Aging is a complex biological process influenced not only by genetic predispositions but also significantly shaped by environmental factors. This review synthesizes experimental evidence from model systems elucidating how environmental exposures modulate genetic aging processes. Studies in organisms such as Caenorhabditis elegans, mice, and human cellular models demonstrate that external conditions including diet, psychosocial stress, pollutants, and physical activity interact dynamically with genetic and epigenetic regulators to influence lifespan and healthspan. Advances in molecular biology and omics technologies reveal mechanisms such as DNA methylation alterations, histone modifications, telomere attrition, oxidative stress, and cellular senescence as critical mediators of gene-environment crosstalk in aging. Genetic manipulation tools like CRISPR and RNA interference enable precise interrogation of genes implicated in environmental responses, deepening understanding of aging pathways. While model organisms provide invaluable platforms to dissect these interactions, challenges remain in translating findings to human aging due to complexity and heterogeneity. Future directions highlight emerging single-cell multiomics, organ-on-chip systems, and artificial intelligence integration to unravel aging's multifactorial nature. The review underscores the necessity of multidisciplinary approaches combining genetics, environmental sciences, and computational biology to develop therapeutic strategies aimed at modulating environmental factors to promote healthy aging. These insights pave the way for personalized interventions targeting both genetic susceptibilities and modifiable environmental risks, ultimately advancing longevity and well-being.
| Published in | European Journal of Clinical and Biomedical Sciences (Volume 11, Issue 4) |
| DOI | 10.11648/j.ejcbs.20251104.12 |
| Page(s) | 49-59 |
| Creative Commons |
This is an Open Access article, distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution and reproduction in any medium or format, provided the original work is properly cited. |
| Copyright |
Copyright © The Author(s), 2025. Published by Science Publishing Group |
Epigenetic Modifications, Model Organisms, Aging Biomarkers, Genetic Manipulation, Environmental Exposures
| [1] | J. Guo et al. Aging and aging-related diseases: from molecular mechanisms to interventions and treatments, Signal Transduct. Target. Ther., vol. 7, no. 1, 2022. |
| [2] | F. Sanada, S. Hayashi, and R. Morishita. Targeting the hallmarks of aging: mechanisms and therapeutic opportunities, Front. Cardiovasc. Med., vol. 12, pp. 1–6, 2025. |
| [3] | M. A. Argentieri et al. Integrating the environmental and genetic architectures of aging and mortality, Nature Medicine, Vol. 31, pp. 1016–1025, 2025. |
| [4] | X. Ding and S. Le Clerc. Editorial: Interaction between genes and the environment in skin aging, Front. Aging, vol. 6, pp. 2024–2026, 2025. |
| [5] | T. Pandics et al. Exposome and unhealthy aging: environmental drivers from air pollution to occupational exposures, GeroScience, vol. 45, no. 6, pp. 3381–3408, 2023. |
| [6] | D. Clancy, S. Chtarbanova, and S. Broughton. Editorial: Model organisms in aging research: Drosophila melanogaster, Front. Aging, vol. 3, pp. 1–3, 2022. |
| [7] | Y. Shen, M. Stanislauskas, G. Li, D. Zheng, and L. Liu. Epigenetic and genetic dissections of UV-induced global gene dysregulation in skin cells through multi-omics analyses, Sci. Rep., vol. 7, pp. 1–12, 2017. |
| [8] | S. Jinesh, B. Özüpek, and P. Aditi. Premature aging and metabolic diseases: the impact of telomere attrition, Front. Aging, vol. 6, pp. 1–20, 2025. |
