Open Access

Roles of ten‑eleven translocation family proteins and their O‑linked β‑N‑acetylglucosaminylated forms in cancer development (Review)

  • Authors:
    • Hong-Jiao Li
    • Yi Wang
    • Bing-Xin Li
    • Yang Yang
    • Feng  Guan
    • Xing-Chen Pang
    • Xiang Li
  • View Affiliations

  • Published online on: November 3, 2020     https://doi.org/10.3892/ol.2020.12262
  • Article Number: 1
  • Copyright: © Li et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Members of the ten‑eleven translocation (TET) protein family of which three mammalian TET proteins have been discovered so far, catalyze the sequential oxidation of 5‑methylcytosine to 5‑hydroxymethylcytosine, 5‑formylcytosine, and 5‑carboxylcytosine which serve an important role in embryonic development and tumor progression. O‑GlcNAcylation (O‑linked β‑N‑acetylglucosaminylation) is a reversible post‑translational modification known to serve important roles in tumorigenesis and metastasis especially in hematopoietic malignancies such as myelodysplastic syndromes, chronic myelomonocytic leukemia and acute myeloid leukemia. O‑GlcNAcylation activity requires only two enzymes: O‑GlcNAc transferase (OGT) and O‑GlcNAcase (OGA). OGT catalyzes attachment of GlcNAc sugar to serine, threonine and cytosine residues in proteins, while OGA hydrolyzes O‑GlcNAc attached to proteins. Numerous recent studies have demonstrated that TETs can be O‑GlcNAcylated by OGT, with consequent alteration of TET activity and stability. The present review focuses on the cellular, biological and biochemical functions of TET and its O‑GlcNAcylated form and proposes a model of the role of TET/OGT complex in regulation of target proteins during cancer development. In addition, the present review provides directions for future research in this area.

Introduction

Methylation of cytosines, a common epigenetic modification in eukaryotic cells, serves an important role in a variety of genetic processes, including gene stability and expression, chromosome accessibility and inactivation, and nucleosome positioning (1). 5-methylcytosine (5mC) is produced by DNA methyltransferase activity and is located in CG dinucleotides in DNA (2). Ten-eleven translocation (TET) family proteins participate in oxidation reactions of 5mC to 5-hydroxymethylcytosine, 5-formylcytosine, and 5-carboxylcytosine (5hmC, 5fC, 5caC), which further decrease DNA methylation patterns (2).

Post-translational modifications (PTMs) of proteins facilitate immediate responses of cells to intracellular or extracellular environmental stimuli by modifying functions of targeted proteins (3). PTMs are involved in various pathological processes such as proliferation, apoptosis and migration in tumors (3). O-linked β-N-acetylglucosaminylation (O-GlcNAcylation) is an atypical, dynamic and reversible PTM consisting of addition of N-acetyl-D-glucosamine, a unique non-elongated monosaccharide on proteins (4). Unlike classical glycosylation present in the endoplasmic reticulum and Golgi apparatus, O-GlcNAcylation takes place in the cytoplasm, nucleus and mitochondria and is implicated in a wide range of effects on cellular function and signaling in metabolic diseases and cancer (5). Compared to complex glycosyltransferase and glycosidase system of classical glycosylation, O-GlcNAcylation is only regulated by two enzymes: The glycosyltransferase OGT (O-GlcNAc transferase) and the glycoside hydrolase OGA (O-GlcNAcase) (3). Numerous recent studies indicate a close connection between OGT and TET (57). OGT can catalyze TET to form O-GlcNAcylated TET and can also interact with TET to form a complex with the ability to further modify chromatin which participated in regulating embryonic development and cancer progression (6,7). The present review focuses on the functional roles of TET family proteins and O-GlcNAcylation in cancer progression, with focus on the connection between TET proteins and OGT to clarify the effects of these proteins on cancer development.

TET family proteins

Epigenetic modifications, which include DNA methylation and histone modifications can alter gene expression but cannot change the primary sequence of DNA (8) Epigenetic modifications has been proven to be widely involved in tumor development (911). DNA hypermethylation is observed in myelodysplastic syndromes (MDS), acute myeloid leukemia (AML), colorectal cancer, hepatocellular carcinoma and ovarian cancer (1214). TET family proteins function as DNA hydroxymethylases in vertebrates and can catalyze conversion of 5mC to 5hmC, and subsequently to 5fC and 5caC (15) (Fig. 1). These three versions of oxidized methylcytosines are all associated with DNA demethylation (16,17).

TET proteins have a common cysteine-rich dioxygenase region and C-terminal region which binds to ferrous iron and α-ketoglutarate and catalyzes an oxidation reaction which involves hydroxylation of 5mC to 5hmC and further to 5fC and 5caC (12). The three TET proteins (TET1, TET2, TET3) have differing N-terminal regions (18). TET1 and TET3 have a CXXC-type zinc finger domain (19). TET2 has no CXXC DNA-binding domain, but can interact with a CXXC domain protein, inhibitor of disheveled Dvl and Axin complex (IDAX) (18).

