Research article

Evidence for novel epigenetic marks within plants

  • Received: 22 May 2019 Accepted: 07 December 2019 Published: 24 December 2019
  • Variation in patterns of gene expression can result from modifications in the genome that occur without a change in the sequence of the DNA; such modifications include methylation of cytosine to generate 5-methylcytosine (5mC) resulting in the generation of heritable epimutation and novel epialleles. This type of non-sequence variation is called epigenetics. The enzymes responsible for generation of such DNA modifications in mammals are named DNA methyltransferases (DNMT) including DNMT1, DNMT2 and DNMT3. The later stages of oxidations to these modifications are catalyzed by Ten Eleven Translocation (TET) proteins, which contain catalytic domains belonging to the 2-oxoglutarate dependent dioxygenase family. In various mammalian cells/tissues including embryonic stem cells, cancer cells and brain tissues, it has been confirmed that these proteins are able to induce the stepwise oxidization of 5-methyl cytosine to 5-hydroxymethylcytosine (5hmC), 5-formylcytosine (5fC), and finally 5-carboxylcytosine (5caC). Each stage from initial methylation until the end of the DNA demethylation process is considered as a specific epigenetic mark that may regulate gene expression. This review discusses controversial evidence for the presence of such oxidative products, particularly 5hmC, in various plant species. Whereas some reports suggest no evidence for enzymatic DNA demethylation, other reports suggest that the presence of oxidative products is followed by the active demethylation and indicate the contribution of possible TET-like proteins in the regulation of gene expression in plants. The review also summarizes the results obtained by expressing the human TET conserved catalytic domain in transgenic plants.

    Citation: Asaad M Mahmood, Jim M Dunwell. Evidence for novel epigenetic marks within plants[J]. AIMS Genetics, 2019, 6(4): 70-87. doi: 10.3934/genet.2019.4.70

    Related Papers:

  • Variation in patterns of gene expression can result from modifications in the genome that occur without a change in the sequence of the DNA; such modifications include methylation of cytosine to generate 5-methylcytosine (5mC) resulting in the generation of heritable epimutation and novel epialleles. This type of non-sequence variation is called epigenetics. The enzymes responsible for generation of such DNA modifications in mammals are named DNA methyltransferases (DNMT) including DNMT1, DNMT2 and DNMT3. The later stages of oxidations to these modifications are catalyzed by Ten Eleven Translocation (TET) proteins, which contain catalytic domains belonging to the 2-oxoglutarate dependent dioxygenase family. In various mammalian cells/tissues including embryonic stem cells, cancer cells and brain tissues, it has been confirmed that these proteins are able to induce the stepwise oxidization of 5-methyl cytosine to 5-hydroxymethylcytosine (5hmC), 5-formylcytosine (5fC), and finally 5-carboxylcytosine (5caC). Each stage from initial methylation until the end of the DNA demethylation process is considered as a specific epigenetic mark that may regulate gene expression. This review discusses controversial evidence for the presence of such oxidative products, particularly 5hmC, in various plant species. Whereas some reports suggest no evidence for enzymatic DNA demethylation, other reports suggest that the presence of oxidative products is followed by the active demethylation and indicate the contribution of possible TET-like proteins in the regulation of gene expression in plants. The review also summarizes the results obtained by expressing the human TET conserved catalytic domain in transgenic plants.


    加载中


    Conflict of interest



    The authors declare no conflict of interest.

