Perspective, Jrgm Vol: 13 Issue: 5
Molecular Pathways of Rejuvenation: Delaying Aging through Epigenetic Regulation.
Tung Hoang*
Institute of Biomedicine, University of Medicine and Pharmacy, Hue University, Viet Nam
*Corresponding Author: Tung Hoang
Institute of Biomedicine, University of Medicine and Pharmacy, Hue University, Viet Nam
E-mail: hoang@huemed-univ.edu.vn
Received: 02-Sep-2024, Manuscript No. JRGM-24-148129
Editor assigned: 03-Sep-2024, PreQC No. JRGM-24-148129 (PQ)
Reviewed: 17-Sep-2024, QC No. JRGM-24-148129
Revised: 23-Sep-2024, Manuscript No. JRGM-24-148129 (R)
Published: 27-Sep-2024, DOI:10.4172/2325-9620.1000332
Citation: Prins C (2022) Contrasting Intensive Trauma-Focused Treatment Outcome on PTSD Indication strength in Older and Younger. J Trauma Stress Disor Treat 11:2.
Copyright: © 2024 Hoang T. 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
Aging is a complex biological process characterized by the gradual decline in physiological functions and an increased risk of age-related diseases. In recent years, the focus has shifted from merely understanding aging to exploring ways to delay or reverse its effects. One promising area of research is epigenetic regulation, which involves changes in gene expression without altering the underlying DNA sequence. This article explores how epigenetic mechanisms influence aging and the potential for rejuvenation through these molecular pathways [1].
Epigenetics refers to modifications on DNA or histone proteins that affect gene expression. These modifications include DNA methylation, histone acetylation, and chromatin remodeling. As individuals age, their epigenetic landscape undergoes significant changes, leading to altered gene expression patterns. These changes are associated with various aging phenotypes, including cellular senescence, reduced regenerative capacity, and increased susceptibility to diseases [2].
DNA methylation involves the addition of a methyl group to the cytosine base of DNA, typically repressing gene expression. During aging, global DNA methylation levels decrease, while specific regions of the genome may become hypermethylated. This altered methylation pattern can disrupt normal gene function and contribute to age-related diseases. For instance, the loss of DNA methylation in stem cells impairs their regenerative potential, while aberrant methylation in specific genes can lead to cancer [3].
Histones are proteins around which DNA is wrapped to form chromatin. Post-translational modifications of histones, such as acetylation, methylation, and phosphorylation, regulate chromatin structure and gene expression. Aging is associated with changes in histone modifications that can lead to altered gene expression profiles. For example, reduced histone acetylation can result in a more closed chromatin structure and decreased gene expression, contributing to cellular senescence [4].
Chromatin remodeling involves the repositioning of nucleosomes and changes in chromatin structure, impacting gene accessibility. Age-related chromatin remodeling can lead to the silencing of genes essential for maintaining cellular function and enhancing inflammation. Studies have shown that restoring proper chromatin structure can ameliorate some age-associated defects, highlighting the potential for interventions targeting chromatin remodeling pathways [5].
Epigenetic clocks are tools used to measure biological age based on DNA methylation patterns. These clocks provide a quantitative assessment of aging and have been used to evaluate the effectiveness of anti-aging interventions. By targeting specific epigenetic marks, researchers aim to "rejuvenate" cells and tissues, effectively reversing some aspects of biological aging [6].
Several strategies have been proposed to delay aging and promote rejuvenation through epigenetic regulation. These include: Small molecules that modulate epigenetic marks, such as histone deacetylase inhibitors or DNA methyltransferase inhibitors, have shown promise in preclinical studies. These compounds can potentially reverse some age-related epigenetic changes and restore cellular function [7].
Techniques like CRISPR/Cas9 allow for precise modifications of epigenetic marks. Researchers are exploring the use of gene editing to correct aberrant DNA methylation or histone modifications associated with aging [8].
Diet and exercise can influence epigenetic marks and contribute to healthy aging. Nutritional factors, such as polyphenols and vitamins, have been shown to affect DNA methylation and histone modifications, highlighting the potential for lifestyle changes to impact epigenetic regulation [9].
Despite the exciting prospects, several challenges remain in the field of epigenetic rejuvenation. The complexity of the epigenetic landscape and the potential for off-target effects with epigenetic therapies necessitate thorough research and cautious application. Additionally, individual variability in epigenetic responses underscores the need for personalized approaches in anti-aging interventions [10].
Conclusion
Epigenetic regulation offers a novel avenue for delaying aging and promoting rejuvenation. By targeting molecular pathways involved in epigenetic modifications, researchers aim to address the underlying mechanisms of aging and improve healthspan. As our understanding of epigenetic processes deepens and new technologies emerge, the potential for epigenetic-based therapies to transform aging and longevity becomes increasingly promising. Continued research in this field holds the key to unlocking innovative strategies for extending health and vitality into older age.
References
- Puck J M (2019) Newborn screening for severe combined immunodeficiency and T-cell lymphopenia. Immunol Rev;287(1):241–52.
- Booth C, Romano R, Roncarolo M G,Thrasher A J (2019) Gene therapy for primary immunodeficiency. Hum Gene Ther; 6(6):709–10.
- Kohn L A, Kohn D B (2021) Gene therapies for primary immune deficiencies. Front Immunol;12:648951.
- Ferrari G, Thrasher A J, Aiuti A (2021) Gene therapy using haematopoietic stem and progenitor cells. Nat Rev Genet; 22(4):216–34.
- Ali M, Pages E, Ducom A, Fontaine A, Guillemot F (2014) Controlling laser-induced jet formation for bioprinting mesenchymal stem cells with high viability and high resolution. Biofabrication 6:045001.
- Feng C, Zhang W, Deng C, Li G, Chang J, et al. (2017) 3D printing of lotus root-like biomimetic materials for cell delivery and tissue regeneration. Adv Sci 4:1700401.
- Ali M, Pages E, Ducom A, Fontaine A, Guillemot F (2014) Controlling laser-induced jet formation for bioprinting mesenchymal stem cells with high viability and high resolution. Biofabrication 6:045001.
- Bruton O. C (1952) Agammaglobulinemia. Pediatrics; 9(6):722–6.
- Dropulic L. K, Lederman H M (2016) Overview of infections in the immunocompromised host. Microbiol Spectr; 4(4):1–43
- Zhang L, Thrasher AJ, Gaspar HB (2013) Current progress on gene therapy for primary immunodeficiencies. Gene Ther;20(10):963-9.
Indexed at, Google Scholar, Cross Ref
Indexed at, Google Scholar, Cross Ref
Indexed at, Google Scholar, Cross Ref
Indexed at, Google Scholar, Cross Ref
Indexed at, Google Scholar, Cross Ref
Indexed at, Google Scholar, Cross Ref
Indexed at, Google Scholar, Cross Ref
Indexed at, Google Scholar, Cross Ref