNA cleavage for adaptive immunity in bacteria. Proc Natl Acad Sci U S A 2012;109:E2579-86.
41 Nishimasu H, Ran FA, Hsu PD, Konermann S, Shehata SI, Dohmae N, et al. Crystal structure of Cas9 in complex with guide RNA and target DNA. Cell 2014;156:935-49.   DOI
42 Garneau JE, Dupuis ME, Villion M, Romero DA, Barrangou R, Boyaval P, et al. The CRISPR/Cas bacterial immune system cleaves bacteriophage and plasmid DNA. Nature 2010;468:67-71.   DOI
43 Hale CR, Zhao P, Olson S, Duff MO, Graveley BR, Wells L, et al. RNA-guided RNA cleavage by a CRISPR RNA-Cas protein complex. Cell 2009;139:945-56.   DOI
44 Adli M. The CRISPR tool kit for genome editing and beyond. Nat Commun 2018;9:1911.   DOI
45 Smith HO, Wilcox KW. A restriction enzyme from Hemophilus influenzae. I. Purification and general properties. J Mol Biol 1970;51:379-91.   DOI
46 Vermersch E, Jouve C, Hulot JS. CRISPR/Cas9 gene-editing strategies in cardiovascular cells. Cardiovasc Res 2020;116:894-907.   DOI
47 Nguyen Q, Lim KRQ, Yokota T. Genome editing for the understanding and treatment of inherited cardiomyopathies. Int J Mol Sci 2020;21:733.   DOI
48 Smithies O, Gregg RG, Boggs SS, Koralewski MA, Kucherlapati RS. Insertion of DNA sequences into the human chromosomal beta-globin locus by homologous recombination. Nature 1985;317:230-4.   DOI
49 Capecchi MR. Altering the genome by homologous recombination. Science 1989;244:1288-92.   DOI
50 Lin FL, Sperle K, Sternberg N. Recombination in mouse L cells between DNA introduced into cells and homologous chromosomal sequences. Proc Natl Acad Sci U S A 1985;82:1391-5.   DOI
51 Theis JL, Zimmermann MT, Evans JM, Eckloff BW, Wieben ED, Qureshi MY, et al. Recessive MYH6 mutations in hypoplastic left heart with reduced ejection fraction. Circ Cardiovasc Genet 2015;8:564-71.   DOI
52 Tomita-Mitchell A, Stamm KD, Mahnke DK, Kim MS, Hidestrand PM, Liang HL, et al. Impact of MYH6 variants in hypoplastic left heart syndrome. Physiol Genomics 2016;48:912-21.   DOI
53 Granados-Riveron JT, Ghosh TK, Pope M, Bu'Lock F, Thornborough C, Eason J, et al. Alpha-cardiac myosin heavy chain (MYH6) mutations affecting myofibril formation are associated with congenital heart defects. Hum Mol Genet 2010;19:4007-16.   DOI
