Review
Intragenic DNA methylation in transcriptional regulation, normal differentiation and cancer

https://doi.org/10.1016/j.bbagrm.2013.08.001Get rights and content

Highlights

  • ā€¢

    The role of DNA methylation on gene regulation depends on the genomic context.

  • ā€¢

    Intragenic DNA methylation is involved in multiple gene regulation processes.

  • ā€¢

    Intragenic DNA methylation is modulated during cell differentiation and cancer.

Abstract

Ever since the discovery of DNA methylation at cytosine residues, the role of this so called fifth base has been extensively studied and debated. Until recently, the majority of DNA methylation studies focused on the analysis of CpG islands associated to promoter regions. However, with the upcoming possibilities to study DNA methylation in a genome-wide context, this epigenetic mark can now be studied in an unbiased manner. As a result, recent studies have shown that not only promoters but also intragenic and intergenic regions are widely modulated during physiological processes and disease. In particular, it is becoming increasingly clear that DNA methylation in the gene body is not just a passive witness of gene transcription but it seems to be actively involved in multiple gene regulation processes. In this review we discuss the potential role of intragenic DNA methylation in alternative promoter usage, regulation of short and long non-coding RNAs, alternative RNA processing, as well as enhancer activity. Furthermore, we summarize how the intragenic DNA methylome is modified both during normal cell differentiation and neoplastic transformation.

Introduction

Back in the 1970s, Riggs and Holliday independently suggested that methylation of cytosines in the context of CpG dinucleotides may represent an epigenetic mark associated with gene silencing [1], [2]. Years later, this hypothesis was experimentally demonstrated by several groups [3], [4], [5], [6], [7]. Interestingly, already in the 1980s it was recognized that CpG density is not evenly distributed within the genome, but rather shows a bimodal distribution. Regions with an elevated CpG content are called CpG islands (CGIs), and overlap with transcriptional start sites (TSSs) of approximately 60ā€“70% of all human genes [8], [9]. In contrast, regions with low CpG density are frequently located outside TSS [10], [11]. Furthermore, it was noted in these studies that both DNA methylation and CpG density show a bimodal distribution. In general, CGIs are unmethylated whereas most regions with low CpG density are heavily methylated in normal tissues, something which is now considered general knowledge. Experimental evidence demonstrated that gene expression can be regulated by DNA methylation levels of CGIs in the proximity of TSSs [12], [13]. Based on these data, the majority of cancer-related DNA methylation studies focused on the role of CGI hypermethylation as a mechanism of tumor suppressor gene silencing [14], [15], [16], [17], [18]. Additionally, already in the 1980s, pioneer studies in cancer cells reported that neoplastic transformation was also associated with global and gene specific loss of DNA methylation [19], [20], [21], [22]. Collectively, the studies published so far in cancer epigenomics point to a massive disruption of the DNA methylome in tumor cells as compared to their normal cell counterparts.

DNA methylation can be analyzed by methylation sensitive restriction enzyme digestion, bisulfite conversion based techniques or affinity enrichment of methylated DNA [23], [24], [25]. A comprehensive overview of the advantages and disadvantages of these approaches and their applicability is given by Laird [26]. Until recently, these techniques were PCR or microarray-based to analyze single or multiple regions of interest, respectively [27], [28], [29], [30]. As a consequence, the great majority of studies designed to investigate DNA methylation dynamics were covering only a small fraction of the DNA methylome, namely CGIs and promoter regions. Nowadays, the use of high-density microarray analyses, e.g., comprehensive high-throughput arrays for relative methylation (CHARM) and Infinium [31], [32], and next-generation sequencing based DNA methylation analysis, e.g., methylated DNA immunoprecipitation followed by deep-sequencing (MeDIP-seq), CXXC affinity purification followed by deep-sequencing (CAP-seq), reduced representation bisulfite sequencing (RRBS) and whole genome bisulfite sequencing (WGBS) [33], [34], [35], [36], allows us to obtain an unbiased representation of DNA methylation maps throughout the genome. The use of these techniques has started to reveal that DNA methylation has an even broader function than previously anticipated and that its functions may vary in a context-dependent manner [37].

