Review
5-Hydroxymethylcytosine: An epigenetic mark frequently deregulated in cancer

https://doi.org/10.1016/j.bbcan.2015.01.001Get rights and content

Highlights

  • 5hmC levels are frequently deregulated in cancer.

  • Mutations in TET2, IDH1, IDH2, FH or SDH lead to decreased levels of 5hmC.

  • Changes in DNA (hydroxy)methylation are an important target for cancer therapy.

Abstract

The epigenetic mark 5-hydroxymethylcytosine (5hmC) has gained interest since 2009, when it was discovered that Ten-Eleven-Translocation (TET) proteins catalyze the conversion of 5-methylcytosine (5mC) into 5hmC. This conversion appears to be an intermediate step in the active DNA demethylation pathway. Factors that regulate DNA hydroxymethylation are frequently affected in cancer, leading to deregulated 5hmC levels. In this review, we will discuss the regulation of DNA hydroxymethylation, defects in this pathway in cancer, and novel therapies that may correct deregulated (hydroxy)methylation of DNA.

Introduction

DNA methylation is essential for normal development and plays an important role in many processes, including X-chromosome inactivation, imprinting and transcriptional regulation. Methylation of promoter regions is associated with transcriptional repression, while actively transcribed genes may contain high levels of gene body methylation (from the transcription start site to the end of the transcript) [1]. DNA methylation is mediated by DNA methyltransferases (DNMTs) and predominantly occurs on cytosine residues present in CpG context. DNMT3A and DNMT3B are responsible for de novo methylation, while DNMT1 is mainly involved in maintaining methylcytosine marks during DNA replication (Fig. 1) [2].

DNA demethylation may take place as a passive process due to lack of maintenance methylation during DNA replication. In addition, recent studies presented evidence for an active DNA demethylation pathway initiated by the Ten-Eleven Translocation (TET) protein family. The TET proteins are responsible for the conversion of 5-methylcytosine (5mC) into 5-hydroxymethylcytosine (5hmC), 5-formylcytosine (5fC) and 5-carboxylcytosine (5caC) through three consecutive oxidation reactions [3], [4]. 5fC and 5caC marks are recognized by thymine DNA glycosylase (TDG). TDG activates the base excision repair pathway (BER), which replaces the modified cytosine with an unmodified cytosine (Fig. 1) [5].

Many proteins and cofactors control the balance between methylation and demethylation. In cancer, this balance is frequently deregulated, leading to altered methylation patterns. These changes in methylation can lead to repression of tumor suppressor genes or activation of oncogenes. Recently, it has been shown that hydroxymethylation levels are also altered in various types of cancer [6], [7], [8], [9]. Key events that contributed to the elucidation of the link between deregulated 5hmC levels and cancer are summarized in Fig. 2. Several mechanisms have been described that may be involved in these changes in 5hmC, which will be the focus of this review. Insights into these mechanisms may open new possibilities for treatment.

Section snippets

DNA hydroxymethylation and its regulation

5hmC was initially found in the DNA of bacteriophages [10], and was first reported to be present in mammalian DNA in 1972 [11]. After 1972, the interest for this cytosine modification was lost until 2009, when it was shown that TET proteins catalyze the conversion of 5mC to 5hmC [3]. Currently, 5hmC is well-accepted as an intermediate in demethylation. In addition, several studies suggest that it has additional functions as well. First of all, various tissues accumulate substantial levels of

Deregulation of DNA hydroxymethylation in cancer

Several genes that influence hydroxymethylation are mutated in cancer. First of all one of the TET genes, TET2, is affected by mutations in different hematological malignancies. Furthermore, four genes that play a role in the Krebs cycle, namely IDH1, IDH2, SDH and FH, are mutated in hematological malignancies and various types of solid cancers. These 4 genes are able to affect the activity of the TET proteins by changing the levels of metabolites that compete with the TET co-factor αKG. In

Possibilities for treatment

Although the causes and consequences of deregulated 5hmC levels are still under investigation, several opportunities for targeting the underlying mechanisms have already been explored.

