Fact-checked by Grok 2 weeks ago

Cellular senescence

Cellular senescence is a stress-induced, essentially irreversible form of cell-cycle arrest that prevents the proliferation of damaged or aged cells, serving as a protective mechanism against tumorigenesis while also contributing to tissue aging and dysfunction through the secretion of bioactive molecules known as the (SASP). This process was first described in 1961 by and Paul Moorhead, who observed that normal human diploid fibroblasts in culture undergo a finite number of divisions, termed the , before entering a state of replicative due to telomere shortening. Over subsequent decades, research expanded the concept beyond replicative limits to include various triggers such as persistent DNA damage, oncogenic activation, , and therapeutic agents like , all of which activate tumor suppressor pathways involving proteins like and ^INK4a to enforce the arrest. At the molecular level, senescent cells exhibit distinct hallmarks beyond proliferation cessation, including , metabolic alterations, and resistance to , which allow them to persist in tissues for extended periods. The SASP, a defining feature, comprises a heterogeneous array of secreted factors—including (e.g., IL-6, IL-8), , growth factors (e.g., VEGF), and matrix-degrading enzymes—that propagate senescence in neighboring cells via and modulate the tissue microenvironment. These secretions can elicit beneficial effects, such as reinforcing tumor suppression by alerting immune cells to eliminate precancerous lesions or aiding in and embryonic development by clearing damaged cells. However, chronic SASP accumulation drives detrimental outcomes, including sterile , , and stem cell exhaustion, which are implicated in age-related pathologies like , , and neurodegeneration. Physiologically, cellular senescence plays dual roles across the lifespan: it acts as a safeguard during and tissue repair but becomes maladaptive with aging as senescent cells evade immune clearance and accumulate, exacerbating organismal decline. Emerging evidence as of November 2025 identifies a subset of T helper cells that actively eliminate senescent cells, potentially mitigating aging effects. In cancer, senescence initially suppresses tumor growth—famously demonstrated by oncogene-induced senescence () in response to activation—but paradoxically, SASP factors can foster a tumor-permissive niche by promoting , immune evasion, and in surrounding cells. Emerging evidence also highlights context-dependent reversibility, challenging the traditional view of permanence; for instance, transient SASP inhibition or epigenetic modulation can restore proliferative capacity in some senescent states, opening avenues for therapeutic intervention. Therapeutically, targeting senescent cells has gained traction as a strategy to mitigate aging and disease, with senolytics—drugs like and that selectively induce in senescent cells—showing promise in preclinical models of frailty, , and by reducing SASP burden and improving tissue function. Clinical trials are underway to evaluate these approaches, underscoring cellular as a pivotal target in geroscience, though challenges remain in achieving specificity to avoid disrupting beneficial senescence programs.

Introduction

Definition and Types

Cellular senescence is a stable and generally irreversible form of cell cycle arrest that prevents the proliferation of damaged or aged cells while maintaining their metabolic viability. This state is induced by various endogenous and exogenous stresses, including DNA damage, telomere dysfunction, and oncogenic signaling, and is characterized by distinct morphological changes such as enlarged cell size, flattened cytoplasm, and the expression of senescence-associated β-galactosidase (SA-β-gal). Unlike transient responses, senescent cells remain alive but cease to divide, distinguishing this process as a protective mechanism against tumorigenesis. The primary types of cellular senescence include replicative senescence, oncogene-induced senescence (OIS), therapy-induced senescence (TIS), and stress-induced premature senescence (SIPS). Replicative senescence arises from progressive telomere shortening during repeated cell divisions, ultimately leading to a critical limit on proliferation as first observed in human fibroblasts. OIS is triggered by hyperproliferative signals from activated oncogenes, such as RAS, acting as a safeguard against malignant transformation. TIS occurs in response to genotoxic therapies like chemotherapy or radiation, which cause DNA damage and enforce arrest in potentially cancerous cells. SIPS encompasses premature arrest due to acute stresses, including oxidative damage, epigenetic alterations, or other non-telomeric insults that mimic aging processes. Senescent cells differ fundamentally from quiescent cells, which undergo a reversible G0 arrest that can be exited upon mitogenic stimulation, whereas senescence enforces a permanent halt unresponsive to growth factors. In contrast to , a pathway involving activation and cellular dismantling, senescence preserves cell viability and often involves the secretion of pro-inflammatory factors known as the (SASP). Evolutionarily, cellular senescence serves as a conserved tumor-suppressive mechanism across eukaryotes, limiting the propagation of mutations that could lead to cancer by arresting at-risk cells.

Historical Background

The concept of cellular senescence emerged from early observations in experiments. In 1961, and Paul Moorhead demonstrated that normal human diploid fibroblasts, derived from embryonic lung tissue, undergo a finite number of population doublings—approximately 50—before entering a state of irreversible growth arrest, now known as the . This finding challenged the prevailing view that normal cells could divide indefinitely and laid the groundwork for understanding replicative limits in somatic cells. Theoretical and experimental advances in the 1970s and 1980s linked this replicative arrest to telomere biology. In 1971, Alexey Olovnikov proposed the "marginotomy" hypothesis, positing that incomplete replication of linear DNA ends leads to progressive shortening of chromosome termini, eventually triggering cellular senescence. This idea was experimentally supported in the 1980s through work by Elizabeth Blackburn and Jack Szostak, who identified conserved telomeric DNA sequences that protect chromosome ends and demonstrated their role in maintaining linear plasmids in yeast, providing the first evidence of a telomere maintenance mechanism. The marked a shift toward recognizing as a tumor-suppressive mechanism beyond replication. In 1997, Manuel Serrano and colleagues discovered oncogene-induced , showing that ectopic expression of oncogenic Ras in primary human and rodent fibroblasts provokes a permanent G1 resembling replicative , mediated by and p16INK4a pathways. In the , research elucidated 's broader implications for aging and tissue . Judith Campisi's group identified the (SASP) in the late , revealing that senescent cells secrete proinflammatory cytokines, growth factors, and proteases that can promote chronic and tumor progression in surrounding tissues. Concurrently, studies highlighted p16INK4a as a key of in vivo, with its expression increasing in aged tissues and correlating with age-related proliferative defects in stem cells. Recent developments through 2025 have integrated senescence with epigenetic and single-cell analyses. In 2016, epigenetic clock models based on patterns were applied to senescent cells, demonstrating accelerated epigenetic aging in response to replicative stress and linking it to organismal aging. Advances in single-cell sequencing during the 2020s have uncovered significant heterogeneity among senescent populations, revealing diverse transcriptional states in fibroblasts and other cell types that influence SASP variability and therapeutic targeting.

Induction Mechanisms

Replicative Senescence

Replicative senescence represents the progressive loss of a cell's proliferative capacity due to the inherent limitations of at ends. In normal cells, this process is primarily driven by the gradual attrition of telomeres, which act as protective caps preventing chromosomal instability. As cells divide repeatedly, telomeres shorten until they reach a critical length that elicits a persistent DNA damage response, ultimately enforcing a stable arrest. Human telomeres consist of repetitive TTAGGG sequences arrayed in tandem at the ends of linear chromosomes, forming complexes that shield against end-to-end fusions and degradation. During each round of , the end-replication problem—arising from the inability of to fully synthesize the lagging strand—results in the loss of approximately 50-100 base pairs from these telomeric repeats. This progressive shortening occurs in the absence of compensatory mechanisms, limiting the number of population doublings a cell population can achieve.00629-1.pdf) When telomeres erode to a critical length, typically resulting in an average of about 7 with the shortest reaching 1-2 , they lose their protective function, leading to the formation of telomere dysfunction-induced foci (TIFs). TIFs are cytological markers where telomere ends colocalize with DNA damage response factors, such as γ-H2AX and 53BP1, mimicking double-strand breaks and activating checkpoints that halt . This uncapping triggers a DNA damage response, often involving pathway activation, to induce . Telomerase, a ribonucleoprotein that adds TTAGGG repeats to ends using its template, is notably absent or expressed at undetectable levels in most differentiated somatic cells. This lack of activity enforces the replicative limit by permitting unchecked erosion over successive divisions. In contrast, is active in stem cells, germ cells, and most cancer cells, allowing indefinite proliferation.80001-5) The , which describes the finite number of divisions (typically 40-60 for human fibroblasts) before , can be contextualized by the relationship between initial length, the critical threshold, and the shortening rate per division: approximate population doublings ≈ (initial length - critical length) / shortening rate per division. This framework underscores how variations in starting length influence replicative lifespan. Seminal evidence supporting the causal role of telomere shortening in replicative senescence comes from experiments demonstrating that ectopic overexpression of the catalytic subunit (hTERT) in primary fibroblasts stabilizes telomere length and extends their proliferative lifespan by over 20 doublings without altering or inducing tumorigenicity. These telomerase-expressing cells bypassed markers, such as β-galactosidase activity, confirming telomeres as the primary .

