Skip to main content
Chronic Diseases and Translational Medicine logoLink to Chronic Diseases and Translational Medicine
. 2020 Jan 8;5(4):267–272. doi: 10.1016/j.cdtm.2019.12.001

Ten-eleven translocation-2 affects the fate of cells and has therapeutic potential in digestive tumors

Feng Wang 1, Jing Zhang 1, Jian Qi 1,
PMCID: PMC7004935  PMID: 32055786

Abstract

Ten-eleven translocation (TET) methylcytosine dioxygenases catalyze the oxidative reactions of 5-methylcytosine (5-mC) to 5-hydroxymethylcytosine (5-hmC), 5-formylcytosine (5-fC), and 5-carboxylcytosine (5-caC), which are intermediate steps during DNA demethylation. It is reported that somatic mutations of TET2 gene are identified in a variety of human tumors, especially in hematological malignancies. The tendency and mechanism of cellular differentiation in different systems are affected by TET2 via regulation of associated gene expression or maintenance of demethylated state. TET2 acts as a critical driver of tumorigenesis through the conversion of 5-mC to 5-hmC and successive oxidation products. Sometimes, it requires special interactions and cofactors. Here, we reviewed recent advances in understanding the function of TET2 proteins in regulating cell differentiation, and its role in various tumors focusing on several digestive cancers.

Keywords: Demethylation, Ten-eleven translocation-2, Differentiation, Digestive tumors

Introduction

During DNA methylation, direct methylation of cytosine in CG dinucleotides, which is one of epigenetic modifications is believed to be an irreversible epigenetic event related to gene repression and tumorigenesis.1 Ten-eleven translocation (TET) protein family, comprising three members TET1, TET2, and TET3, provided a new perspective about active demethylation mechanism.2,3 TETs catalyze the oxidation of 5-methylcytosine (5-mC) to 5-hydroxymethylcytosine (5-hmC), 5-formylcytosine (5-fC), and 5-carboxylcytosine (5-caC), and then complete demethylation process with thymine-DNA glycosylase (TDG)-mediated base excision repair mechanism.4,5 In human mesenchymal stem cells, TET1 and TET2 demethylases epigenetically regulate insulin-like growth factor 2 mRNA binding protein 1 (IGF2BP1) hydroxymethylation status of its promoter and its expression.6

TET2 mutations have been reported to be frequently observed in various cancers, such as leucocythemia, including acute myelocytic leukemia (AML), chronic myelomonocytic leukemia (CMML), and myelodysplastic syndrome (MDS).7, 8, 9, 10 TET2 mutations were frequently identified in other hematologic malignancies including angioimmunoblastic T-cell lymphoma and mature lymphoid neoplasms.11,12 The TET2 functions in mutation-mediated activities are mediated by interactions of TET2 with O-linked N-acetylglucosamine (O-GlcNAc) transferase (Ogt),13, 14, 15 Wilms Tumor 1 (WT1) protein,16,17 and CXXC finger protein 4 (IDAX).18 Furthermore, recent studies have demonstrated that TET2 significantly increases in diabetic wound and is associated with insulin sensitivity and pathogenesis of diabetic nephropathy.19, 20, 21 Recent reports suggest that TET2 is the key factor of atherosclerosis, and is involved in regulating the transfer of smooth muscle cell phenotype, ameliorating vascular dysfunction, and inhibiting inflammation.22 TET2 is involved in clonal hematopoiesis resulting in a marked increase in atherosclerotic plaque size in mice, correlating with the risk of atherosclerotic cardiovascular disease.23 Another study demonstrated that TET2 played a critical role in immune regulation and DNA repair to promote tolerance or antitumor immunity and maintain genomic stability.24

This review summarizes the recent progress in understanding TET2 as an epigenetic regulator, emphasizing its roles in regulation of cellular differentiation and mediation in digestive tumors.

TETs are important molecules for epigenetic regulation

DNA methylation is a reversible epigenetic modification that is not associated with any change in DNA sequence but determines chromatin states. There are mainly two mechanisms of DNA demethylation, passive and active demethylation. One is lost during DNA replication due to the lack of a mechanism for maintenance of methylation modifications. The other is mediated by TET and TDG, in which the demethylase eliminates methylation modification of DNA.25, 26, 27, 28, 29, 30 TET proteins convert 5-mC to 5-hmC, leading to DNA demethylation, which was first reported in 2009.5 Structural studies suggested that TET proteins contain several parts including a cysteine-rich domain, a conserved double-stranded β-helix (DSBH) domain, and binding sites for the cofactors Fe(II) and 2-oxoglutarate (2-OG), which form the catalytic region in the C terminus binding to cytosine in CpG context. Interestingly, only TET1 and TET3 have an N-terminal CXXC zinc finger domain that can bind DNA but not TET2.18,31,32 The catalytic domains are the C-termini with cysteine-rich domain and DSBH domain, catalyzing the oxidation of 5-mC to 5-hmC, and further oxidation of 5-hmC to 5-fC and 5-caC in DNA CpG dinucleotides.4,33,34

TET2 regulates cells differentiation and maintains immune homeostasis

T cells differentiation and stability are associated with DNA modification including methylation. It was demonstrated that the loss or mutations of TET2 could have an influence on the differentiation and fate of CD4+ T cell,35, 36, 37 CD8+ T cell,38 and invariant natural killer T (iNKT) cells.39 TET2 catalyzes the oxidation of 5-mC to 5-hmC resulting in gene demethylation.4,40 Treg cell-specific hypomethylation pattern established by TET2 for forkhead box P3 (FOXP3) expression is related to Treg cell differentiation, function, and autoimmune disease.35 Impaired Treg cell differentiation, reduced FOXP3 expression, and inflammatory disease were found in TET2-knockout mice.35,36 The loss of TET2 confers abnormal CD4+ T cell proliferation and differentiation.37 TET2 are guardians of Treg cells stability, which maintains immune homeostasis. In an acute viral infection mice model, TET2 loss stimulated memory CD8+ T cell formation.38 Moreover, abnormal development and proliferation of invariant natural killer T (iNKT) cells were examined in TET2-TET3 double-knockout mice. TET2 and TET3 regulated iNKT-cell-lineage specification showing a skewing towards NKT17-like phenotype.39

In the blood, TET2 regulates erythroid and granular monocyte differentiation of human hematopoietic progenitors and affects human erythropoiesis and terminal myeloid differentiation, with an effect on iron metabolism in erythroblasts.41, 42, 43, 44 In addition, there were some reports that TET2-mediated modification was associated with myoblast differentiation, initiation of adipogenic differentiation, retinal neurogenesis, and conversion of pre-B cells into macrophages.45, 46, 47, 48 For mast cell differentiation and proliferation, and cytokine expression, TET2 also acts as a crucial regulator by oxidizing 5-mC to 5-hmC and via further oxidation products.49 TET2 is down-regulated in human embryonic stem cell (ESC) lines and up-regulated during hematopoietic differentiation, binding to NANOG promoter and leading to differentiation. Genome-wide analysis revealed an increase in 5-hmC in neuroectoderm genes in Sirt6 knockout embryonic stem cells. In TET1/TET2 double-knockdown of ESCs carrying Dox-inducible PRDM14 expression units, the expression of Klf2 and Tcl1 decreased in the presence of leukemia inhibitory factor suggesting that TET1 and TET2 were important for the maintenance of ESC-pluripotency by PRDM14.50, 51, 52

The potential role of TET2 in digestive tumors

It was reported that the expression of TET2 was differently dysregulated across digestive tumors. TET2 expression was significantly up-regulated in gastric cancer, hepatocellular carcinoma, and tumor-infiltrating CD4+ T cells colorectal cancer compared to normal tissues, while it was down-regulated in esophageal squamous cell carcinoma.53, 54, 55, 56 There was also a report that TET2 was markedly down-regulated in gastric cancer tissues compared to normal gastric mucosa.57

In tumor-infiltrating CD4+ T cells of colorectal cancer patients, signal transducers and activators of transcription 5 (STAT5) and TET2 expression were significantly up-regulated, binding to the transcription factor FOXP3 Treg-specific demethylated region, which contributed to DNA demethylation and mRNA transcription suggesting that STAT5 and TET2 played important roles in the pathogenesis of colorectal cancer.53 The expression of TET2 was significantly associated with 5-hmC levels in esophageal epithelial cells, subsequently leading to esophageal squamous cell carcinoma development. Notably, it had a key function in oxidation of 5-mC to 5-hmC in esophageal epithelial cells. However, TET2 was not associated with the prognosis of esophageal squamous cell carcinoma patient.54 mRNA levels of TETs and not TET proteins or 5-hmC levels were significantly higher in gastric cancer than in the corresponding normal tissues In addition, mRNA levels were higher in higher-grade gastric cancers than in lower-grade gastric cancer samples. The increased transcripts were significantly involved in clinicopathological implication including tumor invasion depth, clinical stage, lymph node metastasis, as well as poor overall survival of patients. The knock-down of TET2 inhibited gastric cancer, hepatocellular carcinoma growth, and cell proliferation. TET2 mRNA might play an important oncogenic role independent of the protein in gastric cancer by sequestering the miR-26 resulting in Enhancer of Zeste Homolog 2 (EZH2) overexpression.55,57 Besides, it was reported that TET2 mRNA levels were associated with cancer stage and prognosis in gastric cancer patients. In this study, worse survival was significantly associated with low level of the oncogenic long noncoding RNA (lncRNA-ANRIL), and TET2 knock-down markedly up-regulated the expression of lncRNA-ANRIL, inhibitor of cyclin kinase 4a, inhibitor of cyclin kinase 4b, and alternative reading-frame. Therefore, in gastric cancer, lncRNA-ANRIL was correlated with TET2-mediated effects.57 Subsequently, TET2 repressed E-cadherin expression by interacting with histone deacetylase 1 and reducing the levels of H3K9Ac and H4K16Ac, and attenuated β-catenin transactivation in hepatocellular carcinoma cells.56

In terms of potential therapeutic applications in digestive tumors, TET2 may act as a therapeutic target. Down-regulated TET2 may change the hypomethylation state of tumor-infiltrating CD4(+) T cells in colorectal cancer patients.53 The loss of 5-hmC or TET2 might affect the development of esophageal squamous cell carcinoma and facilitate gastric carcinogenesis.54,55 Knockdown of TET2 inhibited hepatocellular carcinoma growth in vitro and in vivo, as well as the invasive potential hepatocellular carcinoma cells.56 In gastric cancer, TET2 played a tumor suppression role, by inhibiting proliferation and inducing apoptosis of gastric cancer cells. Overexpression of TET2 could restrict the development of cancer.57 Regarding the role of TET2 in chemotherapy resistance, there has been crucial progress in colon cancer studies. For p53-null tumor cells, TET2 acted as a positive contributor to chemotherapy resistant properties, and the sensitivity of anti-cancer treatment increased after TET2 deletion.58

In summary, these findings indicate that the expression pattern and the effect on prognosis of TET2 are the potential mediation mechanisms in digestive tumors and may represent a novel therapeutic target (Table 1).

Table 1.

Role of ten-eleven translocation methylcytosine dioxygenase 2 in digestive tumors.

Cancers TET2 Expression level Related genes Effect on survival or prognosis Reference
Colorectal cancer mRNA↑ FOXP3 unclear Ma et al53
protein↑
(in CD4+ T cells)
Esophageal squamous cell carcinoma 5hmC↓ None Not associated with prognosis Murata et al54
mRNA↓
Gastric cancer mRNA↑(N = 76) EZH2 Poor overall survival Deng et al55
mRNA↓(N = 31) lncRNA-ANRIL Favorable survival Deng et al57
Protein↓
Hepatocellular carcinoma mRNA↑ E-cadherin Poor overall survival Yang et al56
protein↑

TET2: Ten-eleven translocation; FOXP3: transcription factor forkhead box P3; EZH2: Enhancer of Zeste Homolog 2; lncRNA-ANRIL: the oncogenic long noncoding RNA.

Frequent mutations of TET2 in other tumors

Mutations of TET2 gene were frequently identified in myeloid malignancies with a frequency of 20% in MDS, 45% in CMML, 20% in AML, and 20% in myeloproliferative neoplasm.59, 60, 61, 62 TET2 mutation showed adverse prognostic effect63 and inferior overall survival64,65 in AML patients. Mutated TET2 showed a higher response rate to azacitidine in MDS and low blast count AMLs.66 TET2 mutations occurred in early disease evolution in MDS.62,67,68 TET2 mutations were also found in T cell lymphomas69,70 and diffuse large B-cell lymphoma.71,72 Interestingly, there were other genes co-mutating with TET2, such as enhancer of zeste 2 polycomb repressive complex 2 (EZH2), additional sex combs like transcriptional regulator 1 (ASXL1), isocitrate dehydrogenase 1 and 2 (IDH1/2), and TET1.73, 74, 75, 76 The above gene mutations may together promote the development of various hematological malignancies.77

Compared to normal ovarian tissues, a significant reduction in TET2 expression and 5-hmC in epithelial ovarian cancer was discovered.78 It was reported that a rare variation of TET2 was associated with prostate carcinoma in African-American.79 Following this discovery, TET2 loss was associated with cancer progression due to change in the expression of some genes.80 Besides, TET2 interacted with androgen receptor (AR) to influence 5-hmC-mediated tumor progression.80,81 The specific mechanism of TET2 and the effect on survival and prognosis of these tumors remain to be explored extensively.

Conclusion

The current mechanistic understanding of TET2 mutations in relation to hematological malignancies has been clarified. It is clear that the catalytic function of TETs yields to 5-hmC, 5-fC, and 5-caC. The structures of TET2 had provided insights into the enzymatic activities. Dysregulated function of TET2 mutation induces immunologic derangement and malignant clones. This review focused on the effect of TET2 on FOXP3 expression, hypomethylation pattern establishment, iron metabolism, etc. leading to the differentiation fate of some cells, such as Treg cells, and CD8+ T cells, as well as erythropoiesis. Moreover, in this review, several detailed mediations of TET2 in digestive tumors were obtained to explore the potential mechanisms and therapies. More comprehensive biological functions and conclusions of TET2 and specific elucidation of digestive tumors remain to be identified. Advanced research via systematic genomic approaches will provide further insights into the mechanisms of abnormal TET2 regulation.

Funding

This study was supported by a grant from the Health Commission Joint Foundation Project of Hubei Province (WJ2019H056).

Conflict of interest

None.

Edited by Yan-Gang Ren and Yi Cui

Footnotes

Peer review under responsibility of Chinese Medical Association.

References

  • 1.Baylin S.B., Jones P.A. A decade of exploring the cancer epigenome - biological and translational implications. Nat Rev Cancer. 2011;11:726–734. doi: 10.1038/nrc3130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Zhu J.K. Active DNA demethylation mediated by DNA glycosylases. Annu Rev Genet. 2009;43:143–166. doi: 10.1146/annurev-genet-102108-134205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Wu X., Zhang Y. TET-mediated active DNA demethylation: mechanism, function and beyond. Nat Rev Genet. 2017;18:517–534. doi: 10.1038/nrg.2017.33. [DOI] [PubMed] [Google Scholar]
  • 4.He Y.F., Li B.Z., Li Z. Tet-mediated formation of 5-carboxylcytosine and its excision by TDG in mammalian DNA. Science. 2011;333:1303–1307. doi: 10.1126/science.1210944. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Tahiliani M., Koh K.P., Shen Y. Conversion of 5-methylcytosine to 5-hydroxymethylcytosine in mammalian DNA by MLL partner TET1. Science. 2009;324:930–935. doi: 10.1126/science.1170116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Mahaira L.G., Katsara O., Pappou E. IGF2BP1 expression in human mesenchymal stem cells significantly affects their proliferation and is under the epigenetic control of TET1/2 demethylases. Stem Cells Dev. 2014;23:2501–2512. doi: 10.1089/scd.2013.0604. [DOI] [PubMed] [Google Scholar]
  • 7.Jeong M., Goodell M.A. New answers to old questions from genome-wide maps of DNA methylation in hematopoietic cells. Exp Hematol. 2014;42:609–617. doi: 10.1016/j.exphem.2014.04.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Shih A.H., Abdel-Wahab O., Patel J.P., Levine R.L. The role of mutations in epigenetic regulators in myeloid malignancies. Nat Rev Cancer. 2012;12:599–612. doi: 10.1038/nrc3343. [DOI] [PubMed] [Google Scholar]
  • 9.Nibourel O., Kosmider O., Cheok M. Incidence and prognostic value of TET2 alterations in de novo acute myeloid leukemia achieving complete remission. Blood. 2010;116:1132–1135. doi: 10.1182/blood-2009-07-234484. [DOI] [PubMed] [Google Scholar]
  • 10.Chiba S. Dysregulation of TET2 in hematologic malignancies. Int J Hematol. 2017;105:17–22. doi: 10.1007/s12185-016-2122-z. [DOI] [PubMed] [Google Scholar]
  • 11.Lemonnier F., Couronné L., Parrens M. Recurrent TET2 mutations in peripheral T-cell lymphomas correlate with TFH-like features and adverse clinical parameters. Blood. 2012;120:1466–1469. doi: 10.1182/blood-2012-02-408542. [DOI] [PubMed] [Google Scholar]
  • 12.Sakata-Yanagimoto M., Enami T., Yokoyama Y., Chiba S. Disease-specific mutations in mature lymphoid neoplasms: recent advances. Cancer Sci. 2014;105:623–629. doi: 10.1111/cas.12408. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Zhang Q., Liu X., Gao W. Differential regulation of the ten-eleven translocation (TET) family of dioxygenases by O-linked β-N-acetylglucosamine transferase (OGT) J Biol Chem. 2014;289:5986–5996. doi: 10.1074/jbc.M113.524140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Vella P., Scelfo A., Jammula S. Tet proteins connect the O-linked N-acetylglucosamine transferase Ogt to chromatin in embryonic stem cells. Mol Cell. 2013;49:645–656. doi: 10.1016/j.molcel.2012.12.019. [DOI] [PubMed] [Google Scholar]
  • 15.Chen Q., Chen Y., Bian C., Fujiki R., Yu X. TET2 promotes histone O-GlcNAcylation during gene transcription. Nature. 2013;493:561–564. doi: 10.1038/nature11742. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Rampal R., Alkalin A., Madzo J. DNA hydroxymethylation profiling reveals that WT1 mutations result in loss of TET2 function in acute myeloid leukemia. Cell Rep. 2014;9:1841–1855. doi: 10.1016/j.celrep.2014.11.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Wang Y., Xiao M., Chen X. WT1 recruits TET2 to regulate its target gene expression and suppress leukemia cell proliferation. Mol Cell. 2015;57:662–673. doi: 10.1016/j.molcel.2014.12.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Ko M., An J., Bandukwala H.S. Modulation of TET2 expression and 5-methylcytosine oxidation by the CXXC domain protein IDAX. Nature. 2013;497:122–126. doi: 10.1038/nature12052. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Bian F., Ma X., Villivalam S.D. TET2 facilitates PPARγ agonist-mediated gene regulation and insulin sensitization in adipocytes. Metabolism. 2018;89:39–47. doi: 10.1016/j.metabol.2018.08.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Wu D., Hu D., Chen H. Glucose-regulated phosphorylation of TET2 by AMPK reveals a pathway linking diabetes to cancer. Nature. 2018;559:637–641. doi: 10.1038/s41586-018-0350-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Yang L., Zhang Q., Wu Q. Effect of TET2 on the pathogenesis of diabetic nephropathy through activation of transforming growth factor β1 expression via DNA demethylation. Life Sci. 2018;207:127–137. doi: 10.1016/j.lfs.2018.04.044. [DOI] [PubMed] [Google Scholar]
  • 22.Liu Y., Peng W., Qu K. TET2: a novel epigenetic regulator and potential intervention target for atherosclerosis. DNA Cell Biol. 2018;37:517–523. doi: 10.1089/dna.2017.4118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Fuster J.J., MacLauchlan S., Zuriaga M.A. Clonal hematopoiesis associated with TET2 deficiency accelerates atherosclerosis development in mice. Science. 2017;355:842–847. doi: 10.1126/science.aag1381. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Feng Y., Li X., Cassady K., Zou Z., Zhang X. TET2 function in hematopoietic malignancies, immune regulation, and DNA repair. Front Oncol. 2019;9:210. doi: 10.3389/fonc.2019.00210. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Franchini D.M., Schmitz K.M., Petersen-Mahrt S.K. 5-Methylcytosine DNA demethylation: more than losing a methyl group. Annu Rev Genet. 2012;46:419–441. doi: 10.1146/annurev-genet-110711-155451. [DOI] [PubMed] [Google Scholar]
  • 26.Pastor W.A., Aravind L., Rao A. TETonic shift: biological roles of TET proteins in DNA demethylation and transcription. Nat Rev Mol Cell Biol. 2013;14:341–356. doi: 10.1038/nrm3589. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Wu H., Zhang Y. Reversing DNA methylation: mechanisms, genomics, and biological functions. Cell. 2014;156:45–68. doi: 10.1016/j.cell.2013.12.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Bhutani N., Burns D.M., Blau H.M. DNA demethylation dynamics. Cell. 2011;146:866–872. doi: 10.1016/j.cell.2011.08.042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Wu H., Zhang Y. Mechanisms and functions of Tet protein-mediated 5-methylcytosine oxidation. Genes Dev. 2011;25:2436–2452. doi: 10.1101/gad.179184.111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Kohli R.M., Zhang Y. TET enzymes, TDG and the dynamics of DNA demethylation. Nature. 2013;502:472–479. doi: 10.1038/nature12750. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Xu Y., Xu C., Kato A. Tet3 CXXC domain and dioxygenase activity cooperatively regulate key genes for Xenopus eye and neural development. Cell. 2012;151:1200–1213. doi: 10.1016/j.cell.2012.11.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Hu L., Li Z., Cheng J. Crystal structure of TET2-DNA complex: insight into TET-mediated 5mC oxidation. Cell. 2013;155:1545–1555. doi: 10.1016/j.cell.2013.11.020. [DOI] [PubMed] [Google Scholar]
  • 33.Ito S., D'Alessio A.C., Taranova O.V., Hong K., Sowers L.C., Zhang Y. Role of Tet proteins in 5mC to 5hmC conversion, ES-cell self-renewal and inner cell mass specification. Nature. 2010;466:1129–1133. doi: 10.1038/nature09303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Ito S., Shen L., Dai Q. Tet proteins can convert 5-methylcytosine to 5-formylcytosine and 5-carboxylcytosine. Science. 2011;333:1300–1303. doi: 10.1126/science.1210597. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Yang R., Qu C., Zhou Y. Hydrogen sulfide promotes Tet1- and Tet2-mediated Foxp3 demethylation to drive regulatory T cell differentiation and maintain immune homeostasis. Immunity. 2015;43:251–263. doi: 10.1016/j.immuni.2015.07.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Yue X., Lio C.J., Samaniego-Castruita D., Li X., Rao A. Loss of TET2 and TET3 in regulatory T cells unleashes effector function. Nat Commun. 2019;10:2011. doi: 10.1038/s41467-019-09541-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Zang S., Li J., Yang H. Mutations in 5-methylcytosine oxidase TET2 and RhoA cooperatively disrupt T cell homeostasis. J Clin Investig. 2017;127:2998–3012. doi: 10.1172/JCI92026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Carty S.A., Gohil M., Banks L.B. The loss of TET2 promotes CD8+ T cell memory differentiation. J Immunol. 2018;200:82–91. doi: 10.4049/jimmunol.1700559. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Tsagaratou A., González-Avalos E., Rautio S. TET proteins regulate the lineage specification and TCR-mediated expansion of iNKT cells. Nat Immunol. 2017;18:45–53. doi: 10.1038/ni.3630. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Hu L., Lu J., Cheng J. Structural insight into substrate preference for TET-mediated oxidation. Nature. 2015;527:118–122. doi: 10.1038/nature15713. [DOI] [PubMed] [Google Scholar]
  • 41.Yan H., Wang Y., Qu X. Distinct roles for TET family proteins in regulating human erythropoiesis. Blood. 2017;129:2002–2012. doi: 10.1182/blood-2016-08-736587. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Inokura K., Fujiwara T., Saito K. Impact of TET2 deficiency on iron metabolism in erythroblasts. Exp Hematol. 2017;49:56–67. doi: 10.1016/j.exphem.2017.01.002. e5. [DOI] [PubMed] [Google Scholar]
  • 43.Garcia-Gomez A., Li T., Kerick M. TET2- and TDG-mediated changes are required for the acquisition of distinct histone modifications in divergent terminal differentiation of myeloid cells. Nucleic Acids Res. 2017;45:10002–10017. doi: 10.1093/nar/gkx666. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Pronier E., Almire C., Mokrani H. Inhibition of TET2-mediated conversion of 5-methylcytosine to 5-hydroxymethylcytosine disturbs erythroid and granulomonocytic differentiation of human hematopoietic progenitors. Blood. 2011;118:2551–2555. doi: 10.1182/blood-2010-12-324707. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Zhong X., Wang Q.Q., Li J.W., Zhang Y.M., An X.R., Hou J. Ten-eleven translocation-2 (Tet2) is involved in myogenic differentiation of skeletal myoblast cells in vitro. Sci Rep. 2017;7:43539. doi: 10.1038/srep43539. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Yoo Y., Park J.H., Weigel C. TET-mediated hydroxymethylcytosine at the Pparγ locus is required for initiation of adipogenic differentiation. Int J Obes (Lond) 2017;41:652–659. doi: 10.1038/ijo.2017.8. [DOI] [PubMed] [Google Scholar]
  • 47.Kallin E.M., Rodríguez-Ubreva J., Christensen J. Tet2 facilitates the derepression of myeloid target genes during CEBPα-induced transdifferentiation of pre-B cells. Mol Cell. 2012;48:266–276. doi: 10.1016/j.molcel.2012.08.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Seritrakul P., Gross J.M. Tet-mediated DNA hydroxymethylation regulates retinal neurogenesis by modulating cell-extrinsic signaling pathways. PLoS Genet. 2017;13:e1006987. doi: 10.1371/journal.pgen.1006987. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Montagner S., Leoni C., Emming S. TET2 regulates mast cell differentiation and proliferation through catalytic and non-catalytic activities. Cell Rep. 2016;15:1566–1579. doi: 10.1016/j.celrep.2016.04.044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Etchegaray J.P., Chavez L., Huang Y. The histone deacetylase SIRT6 controls embryonic stem cell fate via TET-mediated production of 5-hydroxymethylcytosine. Nat Cell Biol. 2015;17:545–557. doi: 10.1038/ncb3147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Okashita N., Sakashita N., Ito K., Mitsuya A., Suwa Y., Seki Y. PRDM14 maintains pluripotency of embryonic stem cells through TET-mediated active DNA demethylation. Biochem Biophys Res Commun. 2015;466:138–145. doi: 10.1016/j.bbrc.2015.08.122. [DOI] [PubMed] [Google Scholar]
  • 52.Langlois T., da Costa Reis Monte-Mor B., Lenglet G. TET2 deficiency inhibits mesoderm and hematopoietic differentiation in human embryonic stem cells. Stem Cells. 2014;32:2084–2097. doi: 10.1002/stem.1718. [DOI] [PubMed] [Google Scholar]
  • 53.Ma H., Gao W., Sun X., Wang W. STAT5 and TET2 cooperate to regulate FOXP3-TSDR demethylation in CD4+ T cells of patients with colorectal cancer. J Immunol Res. 2018;2018:6985031. doi: 10.1155/2018/6985031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Murata A., Baba Y., Ishimoto T. TET family proteins and 5-hydroxymethylcytosine in esophageal squamous cell carcinoma. Oncotarget. 2015;6:23372–23382. doi: 10.18632/oncotarget.4281. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Deng M., Zhang R., He Z. TET-mediated sequestration of miR-26 drives EZH2 expression and gastric carcinogenesis. Cancer Res. 2017;77:6069–6082. doi: 10.1158/0008-5472.CAN-16-2964. [DOI] [PubMed] [Google Scholar]
  • 56.Yang G., Zeng X., Wang M., Wu A. The TET2/E-cadherin/β-catenin regulatory loop confers growth and invasion in hepatocellular carcinoma cells. Exp Cell Res. 2018;363:218–226. doi: 10.1016/j.yexcr.2018.01.011. [DOI] [PubMed] [Google Scholar]
  • 57.Deng W., Wang J., Zhang J., Cai J., Bai Z., Zhang Z. TET2 regulates LncRNA-ANRIL expression and inhibits the growth of human gastric cancer cells. IUBMB Life. 2016;68:355–364. doi: 10.1002/iub.1490. [DOI] [PubMed] [Google Scholar]
  • 58.Zhang J., Tan P., Guo L. p53-dependent autophagic degradation of TET2 modulates cancer therapeutic resistance. Oncogene. 2019;38:1905–1919. doi: 10.1038/s41388-018-0524-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Abdel-Wahab O., Mullally A., Hedvat C. Genetic characterization of TET1, TET2, and TET3 alterations in myeloid malignancies. Blood. 2009;114:144–147. doi: 10.1182/blood-2009-03-210039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Delhommeau F., Dupont S., Della Valle V. Mutation in TET2 in myeloid cancers. N Engl J Med. 2009;360:2289–2301. doi: 10.1056/NEJMoa0810069. [DOI] [PubMed] [Google Scholar]
  • 61.Solary E., Bernard O.A., Tefferi A., Fuks F., Vainchenker W. The Ten-Eleven Translocation-2 (TET2) gene in hematopoiesis and hematopoietic diseases. Leukemia. 2014;28:485–496. doi: 10.1038/leu.2013.337. [DOI] [PubMed] [Google Scholar]
  • 62.Langemeijer S.M., Kuiper R.P., Berends M. Acquired mutations in TET2 are common in myelodysplastic syndromes. Nat Genet. 2009;41:838–842. doi: 10.1038/ng.391. [DOI] [PubMed] [Google Scholar]
  • 63.Ahn J.S., Kim H.J., Kim Y.K. Adverse prognostic effect of homozygous TET2 mutation on the relapse risk of acute myeloid leukemia in patients of normal karyotype. Haematologica. 2015;100:e351–e353. doi: 10.3324/haematol.2015.126227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Aslanyan M.G., Kroeze L.I., Langemeijer S.M. Clinical and biological impact of TET2 mutations and expression in younger adult AML patients treated within the EORTC/GIMEMA AML-12 clinical trial. Ann Hematol. 2014;93:1401–1412. doi: 10.1007/s00277-014-2055-7. [DOI] [PubMed] [Google Scholar]