TET proteins are highly expressed in embryonic stem cells (ESCs) (20). They are essential for ESC differentiation during embryogenesis and help regulate homeostasis of hematopoietic stem cells, mesenchymal stem celsl and progenitor cells (21). TET1 and TET2 are upregulated in ESCs and TET3 in oocytes (22). Expression of TET proteins is closely related to tumor malignancy (2325); and their expression is significantly lower in tumor tissues compared with normal tissues (24,25). TET2 mutation is often observed in hematopoietic neoplasms including myelodysplastic syndromes and chronic myelomonocytic leukemia (2628). TET2 expression enhances self-renewal, proliferation potential, osteoblast differentiation and hematopoietic supportive capacity of bone marrow stem cells in humans and mice (29). Li et al reported somatic mutation frequencies of the TET2 gene as 30% in MDS, 20% in myeloproliferative neoplasms, 42% in chronic myelomonocytic leukemia and 20% in AML (30). In breast cancer cells, TET2 occupies active enhancers and facilitates proper recruitment of estrogen receptor α, which then transcriptionally activates TET2 expression to establish a positive feedback loop between TET2 and estrogen signaling (31). TET2 also exerts tumor-promoting effects in melanoma and osteosarcoma cells (32,33). TET2 expression is enhanced in tumor-associated macrophages and myeloid-derived suppressor cells, and TET2 deletion in myeloid cells results in inhibition of melanoma growth (32). TET2 can target the promoter of interleukin-6 (IL-6) to increase its expression, and elevated IL-6 may promote lung cancer cell metastasis (33).

Studies on aberrant expression of the three types of TET proteins in various types of cancer are summarized in Table I.

Table I.

Aberrant expression of TET proteins in various cancers.

Table I.

Aberrant expression of TET proteins in various cancers.

Name of proteinCancer typesAlterationConsequent effects(Refs.)
TET1Breast cancer, colon cancer, liver cancerDecreased expressionAdvanced cancer stage, nodal metastases and poor overall survival(64,72)
TET2Leukemia, lymphomaMutationAberrant self-renewal and advanced-stage disease(7376)
TET3Ovarian cancerIncreased expressionUnfavorable prognosis(77)
TET3GlioblastomaDecreased expressionPromoted tumorigenesis(78)

[i] TETs, ten-eleven translocation family proteins.

O-GlcNAcylation

O-GlcNAcylation is a reversible PTM that typically targets proteins in the cytoplasm, cell nuclei (34), or mitochondria (35). It can regulate cellular processes at various levels, such as transcription, translation, signal transduction or cell metabolism (36). In general, proteins modified by O-GlcNAcylation are phosphoproteins or parts of macromolecular complexes (phosphoglycerate kinase 1), transcription complexes (p53, c-myc), or nucleopores (transmembrane nucleoporin Pom121, nucleoporin 155) (37). O-GlcNAcylation has also been reported for numerous functional proteins, including epigenetic regulation factors including the TET proteins, the SIN3 transcription regulator family member A-histone deacetylases and the Polycomb group proteins that regulate DNA methylation, chromatin accessibility and chromatin modification (38).

O-GlcNAcylation often affects subcellular localization, stability and function of target proteins (7,39), and in some cases helps modulate protein phosphorylation status, protein stability, enzymatic activity, protein aggregation and interactions with other proteins or DNA (5,36,40). O-GlcNAc activity requires two enzymes: OGT and OGA (41). OGT catalyzes attachment of GlcNAc to serine (Ser), threonine (Thr) and cysteine residues in proteins (40,42).

OGT activity is highly sensitive to the uridine diphosphate GlcNAc level, and is altered by variations of glucose, glutamate or free fatty acid levels in cells (43). OGT activity is associated with epithelial-mesenchymal transition (EMT), p53, Wnt and TGF-β signaling pathways, inflammatory responses and apoptosis in cervical cancer cells (44). Knockdown of OGT in colon cells results in upregulation and altered glycosylation of E-cadherin, an important factor in EMT progression and may disrupt biosynthesis of glycosphingolipids (lactosylceramide, gangliosides and globosides), with consequent reduction of gangliosides (ganglioside 3 and ganglioside 2) but increase of globosides (globoside 3 and globoside 4) (45). Chronic lymphocytic leukemia (CLL) cells demonstrate high expression of O-GlcNAcylated proteins, including p53, c-Myc, and Akt and enhanced protein glycosylation alters intracellular signaling processes (p53 and PI3K/AKT/mTOR signaling pathways) in these cells (46). O-GlcNAcylation increases downstream signaling of toll-like receptors following cytokine stimulation in CLL cells (46). On the other hand, high baseline O-GlcNAc levels inhibit responses to such stimulation, resulting in increased resistance to TLR agonists, chemotherapeutic agents, B cell receptor crosslinking and mitogens (46). Hart et al (36) reported that increased O-GlcNAcylation of Thr58 on c-Myc inhibited c-Myc activity and reduced transformation of non-Hodgkin's lymphoma cells.

OGA has O-GlcNAc hydrolase and associated enzymatic activity of lysine acetyltransferase (4749) and can therefore hydrolyze O-GlcNAc residues from attached proteins (41). Inhibition of OGA expression in rats and mice resulted in increased O-GlcNAcylation of all tissues (50). OGA shows high mRNA expression in lung, colon and breast cancers (49). In colon cell lines, O-GlcNAcylation level was increased by inhibition of OGA but decreased by inhibition of OGT (51). OGA serves an essential role in differentiation of ESCs (52,53). Blocking of O-GlcNAc cycling in mice by OGA knockdown resulted in anatomical defects and notable changes in expression of pluripotency markers such as Nanog, Sox2 and Orthodenticle homeobox 2 (53). OGA knockdown in mouse hematopoietic stem cells reduced progenitor pools, reduced cell stemness of cells, altered transcription of several crucial genes such as hypoxia inducible factor-1α and cyclin dependent kinase inhibitor 1C and increased apoptotic cell number in bone marrow (54).

