open access

Vol 58, No 3 (2020)
Review paper
Submitted: 2020-07-22
Accepted: 2020-09-17
Published online: 2020-09-24
Get Citation

An overview of applications of CRISPR-Cas technologies in biomedical engineering

Saleh Jamehdor123, Kasra Arbabi Zaboli4, Sina Naserian56, Jose Thekkiniath7, Honey Alef Omidy8, Ali Teimoori9, Behrooz Johari10, Amir Hossein Taromchi11, Yu Sasano12, Saeed Kaboli13
·
Pubmed: 32978771
·
Folia Histochem Cytobiol 2020;58(3):163-173.
Affiliations
  1. Department of Virology, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
  2. Department of Molecular Medicine, Institute of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
  3. Department of Biology, Faculty of Sciences, University of Sistan and Baluchestan, Zahedan, Iran
  4. Department of Medical Biotechnology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
  5. INSERM UMR-S-MD 1197/Ministry of the Armed Forces, Biomedical Research Institute of the Armed Forces (IRBA), Paul-Brousse Hospital Villejuif, and CTSA Clamart, France
  6. SivanCell, Sivan Aryo Pharmed, Tehran, Iran
  7. Fuller Laboratories, Fullerton, CA, USA
  8. Department of Biochemistry and Molecular Biology, University of Southern California, Los Angeles, CA, USA
  9. Department of Virology, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
  10. Department of Medical Biotechnology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
  11. Department of Medical Biotechnology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
  12. Department of Applied Microbial Technology, Faculty of Biotechnology and Life Science, Sojo University, Kumamoto, Japan
  13. Department of Medical Biotechnology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran

open access

Vol 58, No 3 (2020)
REVIEW
Submitted: 2020-07-22
Accepted: 2020-09-17
Published online: 2020-09-24

Abstract

Clustered Regulatory Interspaced Short Palindromic Repeats (CRISPR) is one of the major genome editing systems and allows changing DNA levels of an organism. Among several CRISPR categories, the CRISPR-Cas9 system has shown a remarkable progression rate over its lifetime. Recently, other tools including CRISPR-Cas12 and CRISPR-Cas13 have been introduced. CRISPR-Cas9 system has played a key role in the industrial cell factory’s production and improved our understanding of genome function. Additionally, this system has been used as one of the major genome editing systems for the diagnosis and treatment of several infectious and non-infectious diseases. In this review, we discuss CRISPR biology, its versatility, and its application in biomedical engineering.

Abstract

Clustered Regulatory Interspaced Short Palindromic Repeats (CRISPR) is one of the major genome editing systems and allows changing DNA levels of an organism. Among several CRISPR categories, the CRISPR-Cas9 system has shown a remarkable progression rate over its lifetime. Recently, other tools including CRISPR-Cas12 and CRISPR-Cas13 have been introduced. CRISPR-Cas9 system has played a key role in the industrial cell factory’s production and improved our understanding of genome function. Additionally, this system has been used as one of the major genome editing systems for the diagnosis and treatment of several infectious and non-infectious diseases. In this review, we discuss CRISPR biology, its versatility, and its application in biomedical engineering.

Get Citation

Keywords

Genome editing; sgRNA; CRISPR-Cas9; CRISPR-Cas12; CRISPR-Cas13

About this article
Title

An overview of applications of CRISPR-Cas technologies in biomedical engineering

Journal

Folia Histochemica et Cytobiologica

Issue

Vol 58, No 3 (2020)

Article type

Review paper

Pages

163-173

Published online

2020-09-24

Page views

3271

Article views/downloads

2721

DOI

10.5603/FHC.a2020.0023

Pubmed

32978771

Bibliographic record

Folia Histochem Cytobiol 2020;58(3):163-173.

