Skip to main content
Top
Published in: Cancer Cell International 1/2021

Open Access 01-12-2021 | Acute Myeloid Leukemia | Review

The biological function of m6A reader YTHDF2 and its role in human disease

Authors: Jin-yan Wang, Ai-qing Lu

Published in: Cancer Cell International | Issue 1/2021

Login to get access

Abstract

N6-methyladenosine (m6A) modification is a dynamic and reversible post-transcriptional modification and the most prevalent internal RNA modification in eukaryotic cells. YT521-B homology domain family 2 (YTHDF2) is a member of m6A “readers” and its role in human diseases remains unclear. Accumulating evidence suggests that YTHDF2 is greatly implicated in many aspects of human cancers and non-cancers through various mechanisms. YTHDF2 takes a great part in multiple biological processes, such as migration, invasion, metastasis, proliferation, apoptosis, cell cycle, cell viability, cell adhesion, differentiation and inflammation, in both human cancers and non-cancers. Additionally, YTHDF2 influences various aspects of RNA metabolism, including mRNA decay and pre-ribosomal RNA (pre-rRNA) processing. Moreover, emerging researches indicate that YTHDF2 predicts the prognosis of different cancers. Herein, we focus on concluding YTHDF2-associated mechanisms and potential biological functions in kinds of cancers and non-cancers, and its prospects as a prognostic biomarker.
Literature
1.
go back to reference Boccaletto P. et al. MODOMICS: a database of RNA modification pathways. 2017 update. Nucleic Acids Res, 2018. 46(D1): D303-d307. Boccaletto P. et al. MODOMICS: a database of RNA modification pathways. 2017 update. Nucleic Acids Res, 2018. 46(D1): D303-d307.
2.
go back to reference Sánchez-Vásquez E, et al. Emerging role of dynamic RNA modifications during animal development. Mech Dev. 2018;154:24–32.PubMedCrossRef Sánchez-Vásquez E, et al. Emerging role of dynamic RNA modifications during animal development. Mech Dev. 2018;154:24–32.PubMedCrossRef
3.
go back to reference Desrosiers R, Friderici K, Rottman F. Identification of methylated nucleosides in messenger RNA from Novikoff hepatoma cells. Proc Natl Acad Sci U S A. 1974;71(10):3971–5.PubMedPubMedCentralCrossRef Desrosiers R, Friderici K, Rottman F. Identification of methylated nucleosides in messenger RNA from Novikoff hepatoma cells. Proc Natl Acad Sci U S A. 1974;71(10):3971–5.PubMedPubMedCentralCrossRef
4.
go back to reference Maity, A. and B. Das, N6-methyladenosine modification in mRNA: machinery, function and implications for health and diseases. 2016. 283(9): p. 1607–30. Maity, A. and B. Das, N6-methyladenosine modification in mRNA: machinery, function and implications for health and diseases. 2016. 283(9): p. 1607–30.
5.
6.
go back to reference Dominissini D, et al. Topology of the human and mouse m6A RNA methylomes revealed by m6A-seq. Nature. 2012;485(7397):201–6.CrossRefPubMed Dominissini D, et al. Topology of the human and mouse m6A RNA methylomes revealed by m6A-seq. Nature. 2012;485(7397):201–6.CrossRefPubMed
7.
go back to reference Han J, et al. METTL3 promote tumor proliferation of bladder cancer by accelerating pri-miR221/222 maturation in m6A-dependent manner. Mol Cancer. 2019;18(1):110.PubMedPubMedCentralCrossRef Han J, et al. METTL3 promote tumor proliferation of bladder cancer by accelerating pri-miR221/222 maturation in m6A-dependent manner. Mol Cancer. 2019;18(1):110.PubMedPubMedCentralCrossRef
9.
go back to reference Chen S. ALKBH5-mediated m(6)A demethylation of lncRNA PVT1 plays an oncogenic role in osteosarcoma. Stem Cells Int. 2020;20:34. Chen S. ALKBH5-mediated m(6)A demethylation of lncRNA PVT1 plays an oncogenic role in osteosarcoma. Stem Cells Int. 2020;20:34.
