![]()
🧬 The science of aging has evolved dramatically, with researchers now identifying 15 distinct biological hallmarks that govern how our bodies change over time. These interconnected mechanisms form the blueprint of human aging, from DNA damage and shortened telomeres to gut microbiome imbalances and cellular recycling failures. This guide breaks down complex longevity science into actionable insights, revealing how specific lifestyle interventions target these biological pathways. For health-conscious individuals seeking evidence-based approaches to extend their healthspan, understanding these fundamental aging processes offers a potential roadmap to slow biological decline. Discover how cutting-edge research transforms our approach to aging from an inevitable decline into a modifiable biological process that can be optimized through targeted nutrition, physical activity, stress reduction techniques, and emerging scientific interventions.
“He who would eat much must eat little, for by eating less he will live longer, and so be able to eat more.” – Luigi Cornaro (1484–1566)
Historical Perspectives on Aging
Aging and reversing the aging process have intrigued people throughout history. One of the first documented attempts at reversing the aging process came from Venetian nobleman Luigi Cornaro (1467-1566), who published "The Art of Living Long," describing a lifestyle for achieving longevity. In his youth, Cornaro led a self-indulgent life, resulting in numerous health problems by age 35. By changing his lifestyle, he lived to nearly 100 years old.
Mohammad bin Yousuf al-Harawi's first formal study of aging was published by the Ibn Sina Academy of Medieval Medicine Sciences 1582 in his book Ainul Hayat ("The Source of Life"). This book covered behavioral and lifestyle factors influencing aging, including diet, environment, housing conditions, and drugs that may increase or decrease aging rates.
Recent History of Aging Research
The scientific study of aging has accelerated dramatically over the past century:
- 1908: Nobel Prize winner Elie Metchnikoff (1845-1916) discovered phagocytosis, a critical part of the immune system, and coined the term "gerontology"
- 1915-1917: Thomas Osborne conducted the first systematic experiments to determine the effects of food restriction on life duration in rats
- 1930s: Rudolph Schoenheimer discovered isotope labeling of biomolecules, revealing that all constituents of an organism are in a constant state of chemical renewal
- 1956: Denham Harman presented the Free Radical Theory of Aging, proposing that organisms age because they accumulate oxidative damage
- 1962: Nobel Prize for the discovery of DNA's molecular structure by Watson, Crick, and Wilkins
- 1974: Nobel Prize to Christian de Duve for discovering lysosomes, later found critical to autophagy
- 2001: Nobel Prize for discoveries of key regulators of the cell cycle
- 2009: Nobel Prize for the discovery of how chromosomes are protected by telomeres and the enzyme telomerase
- 2012: Nobel Prize for discovering that mature cells can be reprogrammed to become pluripotent
- 2015: Nobel Prize for mapping how cells repair damaged DNA
- 2016: Nobel Prize to Yoshinori Ohsumi for his discoveries of autophagy mechanisms
- 2017: Nobel Prize for discoveries of molecular mechanisms controlling circadian rhythm
- 2019: David Sinclair popularized the science of aging with his book Lifespan: Why We Age and Why We Don't Have To
The Original Nine Hallmarks of Aging
In 2013, Carlos López-Otín and colleagues published a groundbreaking paper identifying and categorizing the cellular and molecular hallmarks of aging. They proposed nine candidate hallmarks contributing to the aging process and collectively determining the aging phenotype.(1)
A hallmark should ideally fulfill three criteria:
- It should manifest during normal aging
- Its experimental aggravation should accelerate aging
- Its experimental amelioration should retard the normal aging process and increase a healthy lifespan
1. Genomic Instability
External physical, chemical, and biological agents continuously challenge DNA's integrity and stability, and internal threats, including DNA replication errors, spontaneous hydrolytic reactions, and the production of reactive oxygen species, eventually lead to the accumulation of genetic damage throughout life.(2-3)
2. Telomere Attrition
Telomeres are particularly susceptible to age-related deterioration. Telomere shortening is observed during normal aging in both humans and mice. Telomeres are bound by a multiprotein complex called shelterin, which prevents access of DNA repair proteins to telomeres. Without it, telomeres would be repaired as DNA breaks, leading to chromosome fusions. DNA damage at telomeres is remarkably constant and highly efficient at inducing senescence.(4-7)
3. Epigenetic Alterations
Many epigenetic alterations affect all cells and tissues throughout life, caused by diet, chemicals, drugs, sunlight, heat/cold, exercise, etc. Epigenetic changes involve alterations in DNA methylation patterns, posttranslational modification of histones, and chromatin remodeling. Members of the sirtuin family (NAD-dependent protein deacetylases and ADP-ribosyltransferases) have been studied extensively as potential anti-aging factors. In humans, at least three members of the sirtuin family (SIRT1, SIRT3, and SIRT6) contribute to healthy aging.(8-10)
4. Loss of Proteostasis
Proteostasis comprises mechanisms for stabilizing correctly folded proteins (especially the heat-shock family of proteins) and mechanisms for degrading proteins by the proteasome and lysosome. Multiple studies have demonstrated that aging alters proteostasis, leading to chronic expression of unfolded, misfolded, and aggregated proteins. These contribute to the development of some age-related degenerative diseases such as Alzheimer's disease.(11-13)

Image: Loss of Proteostasis. Failure to refold or degrade unfolded proteins can lead to their accumulation and aggregation, resulting in proteotoxic effects.