Keywords

Genome editing
sgRNA
CRISPR-Cas9
CRISPR-Cas12
CRISPR-Cas13

Authors

Saleh Jamehdor
Kasra Arbabi Zaboli
Sina Naserian
Jose Thekkiniath
Honey Alef Omidy
Ali Teimoori
Behrooz Johari
Amir Hossein Taromchi
Yu Sasano
Saeed Kaboli

References (117)
  1. Jiang F, Doudna JA. CRISPR-Cas9 Structures and Mechanisms. Annu Rev Biophys. 2017; 46: 505–529.
  2. Ishino Y, Shinagawa H, Makino K, et al. 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(12): 5429–5433.
  3. Nakata A, Amemura M, Makino K. Unusual nucleotide arrangement with repeated sequences in the Escherichia coli K-12 chromosome. J Bacteriol. 1989; 171(6): 3553–3556.
  4. Hermans P, Van So, Bik E, et al. Insertion element IS987 from Mycobacterium bovis BCG is located in a hot-spot integration region for insertion elements in Mycobacterium tuberculosis complex strains. Infect Immun. 1991; 59(8): 2695–705.
  5. Mojica FJ, Juez G, Rodríguez-Valera F. Transcription at different salinities of Haloferax mediterranei sequences adjacent to partially modified PstI sites. Mol Microbiol. 1993; 9(3): 613–621.
  6. Groenen PM, Bunschoten AE, van Soolingen D, et al. Nature of DNA polymorphism in the direct repeat cluster of Mycobacterium tuberculosis; application for strain differentiation by a novel typing method. Mol Microbiol. 1993; 10(5): 1057–1065.
  7. Mojica FJ, Díez-Villaseñor C, Soria E, et al. Biological significance of a family of regularly spaced repeats in the genomes of Archaea, Bacteria and mitochondria. Mol Microbiol. 2000; 36(1): 244–246.
  8. She Q, Singh RK, Confalonieri F, et al. The complete genome of the crenarchaeon Sulfolobus solfataricus P2. Proc Natl Acad Sci U S A. 2001; 98(14): 7835–7840.
  9. Jansen R, Embden JD, Gaastra W, et al. Identification of genes that are associated with DNA repeats in prokaryotes. Mol Microbiol. 2002; 43(6): 1565–1575.
  10. Storici F, Snipe JR, Chan GK, et al. Conservative repair of a chromosomal double-strand break by single-strand DNA through two steps of annealing. Mol Cell Biol. 2006; 26(20): 7645–7657.
  11. Yoshimi K, Kunihiro Y, Kaneko T, et al. ssODN-mediated knock-in with CRISPR-Cas for large genomic regions in zygotes. Nat Commun. 2016; 7: 10431.
  12. Bressan RB, Dewari PS, Kalantzaki M, et al. Efficient CRISPR/Cas9-assisted gene targeting enables rapid and precise genetic manipulation of mammalian neural stem cells. Development. 2017; 144(4): 635–648.
  13. Zhang F, Wen Y, Guo X. CRISPR/Cas9 for genome editing: progress, implications and challenges. Hum Mol Genet. 2014; 23(R1): R40–R46.
  14. Jiang W, Bikard D, Cox D, et al. CRISPR-assisted editing of bacterial genomes. Nat Biotechnol. 2013; 31(3): 233.
  15. Kimura P, Nakane T, Ishitani R, et al. Molecular mechanism of CRISPR. J Appl Crystallogr. 2014; 156: 935–49.
  16. Wu X, Kriz AJ, Sharp PA. Target specificity of the CRISPR-Cas9 system. Quant Biol. 2014; 2(2): 59–70.
  17. Yan F, Wang W, Zhang J. CRISPR-Cas12 and Cas13: the lesser known siblings of CRISPR-Cas9. Cell Biol Toxicol. 2019; 35(6): 489–492.
  18. Cong Le, Ran FA, Cox D, et al. Multiplex genome engineering using CRISPR/Cas systems. Science. 2013; 339(6121): 819–823.
