×

Why We Age: Cellular Senescence

Cellular Senescence is one of the proposed reasons we age.
Why We Age: Cellular Senescence
Date Published: 01/31/2025
Date Modified: 01/31/2025
Cellular Senescence is one of the proposed reasons we age.

Cells, the building blocks of life, meet a variety of fates. Some succumb to necrosis, a chaotic and uncontrolled structural collapse that spills cellular contents and is usually caused by traumatic injury. Others follow the path of apoptosis, a more orderly, self-imposed demise. Imagine a cell sensing its internal flaws and choosing a noble end: “Propagating my DNA could harm my neighbors. Let the immune system clean up this mess.” This programmed death safeguards the body from more significant harm [1].

Then, there’s the peculiar fate of senescence, in which cells neither divide nor perish. Instead, they remain metabolically active but adopt a repurposed role, banned from further replication because their DNA, if propagated, poses a significant cancer risk. Senescence is a fail-safe, ensuring that cells teetering on the edge of malfunction don’t propagate. It is like an eternal timeout, a cellular purgatory to protect the greater organism [2].

This response can be triggered in two primary ways: acute injury and the Hayflick limit. When a cell sustains acute injury from stressors like radiation or toxins, it may enter senescence rather than risk replicating damaged DNA [3]. Alternatively, cells are programmed to divide a finite number of times before halting due to telomere shortening: the gradual erosion of protective caps and subsequent loss of DNA at the ends of chromosomes [4].

While this strategy has its benefits, senescence is a double-edged sword. These cells can act as vigilant guardians, halting the spread of damage or lingering and gradually unleashing chaos through chronic inflammation [2].

Secreted chemicals

Senescent cells don’t go quietly. They secrete a potent mix of signaling molecules, enzymes, and inflammatory compounds called the senescence-associated secretory phenotype (SASP). This cocktail, comprised of cytokines, chemokines, and proteases, acts as a distress signal, summoning immune cells to clear damaged cells and remodel tissues. This response is beneficial in acute situations, as well as in promoting healing and preventing cancer [5].

However, problems arise when the immune system does not clear these cells. Persistent senescent cells pump out the SASP indefinitely, creating a toxic environment that damages neighboring cells, dampens tissue regeneration, and fuels chronic inflammation. This state, often referred to as inflammaging or sterile inflammation, is linked to various age-related conditions, including cardiovascular disease, diabetes, and neurodegenerative disorders. While senescence is nature’s way of protecting against immediate threats, its long-term presence poses significant challenges to the body’s health and resilience [5].

ADVERTISEMENT

Novos-labs Ads 3

Discovery and early understanding

The concept of cellular senescence emerged in 1961 when Leonard Hayflick and Paul Moorhead made a groundbreaking observation: normal human cells can divide only a limited number of times before ceasing replication [4]. This phenomenon, later named the Hayflick limit, challenged the belief that cells could divide indefinitely. It marked a pivotal moment in our understanding of cellular aging and the intrinsic mechanisms that control it.

The driving force behind replicative senescence lies in the shortening of telomeres, the protective caps at the ends of chromosomes. Each time a cell divides, these telomeres erode slightly, acting as a biological clock that counts down to the cell’s final division. When telomeres reach a critically short length, the cell interprets this as DNA damage and halts further replication. This mechanism prevents the propagation of potentially cancerous mutations [6].

Other triggers can also push cells into senescence. DNA damage, whether from radiation, oxidative stress, or replication errors, activates the cell’s DNA damage response pathways. Similarly, activating genes that drive unregulated cell growth (oncogenes) can signal cells to enter senescence as a protective measure against cancer [7, 8].

Additional factors, such as mitochondrial dysfunction, contribute to the induction of senescence. Mitochondria, the cell’s powerhouses, produce reactive oxygen species (ROS) as a byproduct of energy generation. Excessive ROS can damage cellular components, including DNA, accelerating the march toward senescence [9]. Meanwhile, chronic inflammation overwhelms repair systems, creating a feedback loop perpetuating cellular damage and senescence [10].