A number of specific inhibitors for IDH1 and IDH2 mutations have been developed and tested in in vitro and in vivo settings. Inhibitors for IDH1 R132 (AGI-5198 [90], HMS-101 [91], AG1-14100 [92], ML309 [93]) were shown to specifically inhibit the ability of the mutant enzyme to produce 2HG in leukemia and glioma

Concluding remarks and perspectives

Deregulated 5hmC levels have been observed in many types of cancer, both in solid cancers as well as in hematological malignancies. Several mechanisms can be responsible for the altered 5hmC levels, ranging from mutations to deregulated miRNA expression (Table 3, Fig. 4). The fact that altered 5hmC levels are frequently observed in cancer suggests that the (de)methylation pathway might be an important target for cancer therapy, however several unanswered questions still need to be addressed.

Transparency document

Transparency document

Acknowledgments

This work was supported by a grant from the Netherlands Institute for Regenerative Medicine (NIRM, DFES1029161) and ERA-NET/TRANSCAN/FP7 (KUN2013-6395).

References (149)

  • R.J. Molenaar et al.

    The driver and passenger effects of isocitrate dehydrogenase 1 and 2 mutations in oncogenesis and survival prolongation

    Biochim. Biophys. Acta

    (2014)
  • W. Xu et al.

    Oncometabolite 2-hydroxyglutarate is a competitive inhibitor of alpha-ketoglutarate-dependent dioxygenases

    Cancer Cell

    (2011)
  • M.E. Figueroa et al.

    Leukemic IDH1 and IDH2 mutations result in a hypermethylation phenotype, disrupt TET2 function, and impair hematopoietic differentiation

    Cancer Cell

    (2010)
  • A.T. Fathi et al.

    Prospective serial evaluation of 2-hydroxyglutarate, during treatment of newly diagnosed acute myeloid leukemia, to assess disease activity and therapeutic response

    Blood

    (2012)
  • T. Müller et al.

    Nuclear exclusion of TET1 is associated with loss of 5-hydroxymethylcytosine in IDH1 wild-type gliomas

    Am. J. Pathol.

    (2012)
  • T. Shibata et al.

    Mutant IDH1 confers an in vivo growth in a melanoma cell line with BRAF mutation

    Am. J. Pathol.

    (2011)
  • E.K. Oermann et al.

    Alterations of metabolic genes and metabolites in cancer

    Semin. Cell Dev. Biol.

    (2012)
  • E. Letouze et al.

    SDH mutations establish a hypermethylator phenotype in paraganglioma

    Cancer Cell

    (2013)
  • R. Uchiyama et al.

    5-Hydroxymethylcytosine as a useful marker to differentiate between malignant melanomas and benign melanocytic nevi

    J. Dermatol. Sci.

    (2014)
  • A.R. Larson et al.

    Loss of 5-hydroxymethylcytosine correlates with increasing morphologic dysplasia in melanocytic tumors

    Mod. Pathol.

    (2014)
  • J. Cheng et al.

    An extensive network of TET2-targeting microRNAs regulates malignant hematopoiesis

    Cell Rep.

    (2013)
  • S.J. Song et al.

    The oncogenic microRNA miR-22 targets the TET2 tumor suppressor to promote hematopoietic stem cell self-renewal and transformation

    Cell Stem Cell

    (2013)
  • S.J. Song et al.

    MicroRNA-antagonism regulates breast cancer stemness and metastasis via TET-family-dependent chromatin remodeling

    Cell

    (2013)
  • F. Pendino et al.

    Functional involvement of RINF, retinoid-inducible nuclear factor (CXXC5), in normal and tumoral human myelopoiesis

    Blood

    (2009)
  • S. Knappskog et al.

    RINF (CXXC5) is overexpressed in solid tumors and is an unfavorable prognostic factor in breast cancer

    Ann. Oncol.

    (2011)
  • C.J. Mariani et al.

    TET1-mediated hydroxymethylation facilitates hypoxic gene induction in neuroblastoma

    Cell Rep.

    (2014)
  • A. Chaturvedi et al.

    Mutant IDH1 promotes leukemogenesis in vivo and can be specifically targeted in human AML

    Blood

    (2013)
  • M.I. Davis et al.

    Biochemical, cellular, and biophysical characterization of a potent inhibitor of mutant isocitrate dehydrogenase IDH1

    J. Biol. Chem.

    (2014)
  • L. Pleyer et al.

    Azacitidine in CMML: matched-pair analyses of daily-life patients reveal modest effects on clinical course and survival

    Leuk. Res.