Oncogene-Induced Senescence

Oncogene-induced senescence (OIS) represents a critical tumor-suppressive mechanism wherein hyperproliferative signals from activated oncogenes trigger a stable cell cycle arrest in primary cells, preventing malignant transformation. This process is initiated when oncogenes such as RAS or MYC drive excessive cell proliferation, leading to replication stress characterized by hyper-replication of DNA. The resulting stalled replication forks and under-replicated DNA regions activate a DNA damage response (DDR), culminating in premature senescence independent of telomere shortening. Unlike quiescence, which is a reversible growth arrest, OIS enforces an irreversible halt through epigenetic modifications that silence proliferation-associated genes. The molecular cascade begins with oncogenic signaling, exemplified by constitutively active H-RAS^{V12}, which upregulates and transcription factors to promote entry into and accelerate cell cycling. This hyperproliferative state overwhelms replication machinery, causing DNA hyper-replication and accumulation of DNA double-strand breaks, primarily at fragile sites, as detected by γ-H2AX foci. Feedback loops mitigate this deregulation; for instance, promyelocytic leukemia (PML) nuclear bodies recruit / complexes, sequestering E2F1 and repressing its transcriptional activity to enforce G1 arrest. Similarly, overexpression induces replication stress via deregulation of metabolism and , activating pathways that converge on and effectors to stabilize . Seminal experiments in 1997 demonstrated OIS upon retroviral expression of oncogenic H-RAS^{V12} in primary human fibroblasts (e.g., IMR-90) and rodent cells, where transduced cells adopted a flattened, enlarged morphology by day 6 post-infection, ceased DNA synthesis (near 0% BrdU incorporation), and exhibited senescence-associated β-galactosidase activity in up to 60% of cells by the same time point. This arrest was permanent, associated with elevated p53, p21^{CIP1}, and p16^{INK4a} levels (10- to 20-fold increases), and required intact p53 and Rb pathways, as their inactivation allowed proliferation. OIS thus manifests as a subtype of premature senescence, bypassing the need for cumulative cell divisions or telomere erosion seen in replicative senescence, and serves as an early barrier to tumorigenesis in vivo. The irreversibility of distinguishes it from transient quiescence, as it involves stable epigenetic silencing of target genes through senescence-associated heterochromatin foci (SAHF). These structures, marked by trimethylation of at lysine 9 () and recruitment of (HP1), compact proliferation-promoting loci, preventing re-entry into the even upon oncogene withdrawal. In lymphoma models, this chromatin remodeling enforces as an initial restraint on /RAS-driven lymphomagenesis, with escape requiring additional mutations that disrupt these epigenetic locks.

Stress-Induced Senescence

Stress-induced premature senescence (SIPS) arises from various exogenous and endogenous stressors that accumulate cellular damage, distinct from telomere attrition or oncogenic activation. Key triggers include generated by (ROS) from mitochondrial dysfunction, which overwhelms defenses and leads to macromolecular damage. Genotoxic agents such as (UV) radiation and chemotherapeutic drugs like also induce senescence by causing direct DNA lesions. Additionally, epigenetic alterations, such as aberrant or histone modifications triggered by , contribute to instability and enforce arrest. Severe mitogenic signals, including hyperactivation of pathways like MEK/MAPK, can similarly provoke senescence through excessive proliferative pressure and associated damage. At the molecular level, these stresses often converge on the DNA damage response () pathway. Persistent double-strand breaks (DSBs) activate ataxia-telangiectasia mutated () and ataxia-telangiectasia and Rad3-related (ATR) kinases, which phosphorylate histone H2AX to form γ-H2AX foci, marking sites of irreparable damage and sustaining a pro-senescence signal. This persistent prevents repair and amplifies signals. Beyond telomeric regions, stress induces non-telomeric alterations, including loss of marks like , leading to global and reinforcement of the senescent state. Experimental evidence underscores these mechanisms. In 2004, treatment of human fibroblasts with (H₂O₂) was shown to induce a senescent characterized by irreversible growth arrest and β-galactosidase activity, mimicking oxidative damage in aging. Similarly, doxorubicin therapy in cancer s triggers senescence via DSB accumulation, contributing to treatment outcomes but also potential relapse risks. A explains induction: even a small number of persistent unrepaired DSBs, as few as one or two per , can favor senescence over successful repair, , or continued .

Other Triggers

Cellular senescence can also be triggered by cellular hypertrophy, where enlarged cell size in tissues such as the liver, muscle, or heart disrupts normal homeostasis and activates senescence pathways. In cardiomyocytes, for instance, hypertrophy associated with dilated cardiomyopathy leads to increased cell size, which correlates with elevated markers of senescence like p16^INK4a and β-galactosidase activity, as observed in studies from the 2010s examining heart failure models. This process involves dysregulation of the Hippo signaling pathway, where overgrowth signals via YAP/TAZ effectors promote a pro-senescence state, and dilution of cytoplasmic components due to volume expansion impairs proteostasis and metabolic balance, ultimately enforcing cell cycle arrest. Metabolic stress represents another trigger, encompassing conditions like nutrient deprivation or chronic high glucose exposure that dysregulate energy sensing and induce senescence through pathway hyperactivation. Under nutrient scarcity, such as glucose deprivation, inhibition typically promotes survival adaptations, but paradoxical hyperactivation in response to metabolic imbalance—often seen in —drives cellular senescence by enhancing protein synthesis while suppressing , leading to accumulation of damaged organelles. High glucose levels, in particular, trigger hyperactivity in a glutamine-dependent manner, suppressing mTORC2 and promoting senescent phenotypes in endothelial and renal cells. Mechanical cues from the () stiffness can further induce , particularly in conditions where rigid matrices signal through to activate mechanotransduction pathways. In , increased stiffness—due to excessive deposition and cross-linking—engages integrin-mediated focal adhesion kinase (FAK) signaling, which upregulates /TAZ activity and promotes p53-dependent in fibroblasts and epithelial cells. This mechanosensitive response contributes to pathological tissue remodeling, as demonstrated in and liver models where substrate stiffness mimicking diseased states elevates -associated β- and SASP factors. Emerging research highlights additional triggers such as viral infections and , which impose leading to in specific contexts. HIV infection, for example, accelerates cellular in immune cells and tissues through persistent viral proteins like Tat, which upregulate senescence biomarkers via TLR7 and mitochondrial dysfunction, contributing to premature aging in infected individuals. Similarly, in neurodegeneration, protein aggregates like in induce in neurons and glia by activating DNA damage responses and pathways, exacerbating tissue decline. Recent studies also implicate the gut in modulating , where alters metabolite production—such as phenylacetylglutamine—that promotes mitochondrial dysfunction and senescent phenotypes in distant organs like the and .

Molecular Signaling Pathways

p53-Dependent Pathways

Cellular senescence is critically mediated by the tumor suppressor protein p53, which acts as a key sensor of cellular stress, particularly DNA damage, to enforce irreversible cell cycle arrest. Upon detection of DNA double-strand breaks or other genotoxic insults, kinases such as ATM and ATR phosphorylate p53 at multiple sites, including serine 15 and serine 20, stabilizing the protein by disrupting its interaction with the E3 ubiquitin ligase MDM2. This prevents MDM2-mediated ubiquitination and proteasomal degradation of p53, allowing its accumulation in the nucleus where it functions as a transcription factor. In the context of senescence, this stabilization shifts p53 from a transient response to a sustained activation state, distinguishing it from apoptosis or reversible arrest. Activated transcriptionally upregulates several downstream effectors that collectively enforce arrest and reinforce . The inhibitor p21 (encoded by CDKN1A) is a primary target, binding to and inhibiting the CDK2/cyclin E complex to prevent phosphorylation of the (Rb) and block E2F-dependent progression into . Additionally, induces (PAI-1, encoded by SERPINE1), which supports the senescent phenotype by modulating remodeling and reinforcing arrest, and (PML), which forms nuclear bodies that further stabilize through post-translational modifications. These effectors ensure a robust, multilayered barrier to proliferation, with p21 playing a central role in the initial arrest while PAI-1 and PML contribute to long-term maintenance. p53 also promotes epigenetic modifications that lock in the senescent state, including the deposition of lysine 9 trimethylation (), a repressive mark, at promoters of E2F-responsive genes such as cyclins and factors. This formation, often observed in senescence-associated heterochromatin foci (SAHFs), silences proliferation-associated loci and is dependent on sustained activity, which indirectly recruits histone methyltransferases like SUV39H1 via p21-mediated Rb hypophosphorylation. Evidence for 's essential role comes from seminal mouse models: mice, generated in the early 1990s, exhibit normal but dramatically increased susceptibility to spontaneous tumors due to bypassed senescence checkpoints, highlighting 's tumor-suppressive function through this pathway. More recent single-cell sequencing studies from the 2010s onward have revealed heterogeneity in activation across senescent populations, with variable p21 expression correlating to arrest stability and underscoring 's dynamic regulation in vivo. A reinforcing feedback loop exists between and the (SASP), where suppresses pro-inflammatory SASP components like IL-6 while SASP factors such as COX-2-derived prostaglandins enhance stability, amplifying the arrest in a paracrine manner. This ensures senescence persistence but can also propagate senescence in neighboring cells, contributing to tissue-level effects.

