GET THE APP

Exploring Epigenetic Modifications and their Role in Human Disease
..

Journal of Genetics and DNA Research

ISSN: 2684-6039

Open Access

Brief Report - (2024) Volume 8, Issue 3

Exploring Epigenetic Modifications and their Role in Human Disease

Franz Staubach*
*Correspondence: Franz Staubach, Department of Anthropology, University of Helsinki, Yliopistonkatu 4, 00100 Helsinki, Finland, Email:
Department of Anthropology, University of Helsinki, Yliopistonkatu 4, 00100 Helsinki, Finland

Received: 01-May-2024, Manuscript No. jgdr-24-145973; Editor assigned: 02-May-2024, Pre QC No. P-145973; Reviewed: 17-May-2024, QC No. Q-145973; Revised: 22-May-2024, Manuscript No. R-145973; Published: 30-May-2024 , DOI: 10.37421/2684-6039.2024.08.202
Citation: Staubach, Franz. “Exploring Epigenetic Modifications and their Role in Human Disease.” J Genet DNA Res 8 (2024): 202.
Copyright: © 2024 Staubach F. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Introduction

Epigenetics refers to heritable changes in gene expression that do not involve alterations to the DNA sequence. These changes are mediated by a variety of mechanisms, including DNA methylation, histone modification, and the action of non-coding RNAs. Understanding these modifications is critical for elucidating their roles in human health and disease. This article aims to provide a comprehensive overview of epigenetic modifications and their implications for human diseases. Epigenetic modifications, which include DNA methylation, histone modification, and non-coding RNA interactions, play a crucial role in regulating gene expression without altering the underlying DNA sequence. These modifications are essential for normal cellular function and development. Dysregulation of epigenetic mechanisms has been implicated in a range of human diseases, including cancer, cardiovascular diseases, and neurological disorders.

Description

Types of epigenetic modifications

DNA methylation: DNA methylation is a key epigenetic mechanism involving the addition of a methyl group to the cytosine residue in DNA, particularly within CpG dinucleotides. This chemical modification typically results in the repression of gene expression. The presence of a methyl group can prevent the binding of transcription factors and other regulatory proteins to the DNA, thus inhibiting transcription.

DNA methylation patterns are established during development and can be heritable, but they can also be altered in response to environmental factors. Abnormalities in DNA methylation are associated with a range of diseases. For instance, hypermethylation of promoter regions is a common feature in many cancers, leading to the silencing of tumor suppressor genes. Conversely, hypomethylation can activate oncogenes or lead to chromosomal instability [1,2].

Histone modifications: Histone modifications involve the chemical alteration of histone proteins around which DNA is wrapped, influencing the structure of chromatin and the accessibility of the genetic material for transcription. These modifications include acetylation, methylation, phosphorylation, and ubiquitination. Each type of modification can impact gene expression by altering chromatin structure, either making it more open and accessible or more condensed and less accessible.

Acetylation of histones typically correlates with gene activation by reducing the positive charge on histones, leading to a more relaxed chromatin structure. Methylation, depending on the specific amino acid residue and the number of methyl groups added, can either activate or repress gene expression. Phosphorylation often occurs in response to cellular signaling and can impact both gene expression and chromatin dynamics. Ubiquitination is involved in various aspects of chromatin regulation, including the maintenance of histone modification patterns and the response to DNA damage [3].

Non-coding RNAs: Non-coding RNAs play a crucial role in regulating gene expression at various stages, from transcription to post-transcriptional modifications. Unlike protein-coding RNAs, non-coding RNAs do not encode proteins but instead perform regulatory functions through their interactions with other RNA molecules, DNA, or proteins.

MicroRNAs are small non-coding RNAs that typically bind to complementary sequences on target messenger RNAs (mRNAs), leading to their degradation or inhibition of translation. This regulation of mRNA stability and translation affects the expression of genes involved in numerous cellular processes, including development, differentiation, and metabolism. Abnormalities in microRNA expression are associated with a range of diseases, including cancer, where specific microRNAs can act as oncogenes or tumor suppressors [4].

Epigenetics in human disease: Epigenetic modifications have a profound impact on human disease by influencing gene expression patterns without altering the underlying DNA sequence. These modifications can contribute to the development and progression of various diseases through their effects on gene regulation.

In cancer, epigenetic changes are often among the earliest alterations detected and play a critical role in tumorigenesis. Abnormal DNA methylation patterns can lead to the silencing of tumor suppressor genes or the activation of oncogenes. For instance, hypermethylation of CpG islands in the promoter regions of tumor suppressor genes can inhibit their expression, contributing to uncontrolled cell growth and cancer development. Similarly, global hypomethylation can lead to chromosomal instability and activation of oncogenes. Changes in histone modifications also play a role, as altered patterns can lead to the disruption of normal gene expression and chromatin structure, further driving tumor progression.

