Cellular senescence and immunosenescence encompass critical molecular pathways that govern aging and age-related pathologies. Central to cellular senescence are DNA damage response activation, telomere attrition, chromatin remodeling, metabolic reprogramming, and cytoplasmic DNA sensing via cGAS-STING signaling, which collectively drive cell cycle arrest and the pro-inflammatory senescence-associated secretory phenotype (SASP). Immunosenescence involves progressive deterioration of immune cell function characterized by depleted naive lymphocytes, accumulation of dysfunctional senescent immune cells, and chronic inflammation (inflammaging), creating a feedback loop that exacerbates tissue degeneration and systemic aging. Model organisms such as mice and killifish have been indispensable for unraveling these mechanisms, enabling genetic and functional studies that illuminate senescence dynamics and immune clearance processes. Future research, empowered by multi-omics, single cell sequencing, and artificial intelligence, promises deeper dissection of senescence heterogeneity and tissue-specific pathways, offering biomarkers and therapeutic targets with unprecedented precision. Therapeutic strategies aiming to selectively eliminate or modulate senescent cells through senolytics, senomorphics, and immunomodulatory approaches hold promise to extend health span and ameliorate chronic diseases. However, challenges including senescent cell heterogeneity, context-dependent functions, and biomarker limitations necessitate individualized and careful translation of findings into clinical therapies. Continued interdisciplinary efforts integrating molecular biology, systems medicine, and clinical research will be pivotal in harnessing the full potential of senescence targeting for healthy aging and transformative disease management. This review was conducted to comprehensively compile and discuss the intricate molecular mechanisms underlying cellular senescence and immunosenescence, which are critical processes involved in aging and age-related diseases. The aim of this review article is to comprehensively elucidate the molecular mechanisms underlying cellular senescence and immunosenescence, integrating insights gained from model organism research and emerging signaling pathways.
| Published in | American Journal of Biomedical and Life Sciences (Volume 13, Issue 5) |
| DOI | 10.11648/j.ajbls.20251305.12 |
| Page(s) | 98-113 |
| 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 |
Cellular Senescence, Immunosenescence, Senescence-Associated Secretory Phenotype, DNA Damage Response, Senolytic Therapies
| [1] | K. A. Lee, R. R. Flores, I. H. Jang, A. Saathoff, and P. D. Robbins. Immune Senescence, Immunosenescence and Aging, Front. Aging, vol. 3, no. May, pp. 1-7, 2022. |
| [2] | K. Razzaq and G. Dickinson. The aging immune system, A Compr. Guid. to Geriatr. Rehabil. Third Ed., vol. 13, no. 15, pp. 64-67, 2014. |
| [3] | N. Herranz and J. Gil. Mechanisms and functions of cellular senescence,” J. Clin. Invest., vol. 128, no. 4, pp. 1238-1246, 2018. |
| [4] | R. Kumari and P. Jat. Mechanisms of Cellular Senescence: Cell Cycle Arrest and Senescence Associated Secretory Phenotype, Front. Cell Dev. Biol., vol. 9, no. March, pp. 1-24, 2021. |
| [5] | Z. Liu et al. Immunosenescence: molecular mechanisms and diseases, Signal Transduct. Target. Ther., vol. 8, no. 1, 2023. |
| [6] | Z. Liang, X. Dong, Z. Zhang, Q. Zhang, and Y. Zhao. Age-related thymic involution: Mechanisms and functional impact, Aging Cell, vol. 21, no. 8, pp. 1-14, 2022. |
| [7] | L. Prata et al. Senescent cell clearance by the immune system: Emerging therapeutic opportunities, Semin Immunol. Vol. 40: 101275. 2018. |