Rb-Dependent Pathways

The (Rb), a key tumor suppressor, enforces permanent cell cycle exit in cellular senescence primarily through the Rb-dependent pathway, which blocks progression from G1 to . In this mechanism, the cyclin-dependent kinase inhibitor p16^{INK4A}, encoded by the gene, binds to and inhibits cyclin-dependent kinases 4 and 6 (CDK4/6), preventing their association with . This inhibition maintains Rb in its hypophosphorylated state, allowing Rb to remain bound to transcription factors and thereby repressing the expression of genes required for and , such as those involved in the . The resulting blockade is a hallmark of senescence, distinct from reversible quiescence, as hypophosphorylated Rb actively silences proliferative targets in a stable manner. Upregulation of ^{INK4A} is a critical step in activating this pathway and occurs in response to various senescence-inducing stresses, including replicative exhaustion and oncogenic signaling. Stress signals, such as persistent DNA damage or mitogenic cues, activate the p38 (MAPK) pathway, which directly enhances ^{INK4A} transcription and protein levels, thereby amplifying the inhibitory effect on CDK4/6. Additionally, epigenetic modifications, including demethylation of the promoter and at the INK4/ARF locus, contribute to sustained ^{INK4A} expression during onset. Beyond cell cycle repression, promotes senescence through additional functions, including the silencing of proliferation-associated genes like cyclin A via direct interaction with -responsive promoters. In senescent cells, hypophosphorylated recruits histone deacetylases and other chromatin-modifying complexes to target loci, inducing formation and long-term transcriptional repression, which reinforces the irreversible arrest. This is essential for maintaining the senescent state, as loss disrupts these epigenetic marks and allows partial reactivation of proliferative genes. For instance, -mediated at sites, such as senescence-associated heterochromatin foci (SAHF), exemplifies how integrates cell cycle control with stable epigenetic silencing. Experimental evidence underscores the necessity of the - axis for induction. In human fibroblasts, combined disruption of and Rb function prevents replicative , allowing continued despite telomere shortening or stress, as demonstrated in early 2000s studies showing bypass of growth arrest in Rb-deficient cells. Similarly, in murine models impairs oncogene-induced in fibroblasts, highlighting the pathway's role across . In humans, expression positively correlates with chronological aging, accumulating in tissues like T-cells and fibroblasts as a of senescent burden, linking the Rb pathway to age-related functional decline. The Rb-dependent pathway exhibits crosstalk with parallel senescence mechanisms, such as the p53-p21 axis, where hypophosphorylated can stabilize by binding and sequestering , its negative regulator, thereby enhancing -dependent transcription in certain stress contexts. This interaction allows coordinated activation of both pathways during oncogene-induced senescence, though primarily enforces the cell cycle exit independently of in many scenarios.

Additional Pathways

Beyond the core and pathways, the signaling pathway serves as a key modulator of cellular , particularly in response to persistent DNA damage or oncogenic stress. Activation of occurs through an autocrine loop involving interleukin-1α (IL-1α), where cell surface-bound IL-1α triggers nuclear translocation, promoting the transcription of pro-inflammatory genes and enhancing senescent cell survival while driving the (SASP). This mechanism was elucidated in studies from the late 2000s, demonstrating that IL-1α depletion in senescent cells reduces activity and disrupts the maintenance of the senescent state. Consequently, reinforces by integrating damage signals with inflammatory responses, independent of direct cell cycle arrest effectors. The PI3K/AKT/ pathway also contributes to senescence induction and maintenance, often through hyperactivation by growth factors or loss of PTEN tumor suppressor function. This hyperactivation phosphorylates and inhibits FoxO transcription factors, leading to metabolic reprogramming that favors senescence, including increased and reduced . Research from the 2010s highlighted how sustained PI3K/AKT signaling in oncogene-transformed cells triggers -dependent senescence, where activity suppresses feedback mechanisms that would otherwise promote . In metabolic contexts, this pathway links nutrient sensing to senescent arrest, ensuring cells enter a state of permanent quiescence amid aberrant growth signals. Peroxisome proliferator-activated receptor β/δ (PPARβ/δ) plays a supportive role in sustaining the metabolic profile of senescent cells by regulating oxidation (FAO). PPARβ/δ activation upregulates genes involved in β-oxidation, such as those encoding carnitine palmitoyltransferase-1 (CPT1) and (ACOX1), which help maintain in the altered lipid environment of . Epigenetic modifiers further enforce senescent arrest through . (HDAC) inhibitors, such as , induce by increasing histone acetylation at promoters of cell cycle inhibitors like p21, leading to transcriptional activation and irreversible growth arrest. Similarly, the polycomb repressive complex 2 (PRC2) component methylates at lysine 27 (H3K27me3), repressing proliferation-associated genes; however, EZH2 downregulation in response to DNA damage triggers by derepressing these loci without altering global H3K27me3 levels. These modifications create a stable epigenetic landscape that locks cells in , distinct from transient epigenetic changes in quiescence. Recent advances as of 2025 have identified additional pathways modulating senescence. For instance, the TFEB-HKDC1 axis promotes mitophagy and lysosomal repair, thereby inhibiting senescence in various cell types. Similarly, the IL-4-STAT6 signaling pathway upregulates genes to delay senescence in macrophages. These pathways integrate into broader networks that reinforce senescence. For instance, persistent activation in senescent cells inhibits by phosphorylating ULK1 and repressing biogenesis, leading to accumulation of damaged organelles and perpetuating the senescent state. This crosstalk with and PI3K/AKT pathways forms feedback loops, where mTORC1-driven metabolic shifts amplify inflammatory signaling and epigenetic silencing, ensuring robust maintenance of senescence across diverse stressors.

Characteristics of Senescent Cells

Morphological and Functional Changes

Senescent cells undergo profound morphological alterations, becoming enlarged and flattened with an expanded that often appears granular and vacuolated. These changes were first observed in human diploid fibroblasts approaching the end of their replicative lifespan, where cells in late passages exhibited a flattened shape and increased cytoplasmic volume compared to proliferating counterparts. Electron microscopy studies from the 1970s further revealed the presence of autophagic vacuoles and lysosomal structures within the cytoplasm of these senescent cells, contributing to their irregular, irregular morphology. A key biomarker for detecting senescent cells is senescence-associated (SA-β-gal) activity, detectable at pH 6.0 through a histochemical . This activity reflects lysosomal , a functional hallmark where senescent cells accumulate enlarged lysosomes, potentially due to impaired degradation pathways. At the nuclear level, senescent cells form senescence-associated heterochromatin foci (SAHF), compact chromatin domains enriched in repressive marks like and HP1 proteins, which help maintain the proliferative arrest.00183-2) Functionally, senescent cells exhibit a metabolic reprogramming toward increased glycolysis, even in the presence of oxygen, accompanied by reduced oxidative phosphorylation and elevated lactate production.30228-5) This shift supports the heightened biosynthetic demands of the senescent state, including protein synthesis for structural changes, while also contributing to redox balance through the pentose phosphate pathway. Additional biomarkers include the loss of nuclear lamin B1, which disrupts nuclear architecture and correlates with senescence entry, and persistent γ-H2AX foci indicating unresolved DNA damage. These can be quantified via immunofluorescence or western blotting, while upregulated cell cycle inhibitors like p16^INK4a and p21^CIP1 are commonly assessed by qPCR for transcript levels or immunohistochemistry for protein expression. Proteomic analyses in the 2020s have identified approximately 100 proteins upregulated in senescent cells, including those involved in lysosomal function, remodeling, and stress response, underscoring the multifaceted alterations. For instance, of senescent human mammary epithelial cells revealed robust changes in lysosomal and metabolic proteins, validating these shifts at a systems level. Senescent cells also display enhanced resistance to apoptosis, mediated by upregulation of anti-apoptotic members such as , Bcl-W, and , which inhibit mitochondrial outer membrane permeabilization. This persistence allows senescent cells to accumulate in s, influencing surrounding microenvironments beyond their intrinsic changes.

Senescence-Associated Secretory Phenotype (SASP)

The (SASP) is a hallmark feature of senescent cells, characterized by the robust and sustained secretion of a proinflammatory secretome that influences the surrounding microenvironment. This secretome comprises a diverse array of bioactive molecules, including cytokines such as interleukin-6 (IL-6) and IL-8, like (also known as GRO-α), growth factors including (VEGF), matrix metalloproteinases (MMPs) such as MMP-3 and MMP-10, and damage-associated molecular patterns (DAMPs) like high-mobility group box 1 (). These components were first systematically profiled in human fibroblasts and epithelial cells induced to senesce by genotoxic stress, revealing approximately 40 distinct factors associated with , remodeling, and immune modulation. The regulation of SASP involves both transcriptional and post-transcriptional mechanisms. At the transcriptional level, nuclear factor kappa B () and CCAAT/enhancer-binding protein beta (C/EBPβ) act as key drivers, cooperatively inducing the expression of proinflammatory SASP factors like IL-6 and IL-8 in response to persistent DNA damage signaling. Post-transcriptionally, p38 (MAPK) enhances SASP production by stabilizing mRNAs and boosting activity, independent of DNA damage response pathways. This regulated secretion typically develops over 4–7 days following senescence induction and can persist for weeks to months, contributing to the long-term impact of senescent cells. SASP exhibits significant heterogeneity influenced by the type of senescence-inducing stress. For example, oncogene-induced SASP often includes prosurvival factors that reinforce the senescent arrest in an autocrine manner, whereas stress-induced SASP (e.g., from genotoxic or oxidative damage) emphasizes paracrine signaling to propagate senescence or modulate nearby cells. Advances in single-cell RNA sequencing (scRNA-seq) during the 2020s have highlighted this context-specific diversity, identifying distinct SASP subtypes across cell types and inducers, such as enriched inflammatory profiles in therapy-induced senescence versus metabolic regulators in replicative senescence.30892-8) Functionally, SASP reinforces the through autocrine loops that maintain arrest and DNA damage resistance, while its paracrine effects can propagate to neighboring cells. However, prolonged SASP activity from uncleared senescent cells fosters chronic low-grade inflammation by continuously releasing proinflammatory mediators, exacerbating tissue dysfunction over time.31121-3)

Physiological Roles

In Development and Tissue Repair

Cellular senescence plays a critical role in embryonic by contributing to tissue patterning and remodeling. In mammalian embryos, senescence occurs in specific structures such as the endolymphatic sac of the and the mesonephros, where it facilitates proper organ formation by limiting excessive and promoting structured tissue regression. For instance, in models, senescent cells in the endolymphatic sac help establish the balance between and necessary for development, with these cells being cleared after fulfilling their patterning function.01295-6) Similarly, in human placental development, senescence in cells supports villous maturation and nutrient exchange, acting as a programmed mechanism to ensure proper placental architecture without leading to . In , transient enhances tissue repair by coordinating cellular responses. Shortly after , fibroblasts in the wound bed undergo , secreting platelet-derived growth factor AA (PDGF-AA) that stimulates proliferation and migration, thereby accelerating re-epithelialization and closure. This is temporary, resolving within days as the heals, and its absence impairs repair , as demonstrated in excisional models where inhibiting delays closure. Senescence also contributes to tissue remodeling in regenerative processes, such as liver regeneration, where it curbs excessive fibrotic responses. In response to partial hepatectomy in mice, senescent hepatic stellate cells accumulate and limit extracellular matrix deposition, preventing fibrosis and allowing orderly hepatocyte proliferation to restore liver mass. This anti-fibrotic role highlights senescence's function in maintaining tissue homeostasis during repair. Evidence from senescence-deficient mouse models, including those lacking p21, reveals developmental defects such as impaired mesonephros regression and ectodermal ridge abnormalities, underscoring the necessity of senescence for proper embryogenesis and repair.00807-6) Unlike persistent senescence in other contexts, developmental and repair-associated senescence is transient and efficiently resolved, often through clearance mechanisms that prevent long-term accumulation. This resolution ensures that senescence supports positive outcomes without contributing to chronic issues.

