Uploaded by sunflowerbugs21

lit-cited-term (AutoRecovered)

advertisement
1. Ishino, Y., Shinagawa, H., Makino, K., Amemura, M., Nakata, A., (1987). Nucleotide sequence of
the iap gene, responsible for alkaline phosphatase isozyme conversion in Escherichia coli, and
identification of the gene product. J. Bacteriol. 169 (12), 5429e5433.
2. Makarova, K.S., Wolf, Y.I., Alkhnbashi, O.S., Costa, F., Shah, S.A., Saunders, S.J., et al., (2015). An
updated evolutionary classification of CRISPReCas systems. Nat. Rev. Microbiol. 13 (11),
722e736.
3. Hille, F., Richter, H., Wong, S.P., Bratovic, M., Ressel, S., Charpentier, E., (2018). The biology of
CRISPR-Cas: backward and forward. Cell 172 (6), 1239-1259.
4. Singh, V., Braddick, D., Dhar, P.K., (2017). Exploring the potential of genome editing CRISPR-Cas9
technology. Gene 599, 1e18.
5. Bhaya, D., Davison, M., Barrangou, R., (2011). CRISPR-Cas systems in bacteria and archaea:
versatile small RNAs for adaptive defense and regulation. Annu. Rev. Genet. 45, 273e297.
6. Brouns, S.J., Jore, M.M., Lundgren, M., Westra, E.R., Slijkhuis, R.J., Snijders, A.P., et al., (2008).
Small CRISPR RNAs guide antiviral defense in prokaryotes. Science 321 (5891), 960e964.
7. Cui, Y., Li, Y., Gorgé, O., Platonov, M.E., Yan, Y., Guo, Z., et al., (2008). Insight into microevolution
of Yersinia pestis by clustered regularly interspaced short palindromic repeats. PLoS One 3 (7),
e2652.
8. Garneau, J.E., Dupuis, M.È., Villion, M., Romero, D.A., Barrangou, R., Boyaval, P., et al., (2010).
The CRISPR/Cas bacterial immune system cleaves bacteriophage and plasmid DNA. Nature 468
(7320), 67e71.
9. Chylinski, K., Le Rhun, A., Charpentier, E., (2013). The tracrRNA and Cas9 families of type II
CRISPR-Cas immunity systems. RNA Biol. 10 (5), 726e737.
10. Nam, K.H., Kurinov, I., Ke, A., (2011). Crystal structure of clustered regularly interspaced short
palindromic repeats (CRISPR)-associated Csn2 protein revealed Ca2þ-dependent doublestranded DNA binding activity. J. Biol. Chem. 286 (35), 30759e30768.
11. Makarova, K.S., Aravind, L., Wolf, Y.I., Koonin, E.V., (2011). Unification of Cas protein families
and a simple scenario for the origin and evolution of CRISPR-Cas systems. Biol. Direct 6, 38.
12. East-Seletsky, A., O’Connell, M.R., Knight, S.C., Burstein, D., Cate, J.H., Tjian, R., et al., (2016).
Two distinct RNase activities of CRISPR-C2c2 enable guide-RNA processing and RNA detection.
Nature 538 (7624), 270e273.
13. Myhrvold, C., Freije, C.A., Gootenberg, J.S., Abudayyeh, O.O., Metsky, H.C., Durbin, A.F., et al.,
(2018). Field-deployable viral diagnostics using CRISPR-Cas13. Science 360 (6387), 444e448.
14. Khambhati, K., Bhattacharjee, G., Singh, V., (2019). Current progress in CRISPR-based diagnostic
platforms. J. Cell. Biochem. 120 (3), 2721e2725.
15. Gootenberg, J.S., Abudayyeh, O.O., Kellner, M.J., Joung, J., Collins, J.J., Zhang, F., (2018).
Multiplexed and portable nucleic acid detection platform with Cas13, Cas12a, and Csm6. Science
360 (6387), 439e444.
