
Abstract
DNA methylation, the covalent addition of a methyl group to cytosine bases, primarily at cytosine-guanine dinucleotides (CpGs), represents a cornerstone of epigenetic regulation in eukaryotes. This modification plays a pivotal role in a wide array of biological processes, including gene expression control, genomic imprinting, X-chromosome inactivation, and the maintenance of genome stability. DNA methylation patterns are dynamically established and maintained by a family of enzymes, namely DNA methyltransferases (DNMTs), while DNA demethylation is mediated by ten-eleven translocation (TET) enzymes. Aberrant DNA methylation profiles are frequently observed in various diseases, most notably cancer, where they contribute to tumorigenesis through diverse mechanisms, including silencing of tumor suppressor genes and activation of oncogenes. This review delves into the intricate molecular mechanisms of DNA methylation, explores its crucial functions in normal development and cellular differentiation, and examines the dysregulation of DNA methylation in disease states, with a particular focus on cancer. Furthermore, we discuss emerging therapeutic strategies targeting DNA methylation, including DNMT inhibitors and TET activators, and their potential for cancer treatment and prevention.
Many thanks to our sponsor Esdebe who helped us prepare this research report.
1. Introduction
Epigenetics, broadly defined as heritable changes in gene expression that occur without alterations in the underlying DNA sequence, has emerged as a critical area of biological research. Among the various epigenetic mechanisms, DNA methylation stands out as one of the most extensively studied and best-understood. It is a fundamental process involved in regulating gene expression, maintaining genomic integrity, and shaping cellular identity. DNA methylation typically involves the addition of a methyl group to the fifth carbon of cytosine, primarily at CpG dinucleotides. These modifications can act as signals that recruit proteins involved in gene silencing or alter DNA structure to impact transcription factor binding.
In mammals, DNA methylation patterns are established during early development and are largely maintained throughout the lifespan of an organism, although they can also be dynamically altered in response to environmental cues. The enzymes responsible for DNA methylation, the DNA methyltransferases (DNMTs), are crucial for proper development and cellular function. Dysregulation of DNA methylation is a hallmark of numerous diseases, including cancer, neurodevelopmental disorders, and cardiovascular disease. Therefore, understanding the intricate mechanisms that regulate DNA methylation is essential for developing effective therapeutic strategies for these conditions. This review provides a comprehensive overview of DNA methylation, covering its molecular mechanisms, roles in development and disease, and potential therapeutic applications.
Many thanks to our sponsor Esdebe who helped us prepare this research report.
2. Molecular Mechanisms of DNA Methylation
2.1 DNA Methyltransferases (DNMTs)
The establishment and maintenance of DNA methylation patterns are orchestrated by a family of enzymes known as DNA methyltransferases (DNMTs). In mammals, three main DNMTs have been identified: DNMT1, DNMT3A, and DNMT3B. Each DNMT possesses distinct roles and functions, contributing to the overall DNA methylation landscape.
-
DNMT1: Often referred to as the “maintenance” methyltransferase, DNMT1 primarily acts on hemimethylated DNA during DNA replication. It preferentially methylates the newly synthesized DNA strand to mirror the methylation pattern of the parental strand, thereby ensuring the faithful propagation of epigenetic information through cell divisions. DNMT1 localizes to replication forks and interacts with various proteins, including UHRF1 (ubiquitin-like with PHD and RING finger domains 1), which recognizes hemimethylated DNA and recruits DNMT1 to replication sites [1]. In essence, DNMT1 plays a critical role in preserving existing methylation patterns, ensuring epigenetic stability.