| [9] | D. McHugh and J. Gil. Senescence and aging: Causes, consequences, and therapeutic avenues, J. Cell Biol., vol. 217, no. 1, pp. 65–77, 2018. |
| [10] | R. Kumari and P. Jat. Mechanisms of Cellular Senescence: Cell Cycle Arrest and Senescence Associated Secretory Phenotype, Front. Cell Dev. Biol., vol. 9, pp. 1–24, 2021. |
| [11] | S. Sándor and E. Kubinyi. Genetic Pathways of Aging and Their Relevance in the Dog as a Natural Model of Human Aging,” Front. Genet., vol. 10, pp. 1–34, 2019. |
| [12] | A. Vaiserman and D. Krasnienkov. Telomere Length as a Marker of Biological Age: State-of-the-Art, Open Issues, and Future Perspectives, Front. Genet., vol. 11, 2021. |
| [13] | S. Andreu-Sánchez et al. Genetic, parental and lifestyle factors influence telomere length, Commun. Biol., vol. 5, no. 1, pp. 1–14, 2022. |
| [14] | R. Mesnage. Environmental Health Is Overlooked in Longevity Research, Antioxidants, vol. 14, no. 4, pp. 1–13, 2025. |
| [15] | S. N. Palani, D. Sellegounder, P. Wibisono, and Y. Liu. The longevity response to warm temperature is neurally controlled via the regulation of collagen genes, Aging Cell, vol. 22, no. 5, pp. 1–17, 2023. |
| [16] | K. E. Gribble, B. M. Moran, S. Jones, E. L. Corey, and D. B. Mark Welch. Congeneric variability in lifespan extension and onset of senescence suggest active regulation of aging in response to low temperature, Exp. Gerontol., vol. 114, pp. 99–106, 2018. |
| [17] | N. A. Ruprecht, S. Singhal, D. Sens, and S. K. Singhal. Translating genetic findings to epigenetics: identifying the mechanisms associated with aging after high-radiation exposure on earth and in space, Front. Public Heal., vol. 12, 2024. |
| [18] | M. Portillo-Esnaola et al. Formation of cyclobutane pyrimidine dimers after UVA exposure (Dark-CPDs) is inhibited by an hydrophilic extract of polypodium leucotomos, Antioxidants, vol. 10, no. 12, 2021. |
| [19] | H. M. Han, S. Y. Kim, and D. H. Kim. Mechanotransduction for therapeutic approaches: Cellular aging and rejuvenation, APL Bioeng., vol. 9, no. 2, pp. 1–23, 2025. |
| [20] | K. M. C. Malecki et al. Integrating Environment and Aging Research: Opportunities for Synergy and Acceleration, Front. Aging Neurosci., vol. 14, pp. 1–19, 2022. |
| [21] | M. Ahmed. Targeting aging pathways with natural compounds: a review of curcumin, epigallocatechin gallate, thymoquinone, and resveratrol, Immun. Ageing, vol. 22, no. 1, 2025. |
| [22] | S. McLean et al. Molecular mechanisms of genotype-dependent lifespan variation mediated by caloric restriction: insight from wild yeast isolates, Front. Aging, vol. 5, pp. 1–19, 2024. |
| [23] | J. L. Dorling, C. K. Martin, and L. M. Redman. Calorie restriction for enhanced longevity: The role of novel dietary strategies in the present obesogenic environment, Ageing Res. Rev., vol. 64, 2020. |
| [24] | E. W. Flanagan, J. Most, J. T. Mey, and L. M. Redman. Calorie Restriction and Aging in Humans, Annu. Rev. Nutr., vol. 40, pp. 105–133, 2020. |
| [25] | L. Best et al. Metabolic modelling reveals the aging-associated decline of host– microbiome metabolic interactions in mice, nature microbiology Article, Vol. 10, pp. 973-991, 2005. |
| [26] | C. N. Brooks, M. E. Wight, O. E. Azeez, R. M. Bleich, and K. A. Zwetsloot. Growing old together: What we know about the influence of diet and exercise on the aging host’s gut microbiome, Front. Sport. Act. Living, vol. 5, pp. 1–9, 2023. |
| [27] | S. Yang, S. Hwang, B. Kim, S. Shin, M. Kim, and S. M. Jeong. Fatty acid oxidation facilitates DNA double-strand break repair by promoting PARP1 acetylation, Cell Death Dis., vol. 14, no. 7, 2023. |
| [28] | B. Cisneros et al. Immune system modulation in aging: Molecular mechanisms and therapeutic targets, Front. Immunol., vol. 13, pp. 1–8, 2022. |
| [29] | M. Kumar et al. Environmental Endocrine-Disrupting Chemical Exposure: Role in Non-Communicable Diseases, Front. Public Heal., vol. 8, pp. 1–28, 2020. |
| [30] | P. V. Carapeto and C. Aguayo-Mazzucato. Effects of exercise on cellular and tissue aging, Aging (Albany. NY)., vol. 13, no. 10, pp. 14522–14543, 2021. |
| [31] | A. Rebelo-Marques et al. Aging hallmarks: The benefits of physical exercise, Front. Endocrinol. (Lausanne)., vol. 9, pp. 1–15, 2018. |
| [32] | J. Ostaiza-Cardenas et al. Epigenetic Modulation by Lifestyle: Advances in Diet, Exercise, and Mindfulness for Disease Prevention and Health Optimization, Front. Nutr., vol. 12, no. 1, p. 1632999. |
| [33] | D. Sorriento, E. Di Vaia, and G. Iaccarino. Physical Exercise: A Novel Tool to Protect Mitochondrial Health, Front. Physiol., vol. 12, pp. 1–14, 2021. |
| [34] | Z. M. Harvanek, N. Fogelman, K. Xu, and R. Sinha. Psychological and biological resilience modulates the effects of stress on epigenetic aging, Transl. Psychiatry, vol. 11, no. 1, pp. 1–9, 2021. |
| [35] | J. M. Lane et al. Genetics of circadian rhythms and sleep in human health and disease, Nat Rev Genet. vol. 24, no. 1, pp. 4–20, 2024. |
| [36] | D. Malhan, B. Schoenrock, M. Yalçin, D. Blottner, and A. Relόgio. Circadian regulation in aging: Implications for spaceflight and life on earth, Aging Cell, vol. 22, no. 9, pp. 1–23, 2023. |
| [37] | S. Garbarino, P. Lanteri, W. G. Sannita, N. L. Bragazzi, and E. Scoditti. Circadian Rhythms, Sleep, Immunity, and Fragility in the Elderly: The Model of the Susceptibility to Infections, Front. Neurol., vol. 11, pp. 10–14, 2020. |
| [38] | V. A. Acosta-Rodríguez, F. Rijo-Ferreira, C. B. Green, and J. S. Takahashi. Importance of circadian timing for aging and longevity, Nat. Commun., vol. 12, no. 1, 2021. |
| [39] | P. Sen, P. P. Shah, R. Nativio, and S. L. Berger. Epigenetic mechanisms regulating longevity and aging, Cell, vol. 166, no. 4, pp. 822–39, 2016. |
| [40] | E. Santiago, D. F. Moreno, and M. Acar. Modeling aging and its impact on cellular function and organismal behavior, Exp. Gerontol., vol. 155, pp. 1–42, 2021. |
| [41] | J. A. N. L. F. Freitas and O. Bischof. Computational modeling of aging-related gene networks: a review,” Front. Appl. Math. Stat., vol. 10, pp. 1–16, 2024. |
| [42] | P. Dhar, S. S. Moodithaya, and P. Patil. Epigenetic alterations, The silent indicator for early aging and age-associated health-risks, Aging Med., vol. 5, no. 4, pp. 287–293, 2022. |
| [43] | J. Song et al. Effects of reactive oxygen species and mitochondrial dysfunction on reproductive aging, Front. Cell Dev. Biol., vol. 12, no. February, pp. 1–12, 2024. |
| [44] | D. Khodasevich et al. Exposome-wide association study of environmental chemical exposures and epigenetic aging in the national health and nutrition examination survey, Aging (Albany. NY)., vol. 17, no. 2, pp. 408–430, 2025. |
| [45] | L. S. Treviño et al. Epigenome environment interactions accelerate epigenomic aging and unlock metabolically restricted epigenetic reprogramming in adulthood, Nat. Commun., vol. 11, no. 1, pp. 1–14, 2020. |
| [46] | C. E. Finch and A. Haghani. Gene-Environment Interactions and Stochastic Variations in the Gero-Exposome, Journals Gerontol. - Ser. A Biol. Sci. Med. Sci., vol. 76, no. 10, pp. 1740–1747, 2021. |
| [47] | K. Wang et al. Epigenetic regulation of aging: implications for interventions of aging and diseases, Signal Transduct. Target. Ther., vol. 7, no. 1, 2022. |
| [48] | J. Yu, T. Li, and J. Zhu. Gene Therapy Strategies Targeting Aging-Related Diseases, Aging Dis., vol. 14, no. 2, pp. 398–417, 2023. |
| [49] | A. Azani, M. Sharafi, R. Doachi, S. Akbarzadeh, and P. Lorestani. Applications of CRISPR ‑ Cas9 in mitigating cellular senescence and age ‑ related disease progression, 2025. |
| [50] | D. Gómez-Domínguez et al. CRISPR/Cas9-mediated elimination of the LMNA c.745C>G pathogenic mutation enhances survival and cardiac function in LMNA-associated congenital muscular dystrophy, bioRxiv, p. 2025.02.13.638060, 2025. |
| [51] | X. Liu et al. A large-scale CRISPR screen and identification of essential genes in cellular senescence bypass, Aging (Albany. NY)., vol. 11, no. 12, pp. 4011–4031, 2019. |
| [52] | A. Caobi et al. The impact of CRISPR-Cas9 on age-related disorders: From pathology to therapy, Aging Dis., vol. 11, no. 4, pp. 895–915, 2020. |
| [53] | Michael Boettcher and Michael T. McManus. Choosing the Right Tool for the Job: RNAi, TALEN or CRISPR, Mol Cell. vol. 176, no. 1, pp. 139–148, 2017. |
| [54] | L. Lu, X. Yu, Y. Cai, M. Sun, and H. Yang. Application of CRISPR/Cas9 in Alzheimer’s Disease, Front. Neurosci., vol. 15, pp. 1–14, 2021. |
| [55] | X. Shen et al. Nonlinear dynamics of multi-omics profiles during human aging, nature aging, vol. 4, pp. 1619-1634, 2024. |
| [56] | A. Perez-Gomez, J. N. Buxbaum, and M. Petrascheck. The aging transcriptome: read between the lines, Curr. Opin. Neurobiol., vol. 63, pp. 170–175, 2020. |
| [57] | D. Palmer, F. Fabris, A. Doherty, A. A. Freitas, and J. P. De Magalhães. Ageing transcriptome meta-analysis reveals similarities and, vol. 13, no. 3, 2021. |
| [58] | J. S. Lorusso, O. A. Sviderskiy, and V. M. Labunskyy. Emerging Omics Approaches in Aging Research, Antioxidants Redox Signal., vol. 29, no. 10, pp. 985–1002, 2018. |
| [59] | B. Long et al. Advances in the application of multi-omics analysis in skin aging, Front. Aging, vol. 6, no. July, pp. 1–14, 2025. |
| [60] | S. Mao, J. Su, L. Wang, X. Bo, C. Li, and H. Chen. A transcriptome-based single-cell biological age model and resource for tissue-specific aging measures, Genome Res., vol. 33, no. 8, pp. 1381–1395, 2023. |
| [61] | R. Barrere-Cain and P. Allard. An Understudied Dimension: Why Age Needs to Be Considered When Studying Epigenetic-Environment Interactions, Epigenetics Insights, vol. 13, 2020. |
| [62] | L. R. Polsky, K. E. Rentscher, and J. E. Carroll. Stress-induced biological aging: A review and guide for research priorities, Brain. Behav. Immun., vol. 104, pp. 97–109, 2022. |
| [63] | V. Kalia, D. W. Belsky, A. A. Baccarelli, and G. W. Miller. An exposomic framework to uncover environmental drivers of aging, Exposome, vol. 2, no. 1, 2022. |
| [64] | J. Bortz et al. Biological age estimation using circulating blood biomarkers, Commun. Biol., vol. 6, no. 1, pp. 1–10, 2023. |
| [65] | A. Unnikrishnan et al. The role of DNA methylation in epigenetics of aging, Pharmacol Ther. Vol. 195, pp. 172–185, 2020. |
| [66] | T. Bergsma and E. Rogaeva. DNA Methylation Clocks and Their Predictive Capacity for Aging Phenotypes and Healthspan, Neurosci. Insights, vol. 15, 2020. |
| [67] | X. Tao et al. Biomarkers of Aging and Relevant Evaluation Techniques: A Comprehensive Review, Aging Dis., vol. 15, no. 3, pp. 977–1005, 2024. |
| [68] | L. Chen et al. Associations between biological ageing and the risk of, genetic susceptibility to, and life expectancy associated with rheumatoid arthritis: a secondary analysis of two observational studies, Lancet Heal. Longev., vol. 5, no. 1, pp. e45–e55, 2024. |