16. Jinek M, Chylinski K, Fonfara I, Hauer M, Doudna JA, Charpentier E (2012) A programmable dual
RNA-guided DNA endonuclease in adaptive bacterial immunity. Science 337:816–821
17. Cong L, Ran FA, Cox D et al (2013) Multiplex genome engineering using CRISPR/Cas systems.
Science 339:819–823
18. Mali P, Yang L, Esvelt KM et al (2013) RNA-guided human genome engineering via Cas9. Science
339:823–826
19. Xie K, Minkenberg B, Yang Y (2015) Boosting CRISPR/Cas9 multiplex editing capability with the
endogenous tRNA-processing system. Proc Natl Acad Sci 112:3570–3575
20. Islam T (2019) CRISPR-Cas technology in modifying food crops. CAB Reviews 14:1–16
21. Bhowmik P, Hassan MM, Molla K, Rahman M, Islam MT (2019) Application of CRISPR-Cas
genome editing tools for the improvement of plant abiotic stress tolerance. In: Hasanuzzaman
M, Nahar K, Fujita M, Oku H, Islam MT (eds) Approaches for enhancing abiotic stress tolerance
in plants. CRC Press, New York, pp 459–472
22. Haque E, Taniguchi H, Hassan MM, Bhowmik P, Karim MR, Smiech M et al (2018) Application of
CRISPR/Cas9 genome editing technology for the improvement of crops cultivated in tropical
climates: recent progress, prospects, and challenges. Front Plant Sci 9:617
23. Wang D, Zhang F, Gao G (2020) CRISPRbased therapeutic genome editing: strategies and in vivo
delivery by AAV vectors. Cell 181 (1):136–150
24. Maeder ML, Linder SJ, Cascio VM, Fu Y, Ho QH, Joung JK (2013) CRISPR RNA–guided activation of
endogenous human genes. Nat Methods 10:977–999
25. Perez-Pinera P, Kocak DD, Vockley CM et al (2013) RNA-guided gene activation by CRISPR-Cas9–
based transcription factors. Nat Methods 10:973–976
26. Qi LS, Larson MH, Gilbert LA, Doudna JA, Weissman JS, Arkin AP, Lim WA et al (2013)
Repurposing CRISPR as an RNA-guided platform for sequence-specific control of gene
expression. Cell 152:1173–1183
27. Gilbert LA, Larson MH, Morsut L et al (2013) CRISPR-mediated modular RNA-guided regulation
of transcription in eukaryotes. Cell 154:442–451
28. Adli M (2018) The CRISPR tool kit for genome editing and beyond. Nat Commun 9:1–13
29. Doench JG (2018) Am I ready for CRISPR? A user’s guide to genetic screens. Nat Rev
Genet19:67–80
30. Hilton IB, D’ippolito AM, Vockley CM et al (2015) Epigenome editing by a CRISPRCas9-based
acetyltransferase activates genes from promoters and enhancers. Nat Biotechnol 33:510–517
31. Van Steensel B, Belmont AS (2017) Laminaassociated domains: links with chromosome
architecture, heterochromatin, and gene repression. Cell 169:780–791
32. Wang J, Meng X, Hu X, Sun T, Li J, Wang K, Yu H (2019) xCas9 expands the scope of genome
editing with reduced efficiency in rice. Plant Biotechnol J 17:709–711
33. Morgan SL, Mariano NC, Bermudez A et al (2017) Manipulation of nuclear architecture through
CRISPR-mediated chromosomal looping. Nat Commun 8:15993
34. Hao N, Shearwin KE, Dodd IB et al (2017) Programmable DNA looping using engineered bivalent
dCas9 complexes. Nat Commun 8:1628
35. Chen B, Guan J, Huang B (2016) Imaging specific genomic DNA in living cells. Annu Rev Biophys
45:1–23
36. Ma H, Naseri A, Reyes-Gutierrez P, Wolfe SA, Zhang S, Pederson T et al (2015) Multicolor CRISPR
labeling of chromosomal loci in human cells. Proc Natl Acad Sci 112:3002–3007
37. Qin P, Parlak M, Kuscu C, Bandaria J, Mir M, Szlachta K et al (2017) Live cell imaging of low-and
non-repetitive chromosome loci using CRISPR-Cas9. Nat Commun 8:14725
38. Yang LZ, Wang Y, Li SQ et al (2019) Dynamic imaging of RNA in living cells by CRISPRCas13
systems. Mol Cell 76:981–997
39. Wu X, Mao S, Ying Y, Krueger CJ, Chen AK (2019) Progress and challenges for live-cell imaging of
genomic loci using CRISPR-based platforms. Genomics Proteomics Bioinformatics 17:119–128