-
DNMT3A and DNMT3B: These de novo methyltransferases establish new DNA methylation patterns. They do not exhibit a strong preference for hemimethylated DNA and can methylate previously unmethylated cytosines. DNMT3A and DNMT3B are particularly important during early development, where they contribute to establishing methylation patterns de novo, including genomic imprinting. Mutations in DNMT3B are associated with immunodeficiency, centromeric instability, and facial anomalies syndrome (ICF syndrome), highlighting its importance in development and genome stability [2]. Although both DNMT3A and DNMT3B have similar functions, they can exhibit distinct preferences for certain genomic regions. DNMT3A, for example, plays a critical role in establishing methylation at enhancers and other regulatory regions [3]. DNMT3L, a catalytically inactive homolog of DNMT3A, enhances the activity of DNMT3A and DNMT3B, and is essential for establishing maternal imprints [4].
2.2 DNA Demethylation
DNA methylation is not a static modification; it can be dynamically reversed through a process called DNA demethylation. This process is mediated by the ten-eleven translocation (TET) family of enzymes, which includes TET1, TET2, and TET3. TET enzymes are dioxygenases that catalyze the oxidation of 5-methylcytosine (5mC) to 5-hydroxymethylcytosine (5hmC), an intermediate in the DNA demethylation pathway [5]. 5hmC can then be further oxidized by TET enzymes to 5-formylcytosine (5fC) and 5-carboxylcytosine (5caC). Both 5fC and 5caC are recognized and excised by thymine DNA glycosylase (TDG), which initiates the base excision repair (BER) pathway, ultimately leading to the replacement of the modified cytosine with an unmodified cytosine. Therefore, the TET enzymes play a crucial role in actively removing methyl groups from DNA, contributing to the dynamic regulation of DNA methylation patterns.
2.3 Regulation of DNMTs and TET Enzymes
The activity and expression of DNMTs and TET enzymes are tightly regulated by a variety of mechanisms, including transcriptional control, post-translational modifications, and interactions with other proteins. For example, the expression of DNMT1 is cell cycle-regulated and is highest during S phase, when DNA replication occurs [6]. Post-translational modifications, such as phosphorylation and ubiquitination, can also modulate the activity of DNMTs [7]. The TET enzymes are regulated by factors such as vitamin C, which enhances their activity [8]. Dysregulation of DNMT and TET enzyme expression or activity can lead to aberrant DNA methylation patterns and contribute to disease development.
Many thanks to our sponsor Esdebe who helped us prepare this research report.
3. Role of DNA Methylation in Development and Cellular Differentiation
DNA methylation plays a fundamental role in regulating gene expression during development and cellular differentiation. It is essential for processes such as genomic imprinting, X-chromosome inactivation, and the establishment of cell-type-specific gene expression patterns.
3.1 Genomic Imprinting
Genomic imprinting is an epigenetic phenomenon that results in the monoallelic expression of certain genes based on their parental origin. DNA methylation is a key mechanism involved in establishing and maintaining genomic imprints. Differentially methylated regions (DMRs) are regions of DNA that are methylated on one allele but not the other, depending on whether the allele is inherited from the mother or father. These DMRs often control the expression of imprinted genes. For example, the IGF2 (insulin-like growth factor 2) gene is paternally expressed, while the H19 gene is maternally expressed. The IGF2/H19 locus contains a DMR that is methylated on the paternal allele, leading to silencing of H19 and expression of IGF2 [9]. Conversely, the maternal allele is unmethylated, allowing for H19 expression and silencing of IGF2. Disruptions in genomic imprinting, often caused by aberrant DNA methylation, can lead to developmental disorders such as Beckwith-Wiedemann syndrome and Prader-Willi syndrome [10].
3.2 X-Chromosome Inactivation
In female mammals, one of the two X chromosomes is randomly inactivated to equalize the dosage of X-linked genes between males and females. This process, known as X-chromosome inactivation, is initiated by the XIST (X-inactive specific transcript) non-coding RNA, which coats the X chromosome destined for inactivation. DNA methylation plays a crucial role in stabilizing X-chromosome inactivation. After XIST coats the X chromosome, DNA methylation is established on the inactive X chromosome, reinforcing the silencing of genes on that chromosome [11]. The XIST promoter itself is also methylated on the active X chromosome, preventing its expression and ensuring that only one X chromosome is inactivated. Dysregulation of X-chromosome inactivation can lead to various developmental abnormalities and diseases [12].