While Hayflick’s discovery provided the foundational framework, research over the decades has illuminated the complexity of senescence. Far from being a simple endpoint, senescence represents a dynamic state with profound implications for aging and disease. Its dual nature, protective in youth and destructive in age (antagonistic pleiotropy), captures the delicate balance cells must maintain to safeguard the organism [11].

Dual nature

Senescent cells embody a paradox: they are both protectors and potential saboteurs of the body. On one hand, these cells act as a frontline defense against cancer. They prevent compromised cells from becoming malignant by halting cell division in response to DNA damage or other stressors. This protective mechanism is vital during wound healing, in which senescent cells release signaling molecules to recruit immune cells and promote tissue repair. For instance, following an injury, the inflammatory response that they provoke helps clear out damaged tissue and paves the way for regeneration [12].

However, with the passage of time, this well-intentioned safeguard begins to overstay its welcome. When senescent cells fail to be cleared by the immune system, they persist, accumulating in tissues and secreting the same inflammatory SASP molecules that were once beneficial. Over time, this pro-inflammatory environment transforms from a healing balm to a corrosive toxin, impairing tissue function and increasing the risk of chronic conditions. Persistent senescence contributes to fibrosis, diabetes, neurodegenerative disorders, and age-related frailty [13-16]. The SASP, now working against the body, encourages neighboring cells to adopt the same senescent state, exacerbating inflammation and tissue degeneration [17].

This duality of senescent cells, a critical defense in youth and a source of harm in old age, lies at the heart of the “friend or foe” dilemma. This tension has fueled research into strategies for selectively eliminating senescent cells or modifying their behavior, aiming to strike a balance between their protective and destructive roles.

Mechanisms

At their core, molecular pathways that function as cellular circuit breakers enforce senescence, halting growth in response to stress or damage. One of the key mechanisms involves the DNA damage response (DDR), a cellular alarm system that detects DNA damage and activates tumor suppressor proteins like p53 and p16INK4a [18, 19]. These proteins work in tandem to trigger a permanent cell cycle arrest, ensuring that damaged cells cannot divide and propagate errors.

When DNA damage is detected, p53 becomes activated, inducing the expression of p21, which blocks cell cycle progression [20, 21]. This acts as an immediate stop sign for damaged cells. p16INK4a reinforces this arrest by inhibiting cyclin-dependent kinases (CDKs), enzymes critical for cell division. Together, p53 and p16INK4a create a robust barrier to uncontrolled growth [18].

However, senescence is not just a matter of halting replication, it also involves active changes in the cell’s behavior. The SASP is driven by signaling pathways like NF-κB [22], JAK-STAT [23], and inflammasomes [22].

The NF-κB pathway acts as a master switch for inflammatory cytokines, such as IL-6 and IL-8, perpetuating senescence by affecting neighboring cells. The JAK-STAT pathway amplifies the production of SASP factors [23], sustaining a chronic inflammatory environment. Inflammasomes, protein complexes that detect cellular stress and activate inflammatory responses, further reinforce the senescent state.

On a broader scale, senescence is maintained by metabolic and epigenetic changes. Dysfunctional mitochondria produce excessive reactive oxygen species (ROS), leading to oxidative stress and further DNA damage [9]. Meanwhile, epigenetic alterations, such as changes in chromatin structure, lock the cell into a state of inactivity. Regions of chromatin become compacted into senescence-associated heterochromatin foci (SAHF), silencing genes related to cell division while promoting the expression of SASP-related genes [24]. These molecular mechanisms create a feedback loop where senescent cells maintain their state and influence their environment, promoting inflammation and senescence in surrounding tissues.