O-GlcNAcylation of TETs

TET proteins mediate DNA demethylation, while OGT mediates protein O-GlcNAcylation (39,55). These two enzymatic activities may seem to be independent of each other. However, several recent studies have revealed the physical and functional interactions between TETs and OGT.

Firstly, TETs can be O-GlcNAcylated by OGT (6,5658). Addition of a GlcNAc group to Ser and Thr residues of TET proteins inhibits TET phosphorylation, since Ser and Thr are potential phosphorylation sites (59). Cross-talk between modified Ser and Thr residues facilitates rapid adaptation of TET protein localization, activity, or targeting in response to altered environmental conditions or other external stimuli (6,59). Secondly, TETs preferentially associate with or bind to OGT in certain gene promoters located close to CpG-rich transcription start sites, hence regulating transcriptional levels of these genes through epigenetic modification (6). A large proportion of nuclear OGT is complexed with TETs (60). Such TET/OGT-occupied promoter regions are characterized by low levels of DNA modification, suggesting that TET demethylation activity serves a role in regulation of CpG island methylation (6). OGT in TET/OGT complexes also mediates O-GlcNAcylation of nearby histone H2B at Ser112, thereby facilitating lysine120 ubiquitination of H2B and transcriptional activation (61), particularly near transcription start sites (62). Thirdly, the TET/OGT complex can serve as a scaffold for epigenetic complexes, in addition to its own demethylation and O-GlcNAcylation activities (57,63). Host Cell Factor 1, a component of the H3K4 methyltransferase SET1/COMPASS complex (63), can be O-GlcNAcylated by OGT and bind further to the TET2/3/OGT complex to mediate transcriptional activation through methylation on histone 3 lysine 4 (6). TET/OGT complex can also interact with chromatin regulator SIN3 transcription regulator family member A and with several components of the nucleosome remodeling and deacetylase complex, hence enhancing expression of downstream genes, such as single stranded DNA binding protein 2 and LIM homeobox 2 regulated by TET and maintaining ESC pluripotency (57).

Although TET proteins and OGT have been hot topics of research in recent years, very limited knowledge of TET function and TET O-GlcNAcylation in cancer development and progression exists. TET/OGT complex contributes to certain epigenetic modifications, such as DNA demethylation, histone O-GlcNAcylation, histone methylation associated with positive regulation of gene expression, hence providing a direct link between epigenetics and cellular metabolism (62). Hsu et al (64) reported that TET1 demonstrated reduced expression in prostate and breast cancers, and suppresses cancer cell invasion by promoting expression of tissue inhibitors of metalloproteinases. In a study of cervical cancer cells, Guan et al (65) observed that nuclear localization and O-GlcNAcylation of TET3 were modulated by glucose metabolism, and that gene expression was regulated through TET/OGT-mediated epigenetic changes in response to nutrient availability. The role of O-GlcNAcylated TET proteins in cancer progression is an exciting topic for future study. Based on current finding in the field, a working model of the role of TET/OGT complex in regulation of target proteins during cancer development may be proposed (Fig. 2).

Discussion

The TET proteins (TET1, TET2, TET3) catalyze conversion of 5mC to 5hmC. O-GlcNAcylation is a reversible PTM and it can O-GlcNAcylate TETs (59). OGT and O-GlcNAcylation have been clearly demonstrated to serve an important role in tumorigenesis and metastasis (66). TET proteins can recruit the OGT to chromatin, which promotes post-transcriptional modifications of histones and facilitates gene expression (40). It was reported that TET2 mediates OGT modification on H2B Ser112 and is associated with highly transcribed genes (62). In addition, TET/OGT complex can serve as the scaffold for epigenetic complexes (7,63).

However, the present review had some limitations, such as the research of TET/OGT complex mainly focused on the function during embryonic development (6,7,55). The role of TETs and their O-GlcNAcylation in cancer development is largely unknown (60). The essential characteristic of cancer is uncontrolled cell proliferation resulting from accumulated alterations of cell metabolism and signaling pathways (67). One trait of cancer initiation is the dynamics of O-GlcNAcylation are highly sensitive to availability of nutrients and oxygen, determined by the cellular microenvironment (68). Aberrant glucose metabolism in cancer cells may alter O-GlcNAcylation of TET proteins and therefore affect their stability; conversely, TET loss-of function in cancer may influence the nuclear and/or cytoplasmic distribution of OGT, which in turn may affect the stability of tumor suppressors and oncogenes such as p53 (69), MYC (70), and β-catenin (71). The dysregulated expression and loss-of-function mutation of TET family proteins participated in the progress of a variety of cancers especially hematopoietic malignancies (29). Hence, it is logical to raise the question about whether TET/OGT is involved in cancer development and how they get involved. TETs can be post-translationally modified by the nutrient-sensing enzyme OGT, also suggesting a connection between metabolism and the epigenome (6,62). In addition to suggesting a broader role for the TET/OGT complex, the present review provides information about the interaction between OGT and TET proteins, which may provide new insights into the development of cancer.

Conclusions

TET proteins can interact with and undergo O-GlcNAcylation by OGT and O-GlcNAcylation can alter properties of TET enzymes (62). TET/OGT complexes are primarily targeted to promoter regions through interaction of TET with DNA, and TET-linked OGT can O-GlcNAcylate a wide variety of proteins (58). Relationships between OGT and TETs during cancer pathological processes remain to be elucidated. Identification of modified proteins present upstream and downstream of TET/OGT complex will be useful in this regard. The functions of TETs and their O-GlcNAcylation in cancer development is an important topic for future studies.