| [69] | N. A. Koemel and M. R. Skilton. Epigenetic Aging in Early Life: Role of Maternal and Early Childhood Nutrition, Curr. Nutr. Rep., vol. 11, no. 2, pp. 318–328, 2022. |
| [70] | F. Bertile, S. Matallana-Surget, A. Tholey, S. Cristobal, and J. Armengaud. Diversifying the concept of model organisms in the age of -omics, Commun. Biol., vol. 6, no. 1, pp. 4–7, 2023. |
| [71] | S. Holtze et al. Alternative Animal Models of Aging Research, Front. Mol. Biosci., vol. 8, no. May, 2021. |
| [72] | M. Bolster et al. Ageing well in the urban environment: meeting the health and social needs of older Adults - study protocol for a prospective, longitudinal mixed-methods study, BMC Geriatr., vol. 25, no. 1, 2025. |
| [73] | X. Wu et al. Single-cell sequencing to multi-omics: technologies and applications, Biomark. Res., vol. 12, no. 1, pp. 1–28, 2024. |
| [74] | D. M. Ruden, A. Singh, and D. A. Rappolee. Pathological epigenetic events and reversibility review: The intersection between hallmarks of aging and developmental origin of health and disease, Epigenomics, vol. 15, no. 14, pp. 741–754, 2023. |
| [75] | L. Zwi-Dantsis, V. Jayarajan, G. M. Church, R. D. Kamm, J. P. de Magalhães, and E. Moeendarbary. Aging on Chip: Harnessing the Potential of Microfluidic Technologies in Aging and Rejuvenation Research, Adv. Healthc. Mater., vol. 2500217, 2025. |
| [76] | S. Kadyan et al. Microbiome-based therapeutics towards healthier aging and longevity, Genome Med., vol. 17, no. 1, 2025. |
| [77] | N. Reich and C. Hölscher. The neuroprotective effects of glucagon-like peptide 1 in Alzheimer’s and Parkinson’s disease: An in-depth review, Front. Neurosci., vol. 16, no. September, pp. 1–55, 2022. |
| [78] | M. Bahri, K. Epstein, E. Stevens, A. E. Rosko, S. Maturu, and Y. Zhang. Implementing a multidisciplinary approach for older adults with multiple sclerosis: Geriatric neurology in practice, 2025. |
| [79] | D. Wilczok. Deep learning and generative artificial intelligence in aging research and healthy longevity medicine, Aging (Albany. NY)., vol. 17, no. 1, pp. 251–275, 2025. |
| [80] | T. J. Phua. The human biological clock and aging a comprehensive approach integrating reductionism, holism, and geromedicine for proactive healthspan strategies, Front. Aging, vol. 6, pp. 1–7, 2025. |
APA Style
Molla, A. (2025). Environmental Influences on Genetic Aging Processes: Experimental Evidence from Model Systems. European Journal of Clinical and Biomedical Sciences, 11(4), 49-59. https://doi.org/10.11648/j.ejcbs.20251104.12
ACS Style
Molla, A. Environmental Influences on Genetic Aging Processes: Experimental Evidence from Model Systems. Eur. J. Clin. Biomed. Sci. 2025, 11(4), 49-59. doi: 10.11648/j.ejcbs.20251104.12
@article{10.11648/j.ejcbs.20251104.12,
author = {Alebachew Molla},
title = {Environmental Influences on Genetic Aging Processes: Experimental Evidence from Model Systems
},
journal = {European Journal of Clinical and Biomedical Sciences},
volume = {11},
number = {4},
pages = {49-59},
doi = {10.11648/j.ejcbs.20251104.12},
url = {https://doi.org/10.11648/j.ejcbs.20251104.12},
eprint = {https://article.sciencepublishinggroup.com/pdf/10.11648.j.ejcbs.20251104.12},
