Skip to content

This report is written by MaltSci based on the latest literature and research findings


What are the hallmarks of aging?

Abstract

Aging is a complex biological phenomenon marked by a gradual decline in physiological functions, leading to increased vulnerability to age-related diseases. Understanding the biological mechanisms of aging is essential for enhancing healthspan and developing therapeutic interventions. This review focuses on the hallmarks of aging, originally proposed by López-Otín et al. and subsequently expanded, which include genomic instability, telomere attrition, epigenetic alterations, loss of proteostasis, deregulated nutrient sensing, mitochondrial dysfunction, cellular senescence, stem cell exhaustion, altered intercellular communication, chronic inflammation, and dysbiosis. Each hallmark is intricately linked, influencing and exacerbating one another, thereby complicating the aging process. For instance, genomic instability can arise from telomere shortening and epigenetic changes, while mitochondrial dysfunction can further contribute to cellular senescence. This review discusses the mechanisms underlying each hallmark, their impact on aging and disease, and the potential for therapeutic targeting. By synthesizing current knowledge, the review aims to inspire future research endeavors that can lead to effective anti-aging interventions, ultimately promoting healthy longevity.

Outline

This report will discuss the following questions.

  • 1 Introduction
  • 2 Genomic Instability
    • 2.1 Mechanisms of Genomic Instability
    • 2.2 Impact on Aging and Disease
  • 3 Telomere Attrition
    • 3.1 Role of Telomeres in Cellular Aging
    • 3.2 Telomere Length and Age-Related Diseases
  • 4 Epigenetic Alterations
    • 4.1 Changes in DNA Methylation Patterns
    • 4.2 Epigenetic Reprogramming and Aging
  • 5 Loss of Proteostasis
    • 5.1 Protein Folding and Quality Control
    • 5.2 Consequences of Proteostasis Failure
  • 6 Deregulated Nutrient Sensing
    • 6.1 Insulin/IGF-1 Signaling Pathway
    • 6.2 Nutrient Sensing and Longevity
  • 7 Mitochondrial Dysfunction
    • 7.1 Mitochondrial Biogenesis and Dynamics
    • 7.2 Role in Aging and Metabolic Disorders
  • 8 Cellular Senescence
    • 8.1 Mechanisms of Cellular Senescence
    • 8.2 Impact on Tissue Function and Aging
  • 9 Stem Cell Exhaustion
    • 9.1 Role of Stem Cells in Tissue Homeostasis
    • 9.2 Consequences of Stem Cell Exhaustion in Aging
  • 10 Summary

1 Introduction

Aging is a multifaceted biological phenomenon characterized by a gradual decline in physiological functions, leading to increased vulnerability to various age-related diseases such as cancer, cardiovascular disorders, and neurodegenerative conditions [1]. As the global population ages, understanding the biological mechanisms underlying aging has become paramount, not only to enhance healthspan—the period of life spent in good health—but also to devise effective therapeutic interventions that can mitigate the impacts of aging [2]. This review focuses on the hallmarks of aging, a concept that encapsulates the diverse molecular and cellular processes that contribute to the aging trajectory.

The identification of these hallmarks has evolved significantly since their initial proposal in 2013 by López-Otín et al., who delineated nine key hallmarks: genomic instability, telomere attrition, epigenetic alterations, loss of proteostasis, deregulated nutrient sensing, mitochondrial dysfunction, cellular senescence, stem cell exhaustion, and altered intercellular communication [1]. More recent advancements have expanded this list to include additional hallmarks such as chronic inflammation and dysbiosis, highlighting the interconnectedness of various biological processes that drive aging [2]. Understanding these hallmarks not only elucidates the aging process but also identifies potential targets for therapeutic interventions aimed at prolonging healthspan and reducing the burden of age-associated diseases [3].

Current research underscores the complexity of aging, revealing that these hallmarks do not operate in isolation but rather interact in multifaceted ways that can influence one another [4]. For instance, mitochondrial dysfunction may exacerbate genomic instability, while cellular senescence can impact tissue function and contribute to stem cell exhaustion [5]. This intricate interplay necessitates a holistic approach to aging research, one that considers the cumulative effects of these hallmarks on the aging organism [6].

The organization of this review will follow the structure of the hallmarks of aging, beginning with an in-depth exploration of genomic instability and its mechanisms, followed by a discussion on telomere attrition and its implications for cellular aging. Subsequent sections will address epigenetic alterations, loss of proteostasis, deregulated nutrient sensing, mitochondrial dysfunction, cellular senescence, and stem cell exhaustion. Each section will not only define the hallmark but will also discuss its role in aging and its potential as a therapeutic target. The final section will synthesize these findings, emphasizing the need for further research to develop effective anti-aging interventions and improve the quality of life for the aging population [7].

In summary, this review aims to provide a comprehensive overview of the hallmarks of aging, emphasizing their significance in understanding the aging process and their potential as targets for therapeutic strategies. By synthesizing current knowledge in this field, we hope to contribute to the ongoing discourse on aging and inspire future research endeavors aimed at promoting healthy longevity.

2 Genomic Instability

2.1 Mechanisms of Genomic Instability

Aging is characterized by a progressive loss of physiological integrity, leading to impaired function and increased vulnerability to various diseases. One of the primary hallmarks of aging is genomic instability, which encompasses a range of mutational alterations in the genome, including point mutations, chromosomal rearrangements, and loss of genomic integrity. This instability is not only a marker of aging but also contributes significantly to the aging process itself.

Genomic instability arises from several mechanisms, primarily due to the accumulation of DNA damage over time. This damage can result from both endogenous factors, such as oxidative stress and replication errors, and exogenous factors, including environmental mutagens and radiation. The failure of DNA repair mechanisms exacerbates this instability, leading to an increased mutation load in somatic cells, which is a key contributor to age-related diseases such as cancer, neurodegenerative disorders, and cardiovascular diseases [8].