Acknowledgements

Not applicable.

Funding

This study was supported by grants from the National Natural Science Foundation of China (grant nos. 81470294, 81770123 and 32071274), National Science and Technology Major Project of China (grant no. 2018ZX10302205), 13115 Key Projects of Scientific and Technical Innovation of Shaanxi Province (grant no. 2010ZDKG-53), Natural Science Foundation of Shaanxi Province (grant no. 2018JM3014), Youth Innovation Team of Shaanxi Universities, and Hundred-Talent Program of Shaanxi Province.

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

XL, XCP, HJL, and BXL designed the study and co-wrote the manuscript. YW, FG, and YY were involved in study conception and design, and revised the manuscript for important intellectual content. All authors have read and approved the manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

TET protein

ten-eleven translocation family protein

OGT

O-GlcNAc transferase

OGA

O-GlcNAcase

PTM

post-translational modification

5mC

5-methylcytosine

5hmC

5-hydroxymethylcytosine

MDS

myelodysplastic syndromes

AML

acute myeloid leukemia

ESC

embryonic stem cell

IL-6

interleukin-6

Ser

serine

Thr

threonine

O-GlcNAcylation

O-linked β-N-acetylglucosaminylation

EMT

epithelial-mesenchymal transition

CLL

chronic lymphocytic leukemia

References

1 

Schubeler D: Function and information content of DNA methylation. Nature. 517:321–326. 2015. View Article : Google Scholar : PubMed/NCBI

2 

Scott-Browne JP, Lio CJ and Rao A: TET proteins in natural and induced differentiation. Curr Opin Genet Dev. 46:202–208. 2017. View Article : Google Scholar : PubMed/NCBI

3 

Yang X and Qian K: Protein O-GlcNAcylation: Emerging mechanisms and functions. Nat Rev Mol Cell Biol. 18:452–465. 2017. View Article : Google Scholar : PubMed/NCBI

4 

Wells L, Vosseller K and Hart GW: Glycosylation of nucleocytoplasmic proteins: Signal transduction and O-GlcNAc. Science. 291:2376–2378. 2001. View Article : Google Scholar : PubMed/NCBI

5 

Hart GW, Housley MP and Slawson C: Cycling of O-linked beta-N-acetylglucosamine on nucleocytoplasmic proteins. Nature. 446:1017–1022. 2007. View Article : Google Scholar : PubMed/NCBI

6 

Vella P, Scelfo A, Jammula S, Chiacchiera F, Williams K, Cuomo A, Roberto A, Christensen J, Bonaldi T, Helin K and Pasini D: Tet proteins connect the O-linked N-acetylglucosamine transferase Ogt to chromatin in embryonic stem cells. Mol Cell. 49:645–656. 2013. View Article : Google Scholar : PubMed/NCBI

7 

Hrit J, Goodrich L, Li C, Wang BA, Nie J, Cui X, Martin EA, Simental E, Fernandez J, Liu MY, et al: OGT binds a conserved C-terminal domain of TET1 to regulate TET1 activity and function in development. Elife. 7:e348702018. View Article : Google Scholar : PubMed/NCBI

8 

Baylin SB and Jones PA: A decade of exploring the cancer epigenome-biological and translational implications. Nat Rev Cancer. 11:726–734. 2011. View Article : Google Scholar : PubMed/NCBI

9 

Darılmaz Yüce G and Ortaç Ersoy E: Lung cancer and epigenetic modifications. Tuberk Toraks. 64:163–170. 2016.(In Turkish). View Article : Google Scholar : PubMed/NCBI

10 

Sasanakietkul T, Murtha TD, Javid M, Korah R and Carling T: Epigenetic modifications in poorly differentiated and anaplastic thyroid cancer. Mol Cell Endocrinol. 469:23–37. 2018. View Article : Google Scholar : PubMed/NCBI

11 

Alam R, Abdolmaleky HM and Zhou JR: Microbiome, inflammation, epigenetic alterations, and mental diseases. Am J Med Genet B Neuropsychiatr Genet. 174:651–660. 2017. View Article : Google Scholar : PubMed/NCBI

12 

Ciesielski P, Jozwiak P and Krzeslak A: TET proteins and epigenetic modifications in cancers. Postepy Hig Med Dosw (Online). 69:1371–1383. 2015.(In Polish). View Article : Google Scholar : PubMed/NCBI

13 

Li D and Zeng Z: Epigenetic regulation of histone H3 in the process of hepatocellular tumorigenesis. Biosci Rep. 39:BSR201918152019. View Article : Google Scholar : PubMed/NCBI

14 

Losi L, Lauriola A, Tazzioli E, Gozzi G, Scurani L, D'Arca D and Benhattar J: Involvement of epigenetic modification of TERT promoter in response to all-trans retinoic acid in ovarian cancer cell lines. J Ovarian Res. 12:622019. View Article : Google Scholar : PubMed/NCBI

15 

Tahiliani M, Koh KP, Shen Y, Pastor WA, Bandukwala H, Brudno Y, Agarwal S, Iyer LM, Liu DR, Aravind L and Rao A: Conversion of 5-methylcytosine to 5-hydroxymethylcytosine in mammalian DNA by MLL partner TET1. Science. 324:930–935. 2009. View Article : Google Scholar : PubMed/NCBI