    [1] C Song, C He (2011) The hunt for 5-hydroxymethylcytosine: The sixth base. Epigenomics 3: 521–523. doi: 10.2217/epi.11.74
    [2] Paszkowski J, Whitham SA (2001) Gene silencing and DNA methylation processes. Current Opinion Plant Biol 4: 123–129. doi: 10.1016/S1369-5266(00)00147-3
    [3] Bender J (2004) DNA methylation and epigenetics. Annu Rev Plant Biol 55: 41–68. doi: 10.1146/annurev.arplant.55.031903.141641
    [4] Holoch D, Moazed D (2015) RNA-mediated epigenetic regulation of gene expression. Nat Rev Genet 16: 71. doi: 10.1038/nrg3863
    [5] Zhang X, Yazaki J, Sundaresan A, et al. (2006) Genome-wide high-resolution mapping and functional analysis of DNA methylation in Arabidopsis. Cell 126: 1189–1201. doi: 10.1016/j.cell.2006.08.003
    [6] Zhang J, Mei L, Liu R, et al. (2014) Possible involvement of locus-specific methylation on expression regulation of LEAFY homologous gene (CiLFY) during precocious trifoliate orange phase change process. PloS One 9: e88558. doi: 10.1371/journal.pone.0088558
    [7] Yang H, Liu Y, Bai F, et al. (2013) Tumor development is associated with decrease of TET gene expression and 5-methylcytosine hydroxylation. Oncogene 32: 663–669. doi: 10.1038/onc.2012.67
    [8] Ho-Shing O, Dulac C (2019) Influences of genomic imprinting on brain function and behavior. Current Opinion Behav Sci 25: 66–76. doi: 10.1016/j.cobeha.2018.08.008
    [9] Feil R, Berger F (2007) Convergent evolution of genomic imprinting in plants and mammals. Trends Genet 23: 192–199. doi: 10.1016/j.tig.2007.02.004
    [10] Klosinska M, Picard CL, Gehring M (2016) Conserved imprinting associated with unique epigenetic signatures in the Arabidopsis genus. Nat Plants 2: 16145. doi: 10.1038/nplants.2016.145
    [11] Scott RJ, Spielman M (2004) Epigenetics: Imprinting in plants and mammals-the same but different? Current Biol 14: R201–R203. doi: 10.1016/j.cub.2004.02.022
    [12] Feng S, Jacobsen SE (2011) Epigenetic modifications in plants: An evolutionary perspective. Current Opinion Plant Biol 14: 179–186. doi: 10.1016/j.pbi.2010.12.002
    [13] Reinders J, Wulff BB, Mirouze M, et al. (2009) Compromised stability of DNA methylation and transposon immobilization in mosaic Arabidopsis epigenomes. Genet Dev 23: 939–950. doi: 10.1101/gad.524609
    [14] Zemach A, McDaniel IE, Silva P, et al. (2010) Genome-wide evolutionary analysis of eukaryotic DNA methylation. Science 328: 916–919. doi: 10.1126/science.1186366
    [15] Zhang X, Shiu S, Cal A, et al. (2008) Correction: Global analysis of genetic, epigenetic and transcriptional polymorphisms in Arabidopsis thaliana using whole genome tiling arrays. PLoS Genet 4.
    [16] Cao X, Jacobsen SE (2002) Locus-specific control of asymmetric and CpNpG methylation by the DRM and CMT3 methyltransferase genes. Proc Nat Acad Sci 99: 16491–16498. doi: 10.1073/pnas.162371599
    [17] Feng S, Jacobsen SE, Reik W (2010) Epigenetic reprogramming in plant and animal development. Science 330: 622–627. doi: 10.1126/science.1190614
    [18] Huang SsC, Ecker JR (2018) Piecing together cis‐regulatory networks: Insights from epigenomics studies in plants. Wiley Interdiscip Rev: Syst Biol Med 10: e1411.
    [19] Luo C, Hajkova P, Ecker JR (2018) Dynamic DNA methylation: in the right place at the right time. Science 361: 1336–1340. doi: 10.1126/science.aat6806