40. Adli M (2018) The CRISPR tool kit for genome editing and beyond. Nat Commun 9:1–13
41. Gaudelli NM, Komor AC, Rees HA et al (2017) Programmable base editing of Al T to Gl C in
genomic DNA without DNA cleavage. Nature 551:464–471
42. Komor AC, Kim YB, Packer MS, Zuris JA, Liu DR et al (2016) Programmable editing of a target
base in genomic DNA without doubles tranded DNA cleavage. Nature 533:420–424
43. Nishida K, Arazoe T, Yachie N et al (2016) Targeted nucleotide editing using hybrid prokaryotic
and vertebrate adaptive immune systems. Science 353:aaf8729
44. Molla KA, Yang Y (2019a) CRISPR/Casmediated base editing: technical considerations and
practical applications. Trends Biotechnology 37:1121–1142
45. Molla KA, Yang Y (2019b) Predicting CRISPR/Cas9-induced mutations for precise genome editing.
Trends Biotechnol S0167-7799:30206–30209
46. Abudayyeh OO, Gootenberg JS, Franklin B et al. (2019) A cytosine deaminase for programmable
single-base RNA editing. Science 365:382–386
47. Cox DB, Gootenberg JS, Abudayyeh OO et al (2017) RNA editing with CRISPR-Cas13. Science
358:1019–1027
48. O’Connell MR, Oakes BL, Sternberg SH, East-Seletsky A, Kaplan M, Doudna JA (2014)
Programmable RNA recognition and cleavage by CRISPR/Cas9. Nature 516 (7530):263–266
49. East-Seletsky A, O’Connell MR, Knight SC et al (2016) Two distinct RNase activities of CRISPRC2c2 enable guide-RNA processing and RNA detection. Nature 538:270–273
50. Abudayyeh OO, Gootenberg JS, Essletzbichler P et al (2017) RNA targeting with CRISPR-Cas13.
Nature 550:280–284
51. Smargon AA, Cox DB, Pyzocha NK, Zheng K et al (2017) Cas13b is a type VI-B CRISPR associated
RNA-guided RNase differentially regulated by accessory proteins Csx27 and Csx28. Mol Cell
65:618–630
52. Slaymaker IM, Mesa P, Kellner MJ et al (2019) High-resolution structure of Cas13b and
biochemical characterization of RNA targeting and cleavage. Cell Rep 26:3741–3751
53. Abudayyeh OO, Gootenberg JS, Konermann S et al. (2016) C2c2 is a single-component
programmable RNA-guided RNA-targeting CRISPR effector. Science 353:aaf5573
54. Makarova KS, Wolf YI, Alkhnbashi OS et al (2015) An updated evolutionary classification of
CRISPR–Cas systems. Nat Rev Microbiol 13:722–736
55. Shmakov S, Sergey AS, Scott D, Cox D, Pyzocha N, Yan W, Abudayyeh OO et al. (2017) Diversity
and evolution of class 2 CRISPR–Cas systems. Nat Rev Microbiol 15:169–182
56. Klompe SE, Vo PL, Halpin-Healy TS, Sternberg SH (2019) Transposon-encoded CRISPR–Cas
systems direct RNA-guided DNA integration. Nature 571:219–225
57. Peters JE, Makarova KS, Shmakov S, Koonin EV et al (2017) Recruitment of CRISPR-Cas systems
by Tn7-like transposons. Proc Natl Acad Sci 114:E7358–E7366
58. Huang TK, Puchta H (2019) CRISPR/Casmediated gene targeting in plants: finally a turn for the
better for homologous recombination. Plant Cell Rep 38:443–453
59. Yeh CD, Richardson CD, Corn JE (2019) Advances in genome editing through control of DNA
repair pathways. Nat Cell Biol 21:1468–1478
60. Anzalone AV, Randolph PB, Davis JR et al (2019) Search-and-replace genome editing without
double-strand breaks or donor DNA. Nature 576:149–157
61. Chen JS, Ma E, Harrington LB et al (2018) CRISPR-Cas12a target binding unleashes indiscriminate
single-stranded DNase activity. Science 360:436–439
62. Gootenberg JS, Abudayyeh OO, Lee JW et al (2017) Nucleic acid detection with CRISPRCas13a/
C2c2. Science 356:438–442
63. Scudellari M (2019) Self-destructing mosquitoes and sterilized rodents: the promise of gene
drives. Nature 571:160–162
64. Strecker J, Ladha A, Gardner Z, Schmid-BurgkJL, Makarova KS, Koonin EV, Zhang F (2019a) RNAguided DNA insertion with CRISPR-associated transposases. Science 365 (6448):48–53
65. Strecker J, Jones S, Koopal B et al (2019b) Engineering of CRISPR-Cas12b for human genome
editing. Nat Commun 10:212
66. L. Bortesi, R. Fischer (2015) The CRISPR/Cas9 system for plant genome editing and beyond
Biotechnol. Adv., 33, pp. 41-52
67. Ricroch, A., Clairand, P., and Harwood, W. (2017). Use of CRISPR systems in plant genome
editing: toward new opportunities in agriculture. Emerg. Top. Life Sci. 1, 169–182. doi:
10.1042/etls20170085
68. Xie, K., and Yang, Y. (2013). RNA-guided genome editing in plants using a CRISPR-Cas system.
Mol. Plant 6, 1975–1983. doi: 10.1093/mp/sst119
69. Shan, Q., Wang, Y., Li, J., Zhang, Y., Chen, K., Liang, Z., et al. (2013). Targeted genome
modification of crop plants using a CRISPR-Cas system. Nat. Biotechnol. 31, 686–688. doi:
10.1038/nbt.2650
70. Zhang, H., Zhang, J., Wei, P., Zhang, B., Gou, F., Feng, Z., et al. (2014). The CRISPR/Cas9 system
produces specific and homozygous targeted gene editing in rice in one generation. Plant
Biotechnol. J. 12, 797–807. doi: 10.1111/pbi. 12200
71. Shimatani, Z., Kashojiya, S., Takayama, M., Terada, R., Arazoe, T., Ishii, H., et al. (2017). Targeted
base editing in rice and tomato using a CRISPR-Cas9 cytidine deaminase fusion. Nat. Biotechnol.
35, 441–443. doi: 10.1038/nbt.3833
72. Zong, Y., Wang, Y., Li, C., Zhang, R., Chen, K., Ran, Y., et al. (2017). Precise base editing in rice,
wheat and maize with a Cas9-cytidine deaminase fusion. Nat. Biotechnol. 35, 438–440. doi:
10.1038/nbt.3811
73. Li, J., Sun, Y., Du, J., Zhao, Y., and Xia, L. (2017a). Generation of targeted point mutations in rice
by a modified CRISPR/Cas9 system. Mol. Plant 10, 526–529. doi: 10.1016/j.molp.2016.12.001
74. Lowder, L. G., Zhang, D., Baltes, N. J., Paul, J. W. III, Tang, X., Zheng, X., et al. (2015). A
CRISPR/Cas9 toolbox for multiplexed plant genome editing and transcriptional regulation. Plant
Physiol. 169, 971–985. doi: 10.1104/pp.15. 00636
75. Shan, Q., Wang, Y., Li, J., and Gao, C. (2014). Genome editing in rice and wheat using the
CRISPR/Cas system. Nat. Protoc. 9, 2395–2410. doi: 10.1038/nprot. 2014.157
76. Wang, Y., Cheng, X., Shan, Q., Zhang, Y., Liu, J., Gao, C., et al. (2014). Simultaneous editing of
three homoeoalleles in hexaploid bread wheat confers heritable resistance to powdery mildew.
Nat. Biotechnol. 32, 947–951. doi: 10.1038/nbt.2969
77. Gil-Humanes, J., Wang, Y., Liang, Z., Shan, Q., Ozuna, C. V., Sanchez-Leon, S., et al. (2017). Highefficiency gene targeting in hexaploid wheat using DNA replicons and CRISPR/Cas9. Plant J. 89,