| [8] | O. Perdaens and V. van Pesch. Molecular Mechanisms of Immunosenescene and Inflammaging: Relevance to the Immunopathogenesis and Treatment of Multiple Sclerosis, Front. Neurol., vol. 12, pp. 1-29, 2022. |
| [9] | C. Li, Y. Yuan, Y. D. Jia, Q. Zhou, Q. Wang, and X. Jiang. Cellular senescence: from homeostasis to pathological implications and therapeutic strategies, Front. Immunol., vol. 16, no. February, pp. 1-24, 2025. |
| [10] | O. Lushchak, M. Schosserer, and J. Grillari. Senopathies Diseases Associated with Cellular Senescence, Biomolecules, vol. 13, no. 6, pp. 1-10, 2023. |
| [11] | S. Wang et al. Recent Advances in Aging and Immunosenescence: Mechanisms and Therapeutic Strategies, Cells, vol. 14, no. 7, pp. 1-22, 2025. |
| [12] | J. L. St. Sauver et al. Biomarkers of cellular senescence and risk of death in humans, Aging Cell, vol. 22, no. 12, pp. 1-10, 2023. |
| [13] | R. A. Fielding et al. Biomarkers of Cellular Senescence Predict the Onset of Mobility Disability and Are Reduced by Physical Activity in Older Adults, Journals Gerontol. - Ser. A Biol. Sci. Med. Sci., vol. 79, no. 3, pp. 1-9, 2024. |
| [14] | S. Yamauchi and A. Takahashi. Cellular senescence: mechanisms and relevance to cancer and aging, J. Biochem., vol. 177, no. 3, pp. 163-169, 2025. |
| [15] | D. R. Valenzano, A. Aboobaker, A. Seluanov, and V. Gorbunova. Non‐canonical aging model systems and why we need them, EMBO J., vol. 36, no. 8, pp. 959-963, 2017. |
| [16] | G. Taormina, F. Ferrante, S. Vieni, N. Grassi, A. Russo, and M. G. Mirisola. Longevity: Lesson from model organisms, Genes (Basel)., vol. 10, no. 7, 2019. |
| [17] | M. Mijit, V. Caracciolo, A. Melillo, F. Amicarelli, and A. Giordano. Role of p53 in the regulation of cellular senescence, Biomolecules, vol. 10, no. 3, pp. 1-16, 2020. |
| [18] | K. Engeland. Cell cycle regulation: p53-p21-RB signaling, Cell Death Differ., vol. 29, no. 5, pp. 946-960, 2022. |
| [19] | X. Wu, X. Zhou, S. Wang, and G. Mao. DNA damage response (DDR): a link between cellular senescence and human cytomegalovirus, Virol. J., vol. 20, no. 1, pp. 1-14, 2023. |
| [20] | S. Nikfarjam and K. K. Singh. DNA damage response signaling: A common link between cancer and cardiovascular diseases, Cancer Med., vol. 12, no. 4, pp. 4380-4404, 2023. |
| [21] | Y. Abuetabh et al. DNA damage response revisited: the p53 family and its regulators provide endless cancer therapy opportunities, Exp. Mol. Med., vol. 54, no. 10, pp. 1658-1669, 2022. |
| [22] | V. Janelle et al. p16INK4a Regulates Cellular Senescence in PD-1-Expressing Human T Cells, Front. Immunol., vol. 12, pp. 1-17, 2021. |
| [23] | S. Victorelli and J. F. Passos. Telomeres and Cell Senescence - Size Matters Not, EBioMedicine, vol. 21, pp. 14-20, 2017. |
| [24] | E. Bonnell, E. Pasquier, and R. J. Wellinger. Telomere Replication: Solving Multiple End Replication Problems, Front. Cell Dev. Biol., vol. 9, pp. 1-17, 2021. |
| [25] | A. Harman and T. M. Bryan. Telomere maintenance and the DNA damage response: a paradoxical alliance, Front. Cell Dev. Biol., vol. 12, pp. 1-10, 2024. |
| [26] | I. Rodriguez-Brenes et al. The role of telomere shortening in carcinogenesis: A hybrid stochastic-deterministic approach, J Theor Biol. vol. 176, no. 10, pp. 139-148, 2017, |
| [27] | H. A. Shaban and S. M. Gasser. Dynamic 3D genome reorganization during senescence: defining cell states through chromatin, Cell Death Differ., vol. 32, no. 1, pp. 9-15, 2025. |
| [28] | M. Dhankhar et al. Revealing the biophysics of lamina-associated domain formation by integrating theoretical modeling and high-resolution imaging, Nat. Commun., vol. 16, no. 1, 2025. |