    [20] Rangwala SH, Richards EJ (2004) The value-added genome: Building and maintaining genomic cytosine methylation landscapes. Current Opinion Genet Dev 14: 686–691. doi: 10.1016/j.gde.2004.09.009
    [21] He Y, Li B, Li Z, et al. (2011) Tet-mediated formation of 5-carboxylcytosine and its excision by TDG in mammalian DNA. Science 333: 1303–1307. doi: 10.1126/science.1210944
    [22] Torres MA, Dangl JL (2005) Functions of the respiratory burst oxidase in biotic interactions, abiotic stress and development. Current Opinion Plant Biol 8: 397–403. doi: 10.1016/j.pbi.2005.05.014
    [23] Feinberg AP, Koldobskiy MA, Göndör A (2016) Epigenetic modulators, modifiers and mediators in cancer aetiology and progression. Nature Rev Genet 17: 284. doi: 10.1038/nrg.2016.13
    [24] Piccolo FM, Fisher AG (2014) Getting rid of DNA methylation. Trends Cell Biol 24: 136–143. doi: 10.1016/j.tcb.2013.09.001
    [25] Hardwick JS, Lane AN, Brown T (2018) Epigenetic modifications of cytosine: Biophysical properties, regulation, and function in mammalian DNA. BioEssays 40.
    [26] Kondo H, Shiraya T, Wada KC, et al. (2010) Induction of flowering by DNA demethylation in Perilla frutescens and Silene armeria: Heritability of 5-azacytidine-induced effects and alteration of the DNA methylation state by photoperiodic conditions. Plant Sci 178: 321–326. doi: 10.1016/j.plantsci.2010.01.012
    [27] Marfil CF, Asurmendi S, Masuelli RW (2012) Changes in micro RNA expression in a wild tuber-bearing Solanum species induced by 5-Azacytidine treatment. Plant Cell Rep 31: 1449–1461. doi: 10.1007/s00299-012-1260-x
    [28] Law JA, Jacobsen SE (2010) Establishing, maintaining and modifying DNA methylation patterns in plants and animals. Nature Rev Genet 11: 204. doi: 10.1038/nrg2719
    [29] Kohli RM, Zhang Y (2013) TET enzymes, TDG and the dynamics of DNA demethylation. Nature 502: 472. doi: 10.1038/nature12750
    [30] Krokan HE, Bjørås M (2013) Base excision repair. Cold Spring Harbor Perspect Biol 5: a012583.
    [31] Ooi SK, Bestor TH (2008) The colorful history of active DNA demethylation. Cell 133: 1145–1148. doi: 10.1016/j.cell.2008.06.009
    [32] Iwan K, Rahimoff R, Kirchner A, et al. (2018) 5-Formylcytosine to cytosine conversion by C–C bond cleavage in vivo. Nature Chem Biol 14: 72. doi: 10.1038/nchembio.2531
    [33] Pfaffeneder T, Hackner B, Truß M, et al. (2011) The discovery of 5‐formylcytosine in embryonic stem cell DNA. Angew Chemie Int Ed 50: 7008–7012. doi: 10.1002/anie.201103899
    [34] Mohr F, Döhner K, Buske C, et al. (2011) TET genes: New players in DNA demethylation and important determinants for stemness. Exp Hematol 39: 272–281. doi: 10.1016/j.exphem.2010.12.004
    [35] Shen L, Zhang Y (2013) 5-Hydroxymethylcytosine: Generation, fate, and genomic distribution. Current Opinion Cell Biol 25: 289–296. doi: 10.1016/j.ceb.2013.02.017
    [36] Verrijzer CP, Tjian R (1996) TAFs mediate transcriptional activation and promoter selectivity. Trends Biochem Sci 21: 338–342. doi: 10.1016/0968-0004(96)10044-X
    [37] Ito S, D'alessio AC, Taranova OV, et al. (2010) Role of Tet proteins in 5mC to 5hmC conversion, ES-cell self-renewal and inner cell mass specification. Nature 466: 1129. doi: 10.1038/nature09303
    [38] Szwagierczak A, Bultmann S, Schmidt CS, et al. (2010) Sensitive enzymatic quantification of 5-hydroxymethylcytosine in genomic DNA. Nucleic Acid Res 38: e181–e181. doi: 10.1093/nar/gkq684