In Tumor Suppression

Cellular senescence serves as a critical barrier to tumorigenesis by inducing a stable arrest in response to oncogenic stress, thereby preventing the proliferation of premalignant cells. Oncogene-induced senescence (OIS), first demonstrated with oncogenic , activates the and tumor suppressor pathways to enforce this arrest, halting the expansion of early lesions. In models, activation of oncogenes like K-Ras in epithelial cells triggers senescence in premalignant adenomas, limiting tumor progression until evasion occurs. Similarly, in human tissues, evidence of senescence enforcement through /Rb activation is observed, particularly in premalignant stages.00111-X) Genetic evidence underscores 's tumor-suppressive role. Knockout of the Ink4a/Arf locus, which encodes p16^INK4a (an activator) and p14^ARF (a stabilizer), leads to spontaneous development of sarcomas, lymphomas, and other tumors in mice within months of birth, demonstrating that loss of predisposes to . In humans, benign nevi contain senescent melanocytes harboring BRAF^V600E mutations, where mediated by p16^INK4a and -associated β-galactosidase activity prevents progression to . These examples illustrate how confines oncogenic lesions, with markers like p16^INK4a and SA-β-gal prominently expressed in benign or premalignant tissues but absent in fully malignant tumors. Cancer cells evade senescence to promote tumorigenesis, often through mutations disrupting or pathways. Inactivating mutations occur in approximately 50% of human cancers, disabling DNA damage-induced senescence and allowing survival of genotoxically stressed cells. Likewise, p16^INK4a loss via deletion or epigenetic silencing is frequent in cancers such as and pancreatic , bypassing Rb-dependent arrest and enabling unchecked proliferation. This evasion is evident in the transition from senescent premalignant lesions to invasive carcinomas, where inactivation of senescence effectors like or Ink4a/Arf abolishes the growth arrest.00247-3) The senescence-associated secretory phenotype (SASP) contributes to tumor suppression by recruiting immune cells for "senescence surveillance," where natural killer cells and macrophages eliminate senescent premalignant cells via innate immune recognition. Therapeutic strategies exploit this by inducing senescence in tumors with chemotherapy or targeted agents, enhancing immune-mediated clearance and improving outcomes in preclinical models.00121-1) However, chronic SASP can paradoxically drive tumor progression by promoting epithelial-mesenchymal transition (EMT) in adjacent nonsenescent cells through proinflammatory cytokines like IL-6 and TGF-β. Thus, while senescence initially curbs cancer, its evasion or persistence underscores its dual-edged nature in oncogenesis.

In Aging and Disease

Cellular senescence contributes to the aging process through the progressive accumulation of senescent cells in various tissues, which promotes chronic low-grade inflammation known as inflammaging primarily via the senescence-associated secretory phenotype (SASP). This accumulation impairs tissue homeostasis and function, as senescent cells resist apoptosis and secrete pro-inflammatory cytokines, chemokines, and matrix-degrading enzymes that disrupt neighboring cells and extracellular matrix integrity. Studies in human tissues demonstrate that senescent cell burden increases with chronological age, varying by tissue type but reaching notable levels in adipose, skin, and vascular tissues by late adulthood, thereby accelerating age-related decline. In age-related diseases, senescent cells play a pathogenic role by exacerbating tissue dysfunction and pathology. In , senescence of vascular endothelial cells and smooth muscle cells promotes plaque formation, , and arterial stiffening through SASP-mediated and impaired . Similarly, in , senescent chondrocytes contribute to degradation by upregulating matrix metalloproteinases and inflammatory factors, leading to joint degeneration. In neurodegenerative disorders such as Alzheimer's and Parkinson's diseases, neuronal and glial cell senescence drives , amyloid-beta accumulation, and synaptic loss, worsening cognitive decline. These examples illustrate how localized senescence amplifies disease progression across organ systems. Progeroid syndromes, rare genetic disorders mimicking accelerated aging, often involve defects that induce premature cellular senescence, providing insights into senescence's role in aging pathologies. Key examples include: Hutchinson-Gilford progeria syndrome (caused by LMNA mutations, featuring growth retardation, alopecia, scleroderma-like skin changes, and fatal ); Werner syndrome (WRN helicase deficiency, with bilateral cataracts, , , and high cancer risk); Bloom syndrome (BLM mutations, characterized by short stature, , and elevated cancer incidence); Rothmund-Thomson syndrome (RECQL4 defects, involving , skeletal dysplasias, and cataracts); Cockayne syndrome (ERCC6 or ERCC8 mutations, marked by progressive neurodegeneration, , and ); trichothiodystrophy (mutations in DNA repair genes like GTF2H5, presenting with brittle hair, , and ); and ataxia-telangiectasia ( gene alterations, leading to , telangiectasias, , and predisposition). These syndromes highlight how and lamin defects trigger senescence-like phenotypes, resulting in multi-system premature aging. Evidence from experimental models and human studies underscores senescence's causal role in aging and disease. In a seminal 2011 study, genetic clearance of p16^INK4a-positive senescent cells in progeroid mice delayed age-related disorders, demonstrating that reducing senescent burden ameliorates aging phenotypes. Human biopsies from patients with (IPF) reveal elevated p16-positive senescent cells in lung tissue, correlating with disease severity and fibrosis progression. These findings support the notion that senescent cells drive pathology through persistent presence. The mechanisms underlying senescence's detrimental effects in aging involve impaired immune-mediated clearance, leading to senescent cell persistence and amplified tissue damage. Failed clearance mechanisms, such as reduced activity with age, allow senescent cells to accumulate, exacerbating via SASP-induced remodeling and contributing to exhaustion by creating hostile microenvironments that impair regeneration. This vicious cycle perpetuates organ dysfunction and frailty in aging. Recent 2025 research has linked cellular to long symptoms, where therapy-induced or virus-triggered senescence in epithelial and endothelial cells contributes to persistent , , and multi-organ sequelae in long-haul patients, suggesting senescence as a mediator of post-viral aging-like syndromes.

Clearance and Persistence

Immune-Mediated Clearance

The plays a crucial role in recognizing and eliminating senescent cells to preserve tissue homeostasis, primarily through innate effectors like natural killer () cells and macrophages. Senescent cells express surface ligands such as and MICB, which bind to the activating receptor on cells, facilitating their identification. Additionally, components of the (SASP), including cytokines like IL-6, act as chemoattractants to recruit cells and macrophages to sites of senescence. These mechanisms ensure targeted surveillance, though senescent cells can upregulate inhibitory signals like to evade detection in certain contexts. Once recognized, NK cells induce in senescent cells through granule , releasing perforin to permeabilize the target and granzymes to activate cascades. Macrophages, particularly the pro-inflammatory subtype, contribute by engulfing senescent cells via , often mediated by "eat-me" signals such as exposed on the senescent cell surface, which binds to low-density lipoprotein receptor-related protein 1 () on macrophages. This process is supported by + T cells, which provide helper functions to amplify the response; for instance, in mouse models of oncogenic in the liver, + T cells were essential for the efficient recruitment and activation of cytotoxic effectors to clear premalignant senescent hepatocytes. In young organisms, this clearance is highly efficient, with senescent cells exhibiting a of approximately 5 days due to robust immune . However, with advancing age, immune exhaustion—characterized by reduced cell cytotoxicity and macrophage phagocytic capacity—prolongs this to weeks or longer, allowing senescent cell accumulation. Evidence from studies supports the potential of to restore clearance; for example, chimeric antigen receptor () T cells engineered to target uPAR on senescent cells have shown enhanced elimination in preclinical models relevant to age-related , with potential for future clinical translation in conditions like . Regulation of this process balances against risks like , with TGF-β secreted by senescent cells inhibiting excessive and T cell activation to limit collateral tissue damage. This inhibitory feedback, while protective in , contributes to impaired clearance in aging by dampening immune responsiveness.