One of the major findings derived from whole-genome studies is that DNA methylation levels in the gene body, i.e. intragenic DNA methylation, widely change during cell differentiation and carcinogenesis. Although the precise role of DNA methylation within the gene body is still far from being well understood, recent publications indicate that it may be involved in the regulation of multiple processes, such as transcript elongation, expression of intragenic coding and non-coding transcripts, alternative splicing and enhancer activation. The goal of this review is to provide a comprehensive overview of the current knowledge on intragenic DNA methylation and its association with gene regulation in the context of cell differentiation and neoplastic transformation.

Section snippets

DNA methylation and gene expression regulation: General aspects

The molecular mechanisms by which DNA methylation mediates gene silencing have been mainly studied in the context of genes containing CGIs in their promoter region. Many reports have shown that inducing hypomethylation of CGIs, either by genetic deletions of DNA methyltransferases DNMT1, DNMT3A and DNMT3B or by pharmacologic interventions through demethylating agents, results in gene reactivation [7], [13], [38], [39], [40], [41]. How does CGI methylation induce gene silencing? One of the most

Association between intragenic DNA methylation and gene expression

The negative correlation between gene expression and CGI methylation at TSSs is well established. However, this association cannot be extrapolated to other genomic contexts such as CpGs located in the gene body. In general, DNA methylation is thought to block transcription initiation but not elongation. In fact, intragenic nucleosomes with trimethylation of H3K36 (H3K36me3), which is associated with transcript elongation, seem to recruit DNMTs, thus facilitating the methylation of intragenic

Regulation of alternative intragenic promoters

An initial study from 1999 linked the DNA methylation status of an alternative promoter in the TGF-Ī²3 gene to expression of an alternative transcript [67]. Furthermore, Shmelkov et al. showed that DNA methylation of alternative promoters of the AC133 gene suppresses their activity in a luciferase reporter assay [68]. Considering the fact that a high number of transcripts are transcribed from alternative promoters [69], the regulation of alternative intragenic promoters through DNA methylation

Regulation of intragenic non-coding RNAs and transposable elements

Another role of gene-body DNA methylation is the regulation of intragenic non-coding RNAs (ncRNAs) present in the intronic region of numerous genes. They can be co-expressed with the host gene or can have their own promoter. Non-coding RNAs are known to be key regulators in important processes such as proliferation, differentiation, apoptosis and cell development, therefore it is expected that their expression is highly regulated via epigenetic mechanisms [72], [73]. The best studied ncRNAs are

DNA methylation and RNA processing

An additional function of intragenic DNA methylation is related to RNA processing mechanisms such as alternative splicing and alternative polyadenylation [90]. It is commonly accepted that alternative splicing is regulated by splicing enhancers, silencers and specific binding of splicing factors [91], [92]. Furthermore, several studies have identified a link between epigenetics and alternative splicing [93], [94], [95]. Initial indications for the existence of this phenomenon came from a global

Epigenetic composition of enhancers and correlation with gene expression

Enhancers are multidimensional signal integrators that regulate the spatial and temporal patterns of gene expression during development and cell fate specification [113]. Over the last years, the application of next-generation sequencing has led to a better understanding of the epigenetic composition and function of enhancers. Enhancer regions tend to have a low nucleosome occupancy and are enriched for particular histone variants (i.e. H2A.Z and H3.3), while surrounding nucleosomes are

Intragenic DNA methylation in normal differentiation and malignant transformation