    [39] Koh KP, Yabuuchi A, Rao S, et al. (2011) Tet1 and Tet2 regulate 5-hydroxymethylcytosine production and cell lineage specification in mouse embryonic stem cells. Cell Stem Cell 8: 200–213. doi: 10.1016/j.stem.2011.01.008
    [40] Zhao H, Chen T (2013) Tet family of 5-methylcytosine dioxygenases in mammalian development. J Human Genet 58: 421. doi: 10.1038/jhg.2013.63
    [41] Dunwell JM, Purvis A, Khuri S (2004) Cupins: The most functionally diverse protein superfamily? Phytochemistry 65: 7–17. doi: 10.1016/j.phytochem.2003.08.016
    [42] Brooks SC, Fischer RL, Huh JH, et al. (2014) 5-methylcytosine recognition by Arabidopsis thaliana DNA glycosylases DEMETER and DML3. Biochemistry 53: 2525–2532. doi: 10.1021/bi5002294
    [43] Dunwell TL, Holland PW (2017) A sister of NANOG regulates genes expressed in pre-implantation human development. Open Biol 7: 170027. doi: 10.1098/rsob.170027
    [44] Jin S, Zhang Z, Dunwell TL, et al. (2016) Tet3 reads 5-carboxylcytosine through its CXXC domain and is a potential guardian against neurodegeneration. Cell Rep 14: 493–505. doi: 10.1016/j.celrep.2015.12.044
    [45] Wu X, Zhang Y (2017) TET-mediated active DNA demethylation: Mechanism, function and beyond. Nature Rev Genet 18: 517.
    [46] Koivunen P, Laukka T (2018) The TET enzymes. Cell Mol Life Sci 75: 1339–1348. doi: 10.1007/s00018-017-2721-8
    [47] Bhutani N, Brady JJ, Damian M, et al. (2010) Reprogramming towards pluripotency requires AID-dependent DNA demethylation. Nature 463: 1042. doi: 10.1038/nature08752
    [48] Jin S, Wu X, Li AX, et al. (2011) Genomic mapping of 5-hydroxymethylcytosine in the human brain. Nucleic Acid Res 39: 5015–5024. doi: 10.1093/nar/gkr120
    [49] Ficz G, Branco MR, Seisenberger S, et al. (2011) Dynamic regulation of 5-hydroxymethylcytosine in mouse ES cells and during differentiation. Nature 473: 398. doi: 10.1038/nature10008
    [50] Pastor WA, Pape UJ, Huang Y, et al. (2011) Genome-wide mapping of 5-hydroxymethylcytosine in embryonic stem cells. Nature 473: 394. doi: 10.1038/nature10102
    [51] Williams K, Christensen J, Helin K (2012) DNA methylation: TET proteins-guardians of CpG islands? EMBO Rep 13: 28–35.
    [52] Iqbal K, Jin S, Pfeifer GP, et al. (2011) Reprogramming of the paternal genome upon fertilization involves genome-wide oxidation of 5-methylcytosine. Proc Nat Acad Sci 108: 3642–3647. doi: 10.1073/pnas.1014033108
    [53] Robertson J, Robertson AB, Klungland A (2011) The presence of 5-hydroxymethylcytosine at the gene promoter and not in the gene body negatively regulates gene expression. Biochem Biophys Res Commun 411: 40–43. doi: 10.1016/j.bbrc.2011.06.077
    [54] Taylor SE, Smeriglio P, Dhulipala L, et al. (2014) A global increase in 5‐hydroxymethylcytosine levels marks osteoarthritic chondrocytes. Arthritis Rheumatol 66: 90–100. doi: 10.1002/art.38200
    [55] Laird A, Thomson JP, Harrison DJ, et al. (2013) 5-hydroxymethylcytosine profiling as an indicator of cellular state. Epigenomics 5: 655–669. doi: 10.2217/epi.13.69
    [56] Pfeifer GP, Xiong W, Hahn MA, et al. (2014) The role of 5-hydroxymethylcytosine in human cancer. Cell Tissue Res 356: 631–641. doi: 10.1007/s00441-014-1896-7
    [57] Ruzov A, Tsenkina Y, Serio A, et al. (2011) Lineage-specific distribution of high levels of genomic 5-hydroxymethylcytosine in mammalian development. Cell Res 21: 1332. doi: 10.1038/cr.2011.113