16 

Song J, Moscinski L, Zhang H, Zhang X and Hussaini M: Does SF3B1/TET2 double mutation portend better or worse prognosis Than Isolated SF3B1 or TET2 Mutation? Cancer Genomics Proteomics. 16:91–98. 2019. View Article : Google Scholar : PubMed/NCBI

17 

Shen L, Wu H, Diep D, Yamaguchi S, D'Alessio AC, Fung HL, Zhang K and Zhang Y: Genome-wide analysis reveals TET- and TDG-dependent 5-methylcytosine oxidation dynamics. Cell. 153:692–706. 2013. View Article : Google Scholar : PubMed/NCBI

18 

Ko M, An J, Bandukwala HS, Chavez L, Aijö T, Pastor WA, Segal MF, Li H, Koh KP, Lähdesmäki H, et al: Modulation of TET2 expression and 5-methylcytosine oxidation by the CXXC domain protein IDAX. Nature. 497:122–126. 2013. View Article : Google Scholar : PubMed/NCBI

19 

Good CR, Madzo J, Patel B, Maegawa S, Engel N, Jelinek J and Issa JJ: A novel isoform of TET1 that lacks a CXXC domain is overexpressed in cancer. Nucleic Acids Res. 45:8269–8281. 2017. View Article : Google Scholar : PubMed/NCBI

20 

Koh KP, Yabuuchi A, Rao S, Huang Y, Cunniff K, Nardone J, Laiho A, Tahiliani M, Sommer CA, Mostoslavsky G, et al: Tet1 and Tet2 regulate 5-hydroxymethylcytosine production and cell lineage specification in mouse embryonic stem cells. Cell Stem Cell. 8:200–213. 2011. View Article : Google Scholar : PubMed/NCBI

21 

Dawlaty MM, Breiling A, Le T, Barrasa MI, Raddatz G, Gao Q, Powell BE, Cheng AW, Faull KF, Lyko F and Jaenisch R: Loss of Tet enzymes compromises proper differentiation of embryonic stem cells. Dev Cell. 29:102–111. 2014. View Article : Google Scholar : PubMed/NCBI

22 

Gu TP, Guo F, Yang H, Wu HP, Xu GF, Liu W, Xie ZG, Shi L, He X, Jin SG, et al: The role of Tet3 DNA dioxygenase in epigenetic reprogramming by oocytes. Nature. 477:606–610. 2011. View Article : Google Scholar : PubMed/NCBI

23 

Ito S, Shen L, Dai Q, Wu SC, Collins LB, Swenberg JA, He C and Zhang Y: Tet proteins can convert 5-methylcytosine to 5-formylcytosine and 5-carboxylcytosine. Science. 333:1300–1303. 2011. View Article : Google Scholar : PubMed/NCBI

24 

Cheng J, Guo S, Chen S, Mastriano SJ, Liu C, D'Alessio AC, Hysolli E, Guo Y, Yao H, Megyola CM, et al: An extensive network of TET2-targeting microRNAs regulates malignant hematopoiesis. Cell Rep. 5:471–481. 2013. View Article : Google Scholar : PubMed/NCBI

25 

Yang H, Liu Y, Bai F, Zhang JY, Ma SH, Liu J, Xu ZD, Zhu HG, Ling ZQ, Ye D, et al: Tumor development is associated with decrease of TET gene expression and 5-methylcytosine hydroxylation. Oncogene. 32:663–669. 2013. View Article : Google Scholar : PubMed/NCBI

26 

Haferlach T, Nagata Y, Grossmann V, Okuno Y, Bacher U, Nagae G, Schnittger S, Sanada M, Kon A, Alpermann T, et al: Landscape of genetic lesions in 944 patients with myelodysplastic syndromes. Leukemia. 28:241–247. 2014. View Article : Google Scholar : PubMed/NCBI

27 

Fernandez-Mercado M, Yip BH, Pellagatti A, Davies C, Larrayoz MJ, Kondo T, Pérez C, Killick S, McDonald EJ, Odero MD, et al: Mutation patterns of 16 genes in primary and secondary acute myeloid leukemia (AML) with normal cytogenetics. PLoS One. 7:e423342012. View Article : Google Scholar : PubMed/NCBI

28 

Shih AH, Abdel-Wahab O, Patel JP and Levine RL: The role of mutations in epigenetic regulators in myeloid malignancies. Nat Rev Cancer. 12:599–612. 2012. View Article : Google Scholar : PubMed/NCBI

29 

Li R, Zhou Y, Cao Z, Liu L, Wang J, Chen Z, Xing W, Chen S, Bai J, Yuan W, et al: TET2 loss dysregulates the behavior of bone marrow mesenchymal stromal cells and accelerates Tet2−/−Driven myeloid malignancy progression. Stem Cell Reports. 10:166–179. 2018. View Article : Google Scholar : PubMed/NCBI

30 

Li Z, Cai X, Cai CL, Wang J, Zhang W, Petersen BE, Yang FC and Xu M: Deletion of Tet2 in mice leads to dysregulated hematopoietic stem cells and subsequent development of myeloid malignancies. Blood. 118:4509–4518. 2011. View Article : Google Scholar : PubMed/NCBI

31 

Wang L, Ozark PA, Smith ER, Zhao Z, Marshall SA, Rendleman EJ, Piunti A, Ryan C, Whelan AL, Helmin KA, et al: TET2 coactivates gene expression through demethylation of enhancers. Sci Adv. 4:eaau69862018. View Article : Google Scholar : PubMed/NCBI