54 Ogasawara T, Shiba Y. iPS cells as a source of cardiac regeneration. Nihon Rinsho 2016;74 Suppl 6:287-92.
55 Carroll KJ, Makarewich CA, McAnally J, Anderson DM, Zentilin L, Liu N, et al. A mouse model for adult cardiac-specific gene deletion with CRISPR/Cas9. Proc Natl Acad Sci U S A 2016;113:338-43.   DOI
56 Guo Y, VanDusen NJ, Zhang L, Gu W, Sethi I, Guatimosim S, et al. Analysis of cardiac myocyte maturation using CASAAV, a platform for rapid dissection of cardiac myocyte gene function in vivo. Circ Res 2017; 120:1874-88.   DOI
57 Guo Y, Jardin BD, Zhou P, Sethi I, Akerberg BN, Toepfer CN, et al. Hierarchical and stage-specific regulation of murine cardiomyocyte maturation by serum response factor. Nat Commun 2018;9:3837.   DOI
58 Rikhtegar R, Pezeshkian M, Dolati S, Safaie N, Afrasiabi Rad A, Mahdipour M, et al. Stem cells as therapy for heart disease: iPSCs, ESCs, CSCs, and skeletal myoblasts. Biomed Pharmacother 2019;109:304-13.   DOI
59 Martins AM, Vunjak-Novakovic G, Reis RL. The current status of iPS cells in cardiac research and their potential for tissue engineering and regenerative medicine. Stem Cell Rev Rep 2014;10:177-90.   DOI
60 Alankarage D, Szot JO, Pachter N, Slavotinek A, Selleri L, Shieh JT, et al. Functional characterization of a novel PBX1 de novo missense variant identified in a patient with syndromic congenital heart disease. Hum Mol Genet 2020;29:1068-82.   DOI
61 Wang G, McCain ML, Yang L, He A, Pasqualini FS, Agarwal A, et al. Modeling the mitochondrial cardiomyopathy of Barth syndrome with induced pluripotent stem cell and heart-on-chip technologies. Nat Med 2014;20:616-23.   DOI
62 Liu C, Cao R, Xu Y, Li T, Li F, Chen S, et al. Rare copy number variants analysis identifies novel candidate genes in heterotaxy syndrome patients with congenital heart defects. Genome Med 2018;10:40.   DOI
63 Sarkozy A, Carta C, Moretti S, Zampino G, Digilio MC, Pantaleoni F, et al. Germline BRAF mutations in Noonan, LEOPARD, and cardiofaciocutaneous syndromes: molecular diversity and associated phenotypic spectrum. Hum Mutat 2009;30:695-702.   DOI
64 Gripp KW, Lin AE. Costello syndrome: a Ras/mitogen activated protein kinase pathway syndrome (rasopathy) resulting from HRAS germline mutations. Genet Med 2012;14:285-92.   DOI
65 Gripp KW, Hopkins E, Sol-Church K, Stabley DL, Axelrad ME, Doyle D, et al. Phenotypic analysis of individuals with Costello syndrome due to HRAS p.G13C. Am J Med Genet A 2011;155A:706-16.
66 Martinez-Quintana E, Rodriguez-Gonzalez F. LEOPARD syndrome: clinical features and gene mutations. Mol Syndromol 2012;3:145-57.   DOI
67 Olen MM, Baysa SJ, Rossi A, Kanter RJ, Fishberger SB. Wolff-Parkinson-White Syndrome: a stepwise deterioration to sudden death. Circulation 2016;133:105-6.   DOI
68 van der Ven JPG, van den Bosch E, Bogers A, Helbing WA. Current outcomes and treatment of tetralogy of Fallot. F1000Res 2019;8:F1000 Faculty Rev-1530.
69 Fischetto R, Palmieri VV, Tripaldi ME, Gaeta A, Michelucci A, Delvecchio M, et al. Alagille syndrome: a Novel mutation in JAG1 gene. Front Pediatr 2019;7:199.   DOI
70 McDaniell R, Warthen DM, Sanchez-Lara PA, Pai A, Krantz ID, Piccoli DA, et al. NOTCH2 mutations cause Alagille syndrome, a heterogeneous disorder of the notch signaling pathway. Am J Hum Genet 2006;79:169-73.   DOI