It is widely accepted that DNA methylation patterns are dependent on cell type, which is for example shown in a genome-wide methylome study in which clear DNA methylation differences between brain, liver and spleen cells were observed [153]. Furthermore, DNA methylation patterns are dependent on the maturation stage of a particular cell type [56], [154], [155]. Hence, DNA methylation patterns must be acquired during embryonic development and differentiation (Fig.Ā 8). The gradual acquisition of

Intragenic DNA methylation as biomarker in cancer

In the past years, cancer related alterations in DNA methylation have been studied extensively and they are starting to play an important role as biomarkers [163], [164], [165]. Their usefulness as biomarkers can be attributed to the fact that DNA methylation is stable over time, easy to investigate and cell-type specific [165], [166]. DNA methylation profiles in cancer cells seem to be partially based on their cell of origin [56]. As tumors with different cells of origin may differ in

Concluding remarks and perspective

Whole-genome epigenomic studies are starting to reveal that the function of DNA methylation is more variable and context-dependent than previously thought. A new insight derived from these studies is that intragenic DNA methylation is widely modulated during normal cell differentiation and neoplastic transformation. Furthermore, recent functional studies are unraveling the multiple molecular mechanisms through which intragenic DNA methylation affects gene regulation, including alternative

Acknowledgements

The authors' studies on epigenomics are supported by grants of the Spanish Ministerio de EconomĆ­a y Competitividad (project SAF2012-31138 and the ICGC-associated CLL Genome Project) and the European Union's Seventh Framework Programme through the Blueprint Consortium (grant agreement 282510). M.K. is supported by the AgĆØncia de GestiĆ³ d'Ajuts Universitaris i de Recerca of the Generalitat de Catalunya, A.Q. by the Portuguese FundaĆ§Ć£o para a CiĆŖncia e a Tecnologia, R.B. by a Rubicon grant from

References (175)

  • C.P. Ponting et al.

    Evolution and functions of long noncoding RNAs

    Cell

    (2009)
  • R. Stoger et al.

    Maternal-specific methylation of the imprinted mouse Igf2r locus identifies the expressed locus as carrying the imprinting signal

    Cell

    (1993)
  • R.F. Luco et al.

    Epigenetics in alternative pre-mRNA splicing

    Cell

    (2011)
  • R. Holliday et al.

    DNA modification mechanisms and gene activity during development

    Science

    (1975)
  • A.D. Riggs

    X inactivation, differentiation, and DNA methylation

    Cytogenet. Cell Genet.

    (1975)
  • A. Razin et al.

    DNA methylation and gene function

    Science

    (1980)
  • J.D. McGhee et al.

    Specific DNA methylation sites in the vicinity of the chicken beta-globin genes

    Nature

    (1979)
  • R. Stein et al.

    Clonal inheritance of the pattern of DNA methylation in mouse cells

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

    (1982)
  • T. Mohandas et al.

    Reactivation of an inactive human X chromosome: evidence for X inactivation by DNA methylation

    Science

    (1981)
  • S. Saxonov et al.

    A genome-wide analysis of CpG dinucleotides in the human genome distinguishes two distinct classes of promoters

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

    (2006)
  • M.L. Tykocinski et al.

    CG dinucleotide clusters in MHC genes and in 5ā€² demethylated genes

    Nucleic Acids Res.

    (1984)
  • R.S. Hansen et al.

    5-Azacytidine-induced reactivation of the human X chromosome-linked PGK1 gene is associated with a large region of cytosine demethylation in the 5ā€² CpG island

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

    (1990)
  • A. Merlo et al.

    5ā€² CpG island methylation is associated with transcriptional silencing of the tumour suppressor p16/CDKN2/MTS1 in human cancers

    Nat. Med.

    (1995)
  • T. Sakai et al.

    Allele-specific hypermethylation of the retinoblastoma tumor-suppressor gene

    Am. J. Hum. Genet.

    (1991)
  • K. Yoshiura et al.