    [58] Yao B, Jin P (2014) Cytosine modifications in neurodevelopment and diseases. Cellular Mol Life Sci 71: 405–418. doi: 10.1007/s00018-013-1433-y
    [59] Mayer JE, Pfeiffer WH, Beyer P (2008) Biofortified crops to alleviate micronutrient malnutrition. Current Opinion Plant Biol 11: 166–170. doi: 10.1016/j.pbi.2008.01.007
    [60] Ma DK, Jang M, Guo JU, et al. (2009) Neuronal activity-induced Gadd45b promotes epigenetic DNA demethylation and adult neurogenesis. Science 323: 1074–1077. doi: 10.1126/science.1166859
    [61] W Yaish A-LA, Al-Harrasi I, Patankar HV (2018) Genome-wide DNA Methylation analysis in response to salinity in the model plant caliph medic (Medicago truncatula). BMC Genomics 19: 1–17. doi: 10.1186/s12864-017-4368-0
    [62] Al Harrasi I A-YR, Yaish MW (2017) Detection of differential DNA methylation under stress conditions using bisulfite sequence analysis. Plant Stress Tolerance Methods in Molecular Biology. New York, NY: Humana Press, 121–137.
    [63] Gehring M, Henikoff S (2008) DNA methylation and demethylation in Arabidopsis. The Arabidopsis Book/American Society of Plant Biologists 6.
    [64] Gehring M, Huh JH, Hsieh TF, et al. (2006) DEMETER DNA glycosylase establishes MEDEA polycomb gene self-imprinting by allele-specific demethylation. Cell 124: 495–506. doi: 10.1016/j.cell.2005.12.034
    [65] Penterman J, Zilberman D, Huh JH, et al. (2007) DNA demethylation in the Arabidopsis genome. Pro Nat Acad Sci 104: 6752–6757. doi: 10.1073/pnas.0701861104
    [66] Lister R, O'Malley RC, Tonti-Filippini J, et al. (2008) Highly integrated single-base resolution maps of the epigenome in Arabidopsis. Cell 133: 523–536. doi: 10.1016/j.cell.2008.03.029
    [67] Zhang JZ, Mei L, Liu R, et al. (2014) Possible involvement of locus-specific methylation on expression regulation of leafy homologous gene (CiLFY) during precocious trifoliate orange phase change process. PLoS One 9: e88558. doi: 10.1371/journal.pone.0088558
    [68] Gong Z, Zhu JK (2011) Active DNA demethylation by oxidation and repair. Cell Res 21: 1649–1651. doi: 10.1038/cr.2011.140
    [69] Jang H, Shin H, Eichman BF, et al. (2014) Excision of 5-hydroxymethylcytosine by DEMETER family DNA glycosylases. Biochem Biophy Res Comm 446: 1067–1072. doi: 10.1016/j.bbrc.2014.03.060
    [70] Terragni J, Bitinaite J, Zheng Y, et al. (2012) Biochemical characterization of recombinant beta-glucosyltransferase and analysis of global 5-hydroxymethylcytosine in unique genomes. Biochemistry 51: 1009–1019. doi: 10.1021/bi2014739
    [71] Hewitson K, Granatino N, Welford R, et al. (2005) Oxidation by 2-oxoglutarate oxygenases: Non-haem iron systems in catalysis and signalling. Philos Trans R Soc London 363: 807–828. doi: 10.1098/rsta.2004.1540
    [72] Hausinger RP (2015) Biochemical diversity of 2-oxoglutarate-dependent oxygenases. 2-Oxoglutarate-dependent oxygenases. Royal Soc Chem, 1–58.
    [73] Hagel J, Facchini P (2018) Expanding the roles for 2-oxoglutarate-dependent oxygenases in plant metabolism. Natural Prod Rep 35: 721–734. doi: 10.1039/C7NP00060J
    [74] Cheynier V, Comte G, Davies KM, et al. (2013) Plant phenolics: Recent advances on their biosynthesis, genetics, and ecophysiology. Plant Physiol Biochem 72: 1–20. doi: 10.1016/j.plaphy.2013.05.009
    [75] Martens S, Preuß A, Matern U (2010) Multifunctional flavonoid dioxygenases: Flavonol and anthocyanin biosynthesis in Arabidopsis thaliana L. Phytochemistry 71: 1040–1049. doi: 10.1016/j.phytochem.2010.04.016