71 Munger TM, Packer DL, Hammill SC, Feldman BJ, Bailey KR, Ballard DJ, et al. A population study of the natural history of Wolff-ParkinsonWhite syndrome in Olmsted County, Minnesota, 1953-1989. Circulation 1993;87:866-73.   DOI
72 Xie C, Zhang YP, Song L, Luo J, Qi W, Hu J, et al. Genome editing with CRISPR/Cas9 in postnatal mice corrects PRKAG2 cardiac syndrome. Cell Res 2016;26:1099-111.   DOI
73 Hoffman EP, Brown RH Jr, Kunkel LM. Dystrophin: the protein product of the Duchenne muscular dystrophy locus. Cell 1987;51:919-28.   DOI
74 Klug A, Rhodes D. Zinc fingers: a novel protein fold for nucleic acid recognition. Cold Spring Harb Symp Quant Biol 1987;52:473-82.   DOI
75 Hoffman JI, Kaplan S. The incidence of congenital heart disease. J Am Coll Cardiol 2002;39:1890-900.   DOI
76 Collaborators GBDCHD. Global, regional, and national burden of congenital heart disease, 1990-2017: a systematic analysis for the Global Burden of Disease Study 2017. Lancet Child Adolesc Health 2020;4:185-200.   DOI
77 Bruneau BG. The developmental genetics of congenital heart disease. Nature 2008;451:943-8.   DOI
78 Rouet P, Smih F, Jasin M. Introduction of double-strand breaks into the genome of mouse cells by expression of a rare-cutting endonuclease. Mol Cell Biol 1994;14:8096-106.   DOI
79 Jeggo PA. DNA breakage and repair. Adv Genet 1998;38:185-218.   DOI
80 Kim YG, Cha J, Chandrasegaran S. Hybrid restriction enzymes: zinc finger fusions to Fok I cleavage domain. Proc Natl Acad Sci U S A 1996; 93:1156-60.   DOI
81 Smith AT, Sack GH Jr, Taylor GJ. Holt-Oram syndrome. J Pediatr 1979; 95:538-43.   DOI
82 Verhaart IEC, Aartsma-Rus A. Therapeutic developments for Duchenne muscular dystrophy. Nat Rev Neurol 2019;15:373-86.   DOI
83 Long C, McAnally JR, Shelton JM, Mireault AA, Bassel-Duby R, Olson EN. Prevention of muscular dystrophy in mice by CRISPR/Cas9-mediated editing of germline DNA. Science 2014;345:1184-8.   DOI
84 El Refaey M, Xu L, Gao Y, Canan BD, Adesanya TMA, Warner SC, et al. In vivo genome editing restores dystrophin expression and cardiac function in dystrophic mice. Circ Res 2017;121:923-9.   DOI
85 Oda T, Elkahloun AG, Pike BL, Okajima K, Krantz ID, Genin A, et al. Mutations in the human Jagged1 gene are responsible for Alagille syndrome. Nat Genet 1997;16:235-42.   DOI
86 Vanlerberghe C, Jourdain AS, Ghoumid J, Frenois F, Mezel A, Vaksmann G, et al. Holt-Oram syndrome: clinical and molecular description of 78 patients with TBX5 variants. Eur J Hum Genet 2019;27:360-8.   DOI
87 Ewart AK, Morris CA, Atkinson D, Jin W, Sternes K, Spallone P, et al. Hemizygosity at the elastin locus in a developmental disorder, Williams syndrome. Nat Genet 1993;5:11-6.   DOI
88 Lindsay EA, Botta A, Jurecic V, Carattini-Rivera S, Cheah YC, Rosenblatt HM, et al. Congenital heart disease in mice deficient for the DiGeorge syndrome region. Nature 1999;401:379-83.   DOI
89 International HapMap C. The International HapMap Project. Nature 2003;426:789-96.   DOI
90 Wiedenheft B, Sternberg SH, Doudna JA. RNA-guided genetic silencing systems in bacteria and archaea. Nature 2012;482:331-8.   DOI
91 van Kampen SJ, van Rooij E. CRISPR craze to transform cardiac biology. Trends Mol Med 2019;25:791-802.   DOI
92 Ishino Y, Shinagawa H, Makino K, Amemura M, Nakata A. Nucleotide sequence of the iap gene, responsible for alkaline phosphatase isozyme conversion in Escherichia coli, and identification of the gene product. J Bacteriol 1987;169:5429-33.   DOI