As we age, the immune system becomes sluggish and inefficient, leaving senescent cells free to loiter. This age-related decline, known as immunosenescence, dulls the sharp edges of T cells and natural killer (NK) cells, both of which critical for identifying and eliminating rogue cells. Additionally, senescent cells don’t just sit idly, waiting to be removed. They deploy clever survival tactics, downregulating MHC class I proteins (the cellular equivalent of waving a white flag) and suppressing stress signals that usually alert the immune system. Some even express immune checkpoint molecules like PD-L1, effectively telling patrolling immune cells, “Nothing to see here, move along.”

If that weren’t enough, senescent cells weaponize their SASP—a pro-inflammatory secretome that creates a hostile microenvironment, suppressing the immune system further. These evasive maneuvers might seem counterproductive, but they serve a purpose: in the short term, they help senescent cells support tissue repair and suppress tumors. The trouble comes when these cells overstay their welcome. Their prolonged accumulation disrupts tissue function, turning once-vital processes into chronic inflammation and fueling the fires of age-related diseases. It’s a cunning escape act that comes at a steep cost to the body over time [25].

Therapeutic strategies

Scientists are developing strategies to mitigate the harmful effects of cellular senescence while preserving its benefits. At the forefront are senolytics, a class of drugs designed to selectively eliminate senescent cells.

For example, dasatinib and quercetin work by exploiting vulnerabilities unique to senescent cells. These drugs disrupt senescent cells’ survival pathways, effectively clearing them from tissues [26]. Early clinical trials have shown promise in treating conditions like pulmonary fibrosis [27], osteoarthritis, cardiovascular disease [28, 29], and neurodegenerative diseases, improving inflammation and physical function.

An alternative to senolytics are senomorphics, compounds that don’t destroy senescent cells but instead dampen the SASP to mitigate deleterious effects on neighboring tissue. Senomorphics may potentially be better than senolytics because their purpose is to suppress the harmful effects of senescent cells without actually killing the cells, potentially leading to a lower risk of off-target effects and a wider range of potential therapeutic applications compared to senolytics, which could cause unintended cell death in healthy tissues. However, senomorphics may require continuous administration to maintain their effects, and their mechanisms may be more complex to understand and target effectively [28].

Beyond senolytics and senomorphics, emerging therapies like CAR T-cell treatments are pushing the boundaries of precision medicine. CAR T-cells are engineered to recognize specific markers on senescent cells, enabling targeted elimination [30]. These therapies hold the potential for long-lasting effects by leveraging the immune system’s ability to adapt and respond dynamically.

Despite these advances, challenges remain. One major hurdle is ensuring that therapies do not indiscriminately remove all senescent cells, as some play crucial roles in wound healing and cancer suppression. Additionally, researchers are working to develop biomarkers that can accurately detect and monitor senescent cells [31], allowing for precise assessments of treatment effectiveness.

Clinical trials continue to illuminate the potential of senescence-targeting therapies. Early successes suggest that these treatments could transform how we approach aging and age-related diseases, but the journey is far from over. Balancing efficacy, safety, and accessibility are critical.

In addition to senolytics, senomorphics, and CAR-T cell therapies, another promising avenue of treatment is immune checkpoint blockade (ICB). This may be a promising strategy to enhance immune surveillance of senescence, leading to the amelioration of some age-related diseases and tissue dysfunction [25].

Challenges

Senescent cells walk a fine line between hero and villain, and so must the therapies designed to address them. The challenge is clear: how do we clear out the harmful cells without dismantling the good they accomplish? Removing too many senescent cells could impair wound healing and even weaken defenses against cancer while leaving them unchecked contributes to chronic inflammation and disease.

Senescent cells are not a monolith; their behavior varies depending on the tissue and context, meaning a one-size-fits-all solution is unlikely [32]. Researchers must also address safety concerns, optimize dosages, and minimize side effects. Despite these hurdles, current progress in this area inspires optimism, showing that the dream of treating aging at its root is no longer far-fetched.

Literature

[1] Park, W., Wei, S., Kim, B. S., Kim, B., Bae, S. J., Chae, Y. C., Ryu, D., & Ha, K. T. (2023). Diversity and complexity of cell death: a historical review. Experimental & Molecular Medicine, 55(8), 1573–1594.