10.
go back to reference Wang H, Xu B, Shi J. N6-methyladenosine METTL3 promotes the breast cancer progression via targeting Bcl-2. Gene. 2020;722:144076.PubMedCrossRef Wang H, Xu B, Shi J. N6-methyladenosine METTL3 promotes the breast cancer progression via targeting Bcl-2. Gene. 2020;722:144076.PubMedCrossRef
11.
12.
go back to reference Sun T, Wu R, Ming L. The role of m6A RNA methylation in cancer. Biomed Pharmacother. 2019;112:108613.PubMedCrossRef Sun T, Wu R, Ming L. The role of m6A RNA methylation in cancer. Biomed Pharmacother. 2019;112:108613.PubMedCrossRef
13.
go back to reference Jin, D. and J. Guo. m(6)A demethylase ALKBH5 inhibits tumor growth and metastasis by reducing YTHDFs-mediated YAP expression and inhibiting miR-107/LATS2-mediated YAP activity in NSCLC. 2020. 19(1): p. 40. Jin, D. and J. Guo. m(6)A demethylase ALKBH5 inhibits tumor growth and metastasis by reducing YTHDFs-mediated YAP expression and inhibiting miR-107/LATS2-mediated YAP activity in NSCLC. 2020. 19(1): p. 40.
16.
go back to reference Müller S, et al. IGF2BP1 promotes SRF-dependent transcription in cancer in a m6A- and miRNA-dependent manner. Nucleic Acids Res. 2019;47(1):375–90.PubMedCrossRef Müller S, et al. IGF2BP1 promotes SRF-dependent transcription in cancer in a m6A- and miRNA-dependent manner. Nucleic Acids Res. 2019;47(1):375–90.PubMedCrossRef
17.
go back to reference Huang, H., et al. Recognition of RNA N(6)-methyladenosine by IGF2BP proteins enhances mRNA stability and translation. 2018. 20(3): p. 285–295. Huang, H., et al. Recognition of RNA N(6)-methyladenosine by IGF2BP proteins enhances mRNA stability and translation. 2018. 20(3): p. 285–295.
19.
go back to reference Wu R, et al. Epigallocatechin gallate targets FTO and inhibits adipogenesis in an mRNA m(6)A-YTHDF2-dependent manner. Int J Obes (Lond). 2018;42(7):1378–88.CrossRef Wu R, et al. Epigallocatechin gallate targets FTO and inhibits adipogenesis in an mRNA m(6)A-YTHDF2-dependent manner. Int J Obes (Lond). 2018;42(7):1378–88.CrossRef
20.
go back to reference Paris J, et al. Targeting the RNA m(6)A Reader YTHDF2 Selectively Compromises Cancer Stem Cells in Acute Myeloid Leukemia. Genome Biol. 2019;25(1):137-148.e6. Paris J, et al. Targeting the RNA m(6)A Reader YTHDF2 Selectively Compromises Cancer Stem Cells in Acute Myeloid Leukemia. Genome Biol. 2019;25(1):137-148.e6.
21.
go back to reference Xie, H., et al. METTL3/YTHDF2 m(6) A axis promotes tumorigenesis by degrading SETD7 and KLF4 mRNAs in bladder cancer. 2020. 24(7): p. 4092–4104. Xie, H., et al. METTL3/YTHDF2 m(6) A axis promotes tumorigenesis by degrading SETD7 and KLF4 mRNAs in bladder cancer. 2020. 24(7): p. 4092–4104.
22.
go back to reference Fei Q. YTHDF2 promotes mitotic entry and is regulated by cell cycle mediators. Anal Chem. 2020;18(4):e3000664. Fei Q. YTHDF2 promotes mitotic entry and is regulated by cell cycle mediators. Anal Chem. 2020;18(4):e3000664.
23.
go back to reference Dai X, et al. YTHDF2 Binds to 5-Methylcytosine in RNA and Modulates the Maturation of Ribosomal RNA. Protein Cell. 2020;92(1):1346–54. Dai X, et al. YTHDF2 Binds to 5-Methylcytosine in RNA and Modulates the Maturation of Ribosomal RNA. Protein Cell. 2020;92(1):1346–54.