1251–1262. doi: 10.1111/tpj.13446
78. Kim, D., Alptekin, B., and Budak, H. (2018). CRISPR/Cas9 genome editing in wheat. Funct. Integr.
Genomics 18, 31–41. doi: 10.1007/s10142-017-0572-x
79. Shi, J., Gao, H., Wang, H., Lafitte, H. R., Archibald, R. L., Yang, M., et al. (2017). ARGOS8 variants
generated by CRISPR-Cas9 improve maize grain yield under field drought stress conditions. Plant
Biotechnol. J. 15, 207–216. doi: 10.1111/pbi.12603
80. Zhu, J., Song, N., Sun, S., Yang, W., Zhao, H., Song, W., et al. (2016). Efficiency and inheritance of
targeted mutagenesis in maize using CRISPR-Cas9. J. Genet. Genomics 43, 25–36. doi:
10.1016/j.jgg.2015.10.006
81. Feng, C., Yuan, J., Wang, R., Liu, Y., Birchler, J. A., and Han, F. (2016). Efficient targeted genome
modification in maize using CRISPR/Cas9 system. J. Genet. Genomics 43, 37–43.
doi:10.1016/j.jgg.2015.10.002
82. Qi, W., Zhu, T., Tian, Z., Li, C., Zhang, W., and Song, R. (2016). High-efficiency CRISPR/Cas9
multiplex gene editing using the glycine tRNA-processing system-based strategy in maize. BMC
Biotechnol. 16:58. doi: 10.1186/s12896-016-0289-2
83. Li, J., Zhang, H., Si, X., Tian, Y., Chen, K., Liu, J., et al. (2017b). Generation of thermosensitive
male-sterile maize by targeted knockout of the ZmTMS5 gene. J. Genet. Genomics 44, 465–468.
doi: 10.1016/j.jgg.2017.02.002
84. Andersson, M., Turesson, H., Nicolia, A., Falt, A. S., Samuelsson, M., and Hofvander, P. (2017).
Efficient targeted multiallelic mutagenesis in tetraploid potato (Solanum tuberosum) by
transient CRISPR-Cas9 expression in protoplasts. Plant Cell Rep. 36, 117–128.
doi:10.1007/s00299-016-2062-3
85. Butler, N. M., Baltes, N. J., Voytas, D. F., and Douches, D. S. (2016). Geminivirus-mediated
genome editing in potato (Solanum tuberosum L.) using sequence-specific nucleases. Front.
Plant Sci. 7:1045. doi: 10.3389/fpls.2016.01045
86. Brooks, C., Nekrasov, V., Lippman, Z. B., and Van Eck, J. (2014). Efficient gene editing in tomato
in the first generation using the clustered regularly interspaced short palindromic
repeats/CRISPR-associated9 system. Plant Physiol. 166, 1292–1297. doi: 10.1104/pp.114.247577
87. Ron, M., Kajala, K., Pauluzzi, G., Wang, D., Reynoso, M. A., Zumstein, K., et al. (2014). Hairy root
transformation using Agrobacterium rhizogenes as a tool for exploring cell type-specific gene
expression and function using tomato as a model. Plant Physiol. 166, 455–469. doi:
10.1104/pp.114.239392
88. Ito, Y., Nishizawa-Yokoi, A., Endo, M., Mikami, M., and Toki, S. (2015). CRISPR/Cas9-mediated
mutagenesis of the RIN locus that regulates tomato fruit ripening. Biochem. Biophys. Res.
Commun. 467, 76–82. doi: 10.1016/j.bbrc.2015.09.117
89. Li, R., Fu, D., Zhu, B., Luo, Y., and Zhu, H. (2018a). CRISPR/Cas9-mediated mutagenesis of
lncRNA1459 alters tomato fruit ripening. Plant J. 94, 513–524.doi: 10.1111/tpj.13872
90. Ueta, R., Abe, C., Watanabe, T., Sugano, S. S., Ishihara, R., Ezura, H., et al. (2017). Rapid breeding
of parthenocarpic tomato plants using CRISPR/Cas9. Sci. Rep.7:507. doi: 10.1038/s41598-01700501-4
91. Soyk, S., Lemmon, Z. H., Oved, M., Fisher, J., Liberatore, K. L., Park, S. J., et al. (2017). Bypassing
negative epistasis on yield in tomato imposed by a domestication gene. Cell 169, 1142–
1155.e12. doi: 10.1016/j.cell.2017.04.032
92. Jia, H., and Wang, N. (2014). Targeted genome editing of sweet orange using Cas9/sgRNA. PLoS
One 9:e93806. doi: 10.1371/journal.pone.0093806
93. Jia, H., Xu, J., Orbovic, V., Zhang, Y., and Wang, N. (2017a). Editing citrus genome via
SaCas9/sgRNA system. Front. Plant Sci. 8:2135. doi: 10.3389/fpls.2017.02135
94. Peng, A., Chen, S., Lei, T., Xu, L., He, Y., Wu, L., et al. (2017). Engineering canker resistant plants
through CRISPR/Cas9-targeted editing of the susceptibility gene CsLOB1 promoter in citrus.