[2] Wang, B., Han, J., Elisseeff, J. H., & Demaria, M. (2024). The senescence-associated secretory phenotype and its physiological and pathological implications. Nature Reviews Molecular Cell Biology.

[3] Nelson, D. A., Kazanjian, I., Melendez, J. A., & Larsen, M. (2024). Senescence and fibrosis in salivary gland aging and disease. Journal of Oral Biology and Craniofacial Research, 14(3), 231–237.

[4] Hayflick, L. (1965). The limited in vitro lifetime of human diploid cell strains. Experimental Cell Research, 37(3), 614–636.

[5] Coppé, J. P., Desprez, P. Y., Krtolica, A., & Campisi, J. (2010). The senescence-associated secretory phenotype: The dark side of tumor suppression. Annual Review of Pathology: Mechanisms of Disease, 5(Volume 5, 2010), 99–118.

[6] Chakravarti, D., LaBella, K. A., & DePinho, R. A. (2021). Telomeres: history, health, and hallmarks of aging. Cell, 184(2), 306–322.

[7] Courtois-Cox, S., Jones, S. L., & Cichowski, K. (2008). Many roads lead to oncogene-induced senescence. Oncogene 2008 27:20, 27(20), 2801–2809.

[8] Wei, W., & Ji, S. (2018). Cellular senescence: Molecular mechanisms and pathogenicity. Journal of Cellular Physiology, 233(12), 9121–9135.

[9] Miwa, S., Kashyap, S., Chini, E., & von Zglinicki, T. (2022). Mitochondrial dysfunction in cell senescence and aging. The Journal of Clinical Investigation, 132(13).

[10] Kay, J., Thadhani, E., Samson, L., & Engelward, B. (2019). Inflammation-induced DNA damage, mutations and cancer. DNA Repair, 83, 102673.

[11] Austad, S. N., & Hoffman, J. M. (2018). Is antagonistic pleiotropy ubiquitous in aging biology? Evolution, 287–294.

[12] Wilkinson, H. N., & Hardman, M. J. (2020). Senescence in Wound Repair: Emerging Strategies to Target Chronic Healing Wounds. Frontiers in Cell and Developmental Biology, 8(August), 1–13.

[13] Fyhrquist, F., Saijonmaa, O., & Strandberg, T. (2013). The roles of senescence and telomere shortening in cardiovascular disease. Nature Reviews Cardiology 2013 10:5, 10(5), 274–283.

[14] Marcos-Pérez, D., Cruces-Salguero, S., García-Domínguez, E., Araúzo-Bravo, M. J., Gómez-Cabrera, M. C., Viña, J., Vergara, I., & Matheu, A. (2024). Physical Interventions Restore Physical Frailty and the Expression of CXCL-10 and IL-1β Inflammatory Biomarkers in Old Individuals and Mice. Biomolecules, 14(2), 166.

[15] Palmer, J. E., Wilson, N., Son, S. M., Obrocki, P., Wrobel, L., Rob, M., Takla, M., Korolchuk, V. I., & Rubinsztein, D. C. (2025). Autophagy, aging, and age-related neurodegeneration. Neuron, 113(1), 29–48.

[16] Tang, H., Zhou, Y., Ye, Y., Ma, L., Xiao, Q.-X., Tang, J.-Q., & Xu, Y. (2025). SIRT3 alleviates mitochondrial dysfunction and senescence in diabetes-associated periodontitis by deacetylating LRPPRC. Free Radical Biology and Medicine, 227, 407–419.

[17] Paulo, F. L., Mikolaj, S., Olena, O., Julien, K., Miwa, S., Cameron, K., Ishaq, A., Saretzki, G., Nagaraja, S., Glyn, G., & Thomas, N. (2019). The bystander effect contributes to the accumulation of senescent cells in vivo. February 2018.