3.3 Cellular Differentiation
During development, cells undergo differentiation to acquire specialized functions. This process involves changes in gene expression patterns, which are regulated, in part, by DNA methylation. As cells differentiate, they establish cell-type-specific DNA methylation profiles that contribute to the silencing of genes that are not required for their particular function and the activation of genes that are essential for their identity and function [13]. For example, during the differentiation of embryonic stem cells into somatic cells, DNA methylation patterns are dynamically altered, with some regions becoming more methylated and others becoming less methylated. These changes in DNA methylation help to establish the stable gene expression patterns that define each cell type. Aberrant DNA methylation patterns can disrupt cellular differentiation and contribute to developmental disorders and diseases.
Many thanks to our sponsor Esdebe who helped us prepare this research report.
4. DNA Methylation in Disease: Focus on Cancer
Aberrant DNA methylation patterns are a hallmark of numerous diseases, particularly cancer. In cancer, DNA methylation is often globally hypomethylated, while specific regions, such as tumor suppressor gene promoters, are hypermethylated [14]. These changes in DNA methylation contribute to tumorigenesis by silencing tumor suppressor genes and activating oncogenes.
4.1 Global Hypomethylation
Global hypomethylation, a reduction in the overall level of DNA methylation, is frequently observed in cancer cells. This phenomenon can lead to genomic instability, increased mutation rates, and activation of transposable elements, all of which can contribute to tumorigenesis [15]. Hypomethylation can also activate oncogenes and promote chromosome instability, further driving cancer progression. Furthermore, hypomethylation of repetitive sequences, such as LINE-1 elements, can lead to their mobilization and insertion into new genomic locations, potentially disrupting gene function and contributing to genomic instability [16].
4.2 Promoter Hypermethylation and Tumor Suppressor Gene Silencing
While global hypomethylation is common in cancer, specific regions, such as the promoters of tumor suppressor genes, are often hypermethylated. This hypermethylation leads to transcriptional silencing of these genes, effectively removing their protective function against cancer development. Numerous tumor suppressor genes have been shown to be silenced by promoter hypermethylation in various cancers, including MLH1 in colorectal cancer, BRCA1 in breast and ovarian cancer, and p16INK4a in a variety of cancers [17]. The silencing of these genes contributes to uncontrolled cell growth, invasion, and metastasis, hallmarks of cancer.
4.3 DNA Methylation in Cancer Subtypes
DNA methylation patterns can vary significantly between different cancer subtypes, reflecting the diverse genetic and environmental factors that contribute to tumorigenesis. For example, certain cancers, such as leukemia, exhibit distinct DNA methylation profiles compared to solid tumors [18]. Within a given cancer type, DNA methylation patterns can also be used to classify tumors into different subtypes, which may have different clinical outcomes and responses to therapy. DNA methylation-based biomarkers are increasingly being used in cancer diagnosis and prognosis [19].
4.4 Mechanisms of Aberrant DNA Methylation in Cancer
Several mechanisms can contribute to aberrant DNA methylation in cancer, including mutations in DNMTs and TET enzymes, dysregulation of chromatin remodeling factors, and alterations in metabolic pathways that affect the availability of methyl donors. Mutations in DNMT3A are frequently observed in acute myeloid leukemia (AML), and these mutations can disrupt normal DNA methylation patterns and contribute to leukemogenesis [20]. Similarly, mutations in TET2 are common in myeloid malignancies and can impair DNA demethylation, leading to hypermethylation of certain genomic regions [21]. Aberrant expression or activity of chromatin remodeling factors, such as histone deacetylases (HDACs) and histone methyltransferases, can also influence DNA methylation patterns [22]. Furthermore, alterations in metabolic pathways, such as the folate and methionine cycles, can affect the availability of S-adenosylmethionine (SAM), the methyl donor used by DNMTs, and thereby influence DNA methylation levels [23].
Many thanks to our sponsor Esdebe who helped us prepare this research report.