Cardiovascular diseases are also influenced by epigenetic modifications. For example, DNA methylation changes can affect genes involved in inflammation, endothelial function, and vascular remodeling, all of which are crucial in the development of conditions such as atherosclerosis and hypertension. Histone modifications can similarly impact gene expression related to vascular function and cardiac hypertrophy. In addition, non-coding RNAs, such as microRNAs, have been shown to regulate genes associated with cardiovascular disease, providing another layer of epigenetic control.

Therapeutic approaches

Epigenetic drugs: Epigenetic drugs are therapeutic agents designed to modify epigenetic marks and restore normal gene expression patterns that are disrupted in various diseases. These drugs target specific components of the epigenetic machinery, including DNA methylation, histone modifications, and chromatin remodeling.

One class of epigenetic drugs includes DNA methylation inhibitors. These agents, such as azacitidine and decitabine, work by interfering with the addition of methyl groups to DNA, thereby reversing abnormal DNA methylation patterns. This can reactivate silenced tumor suppressor genes and has shown efficacy in treating certain hematological malignancies like Acute Myeloid Leukemia (AML) and myelodysplastic syndromes.

Gene editing technologies: Gene editing technologies have revolutionized our ability to precisely modify genetic sequences, offering new possibilities for research and therapeutic interventions. One of the most notable advancements in this field is CRISPR/Cas9, a tool derived from bacterial immune systems that can target specific DNA sequences for editing. CRISPR/Cas9 works by introducing a guide RNA that binds to a complementary DNA sequence, guiding the Cas9 protein to create a double-strand break at the target location. The cell then attempts to repair this break, which can be harnessed to introduce specific genetic changes or correct mutations.

Beyond CRISPR/Cas9, other gene editing technologies include TALENs (Transcription Activator-Like Effector Nucleases) and ZFNs (Zinc Finger Nucleases). TALENs use engineered DNA-binding proteins to recognize and bind specific DNA sequences, leading to targeted double-strand breaks. ZFNs operate similarly but use zinc finger domains to achieve sequence specificity. Both TALENs and ZFNs can be used to create targeted mutations, insertions, or deletions, although they are generally considered more complex to design compared to CRISPR/Cas9 [5].

Conclusion

Epigenetic modifications are fundamental to gene regulation and are critically involved in the development and progression of various human diseases. Understanding these modifications provides valuable insights into disease mechanisms and opens up new avenues for therapeutic intervention. Continued research into epigenetic regulation will be essential for developing effective treatments and improving patient outcomes in a range of conditions. Gene editing technologies are increasingly being explored for their potential to correct genetic disorders, enhance agricultural practices, and develop new treatments for diseases. Their precision and versatility make them powerful tools for both basic research and clinical applications, although ongoing research is necessary to address challenges related to efficiency, off-target effects, and ethical considerations. Epigenome editing is another emerging area within gene editing technologies. This approach aims to modify epigenetic marks, such as DNA methylation or histone modifications, without altering the underlying DNA sequence. Techniques like CRISPR/dCas9 (dead Cas9) can be engineered to bind specific genomic locations and recruit epigenetic modifiers, thereby introducing or removing epigenetic marks. This method offers the potential to regulate gene expression precisely and reversibly, providing valuable insights into gene function and potential therapeutic applications.

Acknowledgement

None.

Conflict of Interest

Authors declare no conflict of interest.

References

  1. Scott, Gary K., Michael D. Mattie, Crystal E. Berger and Stephen C. Benz, et al. "Rapid alteration of microRNA levels by histone deacetylase inhibition." Cancer Res66 (2006): 1277-1281.

    Google Scholar, Crossref, Indexed at

  2. Zhao, Sha, Yu Wang, Yunsheng Liang and Ming Zhao, et al. "MicroRNA‐126 regulates DNA methylation in CD4+ T cells and contributes to systemic lupus erythematosus by targeting DNA methyltransferase 1." Arthritis Rheum63 (2011): 1376-1386.

    Google Scholar, Crossref, Indexed at

  3. Yao, Qian, Yuqi Chen and Xiang Zhou. "The roles of microRNAs in epigenetic regulation." Curr Opin Chem Biol51 (2019): 11-17.

    Google Scholar, Crossref, Indexed at

  4. J van der Star, Baukje, Daphne YS Vogel, Markus Kipp and Fabiola Puentes, et al. "In vitro and in vivo models of multiple sclerosis."CNS Neurol Disord Drug Targets11 (2012): 570-588.

    Google Scholar, Crossref, Indexed at

  5. Lalive, Patrice H., Oliver Neuhaus, Mahdia Benkhoucha and Danielle Burger, et al. "Glatiramer acetate in the treatment of multiple sclerosis: Emerging concepts regarding its mechanism of action."CNS drugs25 (2011): 401-414.

    Google Scholar, Crossref, Indexed at

Google Scholar citation report
Citations: 3

Journal of Genetics and DNA Research received 3 citations as per Google Scholar report

Journal of Genetics and DNA Research peer review process verified at publons

Indexed In

 
arrow_upward arrow_upward