  19. Ran FA, Hsu PD, Wright J, et al. Genome engineering using the CRISPR-Cas9 system. Nat Protoc. 2013; 8(11): 2281–2308.
  20. Qi LS, Larson MH, Gilbert LA, et al. Repurposing CRISPR as an RNA-guided platform for sequence-specific control of gene expression. Cell. 2013; 152(5): 1173–1183.
  21. Brezgin S, Kostyusheva A, Kostyushev D, et al. Dead Cas Systems: Types, Principles, and Applications. Int J Mol Sci. 2019; 20(23).
  22. Perez-Pinera P, Kocak D, Vockley C, et al. RNA-guided gene activation by CRISPR-Cas9–based transcription factors. Nat Methods. 2013; 10 (10):973-976. .
  23. Gilbert LA, Horlbeck MA, Adamson B, et al. Genome-Scale CRISPR-Mediated Control of Gene Repression and Activation. Cell. 2014; 159(3): 647–661.
  24. Kwon DY, Zhao YT, Lamonica JM, et al. Locus-specific histone deacetylation using a synthetic CRISPR-Cas9-based HDAC. Nat Commun. 2017; 8: 15315.
  25. Pulecio J, Verma N, Mejía-Ramírez E, et al. CRISPR/Cas9-Based Engineering of the Epigenome. Cell Stem Cell. 2017; 21(4): 431–447.
  26. 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(7681): 464–471.
  27. Zhou GS, Su ZY, Cai YuR, et al. Different effects of nanophase and conventional hydroxyapatite thin films on attachment, proliferation and osteogenic differentiation of bone marrow derived mesenchymal stem cells. Biomed Mater Eng. 2007; 17(6): 387–395.
  28. Wang Yu, Wang S, Chen W, et al. CRISPR-Cas9 and CRISPR-Assisted Cytidine Deaminase Enable Precise and Efficient Genome Editing in Klebsiella pneumoniae. Appl Environ Microbiol. 2018; 84(23).
  29. Zhao Y, Tian J, Zheng G, et al. Multiplex genome editing using a dCas9-cytidine deaminase fusion in Streptomyces. Sci China Life Sci. 2019; 63(7): 1053–1062.
  30. Gu T, Zhao S, Pi Y, et al. Highly efficient base editing in Staphylococcus aureus using an engineered CRISPR RNA-guided cytidine deaminase. Chemical Science. 2018; 9(12): 3248–3253.
  31. Zhang He, Pan H, Zhou C, et al. Simultaneous zygotic inactivation of multiple genes in mouse through CRISPR/Cas9-mediated base editing. Development. 2018; 145(20).
  32. Li Q, Li Y, Yang S, et al. CRISPR-Cas9-mediated base-editing screening in mice identifies DND1 amino acids that are critical for primordial germ cell development. Nat Cell Biol. 2018; 20(11): 1315–1325.
  33. Yuan J, Ma Y, Huang T, et al. Genetic Modulation of RNA Splicing with a CRISPR-Guided Cytidine Deaminase. Mol Cell. 2018; 72(2): 380–394.e7.
  34. Chiang TWW, le Sage C, Larrieu D, et al. CRISPR-Cas9(D10A) nickase-based genotypic and phenotypic screening to enhance genome editing. Sci Rep. 2016; 6: 24356.
  35. Rysenkova KD, Semina EV, Karagyaur MN, et al. CRISPR/Cas9 nickase mediated targeting of urokinase receptor gene inhibits neuroblastoma cell proliferation. Oncotarget. 2018; 9(50): 29414–29430.
  36. Dong De, Ren K, Qiu X, et al. The crystal structure of Cpf1 in complex with CRISPR RNA. Nature. 2016; 532(7600): 522–526.
  37. Nishimasu H, Ran FA, Hsu PD, et al. Crystal structure of Cas9 in complex with guide RNA and target DNA. Cell. 2014; 156(5): 935–949.
  38. Safari F, Zare K, Negahdaripour M, et al. CRISPR Cpf1 proteins: structure, function and implications for genome editing. Cell Biosci. 2019; 9: 36.