Accumulation and Consequences

The persistence of senescent cells arises from multiple factors that impair their clearance, including age-related immune , which diminishes the of immune mechanisms. Fibrotic remodeling in tissues can physically shield senescent cells from immune detection, while the (SASP) promotes local by recruiting regulatory T cells and secreting factors. These processes collectively allow senescent cells to evade elimination, leading to their gradual buildup over time. Accumulation of senescent cells disrupts homeostasis, primarily through chronic inflammation and direct interference with neighboring cells. In the niche, senescent cells secrete factors that inhibit progenitor and , resulting in impaired regeneration and functional decline, as observed in aged muscle and hematopoietic tissues. from SASP further propagates senescence to adjacent healthy cells, amplifying the senescent burden in a . Systemically, this contributes to frailty by promoting , with elevated senescent cell levels correlating with reduced physical resilience and increased vulnerability to stressors in older adults. Models estimate that elderly s harbor a low but functionally significant fraction of senescent cells across tissues, representing less than 1% of total cells in most organs. Experimental clearance of senescent cells has been shown to reduce associated biomarkers by 30-50% in human tissues, underscoring the potential impact of this burden. Evidence from human studies highlights these effects; for instance, in extremely obese individuals, senescent preadipocytes accumulate over 30-fold higher in visceral compared to non-obese controls, exacerbating metabolic dysfunction and . Longitudinal studies in the 2020s have correlated higher senescent cell burden, measured via circulating biomarkers like ^{INK4a}, with increased and mortality risk in aging populations. This accumulation creates vicious feedback cycles, as persistent senescent cells intensify SASP production, further suppressing immune clearance and promoting additional in surrounding tissues.

Therapeutic Approaches

Senolytic Drugs

drugs are pharmacological agents that selectively eliminate senescent cells by inducing , targeting their characteristic resistance to . These cells upregulate anti-apoptotic pathways, including those mediated by proteins such as , , and Bcl-w, which senolytics exploit to promote their demise while sparing proliferating cells. This approach relies on the senescent cells' reliance on hyperactivated signals, allowing for selective clearance without broad . Key examples of senolytics include the combination of dasatinib, a Src tyrosine kinase inhibitor, and quercetin, a natural flavonoid polyphenol, first identified in 2015 through a targeted screen for agents that kill human endothelial, mouse embryonic fibroblast, and primary fat cell senescent models. This regimen clears 30-70% of senescent cells in adipose tissue, lung, and other sites in preclinical models, demonstrating potency across multiple cell types. Navitoclax (ABT-263), a small-molecule BH3 mimetic that inhibits Bcl-2, Bcl-xL, and Bcl-w, was discovered as a senolytic in 2016 and effectively reduces senescent cell burden in diverse tissues like bone marrow and lung. Fisetin, another flavonoid, emerged in 2018 as a potent senolytic, selectively inducing apoptosis in senescent but not non-senescent cells in vitro and reducing senescence markers in progeroid and aged mice when administered intermittently. These agents achieve specificity partly through senescent cells' altered SASP, which can sensitize them to apoptosis, though dosing regimens are optimized to minimize effects on healthy cells. Mechanistically, disrupt the pro-survival network in , where pathways like PI3K/AKT and MAPK are chronically active, leading to overexpression that these drugs counteract. For instance, inhibits kinases upregulated in , while modulates heat shock proteins and pro-apoptotic factors; together, they synergize to tip the balance toward . Navitoclax directly binds anti-apoptotic proteins, freeing pro-apoptotic effectors like Bax and Bak to permeabilize mitochondria. similarly engages multiple pathways, including inhibition of anti-apoptotic and modulation of . This targeted vulnerability arises from 's irreversible growth arrest, making these cells "addicted" to survival signals absent in healthy cells. Preclinical evidence highlights senolytics' potential to mitigate age-related decline; in naturally aged mice, intermittent plus reduced senescent cell accumulation, improved physical function, and extended lifespan by 36% (Xu et al., 2018). In progeroid models, it improved healthspan. In naturally aged mice, treatment decreased senescence markers in multiple tissues and enhanced lifespan by 10%. Human trials, primarily I and II, support translation: in (IPF), plus administered intermittently over 3 weeks (3 days/week) cleared senescent cells in lung tissue and improved physical function, including a increase of 21.5 meters (~10%) in 6-minute walk distance and 0.12 m/s in speed, with good tolerability ( et al., 2019). For , a 2024 2 trial with similar regimens in postmenopausal women showed no overall reduction in markers but modest effects in a with higher baseline levels; larger studies are needed. UNITY Biotechnology's navitoclax analog UBX1325 advanced to 2b for diabetic , reporting functional improvements in vision as of 2025; their earlier program did not advance beyond 2. Challenges persist in clinical application, including off-target effects—navitoclax induces dose-limiting thrombocytopenia by depleting platelets via Bcl-xL inhibition—and variable efficacy across tissues due to delivery barriers like the blood-brain barrier. Intermittent "hit-and-run" dosing mitigates toxicity but requires optimization for tissue-specific senescence burdens, and long-term safety data remain limited despite promising short-term results.

Senomorphic Interventions

Senomorphic interventions refer to pharmacological strategies that modulate the deleterious effects of senescent cells by suppressing their harmful outputs, particularly the senescence-associated secretory phenotype (SASP), without inducing cell death. These approaches target key signaling pathways involved in SASP production, such as JAK/STAT, NF-κB, and mTOR, thereby mitigating inflammation and tissue dysfunction associated with senescence accumulation. Unlike senolytics, senomorphic effects are often reversible, allowing senescent cells to persist while restoring their functionality to a more benign state. A primary mechanism of senomorphic action involves inhibiting SASP secretion through interference with transcriptional regulators. For instance, JAK inhibitors block the IL-6/ pathway, reducing pro-inflammatory release from senescent cells. Similarly, inhibitors target epigenetic modifications that drive SASP expression, preventing enhancer remodeling and immune surveillance evasion in senescent cells. These interventions preserve the tumor-suppressive benefits of while attenuating its pathological consequences. Prominent examples include metformin, an AMPK activator that suppresses NF-κB-mediated SASP factors, thereby alleviating senescence-induced and mitochondrial dysfunction in aging cells. Rapamycin, an inhibitor, exemplifies a classic senomorphic agent by inhibiting the secretory phenotype in senescent fibroblasts and other cell types, with effects observed across multiple preclinical models since the 2010s. Additionally, anti-IL-6 antibodies directly neutralize key SASP components, reducing driven by interleukin-6 in senescent microenvironments. Preclinical evidence demonstrates the efficacy of senomorphics in extending healthspan without senescent cell clearance. In aged mouse models, JAK inhibitors administered for 10 weeks reduced , enhanced physical function, and alleviated frailty by suppressing SASP. Rapamycin treatment in mice similarly improved health metrics and extended lifespan by modulating mTOR-dependent SASP secretion, as shown in studies from the mid-2010s onward. These findings highlight senomorphics' potential in age-related conditions, with reversible modulation allowing for sustained therapeutic benefits. Senomorphic interventions offer advantages over cell-eliminating strategies, including a safer profile for chronic use due to lower and the ability to combine with immune-enhancing therapies for synergistic effects on persistence.