| [29] | Y. Liu, Y. Feng, L. Cheng, Y. Xu, A. Wu, and P. Cheng. Profiling with senescence-associated secretory phenotype score identifies GDC-0879 as a small molecule sensitizing glioblastoma to anti-PD1, Cell Death Dis., vol. 16, no. 1, 2025. |
| [30] | M. Takasugi, Y. Yoshida, and N. Ohtani. Cellular senescence and the tumour microenvironment, Mol. Oncol., vol. 16, no. 18, pp. 3333-3351, 2022. |
| [31] | Z. Han, K. Wang, S. Ding, and M. Zhang. Cross-talk of inflammation and cellular senescence: a new insight into the occurrence and progression of osteoarthritis, Bone Res., vol. 12, no. 1, 2024. |
| [32] | K. Kirschner, N. Rattanavirotkul, M. F. Quince, and T. Chandra. Functional heterogeneity in senescence, Biochem. Soc. Trans., vol. 48, no. 3, pp. 765-773, 2020. |
| [33] | X. Han et al. Potential Regulators of the Senescence-Associated Secretory Phenotype During Senescence and Aging, Journals Gerontol. - Ser. A Biol. Sci. Med. Sci., vol. 77, no. 11, pp. 2207-2218, 2022. |
| [34] | N. Dasgupta, R. Arnold, A. Equey, A. Gandhi, and P. D. Adams. The role of the dynamic epigenetic landscape in senescence: orchestrating SASP expression, npj Aging, vol. 10, no. 1, pp. 1-11, 2024. |
| [35] | K. Wang et al. Epigenetic regulation of aging: implications for interventions of aging and diseases, Signal Transduct. Target. Ther., vol. 7, no. 1, 2022. |
| [36] | H. Paluvai, E. Di Giorgio, and C. Brancolini. The histone code of senescence, Cells, vol. 9, no. 2, 2020. |
| [37] | C. Sidler, O. Kovalchuk, and I. Kovalchuk. Epigenetic regulation of cellular senescence and aging, Front. Genet., vol. 8, no. SEP, 2017. |
| [38] | Y. Guan et al. Senescence-activated enhancer landscape orchestrates the senescence-associated secretory phenotype in murine fibroblasts, Nucleic Acids Res., vol. 48, no. 19, pp. 10909-10923, 2020. |
| [39] | 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. |
| [40] | I. J. Rodriguez et al. Immunosenescence Study of T Cells: A Systematic Review,” Front. Immunol., vol. 11, pp. 1-17, 2021. |
| [41] | I. M. Rea, D. S. Gibson, V. McGilligan, S. E. McNerlan, H. Denis Alexander, and O. A. Ross. Age and age-related diseases: Role of inflammation triggers and cytokines, Front. Immunol., vol. 9, no. APR, pp. 1-28, 2018. |
| [42] | J. Hou, Y. Zheng, and C. Gao. Regulation of cellular senescence by innate immunity, Biophys. Reports, vol. 9, no. 6, pp. 338-351, 2023. |
| [43] | C. E. Moss, H. Phipps, H. L. Wilson, and E. Kiss-Toth. Markers of the ageing macrophage: a systematic review and meta-analysis, Front. Immunol., vol. 14, pp. 1-16, 2023. |
| [44] | P. Pontrelli, F. Rascio, G. Castellano, G. Grandaliano, L. Gesualdo, and G. Stallone. The Role of Natural Killer Cells in the Immune Response in Kidney Transplantation, Front. Immunol., vol. 11, pp. 1-10, 2020. |
| [45] | M. Gergues, R. Bari, S. Koppisetti, A. Gosiewska, L. Kang, and R. J. Hariri. Senescence, NK cells, and cancer: navigating the crossroads of aging and disease, Front. Immunol., vol. 16, pp. 1-21, 2025. |
| [46] | M. Camacho-encina et al. Cellular Senescence, Mitochondrial Dysfunction, and Their Link to Cardiovascular Disease, Cells, Vol. 13(353). pp. 1-21, 2024. |
| [47] | A. Ihan. Senescent Polarization of Macrophages and Inflammatory Biomarkers in Cardiovascular Disease, Cells, vol. 14, no. 17, 2025. |
| [48] | N. D. Funk et al. Mechanisms of natural killer cell-mediated clearance of senescent renal tubular epithelial cells, Front. Cell Dev. Biol., vol. 13, pp. 1-10, 2025. |
| [49] | M. Huang et al. T cell senescence: a new perspective on immunotherapy in lung cancer, Front. Immunol., vol. 15, pp. 1-13, 2024. |