    [76] Barboza L, Effgen S, Alonso-Blanco C, et al. (2013) Arabidopsis semidwarfs evolved from independent mutations in GA20ox1, ortholog to green revolution dwarf alleles in rice and barley. Proc Nat Acad Sci: 201314979.
    [77] Monna L, Kitazawa N, Yoshino R, et al. (2002) Positional cloning of rice semidwarfing gene, sd-1: Rice "green revolution gene" encodes a mutant enzyme involved in gibberellin synthesis. DNA Res 9: 11–17. doi: 10.1093/dnares/9.1.11
    [78] Reagon M, Thurber CS, Olsen KM, et al. (2011) The long and the short of it: SD1 polymorphism and the evolution of growth trait divergence in US weedy rice. Mol Ecol 20: 3743–3756. doi: 10.1111/j.1365-294X.2011.05216.x
    [79] Kawai Y, Ono E, Mizutani M (2014) Evolution and diversity of the 2–oxoglutarate‐dependent dioxygenase superfamily in plants. Plant J 78: 328–343. doi: 10.1111/tpj.12479
    [80] Myllyharju J (2003) Prolyl 4-hydroxylases, the key enzymes of collagen biosynthesis. Matrix Biol 22: 15–24. doi: 10.1016/S0945-053X(03)00006-4
    [81] Tiainen P, Myllyharju J, Koivunen P (2005) Characterization of a second Arabidopsis thaliana prolyl 4-hydroxylase with distinct substrate specificity. J Biol Chem 280: 1142–1148. doi: 10.1074/jbc.M411109200
    [82] Zdzisińska B, Żurek A, Kandefer-Szerszeń M (2017) Alpha-ketoglutarate as a molecule with pleiotropic activity: Well-known and novel possibilities of therapeutic use. Arch Immunol Ther Exp 65: 21–36. doi: 10.1007/s00005-016-0406-x
    [83] Vlad F, Spano T, Vlad D, et al. (2007) Arabidopsis prolyl 4‐hydroxylases are differentially expressed in response to hypoxia, anoxia and mechanical wounding. Physiol Plant 130: 471–483. doi: 10.1111/j.1399-3054.2007.00915.x
    [84] Zhu JK (2009) Active DNA demethylation mediated by DNA glycosylases. Annu Rev Genet 43: 143–166. doi: 10.1146/annurev-genet-102108-134205
    [85] Shi DQ, Ali I, Tang J, et al. (2017) New insights into 5hmC DNA modification: Generation, distribution and function. Front Genet 8: 100. doi: 10.3389/fgene.2017.00100
    [86] Yao Q, Song CX, He C, et al. (2012) Heterologous expression and purification of Arabidopsis thaliana VIM1 protein: In vitro evidence for its inability to recognize hydroxymethylcytosine, a rare base in Arabidopsis DNA. Protein Expr Purif 83: 104–111. doi: 10.1016/j.pep.2012.03.003
    [87] Moricova P, Ondrej V, Navratilova B, et al. (2013) Changes of DNA methylation and hydroxymethylation in plant protoplast cultures. Acta Biochim Pol 60: 33–36.
    [88] Liu S, Dunwell TL, Pfeifer GP, et al. (2013) Detection of oxidation products of 5-methyl-2'-deoxycytidine in Arabidopsis DNA. PLoS One 8: e84620. doi: 10.1371/journal.pone.0084620
    [89] Dunwell JM GG (2011) Detection of 5hmC in Arabidopsis using fluorescent antibody method.
    [90] Wang XL, Song SH, Wu YS, et al. (2015) Genome-wide mapping of 5-hydroxymethylcytosine in three rice cultivars reveals its preferential localization in transcriptionally silent transposable element genes. J Exp Bot 66: 6651–6663. doi: 10.1093/jxb/erv372
    [91] Erdmann RM, Souza AL, Clish CB, et al. (2014) 5-hydroxymethylcytosine is not present in appreciable quantities in Arabidopsis DNA. G3 (Bethesda) 5: 1–8.
    [92] Golubov A, Kovalchuk I (2017) Analysis of DNA hydroxymethylation using colorimetric assay. Plant Epigenet: Springer, 89–97.