24.
go back to reference Yu R, et al. m6A Reader YTHDF2 Regulates LPS-Induced Inflammatory Response. Hepatology, 2019. 20(6). Yu R, et al. m6A Reader YTHDF2 Regulates LPS-Induced Inflammatory Response. Hepatology, 2019. 20(6).
25.
go back to reference Wu R, et al. m(6)A methylation controls pluripotency of porcine induced pluripotent stem cells by targeting SOCS3/JAK2/STAT3 pathway in a YTHDF1/YTHDF2-orchestrated manner. Cell Death Dis. 2019;10(3):171.PubMedPubMedCentralCrossRef Wu R, et al. m(6)A methylation controls pluripotency of porcine induced pluripotent stem cells by targeting SOCS3/JAK2/STAT3 pathway in a YTHDF1/YTHDF2-orchestrated manner. Cell Death Dis. 2019;10(3):171.PubMedPubMedCentralCrossRef
26.
go back to reference Zhu T, et al. Crystal structure of the YTH domain of YTHDF2 reveals mechanism for recognition of N6-methyladenosine. RNA. 2014;24(12):1493–6. Zhu T, et al. Crystal structure of the YTH domain of YTHDF2 reveals mechanism for recognition of N6-methyladenosine. RNA. 2014;24(12):1493–6.
27.
go back to reference Ma C, Liao S, Zhu Z. Crystal structure of human YTHDC2 YTH domain. Biochem Biophys Res Commun. 2019;518(4):678–84.PubMedCrossRef Ma C, Liao S, Zhu Z. Crystal structure of human YTHDC2 YTH domain. Biochem Biophys Res Commun. 2019;518(4):678–84.PubMedCrossRef
28.
go back to reference Li F, et al. Structure of the YTH domain of human YTHDF2 in complex with an m(6)A mononucleotide reveals an aromatic cage for m(6)A recognition. Cell Res. 2014;24(12):1490–2.PubMedPubMedCentralCrossRef Li F, et al. Structure of the YTH domain of human YTHDF2 in complex with an m(6)A mononucleotide reveals an aromatic cage for m(6)A recognition. Cell Res. 2014;24(12):1490–2.PubMedPubMedCentralCrossRef
29.
go back to reference Huang G, et al. YTHDF2 destabilizes m(6)A-containing RNA through direct recruitment of the CCR4-NOT deadenylase complex. Cell Res. 2016;7:12626. Huang G, et al. YTHDF2 destabilizes m(6)A-containing RNA through direct recruitment of the CCR4-NOT deadenylase complex. Cell Res. 2016;7:12626.
30.
go back to reference Wang J, et al. Binding to m(6)A RNA promotes YTHDF2-mediated phase separation. 2020. 11(4): p. 304–307. Wang J, et al. Binding to m(6)A RNA promotes YTHDF2-mediated phase separation. 2020. 11(4): p. 304–307.
31.
go back to reference Pardo JC, et al., Moving towards Personalized Medicine in Muscle-Invasive Bladder Cancer: Where Are We Now and Where Are We Going? Int J Mol Sci, 2020. 21(17). Pardo JC, et al., Moving towards Personalized Medicine in Muscle-Invasive Bladder Cancer: Where Are We Now and Where Are We Going? Int J Mol Sci, 2020. 21(17).
32.
go back to reference Li R, et al. Exosome-mediated secretion of LOXL4 promotes hepatocellular carcinoma cell invasion and metastasis. J Cell Physiol. 2019;18(1):18. Li R, et al. Exosome-mediated secretion of LOXL4 promotes hepatocellular carcinoma cell invasion and metastasis. J Cell Physiol. 2019;18(1):18.
33.
go back to reference Chen M, et al. RNA N6-methyladenosine methyltransferase-like 3 promotes liver cancer progression through YTHDF2-dependent posttranscriptional silencing of SOCS2. 2018. 67(6): p. 2254–2270. Chen M, et al. RNA N6-methyladenosine methyltransferase-like 3 promotes liver cancer progression through YTHDF2-dependent posttranscriptional silencing of SOCS2. 2018. 67(6): p. 2254–2270.