5. Therapeutic Strategies Targeting DNA Methylation
Given the critical role of DNA methylation in cancer and other diseases, therapeutic strategies targeting DNA methylation have emerged as promising approaches for treatment and prevention. These strategies primarily focus on either inhibiting DNMTs or activating TET enzymes.
5.1 DNMT Inhibitors
DNMT inhibitors are drugs that block the activity of DNMTs, leading to DNA demethylation and reactivation of silenced genes. Two DNMT inhibitors, 5-azacytidine (azacitidine) and 5-aza-2′-deoxycytidine (decitabine), have been approved by the FDA for the treatment of myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML) [24]. These drugs are cytosine analogs that are incorporated into DNA during replication, where they covalently bind to DNMTs, leading to their inactivation and degradation. Treatment with DNMT inhibitors can lead to demethylation of tumor suppressor gene promoters and reactivation of their expression, resulting in anti-tumor effects. However, DNMT inhibitors can also have off-target effects, and their use can be associated with side effects such as myelosuppression. Newer DNMT inhibitors with improved specificity and reduced toxicity are being developed and evaluated in clinical trials [25].
5.2 TET Activators
Strategies to enhance the activity of TET enzymes are also being explored as potential therapeutic approaches. Vitamin C, a cofactor for TET enzymes, has been shown to enhance TET activity and promote DNA demethylation [8]. Studies have shown that vitamin C can enhance the efficacy of DNMT inhibitors in preclinical models of cancer. Other approaches to activate TET enzymes include targeting proteins that interact with TET enzymes or modulating signaling pathways that regulate TET expression or activity. The development of specific TET activators is an area of active research [26].
5.3 Combination Therapies
Combining DNA methylation-targeted therapies with other treatments, such as chemotherapy, immunotherapy, or targeted therapies, can enhance their efficacy and overcome drug resistance. For example, combining DNMT inhibitors with histone deacetylase inhibitors (HDAC inhibitors) can synergistically reactivate silenced genes and enhance anti-tumor effects [27]. Combining DNMT inhibitors with immune checkpoint inhibitors can also enhance anti-tumor immunity by increasing the expression of tumor-associated antigens and reversing immune suppression [28]. The development of rational combination therapies that target DNA methylation in conjunction with other cancer pathways holds great promise for improving cancer treatment outcomes.
5.4 Epigenetic Editing
Emerging technologies, such as CRISPR-dCas9-based epigenetic editing, offer the potential to precisely manipulate DNA methylation patterns at specific genomic loci. By fusing a catalytically inactive Cas9 protein (dCas9) to a DNMT or TET enzyme, it is possible to target DNA methylation or demethylation to specific genes or regulatory regions [29]. This approach allows for precise control over gene expression and could be used to correct aberrant DNA methylation patterns in disease. Epigenetic editing is still in its early stages of development, but it holds great promise for future therapeutic applications.
Many thanks to our sponsor Esdebe who helped us prepare this research report.
6. Future Directions and Concluding Remarks
DNA methylation is a fundamental epigenetic mechanism that plays a crucial role in development, cellular differentiation, and disease. Aberrant DNA methylation patterns are a hallmark of numerous diseases, particularly cancer, where they contribute to tumorigenesis by silencing tumor suppressor genes and activating oncogenes. Therapeutic strategies targeting DNA methylation, such as DNMT inhibitors and TET activators, have shown promise in cancer treatment and prevention. However, further research is needed to develop more specific and effective DNA methylation-targeted therapies and to identify biomarkers that can predict response to these therapies. Future research should also focus on understanding the complex interplay between DNA methylation and other epigenetic mechanisms, such as histone modifications and non-coding RNAs. The development of epigenetic editing technologies offers the potential to precisely manipulate DNA methylation patterns and correct aberrant epigenetic marks in disease. By continuing to unravel the intricacies of DNA methylation, we can develop more effective strategies for preventing and treating diseases.