| [50] | Jörg J. Goronzy1 and Cornelia M. Weyand. Mechanisms underlying T cell ageingJörg, Nat Rev Immunol, vol. 19, no. 9, pp. 573-583, 2019, |
| [51] | X. Liu, D. F. Hoft, and G. Peng. Senescent T cells within suppressive tumor microenvironments: Emerging target for tumor immunotherapy, J. Clin. Invest., vol. 130, no. 3, pp. 1073-1083, 2020. |
| [52] | L. X. Wang, X. M. Zhu, and Y. M. Yao. Sestrin2: Its Potential Role and Regulatory Mechanism in Host Immune Response in Diseases, Front. Immunol., vol. 10, pp. 1-15, 2019. |
| [53] | P. Laphanuwat, D. C. O. Gomes, and A. N. Akbar. Senescent T cells: Beneficial and detrimental roles, Immunol. Rev., vol. 316, no. 1, pp. 160-175, 2023. |
| [54] | D. Frasca. Senescent B cells in aging and age-related diseases: Their role in the regulation of antibody responses, Exp. Gerontol., vol. 107, pp. 55-58, 2018. |
| [55] | I. Ramirez De Oleo et al. Phenotypic and functional characteristics of murine CD11c+ B cells which is suppressed by metformin, Front. Immunol., vol. 14, no. September, pp. 1-15, 2023. |
| [56] | B. Jiang et al. Global research trends in inflammaging from 2005 to 2024: a bibliometric analysis, Front. Aging, vol. 6, pp. 1-12, 2025. |
| [57] | J. Birch and J. Gil. Senescence and the SASP: Many therapeutic avenues, Genes Dev., vol. 34, no. 23-24, pp. 1565-1576, 2020. |
| [58] | X. Li, C. Li, W. Zhang, Y. Wang, P. Qian, and H. Huang. Inflammation and aging: signaling pathways and intervention therapies, Signal Transduct. Target. Ther., vol. 8, no. 1, 2023. |
| [59] | P. Goyani, R. Christodoulou, and E. Vassiliou. Immunosenescence: Aging and Immune System Decline, Vaccines, vol. 12, no. 12, pp. 1-13, 2024. |
| [60] | S. Alqahtani et al. SASP Modulation for Cellular Rejuvenation and Tissue Homeostasis: Therapeutic Strategies and Molecular Insights, Cells, vol. 14, no. 8, pp. 1-19, 2025. |
| [61] | L. Mansfield et al. Emerging insights in senescence: pathways from preclinical models to therapeutic innovations, npj Aging, vol. 10, no. 1, 2024. |
| [62] | E. Sarygina, A. Kliuchnikova, S. Tarbeeva, E. Ilgisonis, and E. Ponomarenko. Model Organisms in Aging Research: Evolution of Database Annotation and Ortholog Discovery, Genes (Basel)., vol. 16, no. 1, 2025. |
| [63] | B. M. Wasko and M. Kaeberlein. Yeast replicative aging: a paradigm for defining conserved longevity interventions, FEMS Yeast Res.vol. 14, no. 1, pp. 148-159, 2014, |
| [64] | R. Dahiya et al. Insights into the conserved regulatory mechanisms of human and yeast aging, Biomolecules, vol. 10, no. 6, pp. 1-27, 2020. |
| [65] | G. E. Janssens and L. M. Veenhoff. Evidence for the hallmarks of human aging in replicatively aging yeast, Microb. Cell, vol. 3, no. 7, pp. 263-274, 2016. |
| [66] | A. Leonov et al. Caloric restriction extends yeast chronological lifespan via a mechanism linking cellular aging to cell cycle regulation, maintenance of a quiescent state, entry into a non-quiescent state and survival in the non-quiescent state, Oncotarget, vol. 8, no. 41, pp. 69328-69350, 2017. |
| [67] | A. Zimmermann, S. Hofer, T. Pendl, K. Kainz, F. Madeo, and D. Carmona-Gutierrez. Yeast as a tool to identify anti-aging compounds, FEMS Yeast Res., vol. 18, no. 6, pp. 1-16, 2018. |
| [68] | R. Yu, M. C. Jo, and W. Dang. Measuring the Replicative Lifespan of Saccharomyces cerevisiae Using the HYAA Microfluidic Platform, Methods Mol. Biol., vol. 2144, no. 1, pp. 1-6, 2020. |
| [69] | S. Jeayeng, J. Thongsroy, and S. Chuaijit. Caenorhabditis elegans as a Model to Study Aging and Photoaging, Biomolecules, vol. 14, no. 10, pp. 1-14, 2024. |