34.
go back to reference He C, Zhang C. YTHDF2 promotes the liver cancer stem cell phenotype and cancer metastasis by regulating OCT4 expression via m6A RNA methylation. PLoS Biol, 2020. He C, Zhang C. YTHDF2 promotes the liver cancer stem cell phenotype and cancer metastasis by regulating OCT4 expression via m6A RNA methylation. PLoS Biol, 2020.
35.
go back to reference Hou J, et al. YTHDF2 reduction fuels inflammation and vascular abnormalization in hepatocellular carcinoma. Cell Res. 2019;18(1):163. Hou J, et al. YTHDF2 reduction fuels inflammation and vascular abnormalization in hepatocellular carcinoma. Cell Res. 2019;18(1):163.
36.
go back to reference Zhong L, et al. YTHDF2 suppresses cell proliferation and growth via destabilizing the EGFR mRNA in hepatocellular carcinoma. Cancer Lett. 2019;442:252–61.PubMedCrossRef Zhong L, et al. YTHDF2 suppresses cell proliferation and growth via destabilizing the EGFR mRNA in hepatocellular carcinoma. Cancer Lett. 2019;442:252–61.PubMedCrossRef
37.
go back to reference Zhou, S., et al., FTO regulates the chemo-radiotherapy resistance of cervical squamous cell carcinoma (CSCC) by targeting β-catenin through mRNA demethylation. 2018. 57(5): p. 590–597. Zhou, S., et al., FTO regulates the chemo-radiotherapy resistance of cervical squamous cell carcinoma (CSCC) by targeting β-catenin through mRNA demethylation. 2018. 57(5): p. 590–597.
38.
go back to reference Li Z, et al. Knockdown of YTH N(6)-methyladenosine RNA binding protein 2 (YTHDF2) inhibits cell proliferation and promotes apoptosis in cervical cancer cells. Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi. 2020;36(3):255–63.PubMed Li Z, et al. Knockdown of YTH N(6)-methyladenosine RNA binding protein 2 (YTHDF2) inhibits cell proliferation and promotes apoptosis in cervical cancer cells. Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi. 2020;36(3):255–63.PubMed
39.
go back to reference Liu, T. and S. Yang, Dysregulated N6-methyladenosine methylation writer METTL3 contributes to the proliferation and migration of gastric cancer. 2020. 235(1): p. 548–562. Liu, T. and S. Yang, Dysregulated N6-methyladenosine methylation writer METTL3 contributes to the proliferation and migration of gastric cancer. 2020. 235(1): p. 548–562.
40.
go back to reference Zhang J, et al. Knockdown of YTH N(6)-methyladenosine RNA binding protein 2 (YTHDF2) inhibits proliferation and promotes apoptosis in MGC-803 gastric cancer cells. Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi. 2017;33(12):1628–34.PubMed Zhang J, et al. Knockdown of YTH N(6)-methyladenosine RNA binding protein 2 (YTHDF2) inhibits proliferation and promotes apoptosis in MGC-803 gastric cancer cells. Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi. 2017;33(12):1628–34.PubMed
41.
go back to reference Wang JY, et al. Potential regulatory role of lncRNA-miRNA-mRNA axis in osteosarcoma. Biomed Pharmacother. 2020;121:109627.PubMedCrossRef Wang JY, et al. Potential regulatory role of lncRNA-miRNA-mRNA axis in osteosarcoma. Biomed Pharmacother. 2020;121:109627.PubMedCrossRef
42.
go back to reference Yang J, et al. Circular RNA circ_0001105 Inhibits Progression and Metastasis of Osteosarcoma by Sponging miR-766 and Activating YTHDF2 Expression. Onco Targets Ther. 2020;13:1723–36.PubMedPubMedCentralCrossRef Yang J, et al. Circular RNA circ_0001105 Inhibits Progression and Metastasis of Osteosarcoma by Sponging miR-766 and Activating YTHDF2 Expression. Onco Targets Ther. 2020;13:1723–36.PubMedPubMedCentralCrossRef
43.