  39. Teng F, Li J, Cui T, et al. Enhanced mammalian genome editing by new Cas12a orthologs with optimized crRNA scaffolds. Genome Biol. 2019; 20(1): 15.
  40. Strecker J, Jones S, Koopal B, et al. Engineering of CRISPR-Cas12b for human genome editing. Nat Commun. 2019; 10(1): 212.
  41. Choi J, Bae T, Byambasuren N, et al. CRISPR-Cf1 activation of endogenous BMP4 gene for osteogenic differentiation of umbilical cord-derived mesenchymal stem cells. Mol Ther Methods Clin Dev. 2020(17): 309–316.
  42. Świat MA, Dashko S, den Ri, et al. Fn Cpf1: a novel and efficient genome editing tool for Saccharomyces cerevisiae. Nucleic Acids Res. 2017; 45(21): 12585–98.
  43. Zetsche B, Heidenreich M, Mohanraju P, et al. Multiplex gene editing by CRISPR-Cpf1 using a single crRNA array. Nat Biotechnol. 2017; 35(1): 31–34.
  44. Yang Z, Edwards H, Xu P. CRISPR-Cas12a/Cpf1-assisted precise, efficient and multiplexed genome-editing in . Metab Eng Commun. 2020; 10: e00112.
  45. Broughton JP, Deng X, Yu G, et al. CRISPR-Cas12-based detection of SARS-CoV-2. Nat Biotechnol. 2020; 38(7): 870–874.
  46. Liu J-J, Orlova N, Oakes BL, et al. CRISPR-CasX is an RNA-dominated enzyme active for human genome editing. Nature. 2019; 566(7743): 218.
  47. Yan WX, Hunnewell P, Alfonse LE, et al. Functionally diverse type V CRISPR-Cas systems. Science. 2019; 363(6422): 88–91.
  48. Karvelis T, Bigelyte G, Young JK, et al. et al.. PAM recognition by miniature CRISPR-Cas14 triggers programmable double-stranded DNA cleavage. Nucleic Acids Res. 2020; 48(9): 516–5023.
  49. Strecker J, Ladha A, Makarova KS, et al. RNA-guided DNA insertion with CRISPR-associated transposases. Science. 2019; 365(6448): 48–53.
  50. Shmakov S, Abudayyeh OO, Makarova KS, et al. Discovery and Functional Characterization of Diverse Class 2 CRISPR-Cas Systems. Mol Cell. 2015; 60(3): 385–397.
  51. Abudayyeh OO, Gootenberg JS, Konermann S, et al. C2c2 is a single-component programmable RNA-guided RNA-targeting CRISPR effector. Science. 2016; 353(6299): aaf5573.
  52. Smargon AA, Cox DBT, Pyzocha NK, et al. Cas13b Is a Type VI-B CRISPR-Associated RNA-Guided RNase Differentially Regulated by Accessory Proteins Csx27 and Csx28. Mol Cell. 2017; 65(4): 618–630.e7.
  53. Gootenberg JS, Abudayyeh OO, Lee JW, et al. Nucleic acid detection with CRISPR-Cas13a/C2c2. Science. 2017; 356(6336): 438–442.
  54. Yang LZ, Wang Y, Li SQ, et al. Dynamic Imaging of RNA in Living Cells by CRISPR-Cas13 Systems. Mol Cell. 2019; 76(6): 981–997.e7.
  55. Kaminski MM, Alcantar MA, Lape IT, et al. A CRISPR-based assay for the detection of opportunistic infections post-transplantation and for the monitoring of transplant rejection. Nat Biomed Eng. 2020; 4(6): 601–609.
  56. Rees HA, Liu DR, Rees HA, et al. Base editing: precision chemistry on the genome and transcriptome of living cells. Nat Rev Genet. 2018; 19(12): 770–788.
  57. Abudayyeh OO, Gootenberg JS, Essletzbichler P, et al. RNA targeting with CRISPR-Cas13. Nature. 2017; 550(7675): 280–284.
  58. Abudayyeh OO, Gootenberg JS, Franklin B, et al. A cytosine deaminase for programmable single-base RNA editing. Science. 2019; 365(6451): 382–386.
  59. Fry LE, Peddle CF, Barnard AR, et al. RNA editing as a therapeutic approach for retinal gene therapy requiring long coding sequences. Int J Mol Sci. 2020; 21(3).