[18] Tufail, M., Huang, Y.-Q., Hu, J.-J., Liang, J., He, C.-Y., Wan, W.-D., Jiang, C.-H., Wu, H., & Li, N. (2024). Cellular Aging and Senescence in Cancer: A Holistic Review of Cellular Fate Determinants. Aging and Disease

[19] Zglinicki, T. Von, Saretzki, G., Ladhoff, J., Adda, F., & Jackson, S. P. (2005). Human cell senescence as a DNA damage response. 126, 111–117.

[20] Mijit, M., Caracciolo, V., Melillo, A., Amicarelli, F., & Giordano, A. (2020). Role of p53 in the regulation of cellular senescence. Biomolecules, 10(3), 1–16.

[21] Shtutman, M., Chang, B. D., Schools, G. P., & Broude, E. V. (2017). Cellular model of p21-induced senescence. Methods in Molecular Biology, 1534, 31–39.

[22] Nelson, G., Kucheryavenko, O., Wordsworth, J., & von Zglinicki, T. (2018). The senescent bystander effect is caused by ROS-activated NF-κB signalling. Mechanisms of Ageing and Development, 170(August 2017), 30–36.

[23] Zhou, J., Wan, F., Xiao, B., Li, X., Peng, C., & Peng, F. (2024). Metochalcone induces senescence-associated secretory phenotype via JAK2/STAT3 pathway in breast cancer. Oncology Research, 32(5), 943.

[24] Crouch, J., Shvedova, M., Thanapaul, R. J. R. S., Botchkarev, V., & Roh, D. (2022). Epigenetic Regulation of Cellular Senescence. Cells 2022, Vol. 11, Page 672, 11(4), 672.

[25] Wang, T. W., & Nakanishi, M. (2024). Immune surveillance of senescence: potential application to age-related diseases. Trends in Cell Biology.

[26] Chaib, S., Tchkonia, T., & Kirkland, J. L. (2022). Cellular senescence and senolytics: the path to the clinic. Nature Medicine 2022 28:8, 28(8), 1556–1568.

[27] Justice, J. N., Nambiar, A. M., Tchkonia, T., LeBrasseur, N. K., Pascual, R., Hashmi, S. K., Prata, L., Masternak, M. M., Kritchevsky, S. B., Musi, N., & Kirkland, J. L. (2019). Senolytics in idiopathic pulmonary fibrosis: Results from a first-in-human, open-label, pilot study. EBioMedicine, 40, 554–563.

[28] Lagoumtzi, S. M., & Chondrogianni, N. (2021). Senolytics and senomorphics: Natural and synthetic therapeutics in the treatment of aging and chronic diseases. Free Radical Biology and Medicine, 171, 169–190.

[29] Nieto, M., Könisgberg, M., & Silva-Palacios, A. (2024). Quercetin and dasatinib, two powerful senolytics in age-related cardiovascular disease. Biogerontology, 25(1), 71–82.

[30] Amor, C., Fernández-Maestre, I., Chowdhury, S., Ho, Y. J., Nadella, S., Graham, C., Carrasco, S. E., Nnuji-John, E., Feucht, J., Hinterleitner, C., Barthet, V. J. A., Boyer, J. A., Mezzadra, R., Wereski, M. G., Tuveson, D. A., Levine, R. L., Jones, L. W., Sadelain, M., & Lowe, S. W. (2024). Prophylactic and long-lasting efficacy of senolytic CAR T cells against age-related metabolic dysfunction. Nature Aging 2024 4:3, 4(3), 336–349.

[31] Muthamil, S., Kim, H. Y., Jang, H. J., Lyu, J. H., Shin, U. C., Go, Y., Park, S. H., Lee, H. G., & Park, J. H. (2024). Biomarkers of Cellular Senescence and Aging: Current State-of-the-Art, Challenges and Future Perspectives. Advanced Biology, 8(9), 2400079.

[32] Cohn, R. L., Gasek, N. S., Kuchel, G. A., & Xu, M. (2023). The heterogeneity of cellular senescence: insights at the single-cell level. Trends in Cell Biology, 33(1), 9–17.

Related Organizations

Related Community Members