93 Brussow H. Phages of dairy bacteria. Annu Rev Microbiol 2001;55:283-303.   DOI
94 Scambler PJ. The 22q11 deletion syndromes. Hum Mol Genet 2000;9:2421-6.   DOI
95 Lee KJ, Choi H, Choi WH, Kwon SU, Doh JH, Namgung J, et al. The management of cardiovascular abnormalities in patient with LEOPARD syndrome. Korean Circ J 2010;40:339-42.   DOI
96 Zeng Y, Li J, Li G, Huang S, Yu W, Zhang Y, et al. Correction of the Marfan syndrome pathogenic FBN1 mutation by base editing in human cells and heterozygous embryos. Mol Ther 2018;26:2631-7.   DOI
97 Tartaglia M, Mehler EL, Goldberg R, Zampino G, Brunner HG, Kremer H, et al. Mutations in PTPN11, encoding the protein tyrosine phosphatase SHP-2, cause Noonan syndrome. Nat Genet 2001;29:465-8.   DOI
98 Pandit B, Sarkozy A, Pennacchio LA, Carta C, Oishi K, Martinelli S, et al. Gain-of-function RAF1 mutations cause Noonan and LEOPARD syndromes with hypertrophic cardiomyopathy. Nat Genet 2007;39: 1007-12.   DOI
99 Ledford H. CRISPR treatment inserted directly into the body for first time. Nature 2020;579:185.   DOI
100 Basson CT, Cowley GS, Solomon SD, Weissman B, Poznanski AK, Traill TA, et al. The clinical and genetic spectrum of the Holt-Oram syndrome (heart-hand syndrome). N Engl J Med 1994;330:885-91.   DOI
101 Bernstein E, Caudy AA, Hammond SM, Hannon GJ. Role for a bidentate ribonuclease in the initiation step of RNA interference. Nature 2001;409:363-6.   DOI
102 Jiang F, Doudna JA. CRISPR-Cas9 structures and mechanisms. Annu Rev Biophys 2017;46:505-29.   DOI
103 Koonin EV, Makarova KS, Zhang F. Diversity, classification and evolution of CRISPR-Cas systems. Curr Opin Microbiol 2017;37:67-78.   DOI
104 Tomari Y, Matranga C, Haley B, Martinez N, Zamore PD. A protein sensor for siRNA asymmetry. Science 2004;306:1377-80.   DOI
105 Heler R, Samai P, Modell JW, Weiner C, Goldberg GW, Bikard D, et al. Cas9 specifies functional viral targets during CRISPR-Cas adaptation. Nature 2015;519:199-202.   DOI
106 Chen H, Choi J, Bailey S. Cut site selection by the two nuclease domains of the Cas9 RNA-guided endonuclease. J Biol Chem 2014;289:13284-94.   DOI
107 Mojica FJM, Montoliu L. On the origin of CRISPR-Cas technology: from prokaryotes to mammals. Trends Microbiol 2016;24:811-20.   DOI
108 Moerman P, Goddeeris P, Lauwerijns J, Van der Hauwaert LG. Cardiovascular malformations in DiGeorge syndrome (congenital absence of hypoplasia of the thymus). Br Heart J 1980;44:452-9.   DOI
109 Redon R, Ishikawa S, Fitch KR, Feuk L, Perry GH, Andrews TD, et al. Global variation in copy number in the human genome. Nature 2006;444:444-54.   DOI
110 Gaj T, Gersbach CA, Barbas CF 3rd. ZFN, TALEN, and CRISPR/Cas-based methods for genome engineering. Trends Biotechnol 2013;31:397-405.   DOI
111 Jackson SA, McKenzie RE, Fagerlund RD, Kieper SN, Fineran PC, Brouns SJ. CRISPR-Cas: adapting to change. Science 2017;356:eaal5056.   DOI
112 Barrangou R. Diversity of CRISPR-Cas immune systems and molecular machines. Genome Biol 2015;16:247.   DOI
113 Mali P, Yang L, Esvelt KM, Aach J, Guell M, DiCarlo JE, et al. RNA-guided human genome engineering via Cas9. Science 2013;339:823-6.   DOI
114 Sulu A, Baspinar O, Sahin DA. Giant aortic aneurysm due to fibulin- 4 deficiency: case series. Turk Pediatri Ars 2019;54:119-24.
115 Mojica FJ, Juez G, Rodriguez-Valera F. Transcription at different salinities of Haloferax mediterranei sequences adjacent to partially modified PstI sites. Mol Microbiol 1993;9:613-21.   DOI
116 Barrangou R, Fremaux C, Deveau H, Richards M, Boyaval P, Moineau S, et al. CRISPR provides acquired resistance against viruses in prokaryotes. Science 2007;315:1709-12.   DOI
117 Mojica FJM, Diez-Villasenor C, Garcia-Martinez J, Almendros C. Short motif sequences determine the targets of the prokaryotic CRISPR defence system. Microbiology (Reading) 2009;155(Pt 3):733-40.   DOI
118 Andersson ER, Chivukula IV, Hankeova S, Sjoqvist M, Tsoi YL, Ramskold D, et al. Mouse model of alagille syndrome and mechanisms of Jagged1 missense mutations. Gastroenterology 2018;154:1080-95.   DOI
119 Westra ER, van Houte S, Gandon S, Whitaker R. The ecology and evolution of microbial CRISPR-Cas adaptive immune systems. Philos Trans R Soc Lond B Biol Sci 2019;374:20190101.   DOI
120 Richards AA, Garg V. Genetics of congenital heart disease. Curr Cardiol Rev 2010;6:91-7.   DOI
121 Motta BM, Pramstaller PP, Hicks AA, Rossini A. The impact of CRISPR/Cas9 technology on cardiac research: from disease modelling to therapeutic approaches. Stem Cells Int 2017;2017:8960236.
122 Li B, Niu Y, Ji W, Dong Y. Strategies for the CRISPR-based therapeutics. Trends Pharmacol Sci 2020;41:55-65.   DOI
123 Moscou MJ, Bogdanove AJ. A simple cipher governs DNA recognition by TAL effectors. Science 2009;326:1501.   DOI
124 Porteus MH, Baltimore D. Chimeric nucleases stimulate gene targeting in human cells. Science 2003;300:763.   DOI
125 Urnov FD, Miller JC, Lee YL, Beausejour CM, Rock JM, Augustus S, et al. Highly efficient endogenous human gene correction using designed zincfinger nucleases. Nature 2005;435:646-51.   DOI
126 Boch J, Scholze H, Schornack S, Landgraf A, Hahn S, Kay S, et al. Breaking the code of DNA binding specificity of TAL-type III effectors. Science 2009;326:1509-12.   DOI
127 Zhang F, Cong L, Lodato S, Kosuri S, Church GM, Arlotta P. Efficient construction of sequence-specific TAL effectors for modulating mammalian transcription. Nat Biotechnol 2011;29:149-53.   DOI
128 Miller JC, Tan S, Qiao G, Barlow KA, Wang J, Xia DF, et al. A TALE nuclease architecture for efficient genome editing. Nat Biotechnol 2011; 29:143-8.   DOI
129 Porteus M. Genome editing: a new approach to human therapeutics. Annu Rev Pharmacol Toxicol 2016;56:163-90.   DOI
130 Miller JC, Holmes MC, Wang J, Guschin DY, Lee YL, Rupniewski I, et al. An improved zinc-finger nuclease architecture for highly specific genome editing. Nat Biotechnol 2007;25:778-85.   DOI
131 Bhaya D, Davison M, Barrangou R. CRISPR-Cas systems in bacteria and archaea: versatile small RNAs for adaptive defense and regulation. Annu Rev Genet 2011;45:273-97.   DOI
132 Wright AV, Nunez JK, Doudna JA. Biology and applications of CRISPR systems: Harnessing Nature's toolbox for genome engineering. Cell 2016;164:29-44.   DOI
133 Leenay RT, Maksimchuk KR, Slotkowski RA, Agrawal RN, Gomaa AA, Briner AE, et al. Identifying and visualizing functional PAM diversity across CRISPR-Cas systems. Mol Cell 2016;62:137-47.   DOI
134 Anders C, Niewoehner O, Duerst A, Jinek M. Structural basis of PAM-dependent target DNA recognition by the Cas9 endonuclease. Nature 2014;513:569-73.   DOI