  60. Yan WX, Chong S, Zhang H, et al. Cas13d Is a Compact RNA-Targeting Type VI CRISPR Effector Positively Modulated by a WYL-Domain-Containing Accessory Protein. Mol Cell. 2018; 70(2): 327–339.e5.
  61. Abbott TR, Dhamdhere G, Liu Y, et al. Development of CRISPR as an Antiviral Strategy to Combat SARS-CoV-2 and Influenza. Cell. 2020; 181(4): 865–876.e12.
  62. Inui M, Miyado M, Igarashi M, et al. Rapid generation of mouse models with defined point mutations by the CRISPR/Cas9 system. Sci Rep. 2014; 4: 5396.
  63. Chen S, Sanjana NE, Zheng K, et al. Genome-wide CRISPR screen in a mouse model of tumor growth and metastasis. Cell. 2015; 160(6): 1246–60.
  64. Egorova TV, Zotova ED, Reshetov DA, et al. CRISPR/Cas9-generated mouse model of Duchenne muscular dystrophy recapitulating a newly identified large 430 kb deletion in the human gene. Dis Model Mech. 2019; 12(4).
  65. Lee H, Yoon DaE, Kim K. Genome editing methods in animal models. Anim Cells Syst (Seoul). 2020; 24(1): 8–16.
  66. Mehravar M, Shirazi A, Nazari M, et al. Mosaicism in CRISPR/Cas9-mediated genome editing. Dev Biol. 2019; 445(2): 156–162.
  67. Maddalo D, Manchado E, Concepcion CP, et al. In vivo engineering of oncogenic chromosomal rearrangements with the CRISPR/Cas9 system. Nature. 2014; 516(7531): 423–427.
  68. Wen L, Zhao C, Song J, et al. CRISPR/Cas9-Mediated TERT Disruption in Cancer Cells. Int J Mol Sci. 2020; 21(2).
  69. Xu M, Weng Q, Ji J. Applications and advances of CRISPR/Cas9 in animal cancer model. Brief Funct Genomics. 2020; 19(3): 235–241.
  70. Reyes LM, Estrada JL, Wang ZYu, et al. Creating class I MHC-null pigs using guide RNA and the Cas9 endonuclease. J Immunol. 2014; 193(11): 5751–5757.
  71. Sheets TP, Park CH, Park KE, et al. Somatic Cell Nuclear Transfer Followed by CRIPSR/Cas9 Microinjection Results in Highly Efficient Genome Editing in Cloned Pigs. Int J Mol Sci. 2016; 17(12).
  72. Gao Y, Wu H, Wang Y, et al. Single Cas9 nickase induced generation of NRAMP1 knockin cattle with reduced off-target effects. Genome Biol. 2017; 18(1): 13.
  73. Ni W, Qiao J, Hu S, et al. Efficient gene knockout in goats using CRISPR/Cas9 system. PLoS One. 2014; 9(9): e106718.
  74. Bharati J, Punetha M, Kumar BS, et al. Genome editing in animals: an overview. Genomics and Biotechnological Advances in Veterinary, Poultry, and Fisheries. 2020: 75–104.
  75. Niu Y, Zhao X, Zhou J, et al. Efficient generation of goats with defined point mutation (I397V) in GDF9 through CRISPR/Cas9. Reprod Fertil Dev. 2018; 30(2): 307–312.
  76. Li X, Hao F, Hu X, et al. Generation of Tβ4 knock-in Cashmere goat using CRISPR/Cas9. Int J Biol Sci. 2019; 15(8): 1743–1754.
  77. Crispo M, Mulet AP, Tesson L, et al. Efficient Generation of Myostatin Knock-Out Sheep Using CRISPR/Cas9 Technology and Microinjection into Zygotes. PLoS One. 2015; 10(8): e0136690.
  78. Niu Y, Jin M, Li Y, et al. Biallelic β-carotene oxygenase 2 knockout results in yellow fat in sheep via CRISPR/Cas9. Anim Genet. 2017; 48(2): 242–244.
  79. Kalds P, Zhou S, Cai B, et al. Sheep and Goat Genome Engineering: From Random Transgenesis to the CRISPR Era. Front Genet. 2019; 10: 750.