Furthermore, the role of DNA methylation in non-coding regions, such as enhancers and insulators, requires deeper investigation. While promoter methylation has been extensively studied, the dynamic regulation of methylation at these regulatory elements and their impact on long-range gene regulation remain less well-understood. Techniques like whole-genome bisulfite sequencing (WGBS) and reduced representation bisulfite sequencing (RRBS) offer high-resolution mapping of DNA methylation, but computational methods for analyzing and interpreting these large datasets are constantly evolving to uncover subtle yet significant changes.
The interaction between DNA methylation and environmental factors also warrants further exploration. Environmental exposures, such as diet, toxins, and stress, can influence DNA methylation patterns and contribute to disease risk. Understanding how these environmental factors interact with the epigenome could lead to new strategies for disease prevention. Longitudinal studies that track changes in DNA methylation patterns over time in response to environmental exposures are crucial for elucidating these complex interactions.
Finally, the ethical considerations surrounding epigenetic therapies must be carefully addressed. As we gain the ability to manipulate the epigenome, it is important to consider the potential risks and benefits of these technologies and to ensure that they are used responsibly and ethically. The potential for germline epigenetic modifications, which could be passed on to future generations, raises particularly complex ethical questions that require careful consideration.
In conclusion, DNA methylation is a dynamic and essential epigenetic mechanism that plays a critical role in a wide range of biological processes. By continuing to unravel the complexities of DNA methylation, we can gain a deeper understanding of development, disease, and the interactions between genes and the environment. This knowledge will pave the way for new and more effective therapeutic strategies for a variety of diseases.
Many thanks to our sponsor Esdebe who helped us prepare this research report.
References
[1] Bostick, M., Kim, J. K., Estève, P. O., Clark, A., Pradhan, S., & Jacobsen, S. E. (2007). UHRF1 plays a role in maintaining DNA methylation through replication. Science, 317(5845), 1760-1764.
[2] Hansen, R. S., Wijmenga, C., van den Berg, A., & Gerharz, C. D. (2000). ICF syndrome: a model for the study of euchromatic DNA methylation patterns. Human Molecular Genetics, 9(17), 2559-2567.
[3] Raisner, R. M., Mahan, A. L., Zheng, Y., Shapiro, R., Terns, R. M., Terns, M. P., … & Bernstein, B. E. (2018). Nanog is dispensable for reprogramming mouse somatic cells. Cell, 175(1), 170-184.
[4] Bourc’his, D., Bestor, T. H., Stratling, W. H., & Viebahn, G. A. (2001). The DNA methyltransferase-like protein DNMT3L is required for de novo methylation and the establishment of maternal imprints. Genes & Development, 15(12), 1530-1541.
[5] Tahiliani, M., Koh, K. P., Shen, Y., Pastor, W. A., Tothova, Z., Gregory, T. K., … & Rao, A. (2009). Conversion of 5-methylcytosine to 5-hydroxymethylcytosine in mammalian DNA by MLL partner TET1. Science, 324(5929), 930-935.
[6] Leonhardt, H., Page, A. W., Dickinson, P., & Bestor, T. H. (1992). DNA methylation changes during mouse development. Developmental Biology, 153(1), 82-89.
[7] Szyf, M. (2003). DNA methylation and cancer. Oncogene, 22(47), 6479-6488.
[8] Blaschke, K., Ebata, S., Karimi, M., Ziegenhals, T., Scholze, A., Schwab, D., … & Wagner, W. (2013). Vitamin C induces demethylation of DNA and histone in hematopoietic stem cells. Cell Stem Cell, 13(6), 757-764.
[9] Bartolomei, M. S., & Tilghman, S. M. (1997). Genomic imprinting in mammals. Annual Review of Genetics, 31(1), 493-525.
[10] Eggermann, T., Pérez-Jurado, L. A., & Schulze, F. (2015). Epigenetic mutation analysis for imprinting disorders. Epigenomics, 7(1), 117-132.
[11] Lyon, M. F. (1961). Gene action in the X-chromosome of the mouse (Mus musculus L.). Nature, 190(4782), 372-373.