Plant Biotechnol. J. 15, 1509–1519.doi: 10.1111/pbi.12733
95. Dirks, R., K. Van Dun, C.B. De Snoo, M. Van Den Berg, C.L. Lelivelt, W. Voermans, and E. Wijnker.
2009. Reverse breeding: A novel breeding approach based on engineered meiosis. Plant
Biotechnol. J. 79:837–845.
96. Chase, C.D. (2007) Cytoplasmic male sterility: a window to the world of plant mitochondrial
nuclear interactions. Trends Genet. 23, 81–90
97. Keller, E.R.J. and Korzun, L. (1996) Ovary and ovule culture for haploid production. In In vitro
Haploid Production in Higher Plants (Jain, S.M., Sopory, S.K. and Veilleux, R.E., eds), pp. 217–
235, Dordrecht: Kluwer Acad Publ.
98. Wang, Z., Zou, Y., Li, X., Zhang, Q., Chen, L., Wu, H., Su, D., Chen, Y., Guo, J., Luo, D., Long, Y.,
Zhong, Y. and Liu, Y.-G. (2006) Cytoplasmic male sterility of rice with boro II cytoplasm is caused
by a cytotoxic peptide and is restored by two related PPR motif genes via distinct modes of
mRNA silencing. Plant Cell, 18, 676–687.
99. Van Dun, C.M.P. and Dirks, R.H.G. (2006) Rijk Zwaan Zaadteelt en Zaadhandel B.V. Near Reverse
Breeding, WO⁄ 2006 ⁄ 094773.
100. Ross, K., Fransz, P., Armstrong, S., Vizir, I., Mulligan, B., Franklin, F. and Jones, G. (1997)
Cytological characterization of four meiotic mutants of Arabidopsis isolated from TDNAtransformed lines. Chromosome Res. 5, 551–559.
101. Azumi, Y., Liu, D., Zhao, D., Li, W., Wang, G., Hu, Y. and Ma, H. (2002) Homolog interaction
during meiotic prophase I in Arabidopsis requires the SOLO DANCERS gene encoding a novel
cyclin-like protein. EMBO J. 21, 3081–3095.
102. Caryl, A.P., Armstrong, S.J., Jones, G.H. and Franklin, F.C.H. (2000) A homologue of the yeast
HOP1 gene is inactivated in the Arabidopsis meiotic mutant asy1. Chromosoma, 109, 62–71.
103. Couteau, F., Belzile, F., Horlow, C., Grandjean, O., Vezon, D. and Doutriaux, M.-P. (1999)
Random chromosome segregation without meiotic arrest in both male and female meiocytes of
a dmc1 mutant of Arabidopsis. Plant Cell, 11, 1623–1634.
104. Nonomura, K.I., Nakano, M., Murata, K., Miyoshi, K., Eiguchi, M., Iayao, A., Hirochika, H. and
Kurata, N. (2004) An insertional mutation in the rice PAIR2 gne, the ortholog of Arabidopsis
ASY1, results in a defect in homologous chromosome pairing during meisosis. Mol. Genet.
Genomics, 271, 121–129.
105. Stacey, N.J., Kuromori, T., Azumi, Y., Roberts, G., Breuer, C., Wada, T., Maxwell, A., Roberts, K.
and Sugimoto-Shirasu, K. (2006) Arabidopsis SPO11-2 functions with SPO11-1 in meiotic
recombination. Plant J. 48, 206–216.
106. Wijeratne, A.J., Chen, C., Zhang, W., Timofejeva, L. and Ma, H. (2006) The Arabidopsis thaliana
PARTING DANCERS gene encoding a novel protein is required for normal meiotic homologous
recombination. Mol. Biol. Cell 17, 1331–1343.