| [70] | S. Zhang, F. Li, T. Zhou, G. Wang, and Z. Li. Caenorhabditis elegans as a Useful Model for Studying Aging Mutations, Front. Endocrinol. (Lausanne)., vol. 11, no. October, pp. 1-9, 2020. |
| [71] | W. J. Li et al. Insulin signaling regulates longevity through protein phosphorylation in Caenorhabditis elegans, Nat. Commun., vol. 12, no. 1, pp. 1-16, 2021. |
| [72] | D. Clancy, S. Chtarbanova, and S. Broughton. Editorial: Model organisms in aging research: Drosophila melanogaster, Front. Aging, vol. 3, pp. 1-3, 2022. |
| [73] | L. Jia, X. An, Y. Liu, and X. Meng. Unveiling the anti-aging of radix saposhnikoviae: A metabolomic study in Drosophila, PLoS One, vol. 20, pp. 1-28, 2025. |
| [74] | D. L. Hubert et al. Selection for Early Reproduction Leads to Accelerated Aging and Extensive Metabolic Remodeling in Drosophila melanogaster, Genome Biol. Evol., vol. 17, no. 5, pp. 1-18, 2025. |
| [75] | M. A. Sanborn, X. Wang, S. Gao, Y. Dai, and J. Rehman. Unveiling the cell-type-specific landscape of cellular senescence through single-cell transcriptomics using SenePy, Nat. Commun., vol. 16, no. 1, 2025. |
| [76] | C. E. Hansen et al. Endothelial-Ercc1 DNA repair deficiency provokes blood-brain barrier dysfunction, Cell Death Dis., vol. 16, no. 1, pp. 1-14, 2025. |
| [77] | L. He, D. Han, F. Zong, Y. Zhang, Z. Han, and Z. Xu. Recent progress in stem cell and immune cell-based interventions for aging and age-related disorders, Front. Aging, vol. 6, no. July, pp. 1-12, 2025. |
| [78] | L. Zhu et al. Inflammatory aging clock: A cancer clock to characterize the patients’ subtypes and predict the overall survival in glioblastoma, Front. Genet., vol. 13, pp. 1-14, 2022. |
| [79] | H. Zhang, X. Liu, X. Wang, and Y. Jiang. Association of two novel systemic inflammatory biomarkers and frailty based on NHANES 2007-2018, Front. Public Heal., vol. 12, pp. 1-10, 2024. |
| [80] | D. Ramini et al. Replicative Senescence-Associated LINE1 Methylation and LINE1-Alu Expression Levels in Human Endothelial Cells, Cells, vol. 11, no. 23, 2022. |
| [81] | P. Gao, F. Yao, J. Pang, K. Yin, and X. Zhu. m6 A methylation in cellular senescence of age-associated diseases, Acta Biochim. Biophys. Sin. (Shanghai)., vol. 55, no. 8, pp. 1168-1183, 2023. |
| [82] | J. Sun et al. The Potential Role of m6A RNA Methylation in the Aging Process and Aging-Associated Diseases, Front. Genet., vol. 13, pp. 1-18, 2022. |
| [83] | F. Wu et al. Dynamic Alteration Profile and New Role of RNA m6A Methylation in Replicative and H2O2- Induced Premature Senescence of Human Embryonic Lung Fibroblasts, Int. J. Mol. Sci., vol. 23, no. 16, 2022. |
| [84] | X. Wang et al. METTL3-mediated m6A modification of SIRT1 mRNA inhibits progression of endometriosis by cellular senescence enhancing, J. Transl. Med., vol. 21, no. 1, pp. 1-16, 2023. |
| [85] | G. Huang et al. METTL3-mediated m6A modification regulates D-galactose-induced skin fibroblast senescence through miR-208a-5p, Front. Immunol., vol. 16, no. June, pp. 1-14, 2025. |
| [86] | Y. Wang et al. METTL3-mediated m6A modification increases Hspa1a stability to inhibit osteoblast aging, Cell Death Discov., vol. 10, no. 1, pp. 1-12, 2024. |
| [87] | F. Ding et al. The interplay of cellular senescence and reprogramming shapes the biological landscape of aging and cancer revealing novel therapeutic avenues, Front. Cell Dev. Biol., vol. 13, no. April, pp. 1-13, 2025. |
| [88] | S. K. Ghosh-Choudhary, J. Liu, and T. Finkel. The role of mitochondria in cellular senescence, FASEB J., vol. 35, no. 12, pp. 1-16, 2021. |
| [89] | A. Bevilacqua, P. C. Ho, and F. Franco. Metabolic reprogramming in inflammaging and aging in T cells, Life Metab., vol. 2, no. 5, pp. 1-8, 2023. |
| [90] | F. Roth-Walter et al. Metabolic pathways in immune senescence and inflammaging: Novel therapeutic strategy for chronic inflammatory lung diseases. An EAACI position paper from the Task Force for Immunopharmacology, Allergy Eur. J. Allergy Clin. Immunol., vol. 79, no. 5, pp. 1089-1122, 2024. |
| [91] | P. Wei, X. Zhang, C. Yan, S. Sun, Z. Chen, and F. Lin. Mitochondrial dysfunction and aging: multidimensional mechanisms and therapeutic strategies, Biogerontology, vol. 26, no. 4, pp. 1-16, 2025. |
| [92] | Q. Song et al. Integrated multi-omics approach revealed cellular senescence landscape,” Nucleic Acids Res., vol. 50, no. 19, pp. 10947-10963, 2022. |
| [93] | R. L. Cohn, N. S. Gasek, G. A. Kuchel, and M. Xu. The heterogeneity of cellular senescence: insights at the single-cell level, Trends Cell Biol., vol. 33, no. 1, pp. 9-17, 2023. |
| [94] | Y. Ovadya et al. Impaired immune surveillance accelerates accumulation of senescent cells and aging, Nat. Commun., vol. 9, no. 1, 2018. |
| [95] | A. V. Borodkina et al. Social Life’ of Senescent Cells: What Is SASP and Why Study It, Acta Naturae, vol. 9, no. 4, pp. 4-15, 2017, |
| [96] | T. Shreeya et al. Senescence: A DNA damage response and its role in aging and Neurodegenerative Diseases, Front. Aging, vol. 4, no. March, pp. 1-12, 2023. |
| [97] | B. I. Pereira et al. Senescent cells evade immune clearance via HLA-E-mediated NK and CD8+ T cell inhibition, Nat. Commun., vol. 10, no. 1, 2019. |
| [98] | V. Lelarge, R. Capelle, F. Oger, T. Mathieu, and B. Le Calvé. Senolytics: from pharmacological inhibitors to immunotherapies, a promising future for patients’ treatment, npj Aging, vol. 10, no. 1, 2024. |
| [99] | Y. Deng et al. Targeting senescent cells with NKG2D-CAR T cells, Cell Death Discov., vol. 10, no. 1, 2024. |
| [100] | A. Ajoolabady et al. Hallmarks and mechanisms of cellular senescence in aging and disease, Cell Death Discov., vol. 11, no. 1, 2025. |
| [101] | S. Pregizer, T. Vreven, M. Mathur, and L. N. Robinson. Multi-omic single cell sequencing: Overview and opportunities for kidney disease therapeutic development, Front. Mol. Biosci., vol. 10, pp. 1-13, 2023. |
| [102] | 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. |
| [103] | T. J. Ruetz et al. CRISPR-Cas9 screens reveal regulators of ageing in neural stem cells, Nature, vol. 634, no. 8036, pp. 1150-1159, 2024. |
| [104] | D. Saul et al. A new gene set identifies senescent cells and predicts senescence-associated pathways across tissues, Nat. Commun., vol. 13, no. 1, 2022, |
| [105] | J. Y. Kim, I. Atanassov, F. Dethloff, L. Kroczek, and T. Langer. Time-resolved proteomic analyses of senescence highlight metabolic rewiring of mitochondria, Life Sci. Alliance, vol. 6, no. 9, pp. 1-17, 2023. |
| [106] | P. JA, K. Verweij, and et al. The power of proteomics to monitor senescence-associated secretory phenotypes and beyond: toward clinical applications. Expert Rev Proteomics, vol. 21, no. 9, pp. 1161-1170, 2018, |
| [107] | C. Hu, X. Zhang, T. Teng, Z. G. Ma, and Q. Z. Tang. Cellular Senescence in Cardiovascular Diseases: A Systematic Review, Aging Dis., vol. 13, no. 1, pp. 103-128, 2022. |
| [108] | H. M. Ashraf, B. Fernandez, and S. L. Spencer. The intensities of canonical senescence biomarkers integrate the duration of cell-cycle withdrawal, Nat. Commun., vol. 14, no. 1, pp. 1-13, 2023. |
| [109] | M. Malavolta et al. Simple Detection of Unstained Live Senescent Cells with Imaging Flow Cytometry, Cells, vol. 11, no. 16, 2022. |