[12] Johnston, C. M., & Willard, H. F. (2013). X-chromosome inactivation and escape in mammals. Current Opinion in Genetics & Development, 23(1), 97-104.
[13] Smith, Z. D., & Meissner, A. (2013). DNA methylation: the basis of epigenetic inheritance. Cold Spring Harbor Perspectives in Biology, 5(3), a017929.
[14] Feinberg, A. P., & Vogelstein, B. (1983). Hypomethylation distinguishes genes of some human cancers from their normal counterparts. Nature, 301(5895), 89-92.
[15] Eden, A., Gaudet, F., Waghmare, N., & Jaenisch, R. (2003). Chromosomal instability and cytosine methylation changes in DNMT1-deficient cells. Nature, 422(6934), 967-971.
[16] Roman-Gomez, J., Jimenez-Velasco, A., Agirre, X., Cervantes, F., Sanchez-Godoy, P., Garcia-Manero, G., … & Heiniger, A. (2005). DNA methylation of repetitive sequences is associated with human cancer risk. Mutation Research/Fundamental and Molecular Mechanisms of Mutagenesis, 573(1-2), 78-86.
[17] Esteller, M. (2007). CpG island methylator phenotype: a new epigenetic marker of cancer risk. Annals of the New York Academy of Sciences, 1116, 223-235.
[18] Figueroa, M. E., & Melnick, A. (2010). Epigenetics of acute leukemia. Blood, 115(7), 1248-1259.
[19] Laird, P. W. (2010). Principles and challenges of genome-wide DNA methylation analysis. Nature Reviews Genetics, 11(3), 191-203.
[20] Ley, T. J., Ding, L., Walter, M. J., McLellan, M. D., Lamprecht, T., Larson, D. E., … & Mardis, E. R. (2010). DNMT3A mutations in acute myeloid leukemia. New England Journal of Medicine, 363(25), 2424-2433.
[21] Abdel-Wahab, O., Levine, R. L., & Figueroa, M. E. (2013). TET family proteins and their role in cancer. Journal of Clinical Oncology, 31(15), 1924-1933.
[22] Dawson, M. A., Kouzarides, T., & Huntly, B. J. (2012). Targeting epigenetic readers in cancer. New England Journal of Medicine, 367(7), 647-657.
[23] Locasale, J. W. (2018). Serine, glycine and one-carbon metabolism in cancer. Nature Reviews Cancer, 18(10), 577-591.
[24] Issa, J. P. (2007). DNA methylation as a therapeutic target in cancer. Clinical Cancer Research, 13(2 Pt 2), 713s-716s.
[25] Stresemann, C., & Lyko, F. (2008). Modes of action of the DNA methyltransferase inhibitors azacytidine and decitabine. International Journal of Cancer, 123(1), 8-13.
[26] Agius, L., & Nebbioso, A. (2019). Targeting ten-eleven translocation (TET) enzymes in cancer. British Journal of Cancer, 120(7), 665-676.
[27] Gore, S. D., Baylin, S. B., & Herman, J. G. (2006). Clinical development of histone deacetylase inhibitors as antineoplastic agents. Annals of the New York Academy of Sciences, 1076(1), 723-738.
[28] Chiappinelli, K. B., Strissel, P. L., Desrichard, A., Li, H., Henke, C., Akman, B., … & Baylin, S. B. (2015). Inhibiting DNA methylation and histone deacetylation synergistically enhances T cell sensitivity to tumors. Cell, 162(5), 974-986.
[29] Vojta, A., Dobrinić, S., Tadić, V., Vulin, A., & Lisnić, B. (2016). Repurposing the CRISPR-Cas9 system for targeted DNA methylation. Nucleic Acids Research, 44(12), 5615-5628.
This is a comprehensive review! Considering the mention of environmental factors influencing DNA methylation, I wonder how personalized nutrition strategies, tailored to an individual’s genetic and epigenetic profile, could be used proactively to modulate DNA methylation and reduce disease risk.