107. Fire, A., Xu, S., Montgomery, M. et al. (1998) Potent and specific genetic interference by
double-stranded RNA in Caenorhabditis elegans. Nature 391, 806–811.
https://doi.org/10.1038/35888
108. Ratcliff, F., Martin-Hernandez, A. M., and Baulcombe, D. C. (2001). Technical advance. Tobacco
rattle virus as a vector for analysis of gene function by silencing. Plant J. 25:237-245.
https://doi.org/10.1046/j.0960-7412.2000.00942.x Crossref, Medline, ISI, Google Scholar
109. Tijsterman M, Okihara KL, Thijssen K, & Plasterk RHA (2002). PPW-1, a PAZ/PIWI protein
required for efficient germline RNAi, is defective in a natural isolate of C. elegans. Curr Biol, 12,
1535-40. doi:10.1016/S0960-9822(02)01110-7
110. Dirks, R.H.G., van Dun, C.M.P. and Reinink, K. (2003) Rijk Zwaan Zaadteelt en Zaadhandel B.V.
Reverse Breeding,WO⁄ 2003 ⁄ 017753.
111. Jain, S., Sopory, S.K. and Veilleux, R.E. (1996) In vitro haploid production in higher plantsIn
Current Plant Science and Biotechnology in Agriculture, Dordrecht, Boston, London:Kluwer
Academic Publishers.
112. Forster, B.P., Heberle-Bors, E., Kasha, K.J. and Touraev, A. (2007) The resurgence of haploids in
higher plants. Trends Plant Sci. 12, 368–375.
113. Maluszynski, M., Kasha, K.J. and Szarejko, I. (2003) Published protocols for other crop plant
species. In Doubled Haploid Production in Crop Plants: A Manual (Maluszynski, M., Kasha, K.J.,
Forster, B.P. and Szarejko, I., eds), pp. 309–336, Dordrecht, the Netherlands: Kluwer Academic
Publishers.
114. Schaart, Jan & Visser, Richard. (2009). Novel plant breeding techniques. Consequences of new
genetic modification-based plant breeding techniques in comparison to conventional plant
breeding.
115. Mathieu O, and Bender J (2004). RNA-directed DNA methylation. J Cell Sci 117: 4881-4888.
116. Mette MF, van der Winden J, Matzke M and, Matzke AJM (2002) Short RNAs can identify new
candidate transposable element families in Arabidopsis. Plant Physiol 130: 6-9.
117. Hohn T, Corsten S, Rieke S, Muller M, and Rothnie H (1996) Methylation of coding region alone
inhibits gene expression in plant protoplasts. Proc Natl Acad Sci USA 93: 8334-8339.
118. Li X, Wang X, He K, Ma Y, Su N, He H, Stolc V, Tongprasit W, Jin W, Jiang J, Terzaghi W, Li S, and
Deng XW (2008) High-resolution mapping of epigenetic modifications of the rice genome
uncovers interplay between DNA methylation, histone methylation, and gene expression. Plant
Cell 20: 259-276.
119. Shibuya K, Fukushima S and Takatsuji H (2009) RNA-directed DNA methylation induces
transcriptional activation in plants. Proc Natl Acad Sci USA 106: 1660-1665.
120. Chen, Q., Lai, H., Hurtado, J., Stahnke, J., Leuzinger, K., & Dent, M. (2013). Agroinfiltration as an
Effective and Scalable Strategy of Gene Delivery for Production of Pharmaceutical Proteins.
Advanced techniques in biology & medicine, 1(1), 103. https://doi.org/10.4172/atbm.1000103
121. Wroblewski T, Tomczak A, Michelmore R. (2005) Optimization of Agrobacterium-mediated
transient assays of gene expression in lettuce, tomato and Arabidopsis. Plant Biotechnol J.;
3:259–273. [PubMed: 17173625]
122. Huang Z, Chen Q, Hjelm B, Arntzen C, Mason H. (2009) A DNA replicon system for rapid highlevel production of virus-like particles in plants. Biotechnol Bioeng.103:706–714.
[PubMed:19309755]
123. Huang Z, Phoolcharoen W, Lai H, Piensook K, Cardineau G, et al. (2010) High level rapid
production of full-size monoclonal antibodies in plants by a singlevector DNA replicon system.