    [93] Wang X, Guo T, Wang S, et al. (2017) Determination of 5-hydroxymethyl-2′-deoxycytidine in Rice by high-performance liquid chromatography–tandem mass spectrometry with isotope dilution. Anal Lett 50: 2351–2358. doi: 10.1080/00032719.2017.1286668
    [94] Huber SM, van Delft P, Mendil L, et al. (2015) Formation and abundance of 5‐hydroxymethylcytosine in RNA. Chembiochem 16: 752–755. doi: 10.1002/cbic.201500013
    [95] Tang Y, Xiong J, Jiang H, et al. (2014) Determination of oxidation products of 5-methylcytosine in plants by chemical derivatization coupled with liquid chromatography/tandem mass spectrometry analysis. Anal Chem 86: 7764–7772. doi: 10.1021/ac5016886
    [96] Dawlaty MM, Breiling A, Le T, et al. (2013) Combined deficiency of Tet1 and Tet2 causes epigenetic abnormalities but is compatible with postnatal development. Dev Cell 24: 310–323. doi: 10.1016/j.devcel.2012.12.015
    [97] Iyer LM, Tahiliani M, Rao A, et al. (2009) Prediction of novel families of enzymes involved in oxidative and other complex modifications of bases in nucleic acids. Cell Cycle 8: 1698–1710. doi: 10.4161/cc.8.11.8580
    [98] Aravind L, Anantharaman V, Zhang D, et al. (2012) Gene flow and biological conflict systems in the origin and evolution of eukaryotes. Front Cell Infect Microbiol 2: 89.
    [99] Hollwey E, Watson M, Meyer P (2016) Expression of the C-terminal domain of mammalian TET3 DNA dioxygenase in Arabidopsis thaliana induces heritable methylation changes at rDNA loci. Adv Biosci Biotechnol 7: 243–250. doi: 10.4236/abb.2016.75023
    [100] Hollwey E, Out S, Watson MR, et al. (2017) TET 3‐mediated demethylation in tomato activates expression of a CETS gene that stimulates vegetative growth. Plant Direct 1: e00022. doi: 10.1002/pld3.22
    [101] Ji L, Jordan WT, Shi X, et al. (2018) TET-mediated epimutagenesis of the Arabidopsis thaliana methylome. Nature Comm 9: 895. doi: 10.1038/s41467-018-03289-7
    [102] Gallego-Bartolomé J, Gardiner J, Liu W, et al. (2018) Targeted DNA demethylation of the Arabidopsis genome using the human TET1 catalytic domain. Proc Nat Acad Sci 115: E2125–E2134. doi: 10.1073/pnas.1716945115
    [103] Price J, Antunez-Sanchez J, Hussain N, et al. (2019) Importance of parental genome balance in the generation of novel yet heritable epigenetic and transcriptional states during doubled haploid breeding. BioRxiv: 812347.
    [104] Thakore PI, Black JB, Hilton IB, et al. (2016) Editing the epigenome: Technologies for programmable transcription and epigenetic modulation. Nature Methods 13: 127–137. doi: 10.1038/nmeth.3733
    [105] Dunwell JM (2010) Haploids in flowering plants: Origins and exploitation. Plant Biotechnol J8: 377–424.
    [106] Kawakatsu T, Ecker JR (2019) Diversity and dynamics of DNA methylation: Epigenomic resources and tools for crop breeding. Breed Sci: 19005.
    [107] Delatte B, Wang F, Ngoc LV, et al. (2016) Transcriptome-wide distribution and function of RNA hydroxymethylcytosine. Science 351: 282–285. doi: 10.1126/science.aac5253
    [108] Frye M, Harada BT, Behm M, et al. (2018) RNA modifications modulate gene expression during development. Science 361: 1346–1349. doi: 10.1126/science.aau1646
    [109] Dietzsch J, Feineis D, Höbartner C (2018) Chemoselective labeling and site‐specific mapping of 5‐formylcytosine as a cellular nucleic acid modification. FEBS Letters 592: 2032–2047. doi: 10.1002/1873-3468.13058