    Silencing of the E-cadherin invasion-suppressor gene by CpG methylation in human carcinomas

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

    (1995)
  • M. Esteller

    Epigenetics in cancer

    N. Engl. J. Med.

    (2008)
  • P.A. Jones et al.

    The fundamental role of epigenetic events in cancer

    Nat. Rev.

    (2002)
  • A.P. Feinberg et al.

    Hypomethylation distinguishes genes of some human cancers from their normal counterparts

    Nature

    (1983)
  • M.A. Gama-Sosa et al.

    The 5-methylcytosine content of DNA from human tumors

    Nucleic Acids Res.

    (1983)
  • M. Ehrlich

    DNA methylation in cancer: too much, but also too little

    Oncogene

    (2002)
  • C. Waalwijk et al.

    DNA methylation at a CCGG sequence in the large intron of the rabbit beta-globin gene: tissue-specific variations

    Nucleic Acids Res.

    (1978)
  • R.Y. Wang et al.

    Comparison of bisulfite modification of 5-methyldeoxycytidine and deoxycytidine residues

    Nucleic Acids Res.

    (1980)
  • S.H. Cross et al.

    Purification of CpG islands using a methylated DNA binding column

    Nat. Genet.

    (1994)
  • P.W. Laird

    Principles and challenges of genomewide DNA methylation analysis

    Nat. Rev.

    (2010)
  • M. Frommer et al.

    A genomic sequencing protocol that yields a positive display of 5-methylcytosine residues in individual DNA strands

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

    (1992)
  • J.G. Herman et al.

    Methylation-specific PCR: a novel PCR assay for methylation status of CpG islands

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

    (1996)
  • B. Khulan et al.

    Comparative isoschizomer profiling of cytosine methylation: the HELP assay

    Genome Res.

    (2006)
  • M. Weber et al.

    Distribution, silencing potential and evolutionary impact of promoter DNA methylation in the human genome

    Nat. Genet.

    (2007)
  • R.A. Irizarry et al.

    Comprehensive high-throughput arrays for relative methylation (CHARM)

    Genome Res.

    (2008)
  • R.S. Illingworth et al.

    Orphan CpG islands identify numerous conserved promoters in the mammalian genome

    PLoS Genet.

    (2010)
  • R. Lister et al.

    Human DNA methylomes at base resolution show widespread epigenomic differences

    Nature

    (2009)
  • A.K. Maunakea et al.

    Conserved role of intragenic DNA methylation in regulating alternative promoters

    Nature

    (2010)
  • A. Meissner et al.

    Reduced representation bisulfite sequencing for comparative high-resolution DNA methylation analysis

    Nucleic Acids Res.

    (2005)
  • P.A. Jones

    Functions of DNA methylation: islands, start sites, gene bodies and beyond

    Nat. Rev.

    (2012)
  • P.A. Jones et al.

    Cell cycle-specific reactivation of an inactive X-chromosome locus by 5-azadeoxycytidine

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

    (1982)
  • L. Venolia et al.

    Transformation with DNA from 5-azacytidine-reactivated X chromosomes

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

    (1982)
  • I. Rhee et al.

    DNMT1 and DNMT3b cooperate to silence genes in human cancer cells

    Nature

    (2002)
  • M.F. Robert et al.

    DNMT1 is required to maintain CpG methylation and aberrant gene silencing in human cancer cells

    Nat. Genet.

    (2003)
  • E. Ballestar et al.

    Methyl-CpG-binding proteins. Targeting specific gene repression

    Eur. J. Biochem.

    (2001)
  • A. Bird

    DNA methylation patterns and epigenetic memory

    Genes Dev.

    (2002)
  • Cited by (189)

    • Non-thermal plasma promotes boar sperm quality through increasing AMPK methylation

      2024, International Journal of Biological Macromolecules
    • Regulation of gene expression in mammals: an overview

      2022, Post-transcriptional Gene Regulation in Human Disease: Volume 32
    View all citing articles on Scopus
    View full text