go back to reference Xu X, et al. Up-regulation of IGF2BP2 by multiple mechanisms in pancreatic cancer promotes cancer proliferation by activating the PI3K/Akt signaling pathway. J Exp Clin Cancer Res. 2019;38(1):497.PubMedPubMedCentralCrossRef Xu X, et al. Up-regulation of IGF2BP2 by multiple mechanisms in pancreatic cancer promotes cancer proliferation by activating the PI3K/Akt signaling pathway. J Exp Clin Cancer Res. 2019;38(1):497.PubMedPubMedCentralCrossRef
44.
go back to reference Guo X, et al. RNA demethylase ALKBH5 prevents pancreatic cancer progression by posttranscriptional activation of PER1 in an m6A-YTHDF2-dependent manner. Mol Cancer. 2020;19(1):91.PubMedPubMedCentralCrossRef Guo X, et al. RNA demethylase ALKBH5 prevents pancreatic cancer progression by posttranscriptional activation of PER1 in an m6A-YTHDF2-dependent manner. Mol Cancer. 2020;19(1):91.PubMedPubMedCentralCrossRef
45.
go back to reference Wu Y, et al. RNA m(6)A Methyltransferase METTL3 Promotes The Growth Of Prostate Cancer By Regulating Hedgehog Pathway. J Hematol Oncol. 2019;12:9143–52. Wu Y, et al. RNA m(6)A Methyltransferase METTL3 Promotes The Growth Of Prostate Cancer By Regulating Hedgehog Pathway. J Hematol Oncol. 2019;12:9143–52.
46.
go back to reference Li J, et al. Downregulation of N(6)-methyladenosine binding YTHDF2 protein mediated by miR-493-3p suppresses prostate cancer by elevating N(6)-methyladenosine levels. Oncotarget. 2018;9(3):3752–64.PubMedCrossRef Li J, et al. Downregulation of N(6)-methyladenosine binding YTHDF2 protein mediated by miR-493-3p suppresses prostate cancer by elevating N(6)-methyladenosine levels. Oncotarget. 2018;9(3):3752–64.PubMedCrossRef
47.
go back to reference Li Z, et al. FTO Plays an Oncogenic Role in Acute Myeloid Leukemia as a N(6)-Methyladenosine RNA Demethylase. Cancer Cell. 2017;31(1):127–41.PubMedCrossRef Li Z, et al. FTO Plays an Oncogenic Role in Acute Myeloid Leukemia as a N(6)-Methyladenosine RNA Demethylase. Cancer Cell. 2017;31(1):127–41.PubMedCrossRef
48.
go back to reference Nguyen TT, et al. Identification of novel Runx1 (AML1) translocation partner genes SH3D19, YTHDf2, and ZNF687 in acute myeloid leukemia. Genes Chromosomes Cancer. 2006;45(10):918–32.PubMedCrossRef Nguyen TT, et al. Identification of novel Runx1 (AML1) translocation partner genes SH3D19, YTHDf2, and ZNF687 in acute myeloid leukemia. Genes Chromosomes Cancer. 2006;45(10):918–32.PubMedCrossRef
49.
go back to reference Döhner H, Weisdorf DJ, Bloomfield CD. Acute Myeloid Leukemia. N Engl J Med. 2015;373(12):1136–52.PubMedCrossRef Döhner H, Weisdorf DJ, Bloomfield CD. Acute Myeloid Leukemia. N Engl J Med. 2015;373(12):1136–52.PubMedCrossRef
51.
go back to reference Dzierzak E, Bigas A. Blood Development: Hematopoietic Stem Cell Dependence and Independence. Cell Stem Cell. 2018;22(5):639–51.PubMedCrossRef Dzierzak E, Bigas A. Blood Development: Hematopoietic Stem Cell Dependence and Independence. Cell Stem Cell. 2018;22(5):639–51.PubMedCrossRef
52.
go back to reference Walasek MA, van Os R, de Haan G. Hematopoietic stem cell expansion: challenges and opportunities. Ann N Y Acad Sci. 2012;1266:138–50.PubMedCrossRef Walasek MA, van Os R, de Haan G. Hematopoietic stem cell expansion: challenges and opportunities. Ann N Y Acad Sci. 2012;1266:138–50.PubMedCrossRef
53.
go back to reference Li, Z., et al., Suppression of m(6)A reader Ythdf2 promotes hematopoietic stem cell expansion. 2018. 28(9): p. 904–917. Li, Z., et al., Suppression of m(6)A reader Ythdf2 promotes hematopoietic stem cell expansion. 2018. 28(9): p. 904–917.