References

  1. [1]
    Cellular senescence: a key therapeutic target in aging and diseases
    Aug 1, 2022 · Cellular senescence is a hallmark of aging defined by stable exit from the cell cycle in response to cellular damage and stress.
  2. [2]
    Cellular senescence: the good, the bad and the unknown - PMC - NIH
    Aug 3, 2022 · Cellular senescence involves cell-cycle arrest and the release of inflammatory cytokines with autocrine, paracrine and endocrine activities.
  3. [3]
    Mechanisms of Cellular Senescence: Cell Cycle Arrest ... - Frontiers
    Cellular senescence is a stable cell cycle arrest that can be triggered in normal cells in response to various intrinsic and extrinsic stimuli, as well as ...Introduction · Senescence Mediated Cell... · The Senescence Associated...
  4. [4]
    Senescence and the SASP: many therapeutic avenues
    Senescence is a cellular stress response triggered by molecular damage, such as that caused by replicative exhaustion, aberrant oncogene activation (oncogene- ...Senescence And The Sasp... · Targeting The Sasp As An... · Engaging Senescence In...
  5. [5]
    Cellular senescence: Neither irreversible nor reversible
    Feb 22, 2024 · Cellular senescence is a critical stress response program implicated in embryonic development, wound healing, aging, and immunity.
  6. [6]
    Cellular Senescence: Aging, Cancer, and Injury
    Cellular senescence is a permanent state of cell cycle arrest that occurs in proliferating cells subjected to different stresses. Senescence is, therefore ...
  7. [7]
    Cellular Senescence: A Translational Perspective - ScienceDirect.com
    Cellular senescence is a cell fate that involves essentially irreversible replicative arrest, apoptosis resistance, and frequently increased protein synthesis.
  8. [8]
    Mechanisms and functions of cellular senescence - JCI
    Apr 2, 2018 · Cellular senescence was originally identified as a stable exit from the cell cycle caused by the finite proliferative capacity of cultured human ...<|control11|><|separator|>
  9. [9]
    New Horizons in cellular senescence for clinicians - PMC - NIH
    Jul 15, 2023 · Cellular senescence is a cell fate originally defined as an irreversible loss of replicative potential although it is now clear that it can be ...The Cellular Consequences Of... · Senescence As A Driver Of... · Can We Prevent Or Reverse...
  10. [10]
  11. [11]
    Cellular senescence and its effector programs - Genes & Development
    Cellular senescence is a stress response that accompanies stable exit from the cell cycle. Classically, senescence, particularly in human cells, involves the ...
  12. [12]
    Cellular Aging and Senescence in Cancer: A Holistic Review of ...
    Initiated by diverse stressors such as DNA damage, telomere shortening, and oncogene activation, cellular senescence acts as a stress response mechanism [46].
  13. [13]
    The serial cultivation of human diploid cell strains - ScienceDirect
    The isolation and characterization of 25 strains of human diploid fibroblasts derived from fetuses are described. Routine tissue culture techniques were ...
  14. [14]
    The serial cultivation of human diploid cell strains - PubMed
    The serial cultivation of human diploid cell strains. Exp Cell Res. 1961 Dec:25:585-621. doi: 10.1016/0014-4827(61)90192-6. Authors. L HAYFLICK, P S MOORHEAD.
  15. [15]
    Telomeres, Telomerase, and Aging: Origin of the Theory - PubMed
    In 1971 I published a theory in which I first formulated the DNA end replication problem and explained how it could be solved.
  16. [16]
    Cloning yeast telomeres on linear plasmid vectors - PubMed
    We have constructed a linear yeast plasmid by joining fragments from the termini of Tetrahymena ribosomal DNA to a yeast vector.Missing: paper | Show results with:paper
  17. [17]
    Oncogenic ras provokes premature cell senescence associated with ...
    Mar 7, 1997 · Here we show that expression of oncogenic ras in primary human or rodent cells results in a permanent G1 arrest.Missing: paper | Show results with:paper
  18. [18]
    Senescence-Associated Secretory Phenotypes Reveal Cell ...
    This senescence-associated secretory phenotype (SASP) developed slowly over several days and only after DNA damage of sufficient magnitude to induce senescence.
  19. [19]
    The Regulation of INK4/ARF in Cancer and Aging - ScienceDirect.com
    Oct 20, 2006 · Recent data also suggest that expression of p16INK4a induces an age-dependent decrease in the proliferative capacity of certain tissue-specific ...
  20. [20]
    Epigenetic clock analyses of cellular senescence and ageing - PMC
    Feb 14, 2016 · The epigenetic clock is defined as a prediction method of age based on the linear combination of the DNA methylation levels of 353 CpGs ...
  21. [21]
    Single-cell transcriptomic analysis uncovers diverse and dynamic ...
    Apr 19, 2023 · We set out to comprehensively analyze the senescent transcriptome of human diploid fibroblasts at the individual-cell scale by performing single-cell RNA- ...Missing: 2020s | Show results with:2020s
  22. [22]
    Telomere dysfunction in ageing and age-related diseases - Nature
    Feb 14, 2022 · Telomere shortening and damage are recognized causes of cellular senescence and ageing. Several human conditions associated with normal ageing are precipitated ...
  23. [23]
    Telomere Length, Aging, and Somatic Cell Turnover - PMC - NIH
    When these studies were carried out with human fibroblasts, telomeres were found to shorten at a relatively constant rate (50–100 bp/population doubling) ...
  24. [24]
  25. [25]
    Telomere length predicts replicative capacity of human fibroblasts
    TRF length (B) and remaining replicative capacity (mean population doublings, ... At the Hayflick limit (Ml), we assume that one or more telomeres have ...Missing: formula | Show results with:formula
  26. [26]
    Extension of life-span by introduction of telomerase into normal ...
    The telomerase-expressing clones have a normal karyotype and have already exceeded their normal life-span by at least 20 doublings.
  27. [27]
  28. [28]
    Myc and Ras oncogenes engage different energy metabolism ...
    Nov 15, 2014 · Both Myc and Ras oncogenes impact cellular metabolism, deregulate redox homeostasis and trigger DNA replication stress (RS) that compromises ...
  29. [29]
  30. [30]
    Oxidative Stress-Driven Cellular Senescence - PubMed Central - NIH
    This review explores the intricate interplay between redox imbalance and cellular senescence, emphasizing mitochondrial dysfunction, SASP dynamics, and their ...
  31. [31]
    Cellular senescence: a double-edged sword in cancer therapy - PMC
    Sep 12, 2023 · Cellular senescence can be induced by oxidative stress, ionizing radiation, UV stress, telomere shortening, genotoxic agents, and oncogene ...
  32. [32]
    The role of the dynamic epigenetic landscape in senescence - Nature
    Oct 24, 2024 · This review outlines the current understanding of how senescent cells undergo epigenetic changes and how these alterations govern SASP expression.Histone H3 Lysine... · Sasp Expression Is... · Histone Variants
  33. [33]
    Premature senescence involving p53 and p16 is activated in ...
    Premature senescence involving p53 and p16 is activated in response to constitutive MEK/MAPK mitogenic signaling. Athena W. Lin1,; Marta Barradas2,; James C.Missing: severe | Show results with:severe
  34. [34]
    Persistent DNA damage signaling triggers senescence-associated ...
    We show that damaged human cells develop persistent chromatin lesions bearing hallmarks of DNA double-strand breaks (DSBs), which initiate increased secretion ...
  35. [35]
    The loss of heterochromatin is associated with multiscale three ... - NIH
    The loss of heterochromatin is associated with multiscale three-dimensional genome reorganization and aberrant transcription during cellular senescence.Missing: paper | Show results with:paper
  36. [36]
    Loss of proliferative capacity and induction of senescence ... - PubMed
    Nov 19, 2004 · Cellular senescence can result from short, dysfunctional telomeres, oxidative stress, or oncogene expression, and may contribute to aging.
  37. [37]
    Doxorubicin-Induced Cancer Cell Senescence Shows a Time Delay ...
    In this study, we used doxorubicin-treated HeLa cells as a model of in vitro cellular senescence. We found the existence of time delay effect in drug-induced ...
  38. [38]
    Do DNA double-strand breaks drive aging? - PMC - PubMed Central
    DNA DSBs occur in any given cell in the order of 10 to 50 per cell per day, depending on cell cycle and tissue (Vilenchik and Knudson, 2003). DSBs, as well as ...
  39. [39]
    Cardiomyocyte Senescence and Cellular Communications Within ...
    May 21, 2020 · In senescent cardiomyocytes, enhanced expression of hypertrophic genes and enlarged cell size are observed (17). Cardiomyocyte senescence is ...
  40. [40]
    Cell enlargement modulated by GATA4 and YAP instructs ... - Nature
    Feb 17, 2025 · Our study highlights an unexpected instructive role of cell enlargement in modulating the SASP and reveals a mechanical branch in the senescence regulatory ...
  41. [41]
    mTOR as regulator of lifespan, aging and cellular senescence - PMC
    Dec 1, 2017 · This review discusses the potential mechanisms how mTOR signaling controls lifespan and influences aging-related processes such as cellular senescence, ...
  42. [42]
    mTOR: Its Critical Role in Metabolic Diseases, Cancer, and ... - MDPI
    They also showed that high glucose conditions trigger mTORC1 hyperactivity, suppressing mTORC2 in a glutamine-dependent manner. This highlights the important ...
  43. [43]
    Regulation of cellular senescence by extracellular matrix during ...
    Increased ECM stiffness is a feature of fibrosis and is thought to result from the quantity of ECM deposition and the degree of its cross-linking. The ...
  44. [44]
    Extracellular matrix stiffness: mechanisms in tumor progression and ...
    Apr 10, 2025 · Elevated ECM stiffness activates cellular responses through mechanical signal transduction pathways, such as Integrin/FAK and YAP/TAZ signaling ...
  45. [45]
    Substrate stiffness engineered to replicate disease ... - Frontiers
    This study investigated if increased stiffness could modulate markers of senescence and/or fibrosis in primary human lung fibroblasts.<|separator|>
  46. [46]
    HIV-associated cellular senescence: a contributor to accelerated aging
    Dec 23, 2016 · One possible contributor to this accelerated aging is cellular senescence, an age-associated response that can occur prematurely in response to stress.
  47. [47]
    Molecular Mechanisms of Cellular Senescence in ...
    Protein aggregates can induce cellular senescence in brain cells. •. Cellular senescence can be targeted for therapy for neurodegenerative diseases. Abstract.
  48. [48]
    Gut microbes revealed as hidden drivers of aging and cellular decline