Source: López-Otín, C. & Blasco, M. & Partridge, L. & Serrano, M. & Kroemer, G. (2013). The hallmarks of aging. Cell 153 (6): 1194–1217.
5. Deregulated Nutrient Sensing
IGF-1 and insulin signaling, known as the IIS pathway, is the most conserved aging-controlling pathway in evolution. In addition to the IIS pathway that takes part in glucose sensing, there are three additional related and interconnected nutrient-sensing systems: mTOR (sensing high amino acid concentrations), AMPK (sensing low-energy states by detecting high AMP levels), and sirtuins (sensing low-energy states by detecting high NAD+ levels). Firm evidence exists that anabolic signaling (mTOR, high insulin) accelerates aging, while decreased nutrient signaling (AMPK, low insulin) extends longevity.(14-16)
6. Mitochondrial Dysfunction
Mitochondrial dysfunction is found to increase in the aging process. When an organism ages, the cellular respiration chain's efficacy diminishes, leading to electron leakage and reduced ATP generation. The reduced efficiency of mitochondrial bioenergetics with aging may result from multiple intersecting mechanisms, including reduced biogenesis of mitochondria, accumulation of mutations and deletions in mtDNA, oxidative stress to mitochondrial proteins, destabilization of the respiratory chain, changes in the lipid composition of mitochondrial membranes, and alterations in mitochondrial dynamics.(17-19)
7. Cellular Senescence
Because the number of senescent cells increases with age, it is presupposed that senescence contributes to aging. However, senescence is needed to prevent the distribution and proliferation of damaged cells by triggering an immune system response. This cellular checkpoint requires an efficient cell substitution system that involves both the clearance of senescent cells and the mobilization of progenitor cells to restore optimal cell numbers.
Senescent cells express substantial alterations in their secretome, which is particularly enriched in pro-inflammatory cytokines and matrix metalloproteinases. This is hence referred to as the senescence-associated secretory phenotype (SASP).(20)
8. Stem Cell Exhaustion
Adult stem cells can self-renew and differentiate into multiple cell types within a tissue. Although phenotypes and mechanisms vary widely, all stem cell populations' function declines with age. Stem cell depletion is a unifying consequence of various aging-associated damages and likely constitutes one of the ultimate culprits of cellular aging.(21)
Studies in aged mice have revealed an overall decrease in cell-cycle activity of hematopoietic stem cells (HSCs), which correlates with the accumulation of DNA damage and overexpression of cell-cycle inhibitory proteins (e.g., p16INK4a). Telomere shortening is also a significant cause of stem cell decline during aging.(22-24)
9. Altered Intercellular Communication
Cellular aging also occurs at the intercellular communication level. It includes neurohormonal signaling, increased inflammatory reactions, immunosurveillance of pathogens and premalignant cells, and changes in the extracellular environment.
Aging due to inflammation is called inflammaging. It may result from multiple causes, such as the accumulation of pro-inflammatory tissue damage, failure of a dysfunctional immune system to clear pathogens and dysfunctional host cells effectively, or a deficient autophagic response.(25)
Aging changes in one tissue can lead to aging-specific deterioration in neighboring tissue. Senescent cells can induce senescence in their adjacent cells via gap-junction-mediated cell-cell contacts and processes involving reactive oxygen species. This phenomenon is also called the senescent cell bystander effect.(26)
Extracellular Matrix Stiffening: A Potential New Hallmark?
In 2021, researchers Alexander Fedintsev and Alexey Moskalev published a paper examining extracellular matrix (ECM) stiffening and the buildup of crosslinks between long-lived molecules such as collagen and elastin. They suggest that non-enzymatic chemical reactions like glycation, carbamylation, and carbonylation cause ECM stiffening. This could even be an upstream cause of several accepted hallmarks of aging, such as cellular senescence.
These changes lead to the formation of adducts and crosslinks that, in turn, cause inflammation, fibrosis, tissue circadian clock impairment, stem cell aging, etc. It has been previously established that advanced glycation end products (AGEs) have pathogenic significance in various tissues and pathways in the body. Organisms with extraordinarily long lifespans, such as bowhead whales, have exceptionally low rates of AGE accumulation.(27)
Expanding from 9 to 12 Hallmarks of Aging
In their 2023 study "Hallmarks of aging: expanding universe," published in Cell, López-Otín and colleagues added three new hallmarks: dysbiosis, chronic inflammation and disabled macroautophagy.(28)

Image: The hallmarks of aging.
Source: López-Otín, C. & Blasco, M. & Partridge, L. & Serrano, M. & Kroemer, G. (2023). Hallmarks of aging: An expanding universe. Cell 186 (2): P243–278.
10. Dysbiosis
Dysbiosis is an imbalance in the gut microbial community, leading to chronic inflammation and other adverse health outcomes. As we age, the individuality of our gut microbiomes increases, a phenomenon linked to well-known microbial metabolites involved in immune regulation, inflammation, and aging.
In later years, healthy individuals tend to maintain a trajectory toward a distinct microbial composition, whereas those in poorer health exhibit reduced or absent drift. This emphasizes the critical role of gut microbiota in the aging process and suggests that modulating the microbiota may hold promise for maintaining health and preventing age-related diseases.(29)
11. Chronic Inflammation
Chronic or silent inflammation is another newly added hallmark of aging. Chronic inflammation occurs when the immune system is constantly activated, leading to tissue damage and dysfunction. As we age, levels of inflammatory cytokines and blood biomarkers such as CRP tend to rise. This indicates the presence of low-grade inflammation, which is a hallmark of the aging process. Notably, elevated levels of IL-6 in the bloodstream serve as a predictive biomarker for all-cause mortality in aging human populations.(30)
This underscores the importance of monitoring inflammation as a critical factor in aging and its associated diseases. Chronic inflammation contributes to many age-related diseases, including Alzheimer's, cancer and cardiovascular disease.
12. Disabled Macroautophagy
Disabled macroautophagy (or simply autophagy) is the third newly added hallmark of aging. Macroautophagy is a cellular process that clears damaged or dysfunctional organelles and proteins. When macroautophagy is disabled, these damaged components accumulate in cells, leading to cellular dysfunction and disease.
Autophagy contributes to proteostasis and impacts non-proteinaceous macromolecules (e.g., cytosolic DNA, lipids, glycogen) as well as organelles such as mitochondria (targeted by mitophagy), lysosomes (lysophagy), endoplasmic reticulum (reticulophagy), peroxisomes (pexophagy), and invading pathogens (xenophagy). One of the most significant reasons for decreased organelle turnover is the decline in autophagy with age.(31)
Integration of the Hallmarks
In their recent study, López-Otín et al. posit that these twelve fundamental markers of aging are functionally interrelated. These markers indicate key determinants that steer the aging process and are interconnected with eight principal well-being attributes. The latter pertains to a range of organizational features like spatial compartmentalization, maintenance of homeostasis over an extended period, and responses to perturbation.
Furthermore, aging markers are associated with eight suggested strata of organismal organization. The factors mentioned above collectively create a complex multidimensional space of interactions that elucidate some cardinal features of the aging process. By understanding these interconnections and interactions, researchers may uncover fresh insights into the mechanism of aging, thereby leading to the development of more effective tactics for advancing healthy aging and augmenting the quality of life for older individuals.

Image: Integration of hallmarks.
Source: López-Otín, C. & Blasco, M. & Partridge, L. & Serrano, M. & Kroemer, G. (2023). Hallmarks of aging: An expanding universe. Cell 186 (2): P243–278.
Adding More into the Mix: 3 Newest Hallmarks of Aging
As the science of aging and longevity progresses, it is sometimes hard to keep up with all recent research, especially in this developing field of study. That being said, there are three new suggestions to add to the 12 hallmarks of aging presented before:
13. Altered Mechanical Properties
Mechanobiology explores the concept of aging changing the mechanical properties of tissues. It investigates how mechanical forces and changes in the mechanical properties of cells and tissues contribute to disease and aging. Mechanotherapeutics, including self-therapies such as stretching and mobility exercises, may help repair tissue.(32-33)
14. Cellular Enlargement
Cell enlargement (cellular hypertrophy) is linked to aging and cellular senescence. Research has shown that as cells age, they tend to increase in size, and this change can affect cellular function. However, cells function their best when they are not enlarged. Adopting a nutritious diet appears highly beneficial to combat cellular enlargement effectively.(34-35)
15. Splicing Dysregulation
The dysregulation of RNA splicing, a hallmark of aging, relates to transcribing genetic code into messenger RNA (mRNA) before being translated into proteins. With aging, the precision of this splicing process can deteriorate, leading to various age-related diseases such as cancer and type 2 diabetes. Research has shown that the Mediterranean diet positively influences splicing dysregulation-related problems.(36-38)
Conclusion
These 15 hallmarks provide a framework for understanding the complex biological processes of aging. They are crucial for developing natural and medical interventions to slow down and potentially reverse aspects of the aging process.
🔬 Understanding the interconnected nature of these hallmarks is essential for developing comprehensive approaches to healthy aging. As research continues to evolve, our understanding of aging mechanisms will deepen, potentially leading to breakthroughs in extending not just lifespan but healthspan—the period of life spent in good health.
Scientific References:
- López-Otín, C. & Blasco, M. & Partridge, L. & Serrano, M. & Kroemer, G. (2013). The hallmarks of aging. Cell 153 (6): 1194–1217.
- Moskalev, A. et al. (2013). The role of DNA damage and repair in aging through the prism of Koch-like criteria. Ageing Research Reviews 12 (2): 661–684.
- Hoeijmakers, J.(2009). DNA damage, aging, and cancer. New England Journal of Medicine 361 (15): 1475–1485.
- Blackburn, E. &, Greider, C. & Szostak, J. (2006). Telomeres and telomerase: The path from maize, Tetrahymena and yeast to human cancer and aging. Nature Medicine 12 (10): 1133–1138.
- Blasco, M. (2007). Telomere length, stem cells and aging. Nature Chemical Biology 3 (10): 640–649.
- Palm, W, & de Lange, T. (2008). How shelterin protects mammalian telomeres. Annual Review of Genetics 42: 301–334.
- Fumagalli, M. et al. (2012). Telomeric DNA damage is irreparable and causes persistent DNA-damage-response activation. Nature Cell Biology 14 (4): 355–365.
- Talens, R. et al. (2012). Epigenetic variation during the adult lifespan: Cross‐sectional and longitudinal data on monozygotic twin pairs. Aging Cell 11 (4): 694–703.
- Fraga, M. & Esteller, M. (2007). Epigenetics and aging: The targets and the marks. Trends in Genetics 23 (8): 413–418.
- Guarente, L. (2011). Sirtuins, aging, and metabolism. Cold Spring Harbor Symposia On Quantitative Biology 76: 81–90. New York: Cold Spring Harbor Laboratory Press.
- Hartl, F. & Bracher, A. & Hayer-Hartl, M. (2011). Molecular chaperones in protein folding and proteostasis. Nature 475 (7356): 324–332.
- Koga, H. & Kaushik, S. & Cuervo, A. (2011). Protein homeostasis and aging: The importance of exquisite quality control. Ageing Research Reviews 10 (2): 205–215.
- Powers, E. & Morimoto, R. & Dillin, A. & Kelly, J. & Balch, W. (2009). Biological and chemical approaches to diseases of proteostasis deficiency. Annual Review of Biochemistry 78: 959–991.