54.
go back to reference Huang S, et al. Author Correction: Suppression of m(6)A reader Ythdf2 promotes hematopoietic stem cell expansion. Oncogene. 2018;28(10):1042. Huang S, et al. Author Correction: Suppression of m(6)A reader Ythdf2 promotes hematopoietic stem cell expansion. Oncogene. 2018;28(10):1042.
55.
go back to reference Wang, H., et al., Loss of YTHDF2-mediated m(6)A-dependent mRNA clearance facilitates hematopoietic stem cell regeneration. 2018. 28(10): p. 1035–1038. Wang, H., et al., Loss of YTHDF2-mediated m(6)A-dependent mRNA clearance facilitates hematopoietic stem cell regeneration. 2018. 28(10): p. 1035–1038.
56.
go back to reference Takahashi K, et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell. 2007;131(5):861–72.CrossRefPubMed Takahashi K, et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell. 2007;131(5):861–72.CrossRefPubMed
57.
go back to reference Secher JO, et al. Initial embryology and pluripotent stem cells in the pig–The quest for establishing the pig as a model for cell therapy. Theriogenology. 2016;85(1):162–71.PubMedCrossRef Secher JO, et al. Initial embryology and pluripotent stem cells in the pig–The quest for establishing the pig as a model for cell therapy. Theriogenology. 2016;85(1):162–71.PubMedCrossRef
58.
go back to reference Heck AM, et al. YTHDF2 destabilizes m(6)A-modified neural-specific RNAs to restrain differentiation in induced pluripotent stem cells. ACS Chem Biol. 2020;26(6):739–55. Heck AM, et al. YTHDF2 destabilizes m(6)A-modified neural-specific RNAs to restrain differentiation in induced pluripotent stem cells. ACS Chem Biol. 2020;26(6):739–55.
59.
go back to reference Li, M., et al., Ythdf2-mediated m(6)A mRNA clearance modulates neural development in mice. 2018. 19(1): p. 69. Li, M., et al., Ythdf2-mediated m(6)A mRNA clearance modulates neural development in mice. 2018. 19(1): p. 69.
60.
go back to reference Yao Y, et al. METTL3 inhibits BMSC adipogenic differentiation by targeting the JAK1/STAT5/C/EBPβ pathway via an m(6)A-YTHDF2-dependent manner. Faseb j. 2019;33(6):7529–44.PubMedCrossRef Yao Y, et al. METTL3 inhibits BMSC adipogenic differentiation by targeting the JAK1/STAT5/C/EBPβ pathway via an m(6)A-YTHDF2-dependent manner. Faseb j. 2019;33(6):7529–44.PubMedCrossRef
61.
go back to reference Merkestein M, et al. FTO influences adipogenesis by regulating mitotic clonal expansion. Nat Commun. 2015;6:6792.PubMedCrossRef Merkestein M, et al. FTO influences adipogenesis by regulating mitotic clonal expansion. Nat Commun. 2015;6:6792.PubMedCrossRef
62.
go back to reference Wu R, et al. FTO regulates adipogenesis by controlling cell cycle progression via m(6)A-YTHDF2 dependent mechanism. Cell Res. 2018;1863(10):1323–30. Wu R, et al. FTO regulates adipogenesis by controlling cell cycle progression via m(6)A-YTHDF2 dependent mechanism. Cell Res. 2018;1863(10):1323–30.
63.
go back to reference Cai M, et al. Loss of m(6) A on FAM134B promotes adipogenesis in porcine adipocytes through m(6) A-YTHDF2-dependent way. IUBMB Life. 2019;71(5):580–6.PubMedCrossRef Cai M, et al. Loss of m(6) A on FAM134B promotes adipogenesis in porcine adipocytes through m(6) A-YTHDF2-dependent way. IUBMB Life. 2019;71(5):580–6.PubMedCrossRef
65.
go back to reference Martínez-Pérez, M., et al., Arabidopsis m(6)A demethylase activity modulates viral infection of a plant virus and the m(6)A abundance in its genomic RNAs. 2017. 114(40): p. 10755–10760. Martínez-Pérez, M., et al., Arabidopsis m(6)A demethylase activity modulates viral infection of a plant virus and the m(6)A abundance in its genomic RNAs. 2017. 114(40): p. 10755–10760.