32 

Pan W, Zhu S, Qu K, Meeth K, Cheng J, He K, Ma H, Liao Y, Wen X, Roden C, et al: The DNA Methylcytosine Dioxygenase Tet2 sustains immunosuppressive function of Tumor-infiltrating myeloid cells to promote melanoma progression. Immunity. 47:284–297.e5. 2017. View Article : Google Scholar : PubMed/NCBI

33 

Itoh H, Kadomatsu T, Tanoue H, Yugami M, Miyata K, Endo M, Morinaga J, Kobayashi E, Miyamoto T, Kurahashi R, et al: TET2-dependent IL-6 induction mediated by the tumor microenvironment promotes tumor metastasis in osteosarcoma. Oncogene. 37:2903–2920. 2018. View Article : Google Scholar : PubMed/NCBI

34 

Levine ZG and Walker S: The Biochemistry of O-GlcNAc Transferase: Which functions make it essential in mammalian cells? Annu Rev Biochem. 85:631–657. 2016. View Article : Google Scholar : PubMed/NCBI

35 

Ma J, Banerjee P, Whelan SA, Liu T, Wei AC, Ramirez-Correa G, McComb ME, Costello CE, O'Rourke B, Murphy A and Hart GW: Comparative proteomics reveals dysregulated mitochondrial O-GlcNAcylation in diabetic hearts. J Proteome Res. 15:2254–2264. 2016. View Article : Google Scholar : PubMed/NCBI

36 

Hart GW, Slawson C, Ramirez-Correa G and Lagerlof O: Cross talk between O-GlcNAcylation and phosphorylation: Roles in signaling, transcription, and chronic disease. Annu Rev Biochem. 80:825–858. 2011. View Article : Google Scholar : PubMed/NCBI

37 

Love DC and Hanover JA: The hexosamine signaling pathway: Deciphering the ‘O-GlcNAc code’. Sci STKE. 2005:re132005.PubMed/NCBI

38 

Gambetta MC and Muller J: A critical perspective of the diverse roles of O-GlcNAc transferase in chromatin. Chromosoma. 124:429–442. 2015. View Article : Google Scholar : PubMed/NCBI

39 

Bond MR and Hanover JA: O-GlcNAc cycling: A link between metabolism and chronic disease. Annu Rev Nutr. 33:205–229. 2013. View Article : Google Scholar : PubMed/NCBI

40 

Hanover JA, Krause MW and Love DC: Bittersweet memories: Linking metabolism to epigenetics through O-GlcNAcylation. Nat Rev Mol Cell Biol. 13:312–321. 2012. View Article : Google Scholar : PubMed/NCBI

41 

Mulloy B, Dell A, Stanley P and James HP: Structural analysis of glycans. In: Essentials of Glycobiology 3rd. Varki A, Cummings RD, Esko JD, Stanley P, Hart GW, et al: Cold Spring Harbor; NY: pp. 639–652. 2015, PubMed/NCBI

42 

Maynard JC, Burlingame AL and Medzihradszky KF: Cysteine S-linked N-acetylglucosamine (S-GlcNAcylation), A new post-translational modification in mammals. Mol Cell Proteomics. 15:3405–3411. 2016. View Article : Google Scholar : PubMed/NCBI

43 

Berthier A, Vinod M, Porez G, Steenackers A, Alexandre J, Yamakawa N, Gheeraert C, Ploton M, Maréchal X, Dubois-Chevalier J, et al: Combinatorial regulation of hepatic cytoplasmic signaling and nuclear transcriptional events by the OGT/REV-ERBα complex. Proc Natl Acad Sci USA. 115:E11033–E11042. 2018. View Article : Google Scholar : PubMed/NCBI

44 

Gao J, Yang Y, Qiu R, Zhang K, Teng X, Liu R and Wang Y: Proteomic analysis of the OGT interactome: Novel links to epithelial-mesenchymal transition and metastasis of cervical cancer. Carcinogenesis. 39:1222–1234. 2018. View Article : Google Scholar : PubMed/NCBI

45 

Biwi J, Clarisse C, Biot C, Kozak RP, Madunic K, Mortuaire M, Wuhrer M, Spencer DIR, Schulz C, Guerardel Y, et al: OGT Controls the expression and the glycosylation of E-cadherin, and affects glycosphingolipid structures in human colon cell lines. Proteomics. 19:e18004522019. View Article : Google Scholar : PubMed/NCBI

46 

Shi Y, Tomic J, Wen F, Shaha S, Bahlo A, Harrison R, Dennis JW, Williams R, Gross BJ, Walker S, et al: Aberrant O-GlcNAcylation characterizes chronic lymphocytic leukemia. Leukemia. 24:1588–1598. 2010. View Article : Google Scholar : PubMed/NCBI

47 

Hayakawa K, Hirosawa M, Tabei Y, Arai D, Tanaka S, Murakami N, Yagi S and Shiota K: Epigenetic switching by the metabolism- sensing factors in the generation of orexin neurons from mouse embryonic stem cells. J Biol Chem. 288:17099–17110. 2013. View Article : Google Scholar : PubMed/NCBI

48 

Toleman C, Paterson AJ, Whisenhunt TR and Kudlow JE: Characterization of the histone acetyltransferase (HAT) domain of a bifunctional protein with activable O-GlcNAcase and HAT activities. J Biol Chem. 279:53665–53673. 2004. View Article : Google Scholar : PubMed/NCBI

49 

Singh JP, Qian K, Lee JS, Zhou J, Han X, Zhang B, Ong Q, Ni W, Jiang M, Ruan HB, et al: O-GlcNAcase targets pyruvate kinase M2 to regulate tumor growth. Oncogene. 39:560–573. 2020. View Article : Google Scholar : PubMed/NCBI

50 

Macauley MS, Shan X, Yuzwa SA, Gloster TM and Vocadlo DJ: Elevation of Global O-GlcNAc in rodents using a selective O-GlcNAcase inhibitor does not cause insulin resistance or perturb glucohomeostasis. Chem Biol. 17:949–958. 2010. View Article : Google Scholar : PubMed/NCBI

51 

Fuentes-García G, Castañeda-Patlan MC, Vercoutter-Edouart AS, Lefebvre T and Robles-Flores M: O-GlcNAcylation Is Involved in the regulation of stem cell markers expression in colon cancer cells. Front Endocrinol (Lausanne). 10:2892019. View Article : Google Scholar : PubMed/NCBI

52 

Jang H, Kim TW, Yoon S, Choi SY, Kang TW, Kim SY, Kwon YW, Cho EJ and Youn HD: O-GlcNAc regulates pluripotency and reprogramming by directly acting on core components of the pluripotency network. Cell Stem Cell. 11:62–74. 2012. View Article : Google Scholar : PubMed/NCBI

53 

Olivier-Van Stichelen S, Wang P, Comly M, Love DC and Hanover JA: Nutrient-driven O-linked N-acetylglucosamine (O-GlcNAc) cycling impacts neurodevelopmental timing and metabolism. J Biol Chem. 292:6076–6085. 2017. View Article : Google Scholar : PubMed/NCBI

54 

Abramowitz LK, Harly C, Das A, Bhandoola A and Hanover JA: Blocked O-GlcNAc cycling disrupts mouse hematopoeitic stem cell maintenance and early T cell development. Sci Rep. 9:125692019. View Article : Google Scholar : PubMed/NCBI

55 

Delatte B and Fuks F: TET proteins: On the frenetic hunt for new cytosine modifications. Brief Funct Genomics. 12:191–204. 2013. View Article : Google Scholar : PubMed/NCBI

56 

Ito R, Katsura S, Shimada H, Tsuchiya H, Hada M, Okumura T, Sugawara A and Yokoyama A: TET3-OGT interaction increases the stability and the presence of OGT in chromatin. Genes Cells. 19:52–65. 2014. View Article : Google Scholar : PubMed/NCBI

57 

Shi FT, Kim H, Lu W, He Q, Liu D, Goodell MA, Wan M and Songyang Z: Ten-eleven translocation 1 (Tet1) is regulated by O-linked N-acetylglucosamine transferase (Ogt) for target gene repression in mouse embryonic stem cells. J Biol Chem. 288:20776–20784. 2013. View Article : Google Scholar : PubMed/NCBI

58 

Zhang Q, Liu X, Gao W, Li P, Hou J, Li J and Wong J: Differential regulation of the ten-eleven translocation (TET) family of dioxygenases by O-linked β-N-acetylglucosamine transferase (OGT). J Biol Chem. 289:5986–5996. 2014. View Article : Google Scholar : PubMed/NCBI

59 

Bauer C, Gobel K, Nagaraj N, Colantuoni C, Wang M, Müller U, Kremmer E, Rottach A and Leonhardt H: Phosphorylation of TET proteins is regulated via O-GlcNAcylation by the O-linked N-acetylglucosamine transferase (OGT). J Biol Chem. 290:4801–4812. 2015. View Article : Google Scholar : PubMed/NCBI

60 

Singh JP, Zhang K, Wu J and Yang X: O-GlcNAc signaling in cancer metabolism and epigenetics. Cancer Lett. 356:244–250. 2015. View Article : Google Scholar : PubMed/NCBI

61 

Fujiki R, Hashiba W, Sekine H, Yokoyama A, Chikanishi T, Ito S, Imai Y, Kim J, He HH, Igarashi K, et al: GlcNAcylation of histone H2B facilitates its monoubiquitination. Nature. 480:557–560. 2011. View Article : Google Scholar : PubMed/NCBI

62 

Chen Q, Chen Y, Bian C, Fujiki R and Yu X: TET2 promotes histone O-GlcNAcylation during gene transcription. Nature. 493:561–564. 2013. View Article : Google Scholar : PubMed/NCBI

63 

Deplus R, Delatte B, Schwinn MK, Defrance M, Mendez J, Murphy N, Dawson MA, Volkmar M, Putmans P, Calonne E, et al: TET2 and TET3 regulate GlcNAcylation and H3K4 methylation through OGT and SET1/COMPASS. EMBO J. 32:645–655. 2013. View Article : Google Scholar : PubMed/NCBI

64 

Hsu CH, Peng KL, Kang ML, Chen YR, Yang YC, Tsai CH, Chu CS, Jeng YM, Chen YT, Lin FM, et al: TET1 suppresses cancer invasion by activating the tissue inhibitors of metalloproteinases. Cell Rep. 2:568–579. 2012. View Article : Google Scholar : PubMed/NCBI

65 

Guan W, Guyot R, Samarut J, Flamant F, Wong J and Gauthier KC: Methylcytosine dioxygenase TET3 interacts with thyroid hormone nuclear receptors and stabilizes their association to chromatin. Proc Natl Acad Sci USA. 114:8229–8234. 2017. View Article : Google Scholar : PubMed/NCBI