    (2014)
  • R. Bejar et al.

    TET2 mutations predict response to hypomethylating agents in myelodysplastic syndrome patients

    Blood

    (2014)
  • D. Aran et al.

    Replication timing-related and gene body-specific methylation of active human genes

    Hum. Mol. Genet.

    (2011)
  • M. Tahiliani et al.

    Conversion of 5-methylcytosine to 5-hydroxymethylcytosine in mammalian DNA by MLL partner TET1

    Science

    (2009)
  • S. Ito et al.

    Tet proteins can convert 5-methylcytosine to 5-formylcytosine and 5-carboxylcytosine

    Science

    (2011)
  • M.C. Haffner et al.

    Global 5-hydroxymethylcytosine content is significantly reduced in tissue stem/progenitor cell compartments and in human cancers

    Oncotarget

    (2011)
  • M. Ko et al.

    Impaired hydroxylation of 5-methylcytosine in myeloid cancers with mutant TET2

    Nature

    (2010)
  • T.F. Kraus et al.

    Low values of 5-hydroxymethylcytosine (5hmC), the “sixth base,” are associated with anaplasia in human brain tumors

    Int. J. Cancer

    (2012)
  • A.D. Hershey et al.

    Nucleic acid economy in bacteria infected with bacteriophage T2. I. Purine and pyrimidine composition

    J. Gen. Physiol.

    (1953)
  • N.W. Penn et al.

    The presence of 5-hydroxymethylcytosine in animal deoxyribonucleic acid

    Biochem. J.

    (1972)
  • M. Bachman et al.

    5-Hydroxymethylcytosine is a predominantly stable DNA modification

    Nat. Chem.

    (2014)
  • S. Kriaucionis et al.

    The nuclear DNA base 5-hydroxymethylcytosine is present in Purkinje neurons and the brain

    Science

    (2009)
  • M. Ivanov et al.

    Ontogeny, distribution and potential roles of 5-hydroxymethylcytosine in human liver function

    Genome Biol.

    (2013)
  • L.M. Iyer et al.

    Prediction of novel families of enzymes involved in oxidative and other complex modifications of bases in nucleic acids

    Cell Cycle

    (2009)
  • M. Ko et al.

    Modulation of TET2 expression and 5-methylcytosine oxidation by the CXXC domain protein IDAX

    Nature

    (2013)
  • S.M. Langemeijer et al.

    Acquired mutations in TET2 are common in myelodysplastic syndromes

    Nat. Genet.

    (2009)
  • F. Delhommeau et al.

    Mutation in TET2 in myeloid cancers

    N. Engl. J. Med.

    (2009)
  • O. Kosmider et al.

    TET2 gene mutation is a frequent and adverse event in chronic myelomonocytic leukemia

    Haematologica

    (2009)
  • S. Weissmann et al.

    Landscape of TET2 mutations in acute myeloid leukemia

    Leukemia

    (2012)
  • M.G. Aslanyan et al.

    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)
  • M. Ko et al.

    Ten–Eleven-Translocation 2 (TET2) negatively regulates homeostasis and differentiation of hematopoietic stem cells in mice

    Proc. Natl. Acad. Sci. U. S. A.

    (2011)
  • N. Konstandin et al.

    Genomic 5-hydroxymethylcytosine levels correlate with TET2 mutations and a distinct global gene expression pattern in secondary acute myeloid leukemia

    Leukemia

    (2011)
  • Cited by (85)

    • Nutritional Epigenetics in Cancer

      2022, Advances in Nutrition
    • Epigenetic processes-An overview

      2021, Epigenetics of Exercise and Sports: Concepts, Methods, and Current Research
    • TRAIL receptor signaling: From the basics of canonical signal transduction toward its entanglement with ER stress and the unfolded protein response

      2020, International Review of Cell and Molecular Biology
      Citation Excerpt :

      The TET2 enzyme is involved in DNA demethylation and mutations in TET2 mainly cause loss-of-function. Mutations in IDH enzymes alter the reactivity of the enzyme and lead to the generation of 2-hydroxyglutarate, an abnormal metabolite that is a strong and competitive inhibitor of TET enzymes (Kroeze et al., 2015). How these specifically are linked to modified TRAILR expression, however, currently remains understudied.

    View all citing articles on Scopus
    View full text