A critical aspect of genomic instability is its interrelationship with other hallmarks of aging. For instance, telomere attrition—a process where protective caps on the ends of chromosomes shorten with each cell division—can lead to genomic instability by triggering DNA damage responses that ultimately affect cellular senescence and stem cell exhaustion [2]. Furthermore, epigenetic alterations, including changes in DNA methylation and histone modifications, can disrupt normal gene regulation, contributing to genomic instability and age-related functional decline [9].

The mechanisms underlying genomic instability also include impaired DNA repair pathways. As individuals age, the efficiency of various DNA repair systems, such as nucleotide excision repair and mismatch repair, diminishes. This decline leads to an accumulation of unrepaired DNA lesions, further promoting genomic instability [10]. Additionally, the activation of the DNA damage checkpoint can interfere with normal cell cycle progression, contributing to cellular dysfunction and senescence [11].

In the context of specific conditions, such as Cockayne Syndrome, genomic instability manifests through distinct pathophysiological mechanisms, highlighting the interplay between DNA damage accumulation, transcriptional dysregulation, and mitochondrial dysfunction [12]. These interconnections emphasize the complexity of aging and the need for comprehensive research to elucidate the molecular pathways that link genomic instability to age-related phenotypes.

In summary, genomic instability is a multifaceted hallmark of aging that results from various mechanisms, including DNA damage accumulation, impaired repair processes, and epigenetic changes. Understanding these mechanisms is crucial for developing therapeutic strategies aimed at mitigating the effects of aging and promoting healthier aging outcomes.

2.2 Impact on Aging and Disease

Aging is a complex biological process characterized by a progressive decline in physiological integrity, leading to impaired function and increased vulnerability to various diseases. Among the established hallmarks of aging, genomic instability is particularly significant. It refers to the increased susceptibility of the genome to damage and alterations, which accumulate over time and contribute to the aging phenotype and the onset of age-related diseases.

The concept of genomic instability encompasses several critical mechanisms, including DNA damage, the accumulation of mutations, and the failure of DNA repair pathways. As individuals age, their somatic cells are exposed to numerous sources of DNA damage, such as reactive oxygen species, environmental mutagens, and replication errors. Over time, the efficiency of DNA repair mechanisms declines, leading to an increased mutation burden within the genome. This accumulation of genetic damage is a central feature of aging and is believed to be a primary driver of various age-associated diseases, including cancer, cardiovascular disorders, and neurodegenerative diseases [1][2].

Research has shown that genomic instability can manifest in various ways, including telomere attrition, which results from the progressive shortening of telomeres during cell division. Telomeres are protective caps at the ends of chromosomes that prevent genomic degradation; their shortening is directly correlated with cellular aging and the onset of senescence [13][14]. Furthermore, genomic instability is associated with the accumulation of somatic mutations in key regulatory genes, which can disrupt cellular homeostasis and promote the development of diseases [15].

The impact of genomic instability on aging is multifaceted. It contributes to cellular senescence, a state in which cells lose their ability to proliferate and function properly. Senescent cells can secrete pro-inflammatory factors, contributing to a chronic inflammatory state that exacerbates tissue dysfunction and accelerates aging [16]. Moreover, genomic instability is linked to the exhaustion of stem cell populations, which are essential for tissue regeneration and repair. As stem cells accumulate genetic damage, their regenerative capacity diminishes, leading to a decline in tissue function [15].

Additionally, the interplay between genomic instability and other hallmarks of aging, such as epigenetic alterations and mitochondrial dysfunction, further complicates the aging process. Epigenetic changes can influence gene expression patterns and contribute to the phenotypic manifestations of aging, while mitochondrial dysfunction can exacerbate oxidative stress and genomic damage [9][16].

In summary, genomic instability is a critical hallmark of aging that significantly impacts the aging process and the development of age-related diseases. It encompasses a range of mechanisms that lead to the accumulation of genetic damage, cellular senescence, and the decline of regenerative capacity, ultimately contributing to the physiological decline associated with aging and increasing susceptibility to various diseases. Understanding these mechanisms provides valuable insights into potential therapeutic interventions aimed at mitigating the effects of aging and promoting healthier lifespan.

3 Telomere Attrition

3.1 Role of Telomeres in Cellular Aging

Aging is a complex physiological phenomenon characterized by a progressive decline in cellular and organismal function, culminating in increased susceptibility to various diseases. Central to the understanding of aging are the "hallmarks of aging," a concept that identifies key biological processes contributing to this decline. According to López-Otín et al. (2023), the twelve hallmarks of aging include: genomic instability, telomere attrition, epigenetic alterations, loss of proteostasis, disabled macroautophagy, deregulated nutrient-sensing, mitochondrial dysfunction, cellular senescence, stem cell exhaustion, altered intercellular communication, chronic inflammation, and dysbiosis. These hallmarks are interrelated and can be therapeutically targeted to potentially decelerate, stop, or reverse aging processes [2].

Telomere attrition, one of these hallmarks, refers to the progressive shortening of telomeres, the protective DNA sequences located at the ends of chromosomes. Telomeres play a crucial role in maintaining genomic stability by preventing the ends of chromosomes from being recognized as DNA breaks, which could otherwise trigger unwanted repair mechanisms leading to genomic instability [17]. With each cell division, telomeres shorten due to the end-replication problem, which results in cellular senescence or apoptosis when they become critically short [18].

The role of telomeres in cellular aging is profound. As telomeres shorten, they signal cellular aging and contribute to the functional decline of tissues and organs. This process is associated with various age-related diseases, including cardiovascular diseases, cancer, and neurodegeneration [19][20]. The cellular senescence that results from telomere attrition leads to a loss of regenerative capacity in tissues, which is a hallmark of aging [21].

Moreover, telomere length has been proposed as a biomarker for biological aging, reflecting the cumulative effects of genetic, lifestyle, and environmental factors. Studies have shown that lifestyle factors such as physical activity and nutrition can influence telomere length, suggesting potential interventions to mitigate aging [22].

Telomere dysfunction also has implications for gene expression, particularly in subtelomeric regions, where age-related differentially expressed genes have been identified. This suggests that telomere attrition not only leads to cellular senescence but may also alter the expression of genes crucial for maintaining cellular functions [20].

In summary, telomere attrition is a critical hallmark of aging that contributes to cellular senescence and the functional decline associated with aging. The interplay between telomere dynamics and the various hallmarks of aging underscores the importance of telomeres in understanding the biological mechanisms underlying aging and age-related diseases. As research continues to uncover the complexities of telomere biology, it opens new avenues for therapeutic interventions aimed at promoting healthy aging and extending lifespan [17][19].

Aging is characterized by a series of biological changes that lead to a decline in physiological functions and an increased risk of age-related diseases. The concept of "hallmarks of aging" encompasses various interconnected mechanisms that contribute to this process. According to López-Otín et al. (2023), the hallmarks of aging include twelve distinct features: genomic instability, telomere attrition, epigenetic alterations, loss of proteostasis, disabled macroautophagy, deregulated nutrient-sensing, mitochondrial dysfunction, cellular senescence, stem cell exhaustion, altered intercellular communication, chronic inflammation, and dysbiosis[2].

Telomere attrition, specifically, refers to the progressive shortening of telomeres, which are protective caps located at the ends of chromosomes. This shortening occurs with each cell division due to the end replication problem and is closely associated with various age-related disorders, including neurodegenerative diseases and cancer[23]. The biological implications of telomere shortening extend beyond mere cellular aging; they play a significant role in the onset and progression of diseases associated with aging. For instance, telomere length (TL) has emerged as a potential biomarker for aging, as its attrition correlates with cellular senescence and the pathophysiology of age-related diseases[24].

Research has demonstrated that telomere attrition is linked to inflammation and other biological stressors. Baylis et al. (2014) conducted a longitudinal study that found faster telomere attrition was associated with lower grip strength, a marker of physical aging. However, this association was entirely attenuated when adjusted for the burden of systemic inflammation, indicating that inflammaging might drive telomere attrition and partly explain the relationships observed between TL and aging markers[25]. This highlights the intricate interplay between telomere biology and inflammatory processes, suggesting that managing inflammation could potentially mitigate the effects of telomere shortening.

Furthermore, a systematic review and meta-analysis involving 414 study samples indicated a significant negative correlation between TL and chronological age, revealing a non-linear shortening trend across the human lifespan[26]. The findings emphasized the importance of methodological considerations when assessing TL as an aging biomarker, as various biological and methodological factors can influence the observed relationships.

In summary, telomere attrition is a critical hallmark of aging, with significant implications for age-related diseases. Understanding the mechanisms underlying telomere shortening and its association with systemic inflammation and other biological processes is essential for developing potential therapeutic strategies aimed at mitigating the effects of aging and promoting healthy longevity[27]. The exploration of telomere biology in the context of aging continues to be a vital area of research, as it holds promise for interventions that may delay or prevent age-associated diseases[28].

4 Epigenetic Alterations

4.1 Changes in DNA Methylation Patterns

Aging is characterized by a series of biological changes that contribute to the gradual decline in physiological function and increased susceptibility to diseases. One of the critical hallmarks of aging is epigenetic alteration, particularly involving changes in DNA methylation patterns.

Epigenetic alterations refer to modifications that do not change the underlying DNA sequence but affect gene expression and cellular function. Among these, DNA methylation—specifically the addition of methyl groups to cytosine residues in CpG dinucleotides—plays a pivotal role. Research has shown that DNA methylation patterns change significantly with age, a phenomenon referred to as "epigenetic drift." This drift is characterized by global hypomethylation and localized hypermethylation, which can impact genomic stability and contribute to the aging process [29].

In humans, it has been established that specific age-related changes in DNA methylation can serve as reliable biomarkers for biological age. For instance, studies have identified certain CpG sites whose methylation levels correlate strongly with chronological age, allowing for the development of epigenetic clocks that can predict biological aging with high accuracy [30]. In particular, a comprehensive analysis has revealed that a small number of specific CpG sites can predict age with a mean absolute deviation from chronological age of less than five years [30].

Moreover, the aging process is also associated with increased variability in DNA methylation across individuals, suggesting that while the average pattern of methylation changes with age, there is significant inter-individual variability [31]. This variability can influence susceptibility to age-related diseases and overall health outcomes.

Recent studies highlight that the age-related changes in DNA methylation are not merely passive consequences of aging but may actively contribute to the aging process itself. For example, interventions such as caloric restriction and pharmacological treatments have been shown to reverse some age-related DNA methylation changes, suggesting that these epigenetic alterations are reversible and can be targeted for therapeutic purposes [32].

In summary, the hallmark of aging through epigenetic alterations, particularly changes in DNA methylation patterns, reflects a complex interplay of biological mechanisms that underlie the aging process. Understanding these changes not only enhances our comprehension of aging but also opens avenues for potential interventions aimed at promoting healthy aging and extending lifespan.

4.2 Epigenetic Reprogramming and Aging

Aging is characterized by a progressive decline in physiological integrity, which leads to impaired function and increased vulnerability to various diseases. Among the prominent features of aging, epigenetic alterations play a crucial role. These alterations encompass changes in DNA methylation, histone modifications, chromatin remodeling, and the regulation of non-coding RNAs, all of which contribute to the aging process and the development of age-related diseases [33].

The hallmarks of aging, as proposed by López-Otín et al. (2013), include genomic instability, telomere attrition, epigenetic alterations, loss of proteostasis, deregulated nutrient sensing, mitochondrial dysfunction, cellular senescence, stem cell exhaustion, and altered intercellular communication [1]. These hallmarks are interconnected and contribute to the complexity of aging, with epigenetic changes being a significant factor in the regulation of gene expression and cellular identity [34].

Epigenetic reprogramming refers to the ability to reverse age-associated epigenetic changes, which is gaining attention as a potential strategy for combating aging and its related diseases. Recent studies indicate that rejuvenation strategies, such as the use of small molecules to inhibit DNA methyltransferases and histone deacetylases, can effectively reset the aging clock without erasing cellular identity [35]. For instance, partial reprogramming using Yamanaka factors has shown promise in reversing age-related cellular changes [36].

Furthermore, understanding the dynamics of epigenetic changes during aging can lead to novel therapeutic approaches. These approaches may involve manipulating epigenetic mechanisms to enhance healthspan and lifespan by targeting specific epigenetic modifications associated with aging [37]. Research has demonstrated that interventions aimed at correcting disordered epigenetic reprogramming can alleviate age-related organ dysfunctions [38].

In summary, the interplay between epigenetic alterations and the aging process underscores the potential for epigenetic reprogramming as a therapeutic avenue to extend healthy lifespan and mitigate age-related diseases. The ongoing exploration of these mechanisms will likely yield further insights into effective interventions for aging and its associated conditions.

5 Loss of Proteostasis

5.1 Protein Folding and Quality Control

The loss of proteostasis, a critical hallmark of aging, significantly impacts protein folding and quality control within cells. Proteostasis refers to the cellular mechanisms that maintain the balance of protein synthesis, folding, and degradation, which are essential for cellular function and organismal health. As organisms age, the capacity to maintain proteostasis declines, leading to the accumulation of misfolded and aggregated proteins, which are characteristic of age-related diseases, particularly neurodegenerative disorders such as Alzheimer’s and Parkinson’s disease.

Research has established that the decline in proteostasis is marked by a progressive loss of the various components of the proteostasis machinery, including autophagy, the ubiquitin-proteasome system, and protein synthesis pathways (Basisty et al. 2018; Hipp et al. 2019). This deterioration can result in the accumulation of protein aggregates that disrupt cellular function and contribute to the pathology of age-related diseases. For instance, the accumulation of damaged proteins and the resulting cellular dysfunction are attributed to limited protein repair systems and slowed protein turnover, which are exacerbated by aging (Hamilton & Miller 2017).

In model organisms like Caenorhabditis elegans, studies have shown that proteostasis collapses early in adulthood, coinciding with a reduced activation of cytoprotective responses such as the heat shock response and the unfolded protein response (Ben-Zvi et al. 2009). This early collapse of proteostasis amplifies protein damage and contributes to the development of age-associated diseases.

Furthermore, the proteostasis network (PN) not only regulates protein synthesis, folding, and degradation but also plays a role in coordinating these processes across different cellular compartments. The PN includes molecular chaperones and proteolytic machineries that work together to ensure protein homeostasis. Disruption of this network leads to a decline in proteome integrity, particularly in postmitotic cells such as neurons, where the accumulation of misfolded proteins is particularly detrimental (Ruano 2021; Rai et al. 2022).

Recent advances in proteomic methodologies have allowed for the comprehensive measurement of protein turnover at the individual protein level, revealing insights into how protein dynamics change with aging and in age-related diseases (Yang et al. 2019). These studies indicate that the regulation of proteostasis is multifaceted and influenced by various signaling pathways that can either promote or inhibit the maintenance of a healthy proteome.

In summary, the loss of proteostasis is a hallmark of aging that significantly affects protein folding and quality control. The decline in the capacity of the proteostasis network to manage protein homeostasis contributes to the accumulation of misfolded proteins and the onset of age-related diseases, underscoring the importance of understanding and potentially targeting these pathways to mitigate the effects of aging.

5.2 Consequences of Proteostasis Failure

Aging is characterized by a series of hallmarks that contribute to the gradual decline in physiological functions and increased susceptibility to diseases. One of the primary hallmarks of aging is the loss of proteostasis, which refers to the ability of cells to maintain a stable and functional proteome. This decline in proteostasis has significant consequences, particularly as it relates to the development of various age-related diseases.

Loss of proteostasis is marked by a progressive failure of the cellular mechanisms responsible for maintaining protein homeostasis, including protein synthesis, folding, and degradation. This failure leads to the accumulation of misfolded and damaged proteins, which can disrupt cellular functions and contribute to the pathogenesis of several age-related diseases, such as neurodegenerative disorders, cardiac diseases, and cancer. For instance, the accumulation of protein aggregates is a hallmark of neurodegenerative diseases like Alzheimer's and Parkinson's, where the dysfunctional proteostasis network fails to clear damaged proteins effectively (Hipp et al. 2019; Vilchez et al. 2014).

The consequences of proteostasis failure are multifaceted. Firstly, the accumulation of misfolded proteins can trigger cellular stress responses, leading to inflammation and cell death. Senescent cells, which are resistant to apoptosis, can become pro-inflammatory and promote disease progression through the secretion of various inflammatory cytokines (Meller & Shalgi 2021). Additionally, the decline in proteostasis can impair critical cellular functions, particularly in postmitotic cells such as neurons and cardiomyocytes, which are not regularly replenished and are therefore more vulnerable to the effects of proteotoxic stress (Wiersma et al. 2016; Goyal et al. 2024).

Moreover, the disruption of proteostasis is linked to decreased stem cell function, which contributes to tissue degeneration and organismal aging. The accumulation of damaged proteins in stem cells can affect their differentiation and functionality, further exacerbating the aging process (Vilchez et al. 2014).

In summary, the loss of proteostasis represents a crucial hallmark of aging that has profound implications for cellular function and organismal health. It underlies the development of numerous age-related diseases, emphasizing the need for strategies aimed at reinforcing proteostasis as a potential therapeutic approach to mitigate the effects of aging and improve healthspan (Hetz & Dillin 2024; Hamilton & Miller 2017).

6 Deregulated Nutrient Sensing

6.1 Insulin/IGF-1 Signaling Pathway

The hallmarks of aging include a variety of biological processes that contribute to the gradual decline in physiological function and increase in susceptibility to diseases. One of the critical hallmarks is deregulated nutrient sensing, which encompasses the insulin/IGF-1 signaling pathway. This pathway plays a pivotal role in the regulation of growth, metabolism, and longevity across various species.

The insulin/IGF-1 signaling pathway is highly conserved and has been shown to significantly influence aging and longevity. In mammals, alterations in this pathway are linked to age-related diseases, including metabolic disorders and neurodegenerative diseases. For instance, the insulin/IGF-1 signaling pathway regulates insulin sensitivity, which tends to decline with age, contributing to the development of insulin resistance and associated metabolic syndromes [39].

Research indicates that reduced activity of the insulin/IGF-1 signaling pathway can extend lifespan, as seen in model organisms like Caenorhabditis elegans and mice. Disruption of this signaling pathway in these organisms has been associated with increased longevity and improved healthspan [40].

Moreover, the insulin/IGF-1 signaling pathway also interacts with various cellular mechanisms, such as the activation of the FOXO transcription factors, which are crucial for mediating stress responses, metabolism, and longevity. When insulin signaling is reduced, FOXO proteins translocate to the nucleus, promoting the expression of genes that enhance stress resistance and longevity [41].

In the context of pancreatic β-cells, decreased IGF1R signaling has been linked to improved cellular function and reduced senescence, indicating a direct relationship between nutrient sensing and cellular aging processes [42]. This suggests that targeting the insulin/IGF-1 signaling pathway could be a viable strategy for mitigating age-related decline in function and promoting longevity.

Overall, the deregulation of nutrient sensing, particularly through the insulin/IGF-1 signaling pathway, is a significant hallmark of aging that contributes to the complex interplay between metabolism, cellular function, and longevity. Understanding these mechanisms provides insights into potential interventions that could enhance healthspan and delay the onset of age-associated diseases.

6.2 Nutrient Sensing and Longevity

Aging is a complex biological process characterized by a progressive decline in physiological integrity, which leads to impaired function and an increased vulnerability to various diseases. Research has identified several hallmarks of aging, which are common denominators observed across different organisms, particularly in mammals. These hallmarks include:

  1. Genomic Instability: Refers to the accumulation of genetic damage over time, which can lead to cellular dysfunction.
  2. Telomere Attrition: The gradual shortening of telomeres, protective caps on the ends of chromosomes, contributes to cellular aging and limits cell division.
  3. Epigenetic Alterations: Changes in gene expression regulation that do not involve alterations to the underlying DNA sequence can impact cellular function.
  4. Loss of Proteostasis: The inability to maintain the proper folding and function of proteins can lead to cellular stress and disease.
  5. Deregulated Nutrient Sensing: This hallmark involves the disruption of pathways that detect and respond to nutrient availability, which is crucial for energy metabolism and overall health.
  6. Mitochondrial Dysfunction: Mitochondria, the energy-producing organelles, can become less efficient with age, leading to decreased ATP production and increased production of reactive oxygen species (ROS), contributing to oxidative stress.
  7. Cellular Senescence: The process by which cells lose the ability to divide and function, often associated with chronic inflammation and tissue dysfunction.
  8. Stem Cell Exhaustion: The decline in the regenerative capacity of stem cells, which is essential for tissue repair and maintenance.
  9. Altered Intercellular Communication: Changes in the signaling between cells can disrupt tissue homeostasis and contribute to aging-related diseases.

In addition to these nine hallmarks, recent research has expanded the understanding of aging to include factors such as dysbiosis and chronic inflammation, often referred to as "inflammaging," which significantly impact the aging process and the development of age-related diseases [1][43][44].

Deregulated nutrient sensing plays a critical role in the aging process. It affects how the body detects and responds to nutrient availability, which is crucial for maintaining metabolic health. Alterations in nutrient sensing pathways can lead to a decline in insulin sensitivity and energy metabolism, making the organism more susceptible to age-related diseases [6][45]. For instance, the dysregulation of nutrient sensing mechanisms can contribute to chronic inflammation and metabolic dysfunction, exacerbating the aging phenotype [44].

Furthermore, the interplay between nutrition and longevity is a key area of research. Dietary interventions, particularly caloric restriction, have been shown to positively influence nutrient sensing pathways and promote healthy aging. This highlights the potential of dietary strategies as therapeutic targets for enhancing longevity and mitigating age-related conditions [46][47]. Understanding the molecular signaling pathways involved in nutrient sensing is essential for developing effective interventions aimed at promoting healthy aging and extending healthspan [48][49].

In conclusion, the hallmarks of aging encompass a range of biological processes, with deregulated nutrient sensing being a pivotal factor influencing longevity and the development of age-related diseases. Addressing these hallmarks through dietary and lifestyle interventions may provide new avenues for enhancing health in aging populations.

7 Mitochondrial Dysfunction

7.1 Mitochondrial Biogenesis and Dynamics

Aging is a multifaceted biological process characterized by a progressive decline in cellular and organ function, often associated with increased vulnerability to age-related diseases. Central to this process are the hallmarks of aging, which include various molecular and cellular alterations. Among these, mitochondrial dysfunction is a significant hallmark that plays a critical role in the aging process and its associated pathologies.

Mitochondrial dysfunction encompasses a range of alterations, including decreased mitochondrial biogenesis, compromised mitochondrial dynamics, and impaired quality control mechanisms. These dysfunctions manifest as reduced respiratory capacity, diminished mitochondrial membrane potential, and increased production of reactive oxygen species (ROS) [50][51][52]. The accumulation of ROS leads to oxidative stress, which can cause extensive damage to cellular components, including DNA, proteins, and lipids, thereby contributing to the aging phenotype [43][53].

Mitochondrial biogenesis refers to the process by which new mitochondria are formed within cells, a critical mechanism for maintaining mitochondrial health and function. This process is regulated by various factors, including the PGC-1 family of coactivators, which play a pivotal role in stimulating mitochondrial biogenesis and enhancing mitochondrial function [54][55]. During aging, there is a decline in mitochondrial biogenesis, leading to a reduction in mitochondrial mass and function, which exacerbates the aging process and increases susceptibility to age-related diseases [29][56].

Mitochondrial dynamics, which includes the processes of fission and fusion, is crucial for maintaining mitochondrial function and quality. Fission allows for the removal of damaged mitochondria, while fusion helps in the mixing of mitochondrial contents, thereby maintaining their functionality [51][56]. Dysregulation of these dynamics can lead to the accumulation of dysfunctional mitochondria, contributing to the pathophysiology of various age-related disorders [29][57].

Moreover, the interplay between mitochondrial dysfunction and other hallmarks of aging, such as oxidative stress and cellular senescence, highlights the complexity of the aging process [43][53]. Mitochondrial dysfunction is not only a consequence of aging but also acts as a driving force for the progression of age-related diseases, including neurodegenerative disorders, cardiovascular diseases, and metabolic syndromes [52][56].

In conclusion, mitochondrial dysfunction, characterized by impaired biogenesis and dynamics, plays a crucial role in the aging process. Understanding these mechanisms is essential for developing therapeutic strategies aimed at promoting healthy aging and mitigating age-associated diseases.

7.2 Role in Aging and Metabolic Disorders

Aging is characterized by a progressive decline in physiological integrity, function, and increased vulnerability to age-related diseases. Among the hallmarks of aging, mitochondrial dysfunction plays a crucial role, particularly in the context of metabolic disorders. Mitochondria are essential for cellular energy production and are involved in various metabolic processes, including calcium signaling and redox homeostasis. Their dysfunction is a significant contributor to the aging process and is linked to numerous age-related diseases, such as neurodegenerative disorders, cardiovascular diseases, and metabolic syndromes.

The hallmarks of aging have been categorized into nine distinct mechanisms, which include telomere shortening, genomic instability, epigenetic modifications, mitochondrial dysfunction, loss of proteostasis, dysregulated nutrient sensing, stem cell exhaustion, cellular senescence, and altered cellular communication [43]. Among these, mitochondrial dysfunction is particularly notable as it is often accompanied by an increase in reactive oxygen species (ROS) production, impaired oxidative phosphorylation, and the accumulation of mitochondrial DNA (mtDNA) mutations [29].

Mitochondrial dysfunction is linked to the aging process through several mechanisms. For instance, as aging progresses, there is a decline in ATP production alongside elevated ROS levels, leading to oxidative stress that can damage cellular components, including DNA, proteins, and lipids [43]. This oxidative damage contributes to the aging phenotype and the development of metabolic disorders [52].

Mitophagy, the selective degradation of damaged mitochondria, is a critical quality control mechanism that helps maintain mitochondrial function and cellular homeostasis. However, with aging, the efficiency of mitophagy declines, resulting in the accumulation of dysfunctional mitochondria, which exacerbates oxidative stress and cellular senescence [58].

Furthermore, the interplay between mitochondrial dysfunction and metabolic disorders is significant. Aging leads to alterations in mitochondrial dynamics and metabolism, which can result in metabolic reprogramming and contribute to the pathogenesis of age-related diseases. For example, aged hematopoietic stem cells exhibit enhanced mitochondrial oxidative phosphorylation and increased ROS production, correlating with a decline in regenerative capacity and increased disease susceptibility [59]. Additionally, mitochondrial dysfunction is implicated in the aging immune system, contributing to inflamm-aging and decreased immune response [60].

The role of mitochondrial dysfunction in aging and metabolic disorders underscores the importance of maintaining mitochondrial health as a potential therapeutic target for promoting healthy aging and preventing age-related diseases. Interventions aimed at restoring mitochondrial function, enhancing mitophagy, and reducing oxidative stress may provide promising strategies for counteracting the detrimental effects of aging on metabolic health [61]. Overall, understanding the multifaceted roles of mitochondria in aging can lead to novel approaches for improving healthspan and longevity.

8 Cellular Senescence

8.1 Mechanisms of Cellular Senescence

Aging is characterized by a progressive decline in physiological integrity, which is accompanied by a series of biological changes that lead to increased vulnerability to age-related diseases. The concept of "hallmarks of aging" was introduced to categorize these changes, providing a framework for understanding the mechanisms that drive the aging process. The primary hallmarks of aging include genomic instability, telomere attrition, epigenetic alterations, loss of proteostasis, deregulated nutrient sensing, mitochondrial dysfunction, cellular senescence, stem cell exhaustion, and altered intercellular communication [1][5][62].

Cellular senescence is a critical hallmark of aging defined as an irreversible state of cell cycle arrest that occurs in response to various intrinsic and extrinsic stressors, including DNA damage, telomere shortening, and oncogene activation [62][63]. While initially considered a protective mechanism to prevent the proliferation of damaged cells, the accumulation of senescent cells over time contributes significantly to aging and age-related pathologies. Senescent cells are metabolically active but no longer divide, and they secrete a range of pro-inflammatory cytokines, growth factors, and proteases, collectively referred to as the senescence-associated secretory phenotype (SASP). This secretory profile can induce local inflammation and tissue dysfunction, further exacerbating age-related decline [63][64].

The mechanisms underlying cellular senescence are multifaceted. Key processes include:

  1. Telomere Shortening: As cells divide, telomeres, the protective caps at the ends of chromosomes, shorten. When telomeres reach a critical length, they trigger cellular senescence to prevent genomic instability [21].

  2. Genomic Instability: Accumulation of DNA damage and mutations can lead to cellular senescence. Cells with damaged DNA activate checkpoint pathways that halt cell division [62].

  3. Mitochondrial Dysfunction: Mitochondria play a crucial role in cellular energy metabolism and the generation of reactive oxygen species (ROS). Mitochondrial dysfunction is linked to increased oxidative stress, which can damage cellular components and promote senescence [65].

  4. Epigenetic Alterations: Changes in the epigenetic landscape, including DNA methylation and histone modification, can affect gene expression patterns associated with cell cycle regulation and stress responses, contributing to the senescent phenotype [62].