66 

Phoomak C, Silsirivanit A, Park D, Sawanyawisuth K, Vaeteewoottacharn K, Wongkham C, Lam EW, Pairojkul C, Lebrilla CB and Wongkham S: O-GlcNAcylation mediates metastasis of cholangiocarcinoma through FOXO3 and MAN1A1. Oncogene. 37:5648–565. 2018. View Article : Google Scholar : PubMed/NCBI

67 

Liberti MV and Locasale JW: The warburg effect: How does it benefit cancer cells? Trends Biochem Sci. 41:211–228. 2016. View Article : Google Scholar : PubMed/NCBI

68 

Ma Z and Vosseller K: Cancer metabolism and elevated O-GlcNAc in oncogenic signaling. J Biol Chem. 289:34457–34465. 2014. View Article : Google Scholar : PubMed/NCBI

69 

Yang WH, Kim JE, Nam HW, Ju JW, Kim HS, Kim YS and Cho JW: Modification of p53 with O-linked N-acetylglucosamine regulates p53 activity and stability. Nat Cell Biol. 8:1074–1083. 2026. View Article : Google Scholar

70 

Itkonen HM, Minner S, Guldvik IJ, Sandmann MJ, Tsourlakis MC, Berge V, Svindland A, Schlomm T and Mills IG: O-GlcNAc transferase integrates metabolic pathways to regulate the stability of c-MYC in human prostate cancer cells. Cancer Res. 73:5277–5287. 2013. View Article : Google Scholar : PubMed/NCBI

71 

Olivier-Van Stichelen S, Guinez C, Mir AM, Perez-Cervera Y, Liu C, Michalski JC and Lefebvre T: The hexosamine biosynthetic pathway and O-GlcNAcylation drive the expression of β-catenin and cell proliferation. Am J Physiol Endocrinol Metab. 302:E417–E424. 2012. View Article : Google Scholar : PubMed/NCBI

72 

Thomson JP, Ottaviano R, Unterberger EB, Lempiäinen H, Muller A, Terranova R, Illingworth RS, Webb S, Kerr AR, Lyall MJ, et al: Loss of Tet1-Associated 5-hydroxymethylcytosine is concomitant with aberrant promoter hypermethylation in liver cancer. Cancer Res. 76:3097–3108. 2016. View Article : Google Scholar : PubMed/NCBI

73 

Delhommeau F, Dupont S, Della Valle V, James C, Trannoy S, Massé A, Kosmider O, Le Couedic JP, Robert F, Alberdi A, et al: Mutation in TET2 in myeloid cancers. N Engl J Med. 360:2289–2301. 2029. View Article : Google Scholar

74 

Itzykson R, Kosmider O, Renneville A, Gelsi-Boyer V, Meggendorfer M, Morabito M, Berthon C, Adès L, Fenaux P, Beyne-Rauzy O, et al: Prognostic score including gene mutations in chronic myelomonocytic leukemia. J Clin Oncol. 31:2428–2436. 2013. View Article : Google Scholar : PubMed/NCBI

75 

Nibourel O, Kosmider O, Cheok M, Boissel N, Renneville A, Philippe N, Dombret H, Dreyfus F, Quesnel B, Geffroy S, et al: Incidence and prognostic value of TET2 alterations in de novo acute myeloid leukemia achieving complete remission. Blood. 116:1132–1135. 2010. View Article : Google Scholar : PubMed/NCBI

76 

Dominguez PM, Ghamlouch H, Rosikiewicz W, Kumar P, Béguelin W, Fontán L, Rivas MA, Pawlikowska P, Armand M, Mouly E, et al: TET2 deficiency causes germinal center hyperplasia, impairs plasma cell differentiation, and promotes B-cell lymphomagenesis. Cancer Discov. 8:1632–1653. 2018.PubMed/NCBI

77 

Cao T, Pan W, Sun X and Shen H: Increased expression of TET3 predicts unfavorable prognosis in patients with ovarian cancer-a bioinformatics integrative analysis. J Ovarian Res. 12:1012019. View Article : Google Scholar : PubMed/NCBI

78 

Carella A, Tejedor JR, García MG, Urdinguio RG, Bayón GF, Sierra M, López V, García-Toraño E, Santamarina-Ojeda P, Pérez RF, et al: Epigenetic downregulation of TET3 reduces genome-wide 5hmC levels and promotes glioblastoma tumorigenesis. Int J Cancer. 146:373–387. 2020. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2021
Volume 21 Issue 1

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Li H, Wang Y, Li B, Yang Y, Guan F, Pang X and Li X: Roles of ten‑eleven translocation family proteins and their O‑linked β‑N‑acetylglucosaminylated forms in cancer development (Review). Oncol Lett 21: 1, 2021
APA
Li, H., Wang, Y., Li, B., Yang, Y., Guan, F., Pang, X., & Li, X. (2021). Roles of ten‑eleven translocation family proteins and their O‑linked β‑N‑acetylglucosaminylated forms in cancer development (Review). Oncology Letters, 21, 1. https://doi.org/10.3892/ol.2020.12262
MLA
Li, H., Wang, Y., Li, B., Yang, Y., Guan, F., Pang, X., Li, X."Roles of ten‑eleven translocation family proteins and their O‑linked β‑N‑acetylglucosaminylated forms in cancer development (Review)". Oncology Letters 21.1 (2021): 1.
Chicago
Li, H., Wang, Y., Li, B., Yang, Y., Guan, F., Pang, X., Li, X."Roles of ten‑eleven translocation family proteins and their O‑linked β‑N‑acetylglucosaminylated forms in cancer development (Review)". Oncology Letters 21, no. 1 (2021): 1. https://doi.org/10.3892/ol.2020.12262