66.
go back to reference Gonzales-van Horn, S.R. and P. Sarnow, Making the Mark: The Role of Adenosine Modifications in the Life Cycle of RNA Viruses. Cell Host Microbe, 2017. 21(6): p. 661–669. Gonzales-van Horn, S.R. and P. Sarnow, Making the Mark: The Role of Adenosine Modifications in the Life Cycle of RNA Viruses. Cell Host Microbe, 2017. 21(6): p. 661–669.
67.
go back to reference Hesser, C.R. and J. Karijolich, N6-methyladenosine modification and the YTHDF2 reader protein play cell type specific roles in lytic viral gene expression during Kaposi's sarcoma-associated herpesvirus infection. 2018. 14(4): p. e1006995. Hesser, C.R. and J. Karijolich, N6-methyladenosine modification and the YTHDF2 reader protein play cell type specific roles in lytic viral gene expression during Kaposi's sarcoma-associated herpesvirus infection. 2018. 14(4): p. e1006995.
68.
go back to reference Cardelli M, et al. A polymorphism of the YTHDF2 gene (1p35) located in an Alu-rich genomic domain is associated with human longevity. J Gerontol A Biol Sci Med Sci. 2006;61(6):547–56.PubMedCrossRef Cardelli M, et al. A polymorphism of the YTHDF2 gene (1p35) located in an Alu-rich genomic domain is associated with human longevity. J Gerontol A Biol Sci Med Sci. 2006;61(6):547–56.PubMedCrossRef
69.
go back to reference Kanatsu-Shinohara M, Shinohara T. Spermatogonial stem cell self-renewal and development. Annu Rev Cell Dev Biol. 2013;29:163–87.PubMedCrossRef Kanatsu-Shinohara M, Shinohara T. Spermatogonial stem cell self-renewal and development. Annu Rev Cell Dev Biol. 2013;29:163–87.PubMedCrossRef
70.
go back to reference Huang T, et al. YTHDF2 promotes spermagonial adhesion through modulating MMPs decay via m(6)A/mRNA pathway. PLoS Pathog. 2020;11(1):37. Huang T, et al. YTHDF2 promotes spermagonial adhesion through modulating MMPs decay via m(6)A/mRNA pathway. PLoS Pathog. 2020;11(1):37.
71.
go back to reference Ivanova I, et al. The RNA m(6)A Reader YTHDF2 Is Essential for the Post-transcriptional Regulation of the Maternal Transcriptome and Oocyte Competence. Mol Cell. 2017;67(6):1059-1067.e4.PubMedPubMedCentralCrossRef Ivanova I, et al. The RNA m(6)A Reader YTHDF2 Is Essential for the Post-transcriptional Regulation of the Maternal Transcriptome and Oocyte Competence. Mol Cell. 2017;67(6):1059-1067.e4.PubMedPubMedCentralCrossRef
72.
go back to reference Xie L, Zhao BS, He C. “Gamete On” for m(6)A: YTHDF2 Exerts Essential Functions in Female Fertility. J Cell Mol Med. 2017;67(6):903–5.CrossRef Xie L, Zhao BS, He C. “Gamete On” for m(6)A: YTHDF2 Exerts Essential Functions in Female Fertility. J Cell Mol Med. 2017;67(6):903–5.CrossRef
Metadata
Title
The biological function of m6A reader YTHDF2 and its role in human disease
Authors
Jin-yan Wang
Ai-qing Lu
Publication date
01-12-2021
Publisher
BioMed Central
Published in
Cancer Cell International / Issue 1/2021
Electronic ISSN: 1475-2867
DOI
https://doi.org/10.1186/s12935-021-01807-0

Other articles of this Issue 1/2021

Cancer Cell International 1/2021 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine