Your new experience awaits. Try the new design now and help us make it even better

REVIEW article

Front. Neurosci., 01 August 2025

Sec. Neurodegeneration

Volume 19 - 2025 | https://doi.org/10.3389/fnins.2025.1599492

The emerging role of cellular senescence in amyotrophic lateral sclerosis

Xingli TanXingli Tan1Naiyong Gao
Naiyong Gao2*
  • 1School of Basic Medicine Sciences, Shandong Second Medical University, Weifang, China
  • 2Department of Neurology, Linyi People’s Hospital, Linyi, Shandong, China

Cellular senescence is a state of permanent cell cycle arrest and is considered a key contributor to aging and age-related diseases, including amyotrophic lateral sclerosis (ALS). The physiological processes of aging lead to a variety of molecular and cellular phenotypes, and evidence of overlap between ALS and aging-related biomarkers suggests that cell type-specific senescence may be a critical factor in ALS. Senescent microglial cells, astrocytes, and neurons have been detected in ALS patients and animal models. However, while accumulating evidence suggests a potential link between cellular senescence and ALS, this connection remains not yet conclusively established. Importantly, how senescent cells may contribute to the neuropathophysiology of ALS remains largely unknown. Additionally, the growing popularity of anti-aging therapies has highlighted the potential of senescent cell clearance as a promising strategy for treating age-related diseases, including ALS. This review provides an overview of cellular senescence, discusses recent advances in understanding how senescence in different cell types influences ALS pathogenesis, and explores the potential role of anti-senescence therapies in ALS treatment.

1 Introduction

Amyotrophic lateral sclerosis (ALS) is a common neurodegenerative disorder marked by the progressive degeneration of upper and lower motor neurons. It primarily leads to progressive muscle weakness and atrophy, eventually leading to respiratory failure and death within 3 to 5 years (Meyer, 2021; Irwin et al., 2024). Approximately 90% of ALS cases are classified as sporadic ALS (sALS), with the remaining 10% categorized as familial ALS (fALS; Gois et al., 2020). More than 50 genes have been identified as potential contributors to the pathogenesis or modification of ALS, with SOD1, C9orf72, FUS, and TARDBP being the most frequently implicated (Masrori and Van Damme, 2020; Yun and Ha, 2020). The pathogenesis of ALS is complex and currently implicated in excitotoxicity, nucleocytoplasmic transport defects, impaired protein homeostasis, altered RNA metabolism, DNA repair damage, mitochondrial dysfunction, neuroinflammation, axonal transport deficits, and oligodendrocyte dysfunction (Mejzini et al., 2019).

Cellular senescence is defined at the cellular level by a permanent cell cycle arrest, accompanied by alterations in cell shape, epigenetic modifications and secretory phenotype (Csekes and Rackova, 2021; Gorgoulis et al., 2019). Cellular senescence encompasses both replicative senescence (RS) and stress-induced premature senescence (SIPS). RS refers to cellular senescence primarily induced by telomere shortening and dysfunction under physiological conditions (Aravinthan, 2015; Cristofalo et al., 2004; Daios et al., 2022). SIPS refers to cellular senescence triggered by various stress stimuli, such as oxidative stress (Deng et al., 2023), direct and persistent DNA damage (Ou and Schumacher, 2018), activation of tumor suppressors (Liu et al., 2023) and/or oncogenes (Zhu et al., 2020), mitochondrial dysfunction (Miwa et al., 2022), epigenetic dynamics (Zhu et al., 2021), and therapy-induced senescence (TIS; Prasanna et al., 2021). Cellular senescence may arise at various life stages, ranging from embryonic development to adulthood, but it is predominantly linked to the aging process (Gorgoulis et al., 2019; Rhinn et al., 2019). Cellular senescence is a highly regulated process that plays a crucial role in tumor suppression, aging, wound healing, and embryonic development (Calcinotto et al., 2019). Previous studies have confirmed that cell death is a key consequence of cellular senescence (Ogrodnik, 2021). Cellular senescence has beneficial effects, for example, by preventing tumorigenesis through irreversible cell cycle arrest. However, growing evidence suggests that senescent cells accumulate in aging tissues and organs, disrupting normal physiological functions and playing a role in organismal aging and age-related diseases (Hohn et al., 2017; Tacutu et al., 2011; Baker et al., 2011; Gerenu et al., 2017; Sturmlechner et al., 2017).

As a stress response, cellular senescence is associated with degenerative pathologies of aging. This progressive degeneration occurs at the molecular, cellular, tissue, and organ levels (Regulski, 2017). Senescent cells usually display the following key characteristics: loss of proliferative or regenerative capacity, alterations in metabolic function, and resistance to apoptosis (Khosla et al., 2020).

Although the hallmarks of senescent cells have not yet been precisely defined, there is a general consensus on certain key characteristics of senescent cells. Senescent cells become enlarged and flattened morphologically (Khosla et al., 2020; Hernandez-Segura et al., 2018). At the transcriptional level, p16 and p21 are among the most frequently utilized indicators of cellular senescence (Gorgoulis et al., 2019; Gasek et al., 2021). Other markers for identifying senescent cells include elevated activity of senescence-associated β-galactosidase (SA-β-gal), elevated expression of p53, decreased phosphorylation of retinoblastoma protein (pRb), accumulation of lipofuscin in the lysosomes, an increase in senescence-associated DNA damage foci (SDFs), and various nuclear changes, such as the loss of lamin B1 (Figure 1). In addition, senescent cells secrete a characteristic pro-inflammatory cytokine profile known as the senescence-associated secretory phenotype (SASP; Gorgoulis et al., 2019). These characteristics are commonly employed to recognize senescent cells in diverse tissues linked to aging or pathological states. Nonetheless, they are not universally present in all senescent cells and may be inadequate for identifying senescent cells in vivo (Cohn et al., 2023). Therefore, it is recommended to use multiple markers, particularly in vivo, to identify senescent cells.

Figure 1
www.frontiersin.org

Figure 1. The cause and outcome of cell senescence Telomere shortening/dysfunction induced replicative senescence, while oxidative stress, DNA damage, oncogene activation, and chemotherapy-induced stress led to stress-induced premature senescence. Senescent cells showed increased p16, p21, SA-β-gal, and SASP secretion phenotypes, which ultimately led to cell cycle arrest, resistance to apoptosis, and metabolic alterations.

Although cellular senescence is recognized as a key contributor to age-associated diseases (Baker et al., 2011; Ogrodnik et al., 2021), different factors may induce senescence in distinct cell types under different pathological circumstances (Khan et al., 2017). The age of onset of ALS typically ranges from 50 to 75 years. Owing to the growing aging population, the number of ALS patients is rapidly increasing, with an estimated 400,000 individuals worldwide expected to be diagnosed with ALS by 2040 (Chia et al., 2018). The precise etiology and pathogenesis of ALS remain unclear, and effective treatments are not yet available. Research from both human and animal models indicates that cellular senescence is a crucial factor in the progression of various age-related diseases (Gerenu et al., 2017; Sturmlechner et al., 2017; Xu et al., 2018; Schafer et al., 2017). Studies have also linked cellular senescence to the development of ALS (Pandya and Patani, 2020). Transcriptomic analysis of the spinal cord in hSOD1G93A mice (an established mouse model of ALS) demonstrated a connection between cellular senescence and ALS, showing that 90% of age-related spinal cord transcripts were upregulated in ALS (Herskovits et al., 2018). The DNA damage response (DDR) is strongly linked to cellular senescence and can induce senescence in cells (Kang et al., 2015; Jurk et al., 2012). Our previous studies have shown that the DDR remains persistently activated in motor neurons of the hSOD1G93A mice (Wang et al., 2019; Yang et al., 2020).

These findings strongly suggest that cellular senescence mediates the neuropathophysiology of ALS. Compared with other aging-related diseases, the evidence linking cellular senescence to ALS is less established. In this review, we examine the potential roles of cellular senescence in ALS, focusing on different cell types such as microglia, astrocytes, and motor neurons. Finally, we discuss the significance of senescent cell clearance for ALS treatment.

2 Aging of microglia

Recent studies have reported that aging microglia represent a novel therapeutic target for neurodegenerative diseases (Shahidehpour et al., 2024; Antignano et al., 2023). Research involving spatiotemporal RNA sequencing of the mouse brain identified accelerated senescence of microglial cells, particularly in white matter, and suggested that microglia may be the most prone to senescence among brain cells (Hahn et al., 2023). To date, no studies have elucidated why microglia in the white matter are particularly susceptible to aging. The threshold theory of senescent cell accumulation (Chaib et al., 2022) posits that once the quantity of senescent cells in the body exceeds a certain threshold, the immune system and other organs become more susceptible to aging-related diseases (Karin et al., 2019). Although microglia constitute only 5–10% of the total brain cells, the accumulation of senescent microglia may initiate paracrine senescence in surrounding neurons and glial cells through the senescence-associated secretory phenotype (SASP), ultimately contributing to brain-wide aging (Ng et al., 2023). Notably, an analysis of SASP shows that many of its components are biomarkers linked to neuroinflammation, most of which are substances extensively released by microglia (Matsudaira et al., 2023). Furthermore, senescent microglia exhibit altered mTOR signaling and increased oxidative stress (Palmer and Ousman, 2018). The accumulation of senescent microglia impairs their immune roles and communication with other brain cells, potentially playing a role in the initiation and advancement of neurodegenerative disorders (Angelova and Brown, 2019).

Microglia play a critical role in neuroinflammation associated with ALS (Frakes et al., 2014). Neuroinflammation involves the activation of microglia, which can polarize into either the M1 pro-inflammatory phenotype or the M2 anti-inflammatory phenotype in response to different microenvironmental stimuli (Guo et al., 2022). Thus, the role of microglia in neurodegenerative diseases is a double-edged sword. Neuroinflammation may represent a secondary consequence of cellular senescence and also act as a key contributor to brain aging (Rim et al., 2024). Studies have shown that aging of CD4 T cells intensifies neuroinflammation in a late-onset ALS mouse model. Alterations in CD4 T-cell subsets associated with aging were detected, resulting in a higher proportion of effector T cells in the spleen of SOD1G37R mice (Zaccai et al., 2024). Microglia isolated from symptomatic hSOD1G93A mice exhibit characteristics of the SASP, including elevated β-galactosidase activity, increased levels of p16, p53, MMP-1, and nitrotyrosine (Trias et al., 2019). This suggests that microglia senescence may be involved in ALS pathology. However, the mechanisms underlying the involvement of microglia senescence in ALS remain unclear. Genotoxic stress-induced senescent microglia may represent the outcome of microglial activation (Spittau, 2017).

3 Aging of astrocytes

Astrocytes, the predominant glial cells in the central nervous system, are essential for a range of biological functions, including neurotransmitter cycling, synaptogenesis and synaptic elimination, maintenance of the blood–brain barrier, and support for neuronal survival (Vasile et al., 2017). Astrocytes can be classified into two groups based on their response to injury mechanisms. Astrocytes exposed to inflammatory stimuli adopt the A1 phenotype, characterized by the upregulation of genes involved in synapse elimination. Conversely, astrocytes subjected to ischemia adopt the A2 phenotype, marked by the upregulation of genes that promote neurotrophy, repair, and survival (Liddelow and Barres, 2017). Levels of the astrocyte marker glial fibrillary acidic protein (GFAP) are markedly increased in the aging brain, reflecting astrocyte activation and gliosis in the context of neurodegeneration (Verkerke et al., 2021). Studies report that astrocytes in ALS exhibit an A1 reactive phenotype (Clarke et al., 2018), and in aged mice, astrocytes upregulate more A1 reactive genes compared to A2 reactive genes, suggesting that aging is linked to the more harmful A1 astrocyte phenotype (Clarke et al., 2018). Impaired astrocyte function, resulting in the continuous secretion of pro-inflammatory factors like IL-8, IL-1β, IL-6, IL-18, and TNF-α, has been associated with aging and age-related neurodegenerative disorders (Li et al., 2023). Transcriptomic analysis of multiple brain regions in aged mice reveals a downregulation of cholesterol synthesis in aged astrocytes. Cholesterol is a critical component for presynaptic vesicle formation, suggesting that senescent astrocytes may disrupt neuronal synaptic function and predispose to synaptic elimination (Boisvert et al., 2018).

Astrocytes have emerged as critical contributors to ALS pathology through non-cell-autonomous mechanisms, including gain of toxic functions (Nagai et al., 2007) and loss of neuronal support (Tyzack et al., 2017). Growing evidence indicates that astrocyte senescence contributes to the progression of ALS. Δ133p53, a human p53 isoform, is upregulated under low-level ROS stress to support cell survival and delay cellular senescence through upregulating antioxidant genes. Research has shown that Δ133p53 expression is reduced in the brain tissues of patients with ALS (Zhao et al., 2021). Astrocytes derived from iPSCs of patients with sporadic ALS exhibit significantly increased levels of senescence-associated markers (Birger et al., 2019). The brains of ALS patients also exhibit an increased number of senescent astrocytes (Turnquist et al., 2016). Studies have found that glial cells, primarily astrocytes, in the frontal association cortex (FACx) of ALS patients exhibit increased expression of p16 and p21, suggesting that glial cells may contribute to the induction of cellular senescence (Vazquez-Villasenor et al., 2020). Astrocytes exhibit an accelerated onset of senescence in ALS models, followed by a reduced capacity to support motor neurons (Das and Svendsen, 2015). There are numerous similarities between SOD1-overexpressing astrocytes at 150 days of end-stage and wild-type aged astrocytes at 300 days. This reveals striking similarities between ALS and wild-type aging samples (Das and Svendsen, 2015).

Oxidative stress is pivotal in ALS pathogenesis, and the increased vulnerability of senescent astrocytes to oxidative stress may be a key factor in disease progression (Barber and Shaw, 2010). Conversely, oxidative stress, including exposure to hydrogen peroxide, induces a senescent phenotype in astrocytes (Bitto et al., 2010). The interactions between astrocytes and surrounding cells are disrupted during aging, leading to the impairment of neighboring cell types through non-cell-autonomous mechanisms (Lana et al., 2019). The onset of a senescent phenotype in astrocytes can lead to various functional impairments, ultimately compromising their ability to support both themselves and neurons in the context of aging and age-related disease (Bitto et al., 2010). Additionally, astrocytes interact with microglia, the CNS immune cells, influencing their branching and distribution (Lana et al., 2019). However, during aging, these direct interactions are impaired, disrupting microglial morphology and their phagocytic capacity. This dysfunction ultimately leads to the accumulation of toxic pro-inflammatory cellular debris within the central nervous system.

Astrocytes are numerous and functionally diverse, and they undergo significant age-related changes that affect their interactions with neighboring cells, such as neurons and microglia. These alterations ultimately make the central nervous system more susceptible to age-related pathologies and neurodegenerative diseases. If astrocytes in ALS represent a pathological acceleration of the normal aging process, a deeper understanding of astrocyte senescence will provide valuable insights into the pathophysiology of ALS.

4 Aging of motor neurons

The aging of astrocytes and microglia has been documented, and they may disrupt glia–neuron interactions through the SASP, contributing to age-related brain pathologies (Bhat et al., 2012; Safaiyan et al., 2016; Crowe et al., 2016). Neurons are terminally differentiated cells that have left the cell cycle and ceased to proliferate. As a result, they do not undergo typical cellular senescence (Pandya and Patani, 2020). Interestingly, research shows that post-mitotic neurons can also exhibit senescence-associated alterations (Jurk et al., 2012; Piechota et al., 2016). This results in the emergence of senescence-associated neuronal traits, which could expedite neuronal dysfunction and promote the progression of neurodegenerative disorders (Vazquez-Villasenor et al., 2020). Recent studies suggest that neurons experience senescence-like changes when exposed to stress (von Zglinicki et al., 2021; Ohashi et al., 2018), and the senescence of normally aging neurons may impair their viability and increase their vulnerability to additional damage (Pandya and Patani, 2021). Elevated activity of the cellular senescence marker β-galactosidase has been observed in hippocampal and cerebellar granule neurons of aged rats (Geng et al., 2010; Bhanu et al., 2010). Additionally, SASP-related genes are induced, and β-galactosidase activity is elevated in neurons (Ohashi et al., 2018). Lipofuscin aggregates, which are rich in lipids, metals, and misfolded proteins, accumulate in neurons and other post-mitotic, non-proliferative cell types during normal aging (Moreno-Garcia et al., 2018). Furthermore, studies have suggested that the senescence of dopaminergic neurons plays a role in the pathogenesis of Parkinson’s disease (PD; Riessland et al., 2019). Distinct subtypes of neuronal senescence have also been identified in Alzheimer’s disease (AD; Musi et al., 2018; Dehkordi et al., 2021).

In motor neurons of ALS, the expression of p21, a cellular senescence marker, is increased (Vazquez-Villasenor et al., 2020). Neurons derived from induced pluripotent stem cells (iPSCs) of C9orf72+ ALS patients show a significant upregulation of senescence-associated genes (Porterfield et al., 2020). Although not neurotoxic in itself, the ectopic expression of Nav1.8 during aging can make energy-demanding motor neurons more susceptible to neurodegeneration and neuronal pathological progression (Moldovan et al., 2016). The upregulation of matrix metalloproteinases (MMP) during aging may have particular significance in ALS, as overexpression of TDP-43 in neurons accelerates the accumulation of dMMP1 (Azpurua et al., 2018). Both upper and lower motor neurons often exhibit hyperexcitability in the hSOD1G93A mouse and ALS patients (Wainger et al., 2014; Tankisi et al., 2021). Excitotoxicity is characterized by the overactivation of glutamate receptors, leading to neuronal injury or cell death (Dong et al., 2009). Electrophysiological studies in ALS patients have revealed irregularities in sodium and potassium currents, indicating age-associated alterations in membrane depolarization and excitability of motor axons (Bae et al., 2008). Neurons depend on the error-prone non-homologous end joining (NHEJ) pathway for repairing DNA double-strand breaks (DSBs), and they lack the ability to mitigate the consequences of DNA repair mistakes during cell division. As a result, they are especially susceptible to DNA damage and aging (Wang et al., 2021). Research indicates that factors such as high energy demands, elevated DNA damage, the length of axons, error-prone DNA repair processes, increased susceptibility to excitotoxicity, neuronal senescence, and variability in the vulnerability of different motor neuron subtypes may collectively make them more susceptible to aging-related damage (Arnold and Clark, 2019).

Neuronal senescence may therefore represent a critical mechanism underlying the pathogenesis of ALS. Some studies have indicated that as the number of aging motor neurons decreases, the remaining aging motor neurons may be under higher stress (Pandya and Patani, 2020). Normal aging is accompanied by the loss of synaptic input in alpha motor neurons, a shared pathological hallmark with ALS, and leads to mitochondrial dysfunction in motor neurons. Furthermore, the accumulation of DNA damage during the normal aging may be an important risk factor for neurodegenerative diseases and ALS (Ma and Farny, 2023). Therefore, it is possible that normal aging may be a prerequisite for the degeneration of motor neurons in ALS, and aging may make this system vulnerable to the subsequent disease-specific mechanisms of ALS, although further research is needed to clearly solve this problem. Normal aging impacts the number, structure, and functional capacity of motor neurons, suggesting that age-associated changes may contribute significantly to neurodegenerative disorders affecting motor neurons, like ALS (Figure 2).

Figure 2
www.frontiersin.org

Figure 2. The aging of microglia, astrocytes, and motor neurons contributes to the degeneration of motor neurons in ALS.

5 Anti-senescence therapies

Eliminating senescent cells has been reported to alleviate age-related brain diseases (Sikora et al., 2019). In paraquat-induced PD mice, the removal of senescent astrocytes reduced the loss of dopaminergic neurons and improved motor function (Chinta et al., 2018). Similarly, clearing senescent cells has been shown to mitigate chemotherapy-induced peripheral neuropathy (Acklin et al., 2020). Numerous therapeutic studies in animal models have demonstrated the feasibility of delaying aging to extend both lifespan and healthspan (Partridge et al., 2020). In AP20187 transgenic mouse models, systemic clearance of p16-positive senescent cells specifically eliminated senescent microglia and reversed cognitive decline (Zhang et al., 2022). In AD, the removal of senescent cells reduced neuroinflammation, amyloid-β plaque levels, and cognitive deficits (Zhang et al., 2019).

Anti-senescence drugs are compounds designed to target and eliminate senescent cells, thereby reducing the burden of these cells and extending organismal lifespan (Rim et al., 2024). Senolytic drugs can selectively target key proteins and apoptotic signaling molecules, such as PI3K/Akt and mTOR, effectively eliminating senescent cells and diminishing the SASP along with its associated consequences (Dashtmian et al., 2024). For instance, the administration of dasatinib combined with quercetin, where quercetin is a repurposed drug used as a senolytic agent, has been demonstrated to decrease senescent cell accumulation in human adipose tissue and reduce inflammation (Xu et al., 2018). Dasatinib, a broad-spectrum tyrosine kinase inhibitor targeting SRC family members, induces apoptosis of senescent cells by activating caspase-mediated pathways (caspase-3, −7, and −9; Schade et al., 2008). At the cellular level, mTOR inhibition has been shown to reverse senescence traits, including increased granularity, enlarged cell size, β-galactosidase activity, and the spindle-like morphology of fibroblasts (Walters et al., 2016). In a randomized, double-blind, placebo-controlled clinical trial for ALS, rapamycin was found to be safe and well-tolerated. However, further studies are needed to clarify its clinical and biological impacts in ALS (Mandrioli et al., 2023). Furthermore, some anti-senescence drugs that inhibit HSP90 show limited efficacy in preventing disease progression and may even counterproductively endanger degenerating motor neurons (Maximova et al., 2021). Therefore, it is still necessary to further determine the effective targets for the treatment of ALS in order to maximize the therapeutic effect.

During normal aging, cellular function gradually declines, but in ALS, this decline is accelerated. In transgenic hSOD1G93A mice, administration of NMN (a CD38 inhibitor) and NR (a PARP inhibitor) delays aging, promotes stem cell renewal, and extends lifespan (Zhang et al., 2016). In hSOD1G93A mice, resveratrol (an anti-aging compound) prevents motor neuron loss, alleviates muscle atrophy, improves muscle mitochondrial function, and extends survival (Mancuso et al., 2014). In vitro experiments have shown that inhibiting astrocyte senescence reduces the SASP and prevents astrocyte-mediated neurotoxicity (Ungerleider et al., 2021). Therefore, clearing senescent cells or restoring their normal function may be a potential strategy for treating ALS (Maximova et al., 2021).

While preclinical studies showed promising outcomes, clinical translation requires rigorous investigation. Clinical trials must evaluate the efficacy and safety of senolytic drugs in ALS patients, while elucidating how senescence contributes to disease pathology. Identifying senolytics with optimal specificity and safety profiles is essential, particularly those targeting ALS-relevant senescent cells. Large longitudinal studies will determine long-term therapeutic effects. Overcoming these challenges demands interdisciplinary collaboration to develop safe, effective ALS senotherapies.

6 Limitation and future directions

While cellular senescence is a key factor in age-related disorders, such as ALS, the exact triggers of senescence and the mechanisms through which senescent cells drive aging and related diseases are still not fully understood. A major challenge in this area is the detection of senescent cells in vivo and in postmortem tissues, emphasizing the necessity for more precise and sensitive techniques to identify these cells in living organisms. Tools for identifying and tracking senescent cells in vivo have been developed and investigated in tumors, such as the senescent-specific PET probe FPyGal, cell-free DNA (cfDNA) analysis, and fine-tuned nanoparticles for recognizing senescent cells. These advancements provide novel avenues for monitoring senescent cells in vivo in ALS. This review examines the potential contributions of cell type-specific senescence to ALS. Promising outcomes from both animal research and clinical trials support the efficacy of anti-senescence treatments. In models of several aging-related diseases, the pharmacological or genetic elimination of senescent cells has been shown to decelerate aging and mitigate senescence-associated pathophysiological changes. A deeper understanding of the mechanisms driving cellular senescence and the role of senescent cells in ALS pathogenesis could open up new avenues for therapeutic interventions. Additionally, identifying the senescence phenotypes of motor neurons in ALS patients remains a crucial focus of research, especially exploring reliable biomarkers for the senescence of motor neurons. Targeting motor neuron senescence could potentially slow disease progression in its early stages and extend the lifespan of individuals with ALS.

Author contributions

XT: Investigation, Writing – original draft, Conceptualization. NG: Supervision, Funding acquisition, Writing – review & editing.

Funding

The author(s) declare that no financial support was received for the research and/or publication of this article.

Acknowledgments

The authors would like to thank Lu Cui of the Chinese Academy of Agricultural Sciences for assistance and guidance in writing.

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Generative AI statement

The author(s) declare that no Gen AI was used in the creation of this manuscript.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

Acklin, S., Zhang, M., and Du, W. (2020). Depletion of senescent-like neuronal cells alleviates cisplatin-induced peripheral neuropathy in mice. Sci. Rep. 10:14170. doi: 10.1038/s41598-020-71042-6

PubMed Abstract | Crossref Full Text | Google Scholar

Angelova, D. M., and Brown, D. R. (2019). Microglia and the aging brain: are senescent microglia the key to neurodegeneration? J. Neurochem. 151, 676–688. doi: 10.1111/jnc.14860

Crossref Full Text | Google Scholar

Antignano, I., Liu, Y., Offermann, N., and Capasso, M. (2023). Aging microglia. Cell. Mol. Life Sci. 80:126. doi: 10.1007/s00018-023-04775-y

Crossref Full Text | Google Scholar

Aravinthan, A. (2015). Cellular senescence: a hitchhiker's guide. Hum. Cell 28, 51–64. doi: 10.1007/s13577-015-0110-x

Crossref Full Text | Google Scholar

Arnold, W. D., and Clark, B. C. (2019). Faster, higher, farther: outpacing age-related motor neuron losses. J. Physiol. 597, 4867–4868.

Google Scholar

Azpurua, J., Mahoney, R. E., and Eaton, B. A. (2018). Transcriptomics of aged drosophila motor neurons reveals a matrix metalloproteinase that impairs motor function. Aging Cell 17:e12729. doi: 10.1111/acel.12729

Crossref Full Text | Google Scholar

Bae, J. S., Sawai, S., Misawa, S., Kanai, K., Isose, S., Shibuya, K., et al. (2008). Effects of age on excitability properties in human motor axons. Clin. Neurophysiol. 119, 2282–2286. doi: 10.1016/j.clinph.2008.07.005

PubMed Abstract | Crossref Full Text | Google Scholar

Baker, D. J., Wijshake, T., Tchkonia, T., LeBrasseur, N. K., Childs, B. G., van de Sluis, B., et al. (2011). Clearance of p16Ink4a-positive senescent cells delays ageing-associated disorders[J]. Nature 479, 232–236. doi: 10.1038/nature10600

PubMed Abstract | Crossref Full Text | Google Scholar

Barber, S. C., and Shaw, P. J. (2010). Oxidative stress in Als: key role in motor neuron injury and therapeutic target. Free Radic. Biol. Med. 48, 629–641. doi: 10.1016/j.freeradbiomed.2009.11.018

Crossref Full Text | Google Scholar

Bhanu, M. U., Mandraju, R. K., and Bhaskar, C. (2010). Cultured cerebellar granule neurons as an in vitro aging model: topoisomerase Iibeta as an additional biomarker in DNA repair and aging. Toxicol. in Vitro 24, 1935–1945. doi: 10.1016/j.tiv.2010.08.003

PubMed Abstract | Crossref Full Text | Google Scholar

Bhat, R., Crowe, E. P., Bitto, A., Moh, M., Katsetos, C. D., Garcia, F. U., et al. (2012). Astrocyte senescence as a component of Alzheimer's disease. PLoS One 7:e45069. doi: 10.1371/journal.pone.0045069

Crossref Full Text | Google Scholar

Birger, A., Ben-Dor, I., Ottolenghi, M., Turetsky, T., Gil, Y., Sweetat, S., et al. (2019). Human ipsc-derived astrocytes from Als patients with mutated C9orf72 show increased oxidative stress and neurotoxicity. EBioMedicine 50, 274–289. doi: 10.1016/j.ebiom.2019.11.026

Crossref Full Text | Google Scholar

Bitto, A., Sell, C., Crowe, E., Lorenzini, A., Malaguti, M., Hrelia, S., et al. (2010). Stress-induced senescence in human and rodent astrocytes. Exp. Cell Res. 316, 2961–2968. doi: 10.1016/j.yexcr.2010.06.021

Crossref Full Text | Google Scholar

Boisvert, M. M., Erikson, G. A., Shokhirev, M. N., and Allen, N. J. (2018). The aging astrocyte transcriptome from multiple regions of the mouse brain. Cell Rep. 22, 269–285. doi: 10.1016/j.celrep.2017.12.039

Crossref Full Text | Google Scholar

Calcinotto, A., Kohli, J., Zagato, E., Pellegrini, L., Demaria, M., and Alimonti, A. (2019). Cellular senescence: aging, cancer, and injury. Physiol. Rev. 99, 1047–1078. doi: 10.1152/physrev.00020.2018

Crossref Full Text | Google Scholar

Chaib, S., Tchkonia, T., and Kirkland, J. L. (2022). Cellular senescence and senolytics: the path to the clinic. Nat. Med. 28, 1556–1568. doi: 10.1038/s41591-022-01923-y

Crossref Full Text | Google Scholar

Chia, R., Chio, A., and Traynor, B. J. (2018). Novel genes associated with amyotrophic lateral sclerosis: diagnostic and clinical implications. Lancet Neurol. 17, 94–102. doi: 10.1016/S1474-4422(17)30401-5

Crossref Full Text | Google Scholar

Chinta, S. J., Woods, G., Demaria, M., Rane, A., Zou, Y., McQuade, A., et al. (2018). Cellular senescence is induced by the environmental neurotoxin paraquat and contributes to neuropathology linked to Parkinson’s disease. Cell Rep. 22, 930–940. doi: 10.1016/j.celrep.2017.12.092

PubMed Abstract | Crossref Full Text | Google Scholar

Clarke, L. E., Liddelow, S. A., Chakraborty, C., Münch, A. E., Heiman, M., and Barres, B. A. (2018). Normal aging induces A1-like astrocyte reactivity. Proc. Natl. Acad. Sci. USA 115, E1896–E1905. doi: 10.1073/pnas.1800165115

Crossref Full Text | Google Scholar

Cohn, R. L., Gasek, N. S., Kuchel, G. A., and Xu, M. (2023). The heterogeneity of cellular senescence: insights at the single-cell level. Trends Cell Biol. 33, 9–17. doi: 10.1016/j.tcb.2022.04.011

Crossref Full Text | Google Scholar

Cristofalo, V. J., Lorenzini, A., Allen, R. G., Torres, C., and Tresini, M. (2004). Replicative senescence: a critical review. Mech. Ageing Dev. 125, 827–848. doi: 10.1016/j.mad.2004.07.010

Crossref Full Text | Google Scholar

Crowe, E. P., Tuzer, F., Gregory, B. D., Donahue, G., Gosai, S. J., Cohen, J., et al. (2016). Changes in the transcriptome of human astrocytes accompanying oxidative stress-induced senescence. Front. Aging Neurosci. 8:208. doi: 10.3389/fnagi.2016.00208

Crossref Full Text | Google Scholar

Csekes, E., and Rackova, L. (2021). Skin aging, cellular senescence and natural polyphenols. Int. J. Mol. Sci. 22:12641. doi: 10.3390/ijms222312641

Crossref Full Text | Google Scholar

Daios, S., Anogeianaki, A., Kaiafa, G., Kontana, A., Veneti, S., Gogou, C., et al. (2022). Telomere length as a marker of biological aging: a critical review of recent literature. Curr. Med. Chem. 29, 5478–5495. doi: 10.2174/0929867329666220713123750

Crossref Full Text | Google Scholar

Das, M. M., and Svendsen, C. N. (2015). Astrocytes show reduced support of motor neurons with aging that is accelerated in a rodent model of Als. Neurobiol. Aging 36, 1130–1139. doi: 10.1016/j.neurobiolaging.2014.09.020

Crossref Full Text | Google Scholar

Dashtmian, A. R., Darvishi, F. B., and Arnold, W. D. (2024). Chronological and biological aging in amyotrophic lateral sclerosis and the potential of senolytic therapies. Cells 13:928. doi: 10.3390/cells13110928

Crossref Full Text | Google Scholar

Dehkordi, S. K., Walker, J., Sah, E., Bennett, E., Atrian, F., Frost, B., et al. (2021). Profiling senescent cells in human brains reveals neurons with Cdkn2D/p19 and tau neuropathology. Nat. Aging 1, 1107–1116. doi: 10.1038/s43587-021-00142-3

Crossref Full Text | Google Scholar

Deng, Y., Wang, M., He, Y., Liu, F., Chen, L., and Xiong, X. (2023). Cellular senescence: ageing and androgenetic alopecia. Dermatology 239, 533–541. doi: 10.1159/000530681

Crossref Full Text | Google Scholar

Dong, X. X., Wang, Y., and Qin, Z. H. (2009). Molecular mechanisms of excitotoxicity and their relevance to pathogenesis of neurodegenerative diseases. Acta Pharmacol. Sin. 30, 379–387. doi: 10.1038/aps.2009.24

Crossref Full Text | Google Scholar

Frakes, A. E., Ferraiuolo, L., and Haidet-Phillips, A. M. (2014). Microglia induce motor neuron death via the classical Nf-kappaB pathway in amyotrophic lateral sclerosis. Neuron 81, 1009–1023. doi: 10.1016/j.neuron.2014.01.013

Crossref Full Text | Google Scholar

Gasek, N. S., Kuchel, G. A., Kirkland, J. L., and Xu, M. (2021). Strategies for targeting senescent cells in human disease. Nat Aging 1, 870–879. doi: 10.1038/s43587-021-00121-8

PubMed Abstract | Crossref Full Text | Google Scholar

Geng, Y. Q., Guan, J. T., Xu, X. H., and Fu, Y. C. (2010). Senescence-associated Beta-galactosidase activity expression in aging hippocampal neurons. Biochem. Biophys. Res. Commun. 396, 866–869. doi: 10.1016/j.bbrc.2010.05.011

Crossref Full Text | Google Scholar

Gerenu, G., Martisova, E., Ferrero, H., Carracedo, M., Rantamäki, T., Ramirez, M. J., et al. (2017). Modulation of bdnf cleavage by plasminogen-activator inhibitor-1 contributes to Alzheimer's neuropathology and cognitive deficits. Biochim. Biophys. Acta Mol. basis Dis. 1863, 991–1001. doi: 10.1016/j.bbadis.2017.01.023

Crossref Full Text | Google Scholar

Gois, A. M., Mendonça, D., Freire, M., and Santos, J. (2020). In vitro and in vivo models of amyotrophic lateral sclerosis: an updated overview. Brain Res. Bull. 159, 32–43. doi: 10.1016/j.brainresbull.2020.03.012

Crossref Full Text | Google Scholar

Gorgoulis, V., Adams, P. D., Alimonti, A., Bennett, D. C., Bischof, O., Bishop, C., et al. (2019). Cellular senescence: defining a path forward. Cell 179, 813–827. doi: 10.1016/j.cell.2019.10.005

Crossref Full Text | Google Scholar

Guo, S., Wang, H., and Yin, Y. (2022). Microglia polarization from M1 to M2 in neurodegenerative diseases. Front. Aging Neurosci. 14:815347. doi: 10.3389/fnagi.2022.815347

Crossref Full Text | Google Scholar

Hahn, O., Foltz, A. G., and Atkins, M. (2023). Atlas of the aging mouse brain reveals white matter as vulnerable foci. Cell 186, 4117–4133.e22. doi: 10.1016/j.cell.2023.07.027

PubMed Abstract | Crossref Full Text | Google Scholar

Hernandez-Segura, A., Nehme, J., and Demaria, M. (2018). Hallmarks of cellular senescence. Trends Cell Biol. 28, 436–453. doi: 10.1016/j.tcb.2018.02.001

Crossref Full Text | Google Scholar

Herskovits, A. Z., Hunter, T. A., and Maxwell, N. (2018). Sirt1 deacetylase in aging-induced neuromuscular degeneration and amyotrophic lateral sclerosis. Aging Cell 17:e12839. doi: 10.1111/acel.12839

Crossref Full Text | Google Scholar

Hohn, A., Weber, D., and Jung, T. (2017). Happily (n)ever after: aging in the context of oxidative stress, proteostasis loss and cellular senescence. Redox Biol. 11, 482–501. doi: 10.1016/j.redox.2016.12.001

PubMed Abstract | Crossref Full Text | Google Scholar

Irwin, K. E., Sheth, U., Wong, P. C., and Gendron, T. F. (2024). Fluid biomarkers for amyotrophic lateral sclerosis: a review. Mol. Neurodegener. 19:9. doi: 10.1186/s13024-023-00685-6

Crossref Full Text | Google Scholar

Jurk, D., Wang, C., Miwa, S., Maddick, M., Korolchuk, V., Tsolou, A., et al. (2012). Postmitotic neurons develop a p21-dependent senescence-like phenotype driven by a DNA damage response. Aging Cell 11, 996–1004. doi: 10.1111/j.1474-9726.2012.00870.x

Crossref Full Text | Google Scholar

Kang, C., Xu, Q., Martin, T. D., Li, M. Z., Demaria, M., Aron, L., et al. (2015). The DNA damage response induces inflammation and senescence by inhibiting autophagy of Gata4. Science 349:aaa5612. doi: 10.1126/science.aaa5612

Crossref Full Text | Google Scholar

Karin, O., Agrawal, A., Porat, Z., Krizhanovsky, V., and Alon, U. (2019). Senescent cell turnover slows with age providing an explanation for the gompertz law. Nat. Commun. 10:5495. doi: 10.1038/s41467-019-13192-4

Crossref Full Text | Google Scholar

Khan, S. S., Singer, B. D., and Vaughan, D. E. (2017). Molecular and physiological manifestations and measurement of aging in humans. Aging Cell 16, 624–633. doi: 10.1111/acel.12601

Crossref Full Text | Google Scholar

Khosla, S., Farr, J. N., Tchkonia, T., and Kirkland, J. L. (2020). The role of cellular senescence in ageing and endocrine disease. Nat. Rev. Endocrinol. 16, 263–275. doi: 10.1038/s41574-020-0335-y

Crossref Full Text | Google Scholar

Lana, D., Ugolini, F., Wenk, G. L., Giovannini, M. G., Zecchi-Orlandini, S., and Nosi, D. (2019). Microglial distribution, branching, and clearance activity in aged rat hippocampus are affected by astrocyte meshwork integrity: evidence of a novel cell-cell interglial interaction. FASEB J. 33, 4007–4020. doi: 10.1096/fj.201801539R

Crossref Full Text | Google Scholar

Li, X., Li, C., Zhang, W., Wang, Y., Qian, P., and Huang, H. (2023). Inflammation and aging: signaling pathways and intervention therapies. Signal Transduct. Target. Ther. 8:239. doi: 10.1038/s41392-023-01502-8

Crossref Full Text | Google Scholar

Liddelow, S. A., and Barres, B. A. (2017). Reactive astrocytes: production, function, and therapeutic potential. Immunity 46, 957–967. doi: 10.1016/j.immuni.2017.06.006

Crossref Full Text | Google Scholar

Liu, X., Li, X., and Wang, S. (2023). Atoh8 binds Smad3 to induce cellular senescence and prevent Ras-driven malignant transformation. Proc. Natl. Acad. Sci. USA 120:e2086040176. doi: 10.1073/pnas.2208927120

Crossref Full Text | Google Scholar

Ma, Y., and Farny, N. G. (2023). Connecting the dots: neuronal senescence, stress granules, and neurodegeneration. Gene 871:147437. doi: 10.1016/j.gene.2023.147437

Crossref Full Text | Google Scholar

Mancuso, R., Del V, J., and Modol, L. (2014). Resveratrol improves motoneuron function and extends survival in Sod1(G93A) als mice. Neurotherapeutics 11, 419–432. doi: 10.1007/s13311-013-0253-y

Crossref Full Text | Google Scholar

Mandrioli, J., D'amico, R., Zucchi, E., De Biasi, S., Banchelli, F., Martinelli, I., et al. (2023). Randomized, double-blind, placebo-controlled trial of rapamycin in amyotrophic lateral sclerosis. Nat. Commun. 14:4970. doi: 10.1038/s41467-023-40734-8

Crossref Full Text | Google Scholar

Masrori, P., and Van Damme, P. (2020). Amyotrophic lateral sclerosis: a clinical review. Eur. J. Neurol. 27, 1918–1929. doi: 10.1111/ene.14393

Crossref Full Text | Google Scholar

Matsudaira, T., Nakano, S., Konishi, Y., Kawamoto, S., Uemura, K., Kondo, T., et al. (2023). Cellular senescence in white matter microglia is induced during ageing in mice and exacerbates the neuroinflammatory phenotype. Commun. Biol. 6:665. doi: 10.1038/s42003-023-05027-2

Crossref Full Text | Google Scholar

Maximova, A., Werry, E. L., and Kassiou, M. (2021). Senolytics: a novel strategy for neuroprotection in Als? Int. J. Mol. Sci. 22:12078. doi: 10.3390/ijms222112078

Crossref Full Text | Google Scholar

Mejzini, R., Flynn, L. L., Pitout, I. L., Fletcher, S., Wilton, S. D., and Akkari, P. A. (2019). Als genetics, mechanisms, and therapeutics: where are we now? Front. Neurosci. 13:1310. doi: 10.3389/fnins.2019.01310

Crossref Full Text | Google Scholar

Meyer, T. (2021). Amyotrophic lateral sclerosis (Als) - diagnosis, course of disease and treatment options. Dtsch. Med. Wochenschr. 146, 1613–1618.

Google Scholar

Miwa, S., Kashyap, S., Chini, E., and von Zglinicki, T. (2022). Mitochondrial dysfunction in cell senescence and aging. J. Clin. Invest. 132:e158447. doi: 10.1172/JCI158447

Crossref Full Text | Google Scholar

Moldovan, M., Rosberg, M. R., Alvarez, S., Klein, D., Martini, R., and Krarup, C. (2016). Aging-associated changes in motor axon voltage-gated Na(+) channel function in mice. Neurobiol. Aging 39, 128–139. doi: 10.1016/j.neurobiolaging.2015.12.005

Crossref Full Text | Google Scholar

Moreno-Garcia, A., Kun, A., and Calero, O. (2018). An overview of the role of lipofuscin in age-related neurodegeneration. Front. Neurosci. 12:464. doi: 10.3389/fnins.2018.00464

PubMed Abstract | Crossref Full Text | Google Scholar

Musi, N., Valentine, J. M., Sickora, K. R., Baeuerle, E., Thompson, C. S., Shen, Q., et al. (2018). Tau protein aggregation is associated with cellular senescence in the brain. Aging Cell 17:e12840. doi: 10.1111/acel.12840

Crossref Full Text | Google Scholar

Nagai, M., Re, D. B., Nagata, T., Chalazonitis, A., Jessell, T. M., Wichterle, H., et al. (2007). Astrocytes expressing Als-linked mutated Sod1 release factors selectively toxic to motor neurons. Nat. Neurosci. 10, 615–622. doi: 10.1038/nn1876

Crossref Full Text | Google Scholar

Ng, P. Y., Mcneely, T. L., and Baker, D. J. (2023). Untangling senescent and damage-associated microglia in the aging and diseased brain. FEBS J. 290, 1326–1339. doi: 10.1111/febs.16315

Crossref Full Text | Google Scholar

Ogrodnik, M. (2021). Cellular aging beyond cellular senescence: markers of senescence prior to cell cycle arrest in vitro and in vivo. Aging Cell 20:e13338. doi: 10.1111/acel.13338

Crossref Full Text | Google Scholar

Ogrodnik, M., Evans, S. A., Fielder, E., Victorelli, S., Kruger, P., Salmonowicz, H., et al. (2021). Whole-body senescent cell clearance alleviates age-related brain inflammation and cognitive impairment in mice. Aging Cell 20:e13296. doi: 10.1111/acel.13296

Crossref Full Text | Google Scholar

Ohashi, M., Korsakova, E., Allen, D., Lee, P., Fu, K., Vargas, B. S., et al. (2018). Loss of Mecp2 leads to activation of P53 and neuronal senescence. Stem Cell Reports 10, 1453–1463. doi: 10.1016/j.stemcr.2018.04.001

Crossref Full Text | Google Scholar

Ou, H. L., and Schumacher, B. (2018). DNA damage responses and p53 in the aging process. Blood 131, 488–495. doi: 10.1182/blood-2017-07-746396

Crossref Full Text | Google Scholar

Palmer, A. L., and Ousman, S. S. (2018). Astrocytes and aging. Front. Aging Neurosci. 10:337. doi: 10.3389/fnagi.2018.00337

Crossref Full Text | Google Scholar

Pandya, V. A., and Patani, R. (2020). Decoding the relationship between ageing and amyotrophic lateral sclerosis: a cellular perspective. Brain 143, 1057–1072. doi: 10.1093/brain/awz360

Crossref Full Text | Google Scholar

Pandya, V. A., and Patani, R. (2021). Region-specific vulnerability in neurodegeneration: lessons from normal ageing. Ageing Res. Rev. 67:101311. doi: 10.1016/j.arr.2021.101311

Crossref Full Text | Google Scholar

Partridge, L., Fuentealba, M., and Kennedy, B. K. (2020). The quest to slow ageing through drug discovery. Nat. Rev. Drug Discov. 19, 513–532. doi: 10.1038/s41573-020-0067-7

Crossref Full Text | Google Scholar

Piechota, M., Sunderland, P., and Wysocka, A. (2016). Is senescence-associated beta-galactosidase a marker of neuronal senescence? Oncotarget 7, 81099–81109. doi: 10.3389/fcell.2021.623175

Crossref Full Text | Google Scholar

Porterfield, V., Khan, S. S., Foff, E. P., Koseoglu, M. M., Blanco, I. K., Jayaraman, S., et al. (2020). A three-dimensional dementia model reveals spontaneous cell cycle re-entry and a senescence-associated secretory phenotype. Neurobiol. Aging 90, 125–134. doi: 10.1016/j.neurobiolaging.2020.02.011

Crossref Full Text | Google Scholar

Prasanna, P. G., Citrin, D. E., Hildesheim, J., Ahmed, M. M., Venkatachalam, S., Riscuta, G., et al. (2021). Therapy-induced senescence: opportunities to improve anticancer therapy. J. Natl. Cancer Inst. 113, 1285–1298. doi: 10.1093/jnci/djab064

Crossref Full Text | Google Scholar

Regulski, M. J. (2017). Cellular senescence: what, why, and how. Wounds 29, 168–174.

Google Scholar

Rhinn, M., Ritschka, B., and Keyes, W. M. (2019). Cellular senescence in development, regeneration and disease. Development 146:dev151837. doi: 10.1242/dev.151837

Crossref Full Text | Google Scholar

Riessland, M., Kolisnyk, B., and Kim, T. W. (2019). Loss of Satb1 induces p21-dependent cellular senescence in post-mitotic dopaminergic neurons. Cell Stem Cell 25, 514–530.e8. doi: 10.1016/j.stem.2019.08.013

PubMed Abstract | Crossref Full Text | Google Scholar

Rim, C., You, M. J., Nahm, M., and Kwon, M. S. (2024). Emerging role of senescent microglia in brain aging-related neurodegenerative diseases. Transl Neurodegener 13:10. doi: 10.1186/s40035-024-00402-3

PubMed Abstract | Crossref Full Text | Google Scholar

Safaiyan, S., Kannaiyan, N., Snaidero, N., Brioschi, S., Biber, K., Yona, S., et al. (2016). Age-related myelin degradation burdens the clearance function of microglia during aging. Nat. Neurosci. 19, 995–998. doi: 10.1038/nn.4325

Crossref Full Text | Google Scholar

Schade, A. E., Schieven, G. L., Townsend, R., Jankowska, A. M., Susulic, V., Zhang, R., et al. (2008). Dasatinib, a small-molecule protein tyrosine kinase inhibitor, inhibits T-cell activation and proliferation. Blood 111, 1366–1377. doi: 10.1182/blood-2007-04-084814

Crossref Full Text | Google Scholar

Schafer, M. J., White, T. A., and Iijima, K. (2017). Cellular senescence mediates fibrotic pulmonary disease. Nat. Commun. 8:14532. doi: 10.1038/ncomms14532

PubMed Abstract | Crossref Full Text | Google Scholar

Shahidehpour, R. K., Nelson, P. T., Katsumata, Y., and Bachstetter, A. D. (2024). Exploring the link between dystrophic microglia and the spread of Alzheimer's neuropathology. Brain 148, 89–101. doi: 10.1093/brain/awae258

PubMed Abstract | Crossref Full Text | Google Scholar

Sikora, E., Bielak-Zmijewska, A., and Mosieniak, G. (2019). Targeting normal and cancer senescent cells as a strategy of senotherapy. Ageing Res. Rev. 55:100941. doi: 10.1016/j.arr.2019.100941

Crossref Full Text | Google Scholar

Spittau, B. (2017). Aging microglia-phenotypes, functions and implications for age-related neurodegenerative diseases. Front. Aging Neurosci. 9:194. doi: 10.3389/fnagi.2017.00194

Crossref Full Text | Google Scholar

Sturmlechner, I., Durik, M., Sieben, C. J., Baker, D. J., and van Deursen, J. M. (2017). Cellular senescence in renal ageing and disease. Nat. Rev. Nephrol. 13, 77–89. doi: 10.1038/nrneph.2016.183

PubMed Abstract | Crossref Full Text | Google Scholar

Tacutu, R., Budovsky, A., Yanai, H., and Fraifeld, V. E. (2011). Molecular links between cellular senescence, longevity and age-related diseases - a systems biology perspective. Aging (Albany NY) 3, 1178–1191. doi: 10.18632/aging.100413

Crossref Full Text | Google Scholar

Tankisi, H., Nielsen, C. S., Howells, J., Cengiz, B., Samusyte, G., Koltzenburg, M., et al. (2021). Early diagnosis of amyotrophic lateral sclerosis by threshold tracking and conventional transcranial magnetic stimulation. Eur. J. Neurol. 28, 3030–3039. doi: 10.1111/ene.15010

Crossref Full Text | Google Scholar

Trias, E., Beilby, P. R., Kovacs, M., et al. (2019). Emergence of microglia bearing senescence markers during paralysis progression in a rat model of inherited Als. Front. Aging Neurosci. 11:42. doi: 10.3389/fnagi.2019.00042

PubMed Abstract | Crossref Full Text | Google Scholar

Turnquist, C., Horikawa, I., Foran, E., Major, E. O., Vojtesek, B., Lane, D. P., et al. (2016). P53 isoforms regulate astrocyte-mediated neuroprotection and neurodegeneration. Cell Death Differ. 23, 1515–1528. doi: 10.1038/cdd.2016.37

Crossref Full Text | Google Scholar

Tyzack, G. E., Hall, C. E., Sibley, C. R., Cymes, T., Forostyak, S., Carlino, G., et al. (2017). A neuroprotective astrocyte state is induced by neuronal signal EphB1 but fails in ALS models. Nat. Commun. 8:1164. doi: 10.1038/s41467-017-01283-z

Crossref Full Text | Google Scholar

Ungerleider, K., Beck, J., Lissa, D., Turnquist, C., Horikawa, I., Harris, B. T., et al. (2021). Astrocyte senescence and Sasp in neurodegeneration: tau joins the loop. Cell Cycle 20, 752–764. doi: 10.1080/15384101.2021.1909260

Crossref Full Text | Google Scholar

Vasile, F., Dossi, E., and Rouach, N. (2017). Human astrocytes: structure and functions in the healthy brain. Brain Struct. Funct. 222, 2017–2029. doi: 10.1007/s00429-017-1383-5

Crossref Full Text | Google Scholar

Vazquez-Villasenor, I., Garwood, C. J., and Heath, P. R. (2020). Expression of p16 and p21 in the frontal association cortex of Als/Mnd brains suggests neuronal cell cycle dysregulation and astrocyte senescence in early stages of the disease. Neuropathol. Appl. Neurobiol. 46, 171–185. doi: 10.1111/nan.12559

PubMed Abstract | Crossref Full Text | Google Scholar

Verkerke, M., Hol, E. M., and Middeldorp, J. (2021). Physiological and pathological ageing of astrocytes in the human brain. Neurochem. Res. 46, 2662–2675. doi: 10.1007/s11064-021-03256-7

Crossref Full Text | Google Scholar

von Zglinicki, T., Wan, T., and Miwa, S. (2021). Senescence in post-mitotic cells: a driver of aging? Antioxid. Redox Signal. 34, 308–323. doi: 10.1089/ars.2020.8048

Crossref Full Text | Google Scholar

Wainger, B. J., Kiskinis, E., Mellin, C., Wiskow, O., Han, S. S. W., Sandoe, J., et al. (2014). Intrinsic membrane hyperexcitability of amyotrophic lateral sclerosis patient-derived motor neurons. Cell Rep. 7, 1–11. doi: 10.1016/j.celrep.2014.03.019

PubMed Abstract | Crossref Full Text | Google Scholar

Walters, H. E., Deneka-Hannemann, S., and Cox, L. S. (2016). Reversal of phenotypes of cellular senescence by pan-mtor inhibition. Aging (Albany NY) 8, 231–244. doi: 10.18632/aging.100872

Crossref Full Text | Google Scholar

Wang, H., Kodavati, M., Britz, G. W., and Hegde, M. L. (2021). DNA damage and repair deficiency in ALS/FTD-associated neurodegeneration: from molecular mechanisms to therapeutic implication. Front. Mol. Neurosci. 14:784361. doi: 10.3389/fnmol.2021.784361

Crossref Full Text | Google Scholar

Wang, X. D., Zhu, M. W., Shan, D., Wang, S. Y., Yin, X., Yang, Y. Q., et al. (2019). Spy1, a unique cell cycle regulator, alters viability in ALS motor neurons and cell lines in response to mutant Sod1-induced DNA damage. DNA Repair (Amst) 74, 51–62. doi: 10.1016/j.dnarep.2018.12.005

Crossref Full Text | Google Scholar

Xu, M., Pirtskhalava, T., Farr, J. N., Weigand, B. M., Palmer, A. K., Weivoda, M. M., et al. (2018). Senolytics improve physical function and increase lifespan in old age. Nat. Med. 24, 1246–1256. doi: 10.1038/s41591-018-0092-9

Crossref Full Text | Google Scholar

Yang, Y. Q., Zheng, Y. H., Zhang, C. T., Liang, W. W., Wang, S. Y., Wang, X. D., et al. (2020). Wild-type p53-induced phosphatase 1 Down-regulation promotes apoptosis by activating the DNA damage-response pathway in amyotrophic lateral sclerosis. Neurobiol. Dis. 134:104648. doi: 10.1016/j.nbd.2019.104648

Crossref Full Text | Google Scholar

Yun, Y., and Ha, Y. (2020). Crispr/Cas9-mediated gene correction to understand Als. Int. J. Mol. Sci. 21:3801. doi: 10.3390/ijms21113801

Crossref Full Text | Google Scholar

Zaccai, S., Nemirovsky, A., Lerner, L., Alfahel, L., Eremenko, E., Israelson, A., et al. (2024). Cd4 t-cell aging exacerbates neuroinflammation in a late-onset mouse model of amyotrophic lateral sclerosis. J. Neuroinflammation 21:17. doi: 10.1186/s12974-023-03007-1

Crossref Full Text | Google Scholar

Zhang, P., Kishimoto, Y., and Grammatikakis, I. (2019). Senolytic therapy alleviates Abeta-associated oligodendrocyte progenitor cell senescence and cognitive deficits in an Alzheimer's disease model. Nat. Neurosci. 22, 719–728. doi: 10.1038/s41593-019-0372-9

Crossref Full Text | Google Scholar

Zhang, X., Pearsall, V. M., Carver, C. M., Atkinson, E. J., Clarkson, B. D. S., Grund, E. M., et al. (2022). Rejuvenation of the aged brain immune cell landscape in mice through p16-positive senescent cell clearance. Nat. Commun. 13:5671. doi: 10.1038/s41467-022-33226-8

Crossref Full Text | Google Scholar

Zhang, H., Ryu, D., and Wu, Y. (2016). Nad(+) repletion improves mitochondrial and stem cell function and enhances life span in mice. Science 352, 1436–1443. doi: 10.1126/science.aaf2693

Crossref Full Text | Google Scholar

Zhao, T., Ye, S., and Tang, Z. (2021). Loss-of-function of p53 isoform Delta113p53 accelerates brain aging in zebrafish. Cell Death Dis. 12:151. doi: 10.1038/s41419-021-03438-9

PubMed Abstract | Crossref Full Text | Google Scholar

Zhu, H., Blake, S., Kusuma, F. K., Pearson, R. B., Kang, J., and Chan, K. T. (2020). Oncogene-induced senescence: from biology to therapy. Mech. Ageing Dev. 187:111229. doi: 10.1016/j.mad.2020.111229

Crossref Full Text | Google Scholar

Zhu, X., Chen, Z., Shen, W., Huang, G., Sedivy, J. M., Wang, H., et al. (2021). Inflammation, epigenetics, and metabolism converge to cell senescence and ageing: the regulation and intervention. Signal Transduct. Target. Ther. 6:245. doi: 10.1038/s41392-021-00646-9

Crossref Full Text | Google Scholar

Keywords: cellular senescence, amyotrophic lateral sclerosis, aging, anti-senescence, aging of motor neurons

Citation: Tan X and Gao N (2025) The emerging role of cellular senescence in amyotrophic lateral sclerosis. Front. Neurosci. 19:1599492. doi: 10.3389/fnins.2025.1599492

Received: 25 March 2025; Accepted: 20 May 2025;
Published: 01 August 2025.

Edited by:

Daniela Ferrari, University of Milano-Bicocca, Italy

Reviewed by:

Jerry Lorren Dominic, Jackson Memorial Hospital, United States
Anna Sarnowska, Polish Academy of Sciences, Poland

Copyright © 2025 Tan and Gao. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Naiyong Gao, VHp4aW42Njc3QDE2My5jb20=

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.