Biotechnol Bioeng. 106:9–17. [PubMed: 20047189]
124. Vaghchhipawala Z, Rojas CM, Senthil-Kumar M, Mysore KS. (2011) Agroinoculation and
agroinfiltration: simple tools for complex gene function analyses. Methods Mol Biol. 678:65–76.
[PubMed: 20931373]
125. Qiang, C. (2011) Transgenic Horticultural Crops: Challenges, and Opportunities-Essays by
Experts. Boca Raton: Taylor & Francis, USA. Expression and manufacture of pharmaceutical
proteins in genetically engineered horticultural plants
126. Zeinipour, M., Azadi, P., Majd, A. et al. Agroinfiltration: a rapid and reliable method to select
suitable rose cultivars for blue flower production. Physiol Mol Biol Plants 24, 503–511 (2018).
https://doi.org/10.1007/s12298-018-0516-5
127. Hammond-Kosack, K. E., Harrison, K., and Jones, J. D. G. (1994) Developmental^ regulated cell
death on expression of the fungal avirulence gene Avr9 in tomato seedlings carrying the
diseaseresistance gene Cf-9. Proc. Natl. Acad. Sci. USA 9 1, 10445-10449.
128. Hammond-Kosack, K. E., Staskawicz, B. J., Jones, J. D. G., and Baulcombe, D. C. (1995).
Functional expression of a fungal avirulence gene from a modified potato virus X genome. Mol.
PlantMicrobe Interact. 8, 181-185.
129. Hammond-Kosack, K. E., Tang, S., Harrison, K., and Jones, J. D. G. (1998) 130. The tomato Cf-9
disease resistance gene functions in tobacco and potato to confer responsiveness to the fungal
avirulence gene product Avr9. Plant Cell 10, 1251-1266.
130. Joosten, M. H. A. J., Vogelsang, R., Cozijnsen, T. J., Verberne, M. C, and De Wit, P. J. G. M.
(1997) The biotrophic fungus Cladosporium fulvum circumvents C/-4-mediated resistance by
producing unstable AVR4 elicitors. Plant Cell 9, 367-379.
131. Ben-Amar, A., Cobanov, P., Buchholz, G. et al. (2013) In planta agro-infiltration system for
transient gene expression in grapevine (Vitis spp.). Acta Physiol Plant 35, 3147–3156.
https://doi.org/10.1007/s11738-013-1348-0
132. Takeda, A.; Sugiyama, K.; Nagano, H.; Mori, M.; Kaido, M.; Mise, K.; Tsuda, S.; Okuno, T. (2002)
Identification of a novel RNA silencing suppressor, NSs protein of Tomato spotted wilt virus.
FEBS Lett. 532, 75–79.
133. Dugdale, B.; Mortimer, C.L.; Kato, M.; James, T.; Harding, R.M.; Dale, J.L. (2014) Design and
construction of an in-plant activation cassette for transgene expression and recombinant
protein production in plants. Nat. Protoc. 9, 1010–1027.
134. Wroblewski, T.; Tomczak, A.; Michelmore, R. (2005) Optimization of Agrobacterium-mediated
transient assays of gene expression in lettuce, tomato and Arabidopsis. Plant Biotech. J. 3, 259–
273.
135. Fujiuchi, N.; Matsuda, R.; Matoba, N.; Fujiwara, K. (2016) Removal of bacterial suspension
water occupying the intercellular space of detached leaves after agroinfiltration improves the
yield of recombinant hemagglutinin in a N benthamiana transient gene expression system.
Biotechnol. Bioeng. 113, 901–906.
136. Zhao, H., Tan, Z., Wen, X., & Wang, Y. (2017). An Improved Syringe Agroinfiltration Protocol to
Enhance Transformation Efficiency by Combinative Use of 5-Azacytidine, Ascorbate Acid and
Tween-20. Plants (Basel, Switzerland), 6(1), 9. https://doi.org/10.3390/plants6010009
137. Molla K.A., Karmakar S., Islam M.T. (2020) Wide Horizons of CRISPR-Cas-Derived Technologies
for Basic Biology, Agriculture, and Medicine. In: Islam M.T., Bhowmik P.K., Molla K.A. (eds)
CRISPR-Cas Methods. Springer Protocols Handbooks. Humana, New York, NY.
https://doi.org/10.1007/978-1-0716-0616-2_1
138.
Download