  5. Inflammatory Response: The SASP associated with senescent cells can create a pro-inflammatory environment that affects neighboring cells, promoting further senescence and contributing to tissue dysfunction [63].

The interplay between these mechanisms highlights the complexity of cellular senescence as both a protective and detrimental process in aging. Understanding these pathways provides insights into potential therapeutic strategies aimed at targeting senescent cells to mitigate their negative effects on health and longevity [5][43].

8.2 Impact on Tissue Function and Aging

Cellular senescence is recognized as one of the critical hallmarks of aging, characterized by a state in which cells lose their ability to divide and function effectively. This phenomenon has profound implications for tissue function and the overall aging process. The accumulation of senescent cells in tissues contributes to age-related functional decline and increased vulnerability to various diseases.

Senescent cells are often associated with the secretion of pro-inflammatory cytokines, growth factors, and proteases, collectively termed the senescence-associated secretory phenotype (SASP). This secretion can disrupt local tissue microenvironments, leading to chronic inflammation, which is a hallmark of aging. Chronic inflammation can impair tissue repair mechanisms and contribute to the pathogenesis of age-related diseases such as cancer, cardiovascular disorders, and neurodegenerative diseases[43].

Moreover, cellular senescence affects the regenerative capacity of tissues. In tissues such as skeletal muscle, the presence of senescent cells can hinder muscle repair and regeneration, leading to conditions like sarcopenia, which is characterized by loss of muscle mass and strength with aging[66]. The interplay between senescence and other hallmarks of aging, such as genomic instability and mitochondrial dysfunction, further complicates the aging process. For instance, genomic instability can trigger cellular senescence, while senescent cells can exacerbate mitochondrial dysfunction, creating a vicious cycle that accelerates aging[2].

In the context of tissue function, cellular senescence can disrupt normal homeostasis. As tissues age, the accumulation of senescent cells can lead to impaired cellular communication and altered intercellular signaling, which can affect tissue architecture and function[67]. This disruption is particularly evident in tissues such as adipose tissue, where senescence contributes to metabolic dysfunction and inflammation, further impacting systemic health[68].

The therapeutic implications of targeting cellular senescence are significant. Strategies aimed at eliminating senescent cells, known as senolytics, or modulating the SASP could potentially restore tissue function and improve healthspan. Research is ongoing to explore how these interventions can mitigate the effects of aging and promote healthier aging outcomes[2][7].

In summary, cellular senescence plays a pivotal role in the aging process by influencing tissue function through mechanisms of inflammation, impaired regeneration, and disrupted cellular communication. Understanding and addressing the impact of senescence on tissue function is crucial for developing effective strategies to promote healthy aging and combat age-related diseases.

9 Stem Cell Exhaustion

9.1 Role of Stem Cells in Tissue Homeostasis

Aging is a complex biological process characterized by a gradual decline in the functionality of tissues and organs, which is significantly influenced by the exhaustion of stem cells. This phenomenon, referred to as "stem cell exhaustion," is a hallmark of aging that leads to a diminished capacity for tissue regeneration and homeostasis.

Stem cells are crucial for the maintenance and repair of tissues throughout an organism's life. They possess the ability to self-renew and differentiate into various cell types, which is essential for tissue homeostasis. However, with advancing age, the functional capacity of these stem cells is compromised. This decline is associated with various intrinsic and extrinsic factors that collectively contribute to the aging process.

One of the key aspects of stem cell aging is the accumulation of molecular damage and the failure of quality control systems, which can lead to alterations in stem cell functionality. Research has identified several hallmarks of aging that are particularly relevant to stem cells:

  1. Accumulation of Molecular Damage: Over time, stem cells accumulate DNA damage and other forms of cellular stress, which can impair their ability to function effectively. This accumulation is thought to be a principal mechanism underlying age-dependent stem cell decline [69].

  2. Epigenetic Alterations: Aging is accompanied by significant changes in the epigenome of stem cells, leading to deregulation of gene expression. These epigenetic modifications can disrupt developmental pathways and contribute to the functional decline of stem cells [70].

  3. Impaired Autophagy: Autophagy, a cellular process that degrades damaged organelles and proteins, is essential for maintaining stem cell function. Age-related impairments in autophagy have been linked to stem cell exhaustion, particularly in muscle and hematopoietic stem cells [71].

  4. Altered Stem Cell Niches: The microenvironment or niche that supports stem cells also undergoes changes with age. For example, in the Drosophila testis, somatic niche cells exhibit reduced expression of self-renewal signals, which correlates with a decline in germline stem cell numbers [72].

  5. Chronic Inflammation: Age-associated inflammation has been shown to negatively impact stem cell function. In particular, the aging epidermis can disrupt cytokine signaling pathways that are critical for maintaining stem cell homeostasis [73].

  6. Oxidative Stress: Increased oxidative stress is another factor contributing to stem cell aging. The production of reactive oxygen species can lead to cellular damage and compromises the regenerative capacity of stem cells [74].

These hallmarks of aging not only elucidate the mechanisms underlying stem cell exhaustion but also highlight potential therapeutic targets for interventions aimed at rejuvenating stem cell function and promoting healthy aging. Understanding these processes is critical for developing strategies to combat age-related diseases and improve tissue regeneration capabilities as organisms age. The interplay between these factors ultimately underscores the importance of stem cells in maintaining tissue homeostasis and their pivotal role in the aging process.

9.2 Consequences of Stem Cell Exhaustion in Aging

Stem cell exhaustion is recognized as one of the critical hallmarks of aging, characterized by a progressive decline in the regenerative capacity and functionality of stem cells. This phenomenon plays a significant role in the aging process and contributes to various age-related diseases.