    Jan 14, 2025 · Gut microbiota-derived phenylacetylglutamine accelerates aging by driving cellular senescence through mitochondrial dysfunction and DNA ...
  49. [49]
    ATM-dependent phosphorylation of Mdm2 on serine 395: role in p53 ...
    p53 is phosphorylated by ATM shortly after DNA damage, resulting in enhanced stability and activity of p53. The Mdm2 oncoprotein is a pivotal negative regulator ...
  50. [50]
    The complexity of p53 stabilization and activation - Nature
    Apr 7, 2006 · The phosphorylated form of MDM2 appears to be less capable of nucleo-cytoplasmic shuttling of p53, suggesting that the MDM2 phosphorylation ...
  51. [51]
    p53 in senescence – it's a marathon, not a sprint - FEBS Press - Wiley
    Dec 17, 2021 · The tumour suppressor p53, a stress-responsive transcription factor, plays a central role in cellular senescence.Senescence-Associated... · Coordinating P53 Activity... · P53-Mediated...<|control11|><|separator|>
  52. [52]
    Senescence Regulation by the p53 Protein Family - PubMed Central
    Rapid ATM-dependent phosphorylation of MDM2 precedes p53 accumulation in response to DNA damage. ... ATM-dependent phosphorylation of Mdm2 on serine 395: role in ...
  53. [53]
    p16 INK4A Mediates Cyclin Dependent Kinase 4 and 6 Inhibition in ...
    The increased expression of p16 translated into an increased association of p16 with cdk4 and cdk6 complexes in senescent HPECs. In senescent HPECs, as in other ...
  54. [54]
    Irreversibility of cellular senescence: dual roles of p16INK4a/Rb ...
    In human cells, once pRb is activated by p16INK4a, senescence cell cycle arrest becomes irreversible, unlike in murine cells where it can be reversed.Missing: review papers
  55. [55]
    p16INK4a Regulates Cellular Senescence in PD-1-Expressing ...
    Aug 9, 2021 · We show that these T cells accumulate DNA damage and activate the p38MAPK signaling pathway, which preferentially leads to p16INK4a upregulation ...
  56. [56]
    p16INK4a in cellular senescence - PMC - NIH
    Aug 17, 2013 · A crucial mechanism in the establishment of cellular senescence is the activation of the INK4/ARF locus, which is epigenetically regulated ...
  57. [57]
    Rb-Mediated Heterochromatin Formation and Silencing of E2F ...
    Rb family proteins are corepressors of the E2F transcription factors, and their combined activities are required for many aspects of cell cycle progression ( ...
  58. [58]
    The Retinoblastoma Protein Selectively Represses E2F1 Targets via ...
    Results: Rb protein selectively represses specific E2F1 target genes via a TAAC element in senescent cells. Conclusion: Cellular senescence is influenced by ...
  59. [59]
    Regulation of transcription and chromatin structure by pRB - NIH
    The retinoblastoma protein is capable of influencing senescence cell cycle arrest at various levels. First, pRB represses the transcription of genes involved in ...
  60. [60]
    Expression of p16INK4a in peripheral blood T-cells is a biomarker of ...
    Expression of the p16INK4a tumor suppressor sharply increases with age in most mammalian tissues, and contributes to an age-induced functional decline of ...
  61. [61]
    Report Significant Role for p16 INK4a in p53-Independent Telomere ...
    Telomere attrition in primary human fibroblasts induces replicative senescence accompanied by activation of the p53 and p16 INK4a /RB tumor suppressor pathways.
  62. [62]
  63. [63]
    Cell surface-bound IL-1α is an upstream regulator of the ... - PNAS
    IL-1α depletion significantly reduced NF-κB and C/EBPβ DNA binding activities in senescent cells and reduced NF-κB activity in presenescent cells. These ...
  64. [64]
    mTOR kinase leads to PTEN-loss-induced cellular senescence by ...
    Oct 18, 2018 · Multiple lines of evidence indicate that increased PI3K/AKT signaling induces cellular senescence in many cell types [5, 6]. In addition, loss ...
  65. [65]
    Roles of the PI3K/AKT/mTOR signalling pathways in ...
    Apr 1, 2020 · In this review, we briefly summarize the results of research on the PI3K/AKT/mTOR pathway and discuss the structural composition, activation, ...
  66. [66]
    PPARβ/δ: Linking Metabolism to Regeneration - PMC
    It has been shown to play major roles in fatty acid metabolism and energy expenditure and thus also in skeletal and cardiac muscle homeostasis and disease as ...
  67. [67]
    Fatty acid oxidation regulates cellular senescence by modulating the ...
    We report that fatty acid oxidation (FAO) serves as a critical regulator of cellular senescence and uncover the underlying mechanism by which FAO inhibition ...
  68. [68]
    Epigenetic Regulation of Cellular Senescence - MDPI
    Feb 15, 2022 · This review will highlight the changes in chromatin, DNA methylation, and histone alterations that establish and maintain cellular senescence.<|separator|>
  69. [69]
    Regulation of Cellular Senescence by Polycomb Chromatin ... - NIH
    Mar 27, 2018 · Ito et al. show that downregulation of EZH2 rapidly elicits DNA damage and triggers the onset of senescence without loss of H3K27me3 marks.
  70. [70]
    Persistent mTORC1 signaling in cell senescence results from ...
    May 31, 2017 · Further, increased autophagy and high levels of intracellular amino acids may act to support mTORC1 activity in starvation conditions.Introduction · Results and discussion · Materials and methods · Acknowledgments
  71. [71]
    The mTORC1-autophagy pathway is a target for senescent cell ...
    Feb 23, 2019 · In its active form, mTORC1 drives protein translation, lipid and nucleotide synthesis as well as inhibiting the catabolic process of autophagy ...Missing: integration | Show results with:integration
  72. [72]
    THE LIMITED IN VITRO LIFETIME OF HUMAN DIPLOID CELL ...
    THE LIMITED IN VITRO LIFETIME OF HUMAN DIPLOID CELL STRAINS. Exp Cell Res. 1965 Mar:37:614-36. doi: 10.1016/0014-4827(65)90211-9.Missing: morphology | Show results with:morphology
  73. [73]
    Electron microscopy of human fibroblasts in tissue culture during ...
    More subtle changes were the increase in the amount of condensation of the nuclear chromatin, and an increase in the number of autophagic vacuoles. Recommended ...
  74. [74]
    A biomarker that identifies senescent human cells in culture and in ...
    Sep 26, 1995 · G P Dimri , X ... We show that several human cells express a beta-galactosidase, histochemically detectable at pH 6, upon senescence in culture.Missing: paper | Show results with:paper
  75. [75]
    Senescent Human Fibroblasts Show Increased Glycolysis and ...
    Feb 18, 2015 · In summary, senescent cells may exhibit increased PPP flux to help restore redox homeostasis while displaying increased glycolytic metabolism in ...<|control11|><|separator|>
  76. [76]
    Identification of a Proteomic Signature of Senescence in Primary ...
    Jan 12, 2021 · We identified a robust proteomic signature of HMEC senescence consisting of 77 upregulated and 36 downregulated proteins.
  77. [77]
    Strategies targeting cellular senescence - JCI
    Apr 2, 2018 · Studies across different cell types have demonstrated an upregulation of the BCL-2 family members BCL-2, BCL-W, and BCL-XL during senescence (46 ...
  78. [78]
    Cellular senescence in development, regeneration and disease
    Oct 1, 2019 · Senescence was first discovered in primary cells that were grown for extended periods in culture, reaching what became known as a state of ...The Senescence Program · Roles For Senescence In... · Conclusions And Future...
  79. [79]
    Senescence in premalignant tumours - Nature
    Aug 4, 2005 · We conclude that a substantial number of cells in premalignant tumours undergo oncogene-induced senescence, but that cells in malignant tumours ...
  80. [80]
    Tumor Spectrum in ARF-deficient Mice1 | Cancer Research
    Mice lacking INK4a/ARF (7) or ARF alone (8) are highly tumor prone, and fibroblasts explanted from their embryos (mouse embryonic fibroblasts), like those ...
  81. [81]
    associated senescence-like cell cycle arrest of human naevi - Nature
    Aug 4, 2005 · Here we show that sustained BRAF V600E expression in human melanocytes induces cell cycle arrest, which is accompanied by the induction of both p16 INK4a and ...
  82. [82]
    Why are there hotspot mutations in the TP53 gene in human cancers?
    Nov 3, 2017 · The p53 gene contains homozygous mutations in ~50–60% of human cancers. About 90% of these mutations encode missense mutant proteins that ...
  83. [83]
    Loss of p16: A Bouncer of the Immunological Surveillance? - NIH
    Apr 2, 2021 · On one hand, loss of p16 is a common feature of cancer that causes an increase in the proliferative capacity of the cell [12]; on the other hand ...
  84. [84]
    Cellular Senescence Is Immunogenic and Promotes Antitumor ...
    Therapy-induced senescence renders cancer cells highly immunogenic, making them very efficient in triggering protective CD8-dependent antitumor immune.
  85. [85]
    The Dual Role of Cellular Senescence in Developing Tumors ... - NIH
    In most cases the SASP was reported to stimulate tumor growth (Figure 1), but on the other hand immune cells are attracted which participate in the clearance of ...
  86. [86]
    Cellular senescence and chronological age in various human tissues
    Dec 5, 2019 · While senescence is higher in aged tissue samples, the magnitude of senescence varies considerably depending upon tissue type, tissue section, ...
  87. [87]
    Cellular senescence and the senescent secretory phenotype - JCI
    We review the mechanisms that induce senescence and the SASP, their associations with chronic disease and frailty, therapeutic opportunities based on targeting ...
  88. [88]
    Survey of senescent cell markers with age in human tissues
    Senescent cells accumulate in tissues during aging and in a range of disease conditions [7, 8]. In addition to telomere shortening resulting from replicative ...
  89. [89]
    Cells in Atherosclerosis: Focus on Cellular Senescence from Basic ...
    Dec 5, 2023 · In summary, cellular senescence is a known major contributor to the development of atherosclerosis, which affects multiple cell types involved ...
  90. [90]
    Cellular senescence in osteoarthritis pathology - PMC
    Jan 26, 2017 · In this review, we discuss the mechanisms by which senescent cells may predispose synovial joints to the development and/or progression of OA.
  91. [91]
    Cellular senescence in brain aging and neurodegenerative diseases
    Feb 19, 2018 · Cells with features of senescence have been detected in the context of brain aging and neurodegenerative disease, suggesting that they may also promote ...
  92. [92]
    Molecular bases of progeroid syndromes - Oxford Academic
    Progeroid syndromes (PSs) constitute a group of disorders characterized by clinical features mimicking physiological aging at an early age. In some of these ...Pss And Dna Repair Defects · Recq Helicases And Ws · Mitochondria, Miscellaneous...
  93. [93]