| [110] | J. M. Sasi et al. Integrated transcriptomics and miRNAomics provide insights into the complex multi-tiered regulatory networks associated with coleoptile senescence in rice, Front. Plant Sci., vol. 13, pp. 1-22, 2022. |
| [111] | N. Kudlova, J. B. De Sanctis, and M. Hajduch. Cellular Senescence: Molecular Targets, Biomarkers, and Senolytic Drugs, Int. J. Mol. Sci., vol. 23, no. 8, 2022. |
| [112] | E. Goy et al. The out-of-field dose in radiation therapy induces delayed tumorigenesis by senescence evasion, Elife, vol. 11, no. 2022. pp. 1-15, 2022, |
| [113] | T. Feng et al. Cellular senescence in cancer: from mechanism paradoxes to precision therapeutics, Molecular Cancer, vol. 24, no. 1. 2025. |
| [114] | J. Yang, M. Liu, D. Hong, M. Zeng, and X. Zhang. The Paradoxical Role of Cellular Senescence in Cancer, Front. Cell Dev. Biol., vol. 9, 2021. |
| [115] | Z. Yue, L. Nie, P. Zhao, N. Ji, G. Liao, and Q. Wang. Senescence-associated secretory phenotype and its impact on oral immune homeostasis, Front. Immunol., vol. 13, pp. 1-14, 2022. |
| [116] | E. U. Alum et al. Targeting Cellular Senescence for Healthy Aging: Advances in Senolytics and Senomorphics, Drug Design, Development and Therapy, Vol. 19, pp. 8489-8522, 2025. |
APA Style
Molla, A. (2025). Molecular Mechanisms of Cellular Senescence and Immuno-Senescence: Insights from Model Organisms and Emerging Pathways. American Journal of Biomedical and Life Sciences, 13(5), 98-113. https://doi.org/10.11648/j.ajbls.20251305.12
ACS Style
Molla, A. Molecular Mechanisms of Cellular Senescence and Immuno-Senescence: Insights from Model Organisms and Emerging Pathways. Am. J. Biomed. Life Sci. 2025, 13(5), 98-113. doi: 10.11648/j.ajbls.20251305.12
@article{10.11648/j.ajbls.20251305.12,
author = {Alebachew Molla},
title = {Molecular Mechanisms of Cellular Senescence and Immuno-Senescence: Insights from Model Organisms and Emerging Pathways
},
journal = {American Journal of Biomedical and Life Sciences},
volume = {13},
number = {5},
pages = {98-113},
doi = {10.11648/j.ajbls.20251305.12},
url = {https://doi.org/10.11648/j.ajbls.20251305.12},
eprint = {https://article.sciencepublishinggroup.com/pdf/10.11648.j.ajbls.20251305.12},
abstract = {Cellular senescence and immunosenescence encompass critical molecular pathways that govern aging and age-related pathologies. Central to cellular senescence are DNA damage response activation, telomere attrition, chromatin remodeling, metabolic reprogramming, and cytoplasmic DNA sensing via cGAS-STING signaling, which collectively drive cell cycle arrest and the pro-inflammatory senescence-associated secretory phenotype (SASP). Immunosenescence involves progressive deterioration of immune cell function characterized by depleted naive lymphocytes, accumulation of dysfunctional senescent immune cells, and chronic inflammation (inflammaging), creating a feedback loop that exacerbates tissue degeneration and systemic aging. Model organisms such as mice and killifish have been indispensable for unraveling these mechanisms, enabling genetic and functional studies that illuminate senescence dynamics and immune clearance processes. Future research, empowered by multi-omics, single cell sequencing, and artificial intelligence, promises deeper dissection of senescence heterogeneity and tissue-specific pathways, offering biomarkers and therapeutic targets with unprecedented precision. Therapeutic strategies aiming to selectively eliminate or modulate senescent cells through senolytics, senomorphics, and immunomodulatory approaches hold promise to extend health span and ameliorate chronic diseases. However, challenges including senescent cell heterogeneity, context-dependent functions, and biomarker limitations necessitate