  • 65.Yamazaki J., Taby R., Jelinek J. Hypomethylation of TET2 target genes identifies a curable subset of acute myeloid leukemia. J Natl Cancer Inst. 2015;108 doi: 10.1093/jnci/djv323. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Itzykson R., Kosmider O., Cluzeau T. Impact of TET2 mutations on response rate to azacitidine in myelodysplastic syndromes and low blast count acute myeloid leukemias. Leukemia. 2011;25:1147–1152. doi: 10.1038/leu.2011.71. [DOI] [PubMed] [Google Scholar]
  • 67.Idossa D., Lasho T.L., Finke C.M. Mutations and karyotype predict treatment response in myelodysplastic syndromes. Am J Hematol. 2018;93:1420–1426. doi: 10.1002/ajh.25267. [DOI] [PubMed] [Google Scholar]
  • 68.Smith A.E., Mohamedali A.M., Kulasekararaj A. Next-generation sequencing of the TET2 gene in 355 MDS and CMML patients reveals low-abundance mutant clones with early origins, but indicates no definite prognostic value. Blood. 2010;116:3923–3932. doi: 10.1182/blood-2010-03-274704. [DOI] [PubMed] [Google Scholar]
  • 69.da Silva Almeida A.C., Abate F., Khiabanian H. The mutational landscape of cutaneous T cell lymphoma and Sézary syndrome. Nat Genet. 2015;47:1465–1470. doi: 10.1038/ng.3442. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Shimoda K., Shide K., Kameda T. TET2 mutation in adult T-cell leukemia/lymphoma. J Clin Exp Hematop. 2015;55:145–149. doi: 10.3960/jslrt.55.145. [DOI] [PubMed] [Google Scholar]
  • 71.Asmar F., Punj V., Christensen J. Genome-wide profiling identifies a DNA methylation signature that associates with TET2 mutations in diffuse large B-cell lymphoma. Haematologica. 2013;98:1912–1920. doi: 10.3324/haematol.2013.088740. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Kubuki Y., Yamaji T., Hidaka T. TET2 mutation in diffuse large B-cell lymphoma. J Clin Exp Hematop. 2017;56:145–149. doi: 10.3960/jslrt.56.145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Muto T., Sashida G., Oshima M. Concurrent loss of Ezh2 and Tet2 cooperates in the pathogenesis of myelodysplastic disorders. J Exp Med. 2013;210:2627–2639. doi: 10.1084/jem.20131144. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Rocquain J., Carbuccia N., Trouplin V. Combined mutations of ASXL1, CBL, FLT3, IDH1, IDH2, JAK2, KRAS, NPM1, NRAS, RUNX1, TET2 and WT1 genes in myelodysplastic syndromes and acute myeloid leukemias. BMC Cancer. 2010;10:401. doi: 10.1186/1471-2407-10-401. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Zhao Z., Chen L., Dawlaty M.M. Combined loss of Tet1 and Tet2 promotes B cell, but not myeloid malignancies, in mice. Cell Rep. 2015;13:1692–1704. doi: 10.1016/j.celrep.2015.10.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Lin T.L., Nagata Y., Kao H.W. Clonal leukemic evolution in myelodysplastic syndromes with TET2 and IDH1/2 mutations. Haematologica. 2014;99:28–36. doi: 10.3324/haematol.2013.091249. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Bowman R.L., Levine R.L. TET2 in normal and malignant hematopoiesis. Cold Spring Harb Perspect Med. 2017;7 doi: 10.1101/cshperspect.a026518. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Zhang L.Y., Li P.L., Wang T.Z., Zhang X.C. Prognostic values of 5-hmC, 5-mC and TET2 in epithelial ovarian cancer. Arch Gynecol Obstet. 2015;292:891–897. doi: 10.1007/s00404-015-3704-3. [DOI] [PubMed] [Google Scholar]
  • 79.Koboldt D.C., Kanchi K.L., Gui B. Rare variation in TET2 is associated with clinically relevant prostate carcinoma in African Americans. Cancer Epidemiol Biomarkers Prev. 2016;25:1456–1463. doi: 10.1158/1055-9965.EPI-16-0373. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Nickerson M.L., Das S., Im K.M. TET2 binds the androgen receptor and loss is associated with prostate cancer. Oncogene. 2017;36:2172–2183. doi: 10.1038/onc.2016.376. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Takayama K., Misawa A., Suzuki T. TET2 repression by androgen hormone regulates global hydroxymethylation status and prostate cancer progression. Nat Commun. 2015;6:8219. doi: 10.1038/ncomms9219. [DOI] [PubMed] [Google Scholar]

Articles from Chronic Diseases and Translational Medicine are provided here courtesy of Chinese Medical Association

RESOURCES