  80. Eaton SL, Proudfoot C, Lillico SG, et al. CRISPR/Cas9 mediated generation of an ovine model for infantile neuronal ceroid lipofuscinosis (CLN1 disease). Sci Rep. 2019; 9(1): 9891.
  81. Hoban MD, Cost GJ, Mendel MC, et al. Correction of the sickle cell disease mutation in human hematopoietic stem/progenitor cells. Blood. 2015; 125(17): 2597–2604.
  82. Wang C, Jin H, Gao D, et al. A CRISPR screen identifies CDK7 as a therapeutic target in hepatocellular carcinoma. Cell Res. 2018; 28(6): 690–692.
  83. Chai N, Haney MS, Couthouis J, et al. Genome-wide synthetic lethal CRISPR screen identifies FIS1 as a genetic interactor of ALS-linked C9ORF72. Brain Res. 2020; 1728: 146601.
  84. Acharya S, Maiti S, Chakraborty D. CRISPR-Cas9 for therapy: the challenges and ways to overcome them. Genome Engineering via CRISPR-Cas9 System. 2020: 101–110.
  85. Xu L, Wang J, Liu Y, et al. CRISPR-Edited Stem Cells in a Patient with HIV and Acute Lymphocytic Leukemia. N Engl J Med. 2019; 381(13): 1240–1247.
  86. 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(2): 229–233.
  87. Soppe JA, Lebbink RJ. Antiviral Goes Viral: Harnessing CRISPR/Cas9 to Combat Viruses in Humans. Trends Microbiol. 2017; 25(10): 833–850.
  88. Sasano Yu, Nagasawa K, Kaboli S, et al. CRISPR-PCS: a powerful new approach to inducing multiple chromosome splitting in Saccharomyces cerevisiae. Sci Rep. 2016; 6: 30278.
  89. Easmin F, Sasano Yu, Kimura S, et al. CRISPR-PCD and CRISPR-PCRep: Two novel technologies for simultaneous multiple segmental chromosomal deletion/replacement in Saccharomyces cerevisiae. J Biosci Bioeng. 2020; 129(2): 129–139.
  90. Wang T, Wei JJ, Sabatini DM, et al. Genetic screens in human cells using the CRISPR-Cas9 system. Science. 2014; 343(6166): 80–84.
  91. Shalem O, Sanjana NE, Hartenian E, et al. Genome-scale CRISPR-Cas9 knockout screening in human cells. Science. 2014; 343(6166): 84–87.
  92. Joung J, Konermann S, Gootenberg JS, et al. Genome-scale CRISPR-Cas9 knockout and transcriptional activation screening. Nat Protoc. 2017; 12(4): 828–863.
  93. Luo Ji. CRISPR/Cas9: From Genome Engineering to Cancer Drug Discovery. Trends Cancer. 2016; 2(6): 313–324.
  94. Maeder ML, Linder SJ, Cascio VM, et al. CRISPR RNA-guided activation of endogenous human genes. Nat Methods. 2013; 10(10): 977–979.
  95. Smurnyy Y, Cai Mi, Wu H, et al. DNA sequencing and CRISPR-Cas9 gene editing for target validation in mammalian cells. Nat Chem Biol. 2014; 10(8): 623–625.
  96. Zhu S, Li W, Liu J, et al. Genome-scale deletion screening of human long non-coding RNAs using a paired-guide RNA CRISPR-Cas9 library. Nat Biotechnol. 2016; 34(12): 1279–1286.
  97. Shi J, Wang E, Milazzo JP, et al. Discovery of cancer drug targets by CRISPR-Cas9 screening of protein domains. Nat Biotechnol. 2015; 33(6): 661–667.
  98. Nelles DA, Fang MY, O'Connell MR, et al. Programmable RNA Tracking in Live Cells with CRISPR/Cas9. Cell. 2016; 165(2): 488–496.
  99. Gu Bo, Swigut T, Spencley A, et al. Transcription-coupled changes in nuclear mobility of mammalian cis-regulatory elements. Science. 2018; 359(6379): 1050–1055.
  100. Duan J, Lu G, Hong Yu, et al. Live imaging and tracking of genome regions in CRISPR/dCas9 knock-in mice. Genome Biol. 2018; 19(1): 192.
  101. Wang H, Nakamura M, Abbott TR, et al. CRISPR-mediated live imaging of genome editing and transcription. Science. 2019; 365(6459): 1301–1305.
  102. Maus MV, Grupp SA, Porter DL, et al. Antibody-modified T cells: CARs take the front seat for hematologic malignancies. Blood. 2014; 123(17): 2625–2635.
  103. Cyranoski D. CRISPR gene-editing tested in a person for the first time. Nature. 2016; 539(7630): 479.
  104. Choi BD, Yu X, Castano AP, et al. CRISPR-Cas9 disruption of PD-1 enhances activity of universal EGFRvIII CAR T cells in a preclinical model of human glioblastoma. J Immunother Cancer. 2019; 7(1): 304.
  105. Hsu MN, Chang YH, Truong VuA, et al. CRISPR technologies for stem cell engineering and regenerative medicine. Biotechnol Adv. 2019; 37(8): 107447.
  106. Chen G, Cheng Du, Chen B. [Development of CRISPR technology and its application in bone and cartilage tissue engineering]. Nan Fang Yi Ke Da Xue Xue Bao. 2019; 39(12): 1515–1520.
  107. Ali Q, Malik S, Malik A, et al. Role of Modern Technologies in Tissue Engineering. Arch Neurosci. 2020; 7(1).
  108. Aghamiri S, Ghavidel AA, Zandsalimi F, et al. Nanoparticles-mediated CRISPR/Cas9 delivery: Applications in cancer treatment and detection. J Drug Deliv Sci Technol. ; 2020: 101533.
  109. Baylis F, McLeod M. First-in-human Phase 1 CRISPR Gene Editing Cancer Trials: Are We Ready? Curr Gene Ther. 2017; 17(4): 309–319.
  110. González-Romero E, Martínez-Valiente C, García-Ruiz C, et al. CRISPR to fix bad blood: a new tool in basic and clinical hematology. Haematologica. 2019; 104(5): 881–893.
  111. 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.
  112. Kostyusheva A, Brezgin S, Babin Y, et al. CRISPR-Cas Systems for Diagnosing Infectious Diseases. .
  113. Pittermann E, Lachmann N, MacLean G, et al. Gene correction of reversed Kostmann disease phenotype in patient-specific induced pluripotent stem cells. Blood Adv. 2017; 1(14): 903–914.
  114. Charlesworth CT, Deshpande PS, Dever DP, et al. Identification of preexisting adaptive immunity to Cas9 proteins in humans. Nat Med. 2019; 25(2): 249–254.
  115. Zhang XH, Tee LY, Wang XG, et al. Off-target Effects in CRISPR/Cas9-mediated Genome Engineering. Mol Ther Nucleic Acids. 2015; 4: e264.
  116. Lino CA, Harper JC, Carney JP, et al. Delivering CRISPR: a review of the challenges and approaches. Drug Deliv. 2018; 25(1): 1234–1257.
  117. Rahimi H, Salehiabar M, Charmi J, et al. Harnessing nanoparticles for the efficient delivery of the CRISPR/Cas9 system. Nano Today. 2020; 34: 100895.

Regulations

Important: This website uses cookies. More >>

The cookies allow us to identify your computer and find out details about your last visit. They remembering whether you've visited the site before, so that you remain logged in - or to help us work out how many new website visitors we get each month. Most internet browsers accept cookies automatically, but you can change the settings of your browser to erase cookies or prevent automatic acceptance if you prefer.

By VM Media Group sp z o.o., ul. Świętokrzyska 73, 80–180 Gdańsk

tel.:+48 58 320 94 94, faks:+48 58 320 94 60, e-mail:  viamedica@viamedica.pl