    [110] Ecsedi S, Rodríguez-Aguilera J, Hernandez-Vargas H (2018) 5-Hydroxymethylcytosine (5hmC), or how to identify your favorite cell. Epigenomes 2: 3. doi: 10.3390/epigenomes2010003
    [111] Gabrieli T, Sharim H, Nifker G, et al. (2018) Epigenetic optical mapping of 5-hydroxymethylcytosine in nanochannel arrays. ACS Nano 12: 7148–7158. doi: 10.1021/acsnano.8b03023
    [112] Roberts C, Raner G, Isaacs G (2018) High performance liquid chromatography separation of epigenetic cytosine variants. Methods Protocols 1: 10. doi: 10.3390/mps1020010
    [113] Shahal T, Koren O, Shefer G, et al. (2018) Hypersensitive quantification of global 5-hydroxymethylcytosine by chemoenzymatic tagging. Analytica Chimica Acta 1038: 87–96. doi: 10.1016/j.aca.2018.08.035
    [114] Song CX, Yin S, Ma L, et al. (2017) 5-Hydroxymethylcytosine signatures in cell-free DNA provide information about tumor types and stages. Cell Res 27: 1231. doi: 10.1038/cr.2017.106
    [115] Xu Y, Liu S, Li J, et al. (2018) Real-time sensing of TET2-mediated DNA demethylation In Vitro by metal–organic framework-based oxygen sensor for mechanism analysis and stem-cell behavior prediction. Anal Chem 90: 9330–9337. doi: 10.1021/acs.analchem.8b01941
    [116] Sedlazeck FJ, Lee H, Darby CA, et al. (2018) Piercing the dark matter: Bioinformatics of long-range sequencing and mapping. Nature Rev Genet 19: 329. doi: 10.1038/s41576-018-0003-4
    [117] Karemaker ID, Vermeulen M (2018) Single-cell DNA methylation profiling: Technologies and biological applications. Trends Biotechnol 36: 952–965. doi: 10.1016/j.tibtech.2018.04.002
    [118] Jin S, Jiang Y, Qiu R, et al. (2011) 5-Hydroxymethylcytosine is strongly depleted in human cancers but its levels do not correlate with IDH1 mutations. Cancer Res 71: 7360–7365. doi: 10.1158/0008-5472.CAN-11-2023
    [119] Dunwell TL, McGuffin LJ, Dunwell JM, et al. (2013) The mysterious presence of a 5-methylcytosine oxidase in the Drosophila genome: Possible explanations. Cell Cycle 12: 3357–3365. doi: 10.4161/cc.26540
    [120] Wang F, Minakhina S, Tran H, et al. (2018) Tet protein function during Drosophila development. PloS One 13: e0190367. doi: 10.1371/journal.pone.0190367
    [121] Piergiorge RM, de Miranda AB, Guimarães AC, et al. (2017) Functional analogy in human metabolism: Enzymes with different biological roles or functional redundancy? Genome Bioland Evol 9: 1624–1636. doi: 10.1093/gbe/evx119
    [122] Omelchenko MV, Galperin MY, Wolf YI, et al. (2010) Non-homologous isofunctional enzymes: A systematic analysis of alternative solutions in enzyme evolution. Biol Direct 5: 31. doi: 10.1186/1745-6150-5-31
    [123] Sukharnikov LO, Cantwell BJ, Podar M, et al. (2011) Cellulases: Ambiguous nonhomologous enzymes in a genomic perspective. Trends Biotechnol 29: 473–479. doi: 10.1016/j.tibtech.2011.04.008
    [124] Bastard K, Perret A, Mariage A, et al. (2017) Parallel evolution of non-homologous isofunctional enzymes in methionine biosynthesis. Nature Chem Biol 13: 858. doi: 10.1038/nchembio.2397
  • Reader Comments
  • © 2019 the Author(s), licensee AIMS Press. This is an open access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0)
通讯作者: 陈斌, bchen63@163.com
  • 1. 

    沈阳化工大学材料科学与工程学院 沈阳 110142

  1. 本站搜索
  2. 百度学术搜索
  3. 万方数据库搜索
  4. CNKI搜索

Metrics

Article views(4398) PDF downloads(654) Cited by(0)

Article outline

Figures and Tables

Figures(4)  /  Tables(1)

Other Articles By Authors

/

DownLoad:  Full-Size Img  PowerPoint
Return
Return

Catalog