individualized and careful translation of findings into clinical therapies. Continued interdisciplinary efforts integrating molecular biology, systems medicine, and clinical research will be pivotal in harnessing the full potential of senescence targeting for healthy aging and transformative disease management. This review was conducted to comprehensively compile and discuss the intricate molecular mechanisms underlying cellular senescence and immunosenescence, which are critical processes involved in aging and age-related diseases. The aim of this review article is to comprehensively elucidate the molecular mechanisms underlying cellular senescence and immunosenescence, integrating insights gained from model organism research and emerging signaling pathways.
},
year = {2025}
}
TY - JOUR T1 - Molecular Mechanisms of Cellular Senescence and Immuno-Senescence: Insights from Model Organisms and Emerging Pathways AU - Alebachew Molla Y1 - 2025/10/30 PY - 2025 N1 - https://doi.org/10.11648/j.ajbls.20251305.12 DO - 10.11648/j.ajbls.20251305.12 T2 - American Journal of Biomedical and Life Sciences JF - American Journal of Biomedical and Life Sciences JO - American Journal of Biomedical and Life Sciences SP - 98 EP - 113 PB - Science Publishing Group SN - 2330-880X UR - https://doi.org/10.11648/j.ajbls.20251305.12 AB - Cellular senescence and immunosenescence encompass critical molecular pathways that govern aging and age-related pathologies. Central to cellular senescence are DNA damage response activation, telomere attrition, chromatin remodeling, metabolic reprogramming, and cytoplasmic DNA sensing via cGAS-STING signaling, which collectively drive cell cycle arrest and the pro-inflammatory senescence-associated secretory phenotype (SASP). Immunosenescence involves progressive deterioration of immune cell function characterized by depleted naive lymphocytes, accumulation of dysfunctional senescent immune cells, and chronic inflammation (inflammaging), creating a feedback loop that exacerbates tissue degeneration and systemic aging. Model organisms such as mice and killifish have been indispensable for unraveling these mechanisms, enabling genetic and functional studies that illuminate senescence dynamics and immune clearance processes. Future research, empowered by multi-omics, single cell sequencing, and artificial intelligence, promises deeper dissection of senescence heterogeneity and tissue-specific pathways, offering biomarkers and therapeutic targets with unprecedented precision. Therapeutic strategies aiming to selectively eliminate or modulate senescent cells through senolytics, senomorphics, and immunomodulatory approaches hold promise to extend health span and ameliorate chronic diseases. However, challenges including senescent cell heterogeneity, context-dependent functions, and biomarker limitations necessitate individualized and careful translation of findings into clinical therapies. Continued interdisciplinary efforts integrating molecular biology, systems medicine, and clinical research will be pivotal in harnessing the full potential of senescence targeting for healthy aging and transformative disease management. This review was conducted to comprehensively compile and discuss the intricate molecular mechanisms underlying cellular senescence and immunosenescence, which are critical processes involved in aging and age-related diseases. The aim of this review article is to comprehensively elucidate the molecular mechanisms underlying cellular senescence and immunosenescence, integrating insights gained from model organism research and emerging signaling pathways. VL - 13 IS - 5 ER -