135 Makarova KS, Grishin NV, Shabalina SA, Wolf YI, Koonin EV. A putative RNA-interference-based immune system in prokaryotes: computational analysis of the predicted enzymatic machinery, functional analogies with eukaryotic RNAi, and hypothetical mechanisms of action. Biol Direct 2006;1:7.   DOI
136 Deltcheva E, Chylinski K, Sharma CM, Gonzales K, Chao Y, Pirzada ZA, et al. CRISPR RNA maturation by trans-encoded small RNA and host factor RNase III. Nature 2011;471:602-7.   DOI
137 Brouns SJ, Jore MM, Lundgren M, Westra ER, Slijkhuis RJ, Snijders AP, et al. Small CRISPR RNAs guide antiviral defense in prokaryotes. Science 2008;321:960-4.   DOI
138 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;337:816-21.   DOI
139 Cong L, Ran FA, Cox D, Lin S, Barretto R, Habib N, et al. Multiplex genome engineering using CRISPR/Cas systems. Science 2013;339:819-23.   DOI
140 Spencer CT, Bryant RM, Day J, Gonzalez IL, Colan SD, Thompson WR, et al. Cardiac and clinical phenotype in Barth syndrome. Pediatrics 2006;118:e337-46.
141 Kim J, Cho SY, Yang A, Jang JH, Choi Y, Lee JE, et al. An atypical case of Noonan syndrome with KRAS mutation diagnosed by targeted exome sequencing. Ann Pediatr Endocrinol Metab 2017;22:203-7.   DOI
142 Wang Y, Li X, Li R, Yang Y, Du J. Identification of novel causal FBN1 mutations in pedigrees of Marfan syndrome. Int J Genomics 2018;2018:1246516.   DOI
143 Lin J, Vora M, Kane NS, Gleason RJ, Padgett RW. Human Marfan and Marfan-like Syndrome associated mutations lead to altered trafficking of the type II TGFbeta receptor in Caenorhabditis elegans. PLoS One 2019;14:e0216628.   DOI
144 Baltimore D, Berg P, Botchan M, Carroll D, Charo RA, Church G, et al. Biotechnology. A prudent path forward for genomic engineering and germline gene modification. Science 2015;348:36-8.   DOI
145 Bibikova M, Carroll D, Segal DJ, Trautman JK, Smith J, Kim YG, et al. Stimulation of homologous recombination through targeted cleavage by chimeric nucleases. Mol Cell Biol 2001;21:289-97.   DOI
146 Weinstock DM, Richardson CA, Elliott B, Jasin M. Modeling oncogenic translocations: distinct roles for double-strand break repair pathways in translocation formation in mammalian cells. DNA Repair (Amst) 2006;5:1065-74.   DOI
147 Ma H, Marti-Gutierrez N, Park SW, Wu J, Lee Y, Suzuki K, et al. Correction of a pathogenic gene mutation in human embryos. Nature 2017;548:413-9.   DOI
148 Rios-Serna LJ, Diaz-Ordonez L, Candelo E, Pachajoa H. A novel de novo TBX5 mutation in a patient with Holt-Oram syndrome. Appl Clin Genet 2018;11:157-62.   DOI
149 Bamford RN, Roessler E, Burdine RD, Saplakoglu U, dela Cruz J, Splitt M, et al. Loss-of-function mutations in the EGF-CFC gene CFC1 are associated with human left-right laterality defects. Nat Genet 2000;26: 365-9.   DOI
150 Athota JP, Bhat M, Nampoothiri S, Gowrishankar K, Narayanachar SG, Puttamallesh V, et al. Molecular and clinical studies in 107 Noonan syndrome affected individuals with PTPN11 mutations. BMC Med Genet 2020;21:50.   DOI
151 Guo Q, Shen J, Liu Y, Pu T, Sun K, Chen S. Exome sequencing identifies a c.148-1G>C mutation of TBX5 in a Holt-Oram family with unusual genotype-phenotype correlations. Cell Physiol Biochem 2015;37:1066-74.   DOI