abstract = {Aging is a complex biological process influenced not only by genetic predispositions but also significantly shaped by environmental factors. This review synthesizes experimental evidence from model systems elucidating how environmental exposures modulate genetic aging processes. Studies in organisms such as Caenorhabditis elegans, mice, and human cellular models demonstrate that external conditions including diet, psychosocial stress, pollutants, and physical activity interact dynamically with genetic and epigenetic regulators to influence lifespan and healthspan. Advances in molecular biology and omics technologies reveal mechanisms such as DNA methylation alterations, histone modifications, telomere attrition, oxidative stress, and cellular senescence as critical mediators of gene-environment crosstalk in aging. Genetic manipulation tools like CRISPR and RNA interference enable precise interrogation of genes implicated in environmental responses, deepening understanding of aging pathways. While model organisms provide invaluable platforms to dissect these interactions, challenges remain in translating findings to human aging due to complexity and heterogeneity. Future directions highlight emerging single-cell multiomics, organ-on-chip systems, and artificial intelligence integration to unravel aging's multifactorial nature. The review underscores the necessity of multidisciplinary approaches combining genetics, environmental sciences, and computational biology to develop therapeutic strategies aimed at modulating environmental factors to promote healthy aging. These insights pave the way for personalized interventions targeting both genetic susceptibilities and modifiable environmental risks, ultimately advancing longevity and well-being.
},
year = {2025}
}
TY - JOUR T1 - Environmental Influences on Genetic Aging Processes: Experimental Evidence from Model Systems AU - Alebachew Molla Y1 - 2025/10/31 PY - 2025 N1 - https://doi.org/10.11648/j.ejcbs.20251104.12 DO - 10.11648/j.ejcbs.20251104.12 T2 - European Journal of Clinical and Biomedical Sciences JF - European Journal of Clinical and Biomedical Sciences JO - European Journal of Clinical and Biomedical Sciences SP - 49 EP - 59 PB - Science Publishing Group SN - 2575-5005 UR - https://doi.org/10.11648/j.ejcbs.20251104.12 AB - Aging is a complex biological process influenced not only by genetic predispositions but also significantly shaped by environmental factors. This review synthesizes experimental evidence from model systems elucidating how environmental exposures modulate genetic aging processes. Studies in organisms such as Caenorhabditis elegans, mice, and human cellular models demonstrate that external conditions including diet, psychosocial stress, pollutants, and physical activity interact dynamically with genetic and epigenetic regulators to influence lifespan and healthspan. Advances in molecular biology and omics technologies reveal mechanisms such as DNA methylation alterations, histone modifications, telomere attrition, oxidative stress, and cellular senescence as critical mediators of gene-environment crosstalk in aging. Genetic manipulation tools like CRISPR and RNA interference enable precise interrogation of genes implicated in environmental responses, deepening understanding of aging pathways. While model organisms provide invaluable platforms to dissect these interactions, challenges remain in translating findings to human aging due to complexity and heterogeneity. Future directions highlight emerging single-cell multiomics, organ-on-chip systems, and artificial intelligence integration to unravel aging's multifactorial nature. The review underscores the necessity of multidisciplinary approaches combining genetics, environmental sciences, and computational biology to develop therapeutic strategies aimed at modulating environmental factors to promote healthy aging. These insights pave the way for personalized interventions targeting both genetic susceptibilities and modifiable environmental risks, ultimately advancing longevity and well-being. VL - 11 IS - 4 ER -