    Progeroid syndromes: probing the molecular basis of aging? - PMC
    These observations are related to some of the less well characterised progeroid syndromes within the context of the cell senescence hypothesis of aging, a ...
  94. [94]
    positive senescent cells delays ageing-associated disorders - Nature
    Nov 2, 2011 · BubR1H/H mice have a markedly shortened lifespan and exhibit a variety of age-related phenotypes, including infertility, lordokyphosis, ...
  95. [95]
    Digital quantification of p16-positive foci in fibrotic interstitial lung ...
    Jun 7, 2022 · The presence of p16-positive foci was specific for the diagnosis of IPF, where 50% of cases expressed any level of p16 and 26% were p16-high.
  96. [96]
    Cellular Senescence in Kidney Fibrosis - Frontiers
    At the cellular level, accumulation of senescent cells and stem cell exhaustion ... unable to proliferate, ultimately causing tissue fibrosis and organ ...
  97. [97]
    Causes of symptoms and symptom persistence in long COVID and ...
    Jul 30, 2025 · Accelerated cellular senescence. Metabolic abnormalities. Redox imbalance: increased levels of pro-oxidants, decreased levels of antioxidants ...
  98. [98]
    Senescent cells evade immune clearance via HLA-E-mediated NK ...
    Jun 3, 2019 · Moreover, by secreting chemokines and cytokines, senescent cells can recruit immune cells into tissues that enable senescent cell clearance.
  99. [99]
    Senescent cells: Living or dying is a matter of NK cells - Antonangeli
    Feb 27, 2019 · In this review, how senescent cells trigger an immune response and, in particular, the ability of NK cells to recognize and clear senescent cells are discussed.3 Nk Cell-Mediated Clearance... · 5 Type I Interferons At The... · 6 Conclusions And...<|control11|><|separator|>
  100. [100]
    The Phagocytic Code Regulating Phagocytosis of Mammalian Cells
    Surface-exposed calreticulin acts as an eat-me signal to phagocytes via the LRP1 receptor, which mediates macrophage phagocytosis of healthy or apoptotic ...
  101. [101]
    Senescent cell turnover slows with age providing an explanation for ...
    Dec 2, 2019 · SnCs turn over rapidly in young mice, with a half-life of days, but slow their own removal rate to a half-life of weeks in old mice.Missing: youth | Show results with:youth
  102. [102]
    Immune Clearance of Senescent Cells to Combat Ageing and ...
    Mounting evidence indicates that immunotherapy targeting senescent cells combats ageing and chronic diseases and subsequently extends the healthy lifespan.
  103. [103]
    Senescent cell clearance by the immune system - PubMed Central
    Recent studies suggest several ways through which SCs may be cleared by immune cells. In a healthy immune surveillance scenario, SCs produce factors and ...
  104. [104]
    Review Article Cellular senescence and the host immune system in ...
    A recent report indicates that elevated CD47 expression by senescent cells undermines the ability of macrophages to perform optimal efferocytosis, an event that ...
  105. [105]
    Senescent cells: an emerging target for diseases of ageing - PMC
    Numerous age-related diseases have now been associated with cellular senescence, including atherosclerosis, osteoarthritis, cancer, Alzheimer disease, chronic ...
  106. [106]
    Senolytics decrease senescent cells in humans - NIH
    Senolytics decrease senescent cells in humans: Preliminary report from a clinical trial of Dasatinib plus Quercetin in individuals with diabetic kidney disease.
  107. [107]
    Exercise Prevents Diet-Induced Cellular Senescence in Adipose ...
    ... Tchkonia , Jan M van Deursen , James L Kirkland , Nathan K LeBrasseur. Affiliations. 1 Robert and Arlene Kogod Center on Aging, Mayo ...
  108. [108]
    Biomarkers of cellular senescence and risk of death in humans
    Oct 6, 2023 · It is therefore plausible that biomarkers of senescent cell burden may be prognostic for mortality. In older adults with zero or only one ...
  109. [109]
    Senescence and aging: Causes, consequences, and therapeutic ...
    The role of p16INK4a-positive senescent cells in age-related pathologies has been further confirmed by additional studies using other p16INK4a-based senescence ...
  110. [110]
    Targeting senescence induced by age or chemotherapy with a ...
    Jul 1, 2024 · Senomorphic compounds (also referred to as senostatics) modulate the function and morphology of senescent cells, restoring functionality to ...
  111. [111]
    Targeting cellular senescence with senotherapeutics: senolytics and ...
    Jan 11, 2022 · Senomorphic treatment provides an alternative pharmacological approach to target cellular senescence. Senomorphics suppress the detrimental ...
  112. [112]
    JAK inhibition alleviates the cellular senescence-associated ... - PNAS
    Nov 2, 2015 · JAK inhibition alleviates the cellular senescence-associated secretory phenotype and frailty in old age. Ming Xu, Tamara Tchkonia, Husheng Ding ...
  113. [113]
    BRD4 connects enhancer remodeling to senescence immune ... - NIH
    Consequently, BRD4 inhibition disrupts immune cell mediated targeting and elimination of premalignant senescent cells in vitro and in vivo. Our results identify ...Missing: senomorphic | Show results with:senomorphic
  114. [114]
    Senescent macrophages in tumor: phenotypes, roles, and ... - Nature
    Oct 6, 2025 · One of the classic senomorphic agents, rapamycin—an inhibitor of the mTOR—has been widely employed in studies targeting cellular senescence.
  115. [115]
    Metformin alleviates stress-induced cellular senescence of aging ...
    Metformin treatment of aged-donor ASCs decreased oxidative stress and mitochondrial dysfunction resulting in decreased senescence.
  116. [116]
    Rapamycin inhibits the secretory phenotype of senescent cells ... - NIH
    Rapamycin, an mTOR inhibitor, improves health in several animal models and inhibits cellular senescence in multiple cell types including fibroblasts. For ...
  117. [117]
    Senescent cells as a target for anti-aging interventions
    Mar 19, 2025 · Recently, studies revealed conditioned media and EVs isolated from human mesenchymal stem cells (MSCs) are potent in reducing senescent cell ...
  118. [118]
    SASP Modulation for Cellular Rejuvenation and Tissue Homeostasis
    Senomorphic therapies act by inhibiting SASP production through key signaling pathways such as mTOR, NF-κB, and JAK/STAT. Agents like rapamycin, metformin, and ...Sasp Modulation For Cellular... · 1. Introduction · 4. Sasp And Its Impact On...<|separator|>
  119. [119]
  120. [120]
    Senescence as a therapeutically relevant response to CDK4/6 ... - NIH
    Cyclin-dependent kinases 4 and 6 (CDK4/6) phosphorylate and inhibit retinoblastoma (RB) family proteins. Hyperphosphorylated RB releases E2F transcription ...Missing: papers | Show results with:papers
  121. [121]
  122. [122]
    Cellular Senescence in Acute and Chronic Wound Repair - PMC
    Transiently present senescence is important for skin repair, where it accelerates extracellular matrix formation, limits fibrosis, promotes reepithelialization ...Senescence From... · Senescence In Wound Repair... · Senescence In Chronic Wound...
  123. [123]
    ERC accumulation depletes Sir2 from rDNA and induces cellular ...
    Oct 29, 2025 · We propose a new model of cellular senescence in budding yeast, driven by Sir2 depletion and rDNA instability. Graphical Abstract. Graphical ...
  124. [124]
    Cellular Senescence in Yeast Is Regulated by rDNA Noncoding ...
    Sep 23, 2013 · This suggested that SIR2 mediates aging pathways other than the rDNA instability pathway of aging [10], so we decided to revisit the rDNA ...
  125. [125]
    Senescence, ageing and death of the whole plant - Thomas - 2013
    Nov 23, 2012 · This review considers the relationship between the lifespan of an individual plant and the longevity of its component cells, tissues and organs.
  126. [126]
    Programmed senescence of plant organs | Cell Death & Differentiation
    Dec 1, 1997 · Here, we review the current understanding of the processes that regulate leaf, flower and fruit senescence, emphasizing the role that programmed ...
  127. [127]
    Intestine-specific removal of DAF-2 nearly doubles lifespan ... - Nature
    Oct 25, 2022 · Twenty-nine years following the breakthrough discovery that a single-gene mutation of daf-2 doubles Caenorhabditis elegans lifespan, ...
  128. [128]
    Analysis of long-lived C. elegans daf-2 mutants ... - PubMed Central
    elegans daf-2(m41) mutant and control worms, which also carried a temperature-sensitive fer-15 (fertility) mutation to prevent the production of offspring that ...
  129. [129]
    Hydra, a powerful model for aging studies - Taylor & Francis Online
    Cnidarian Hydra polyps escape senescence, most likely due to the robust activity of their three stem cell populations. These stem cells continuously ...
  130. [130]
    Constant mortality and fertility over age in Hydra - PNAS
    Senescence, the increase in mortality and decline in fertility with age after maturity, was thought to be inevitable for all multicellular species capable ...Sign Up For Pnas Alerts · Discussion · Materials And Methods<|separator|>
  131. [131]
    INK4 locus of the tumor-resistant rodent, the naked mole rat ... - NIH
    The naked mole rat is a longest lived and cancer-resistant rodent. Tumor resistance in the naked mole rat is mediated by signals from the extracellular ...Missing: studies | Show results with:studies
  132. [132]
    Evidence for improved DNA repair in long-lived bowhead whale
    Oct 29, 2025 · Bowhead whale displays attenuated SASP. Most human somatic cells lack telomerase activity and as a result undergo replicative senescence with ...
  133. [133]
    HeLa Cells: Key Discoveries and the Science of Their Immortality
    Sep 16, 2024 · HeLa cells are classed as immortal as they can divide multiple times without dying. This ability comes from the cells possessing an overactive version of the ...
  134. [134]
    Absence of replicative senescence in cultured cells from the short ...
    The turquoise killifish Nothobranchius furzeri has the shortest known lifespan of a vertebrate that can be bred in captivity. The short lived GRZ strain only ...
  135. [135]
    Convergent evolution of a genomic rearrangement may explain ...
    Sep 1, 2021 · Genomic rearrangements and cancer resistance in naked-mole rat. We first identified the instances of genomic rearrangements between a cancer ...
  136. [136]
    DNA repair and anti-cancer mechanisms in the long-lived bowhead ...
    Growth characteristics, cellular senescence, and cell death in the bowhead whale. Most human somatic cells lack telomerase activity and as a result undergo ...
  137. [137]
    Trade-Off between Body Mass and Cancer Resistance in Cetaceans ...
    Jul 14, 2025 · Trade-Off between Body Mass and Cancer Resistance in Cetaceans Is Mediated by Cell Cycle-Related Gene Evolution | Molecular Biology and ...