- Barzilai, N. & Huffman, D. & Muzumdar, R. & Bartke, A. (2012). The critical role of metabolic pathways in aging. Diabetes 61 (6): 1315–1322.
- Houtkooper, R. & Williams, R. & Auwerx, J. (2010). Metabolic networks of longevity. Cell 142 (1): 9–14.
- Papadopoli, D. et al. (2019). mTOR as a central regulator of lifespan and aging. F1000Research 8: F1000 Faculty Rev-998.
-
Green, D. & Galluzzi, L. & Kroemer, G. (2011). Mitochondria and the autophagy–inflammation–cell death axis in organismal aging. Science 333 (6046): 1109–1112.
- Sahin, E. & DePinho, R. (2012). Axis of ageing: telomeres, p53 and mitochondria. Nature reviews Molecular Cell Biology 13 (6): 397–404.
- Wang, K. & Klionsky, D. (2011). Mitochondria removal by autophagy. Autophagy 7 (3): 297–300.
- Kuilman, T. & Michaloglou, C. & Mooi, W. & Peeper, D. (2010). The essence of senescence. Genes & Development 24 (22): 2463–2479.
- Schultz, M. & Sinclair, D. (2016). When stem cells grow old: Phenotypes and mechanisms of stem cell aging. Development 143 (1): 3–14.
- Rossi, D. et al. (2007). Deficiencies in DNA damage repair limit the function of haematopoietic stem cells with age. Nature 447 (7145): 725–729.
- Janzen, V. et al. (2006). Stem-cell ageing modified by the cyclin-dependent kinase inhibitor p16INK4a. Nature 443 (7110): 421–426.
- Rossi, D. et al (2007). Deficiencies in DNA damage repair limit the function of haematopoietic stem cells with age. Nature 447 (7145): 725–729.
- Salminen, A. & Kaarniranta, K. & Kauppinen, A. (2012). Inflammaging: Disturbed interplay between autophagy and inflammasomes. Aging 4 (3): 166.
- Nelson, G. et al. (2012). A senescent cell bystander effect: Senescence-induced senescence. Aging Cell 11 (2): 345–349.
- Fedintsev, A. & Moskalev, A. (2020). Stochastic non-enzymatic modification of long-lived macromolecules—a missing hallmark of aging. Ageing Research Reviews 62: 101097.
- López-Otín, C. & Blasco, M. & Partridge, L. & Serrano, M. & Kroemer, G. (2023). Hallmarks of aging: An expanding universe. Cell 186 (2): P243–278.
- Wilmanski, T. et al. (2021). Gut microbiome pattern reflects healthy ageing and predicts survival in humans. Nature Metabolism 3 (2): 274–286.
- Hirata, T. et al. (2020). Associations of cardiovascular biomarkers and plasma albumin with exceptional survival to the highest ages. Nature communications 11 (1): 3820.
- Levine, B. & Kroemer, G. (2019). Biological functions of autophagy genes: A disease perspective. Cell 176 (1-2): 11–42.
- Phillip, J. & Aifuwa, I. & Walston, J. & Wirtz, D. (2015). The mechanobiology of aging. Annual Review of Biomedical Engineering 17: 113–141.
- Tschumperlin, D. & Lagares, D. (2020). Mechano-therapeutics: Targeting mechanical signaling in fibrosis and tumor stroma. Pharmacology & Therapeutics 212: 107575.
- Lengefeld, J. et al. (2021). Cell size is a determinant of stem cell potential during aging. Science Advances 7 (46): eabk0271.
- Davies, D. & van den Handel, K. & Bharadwaj, S. & Lengefeld, J. (2022). Cellular enlargement—A new hallmark of aging? Frontiers in Cell and Developmental Biology 10: 1036602.
- Angarola, B. & Anczuków, O. (2021). Splicing alterations in healthy aging and disease. Wiley Interdisciplinary Reviews: RNA 12 (4): e1643.
- Stegeman, R., & Weake, V. (2017). Transcriptional signatures of aging. Journal of Molecular Biology 429 (16): 2427–2437.
- del Río-Moreno, M. et al. (2020). Dietary intervention modulates the expression of splicing machinery in cardiovascular patients at high risk of type 2 diabetes development: Ffrom the CORDIOPREV study. Nutrients 12 (11): 3528.