As organisms age, somatic stem cells lose their ability to sustain tissue homeostasis and support regeneration. Although stem cells are relatively protected from some aging mechanisms compared to their differentiated progeny, they remain susceptible to both intrinsic and extrinsic stressors, leading to functional decline. This decline is attributed to several factors, including oxidative stress, changes in the stem cell niche, and alterations in the epigenome, which collectively compromise stem cell self-renewal and differentiation capabilities[75].

Consequences of stem cell exhaustion manifest in various ways. One of the most notable effects is the reduced regenerative potential of tissues, which can lead to impaired maintenance and repair mechanisms. For instance, the aging of hematopoietic stem cells (HSCs) is associated with an imbalanced immune response and increased susceptibility to diseases such as myelodysplastic syndromes and acute myeloid leukemia. Aged HSCs typically exhibit features such as myeloid-biased differentiation and loss of polarity, which are linked to increased mitochondrial oxidative phosphorylation and reactive oxygen species (ROS) production, further exacerbating the degenerative process[59].

In addition to hematopoietic tissues, stem cell exhaustion also impacts other systems. For example, adult Sox2+ stem cell depletion in mice has been shown to induce cellular senescence and signs of premature aging, such as increased kyphosis and hair graying[76]. This indicates that stem cell exhaustion can lead to broader physiological declines that compromise overall health and increase vulnerability to age-related pathologies.

Moreover, the mechanisms underlying stem cell exhaustion include oxidative stress, which results from unresolved oxidative damage to cellular macromolecules. This oxidative stress not only diminishes the self-renewal capacity of stem cells but also promotes senescence and apoptosis, leading to functional depletion[74]. Furthermore, changes in the systemic environment and the stem cell niche contribute to the loss of stem cell functionality, which is critical for tissue repair and regeneration[77].

In summary, stem cell exhaustion is a hallmark of aging that significantly impacts tissue maintenance and regenerative capabilities. The consequences of this exhaustion are profound, leading to a decline in health span and an increased risk of age-related diseases. Understanding the underlying mechanisms of stem cell exhaustion is essential for developing strategies to mitigate its effects and promote healthier aging.

10 Conclusion

The review comprehensively explores the hallmarks of aging, emphasizing their interconnectivity and their collective impact on the aging process. The major findings indicate that aging is not a linear process but rather a complex interplay of multiple biological mechanisms, including genomic instability, telomere attrition, epigenetic alterations, loss of proteostasis, deregulated nutrient sensing, mitochondrial dysfunction, cellular senescence, stem cell exhaustion, and altered intercellular communication. Each hallmark contributes uniquely to the aging phenotype and the development of age-related diseases, highlighting the need for a holistic approach in aging research. Current studies have shown that these hallmarks interact in intricate ways, with one hallmark influencing another, such as how mitochondrial dysfunction exacerbates genomic instability and how cellular senescence affects tissue function. Future research directions should focus on elucidating these interactions further and identifying therapeutic targets within these pathways to promote healthy aging. Investigating interventions that can mitigate the effects of these hallmarks could lead to innovative strategies aimed at enhancing healthspan and extending lifespan. Ultimately, understanding the hallmarks of aging not only provides insights into the biological mechanisms of aging but also lays the groundwork for potential therapeutic interventions that can improve the quality of life for the aging population.

References

  • [1] Carlos López-Otín;Maria A Blasco;Linda Partridge;Manuel Serrano;Guido Kroemer. The hallmarks of aging.. Cell(IF=42.5). 2013. PMID:23746838. DOI: .
  • [2] Carlos López-Otín;Maria A Blasco;Linda Partridge;Manuel Serrano;Guido Kroemer. Hallmarks of aging: An expanding universe.. Cell(IF=42.5). 2023. PMID:36599349. DOI: 10.1016/j.cell.2022.11.001.
  • [3] Charlotte Delrue;Reinhart Speeckaert;Marijn M Speeckaert. Rewinding the Clock: Emerging Pharmacological Strategies for Human Anti-Aging Therapy.. International journal of molecular sciences(IF=4.9). 2025. PMID:41096641. DOI: 10.3390/ijms26199372.
  • [4] Yuan Li;Laura Berliocchi;Zhiquan Li;Lene Juel Rasmussen. Interactions between mitochondrial dysfunction and other hallmarks of aging: Paving a path toward interventions that promote healthy old age.. Aging cell(IF=7.1). 2024. PMID:37497653. DOI: 10.1111/acel.13942.
  • [5] Rumiana Tenchov;Janet M Sasso;Xinmei Wang;Qiongqiong Angela Zhou. Aging Hallmarks and Progression and Age-Related Diseases: A Landscape View of Research Advancement.. ACS chemical neuroscience(IF=3.9). 2024. PMID:38095562. DOI: 10.1021/acschemneuro.3c00531.
  • [6] Maryam Keshavarz;Kan Xie;Kristina Schaaf;Daniele Bano;Dan Ehninger. Targeting the "hallmarks of aging" to slow aging and treat age-related disease: fact or fiction?. Molecular psychiatry(IF=10.1). 2023. PMID:35840801. DOI: 10.1038/s41380-022-01680-x.
  • [7] Shailendra Kumar Mishra;Vyshnavy Balendra;Josephine Esposto;Ahmad A Obaid;Ricardo B Maccioni;Niraj Kumar Jha;George Perry;Mahmoud Moustafa;Mohammed Al-Shehri;Mahendra P Singh;Anmar Anwar Khan;Emanuel Vamanu;Sandeep Kumar Singh. Therapeutic Antiaging Strategies.. Biomedicines(IF=3.9). 2022. PMID:36289777. DOI: 10.3390/biomedicines10102515.
  • [8] Jan Vijg;Yousin Suh. Genome instability and aging.. Annual review of physiology(IF=19.1). 2013. PMID:23398157. DOI: 10.1146/annurev-physiol-030212-183715.
  • [9] Susana Gonzalo. Epigenetic alterations in aging.. Journal of applied physiology (Bethesda, Md. : 1985)(IF=3.3). 2010. PMID:20448029. DOI: 10.1152/japplphysiol.00238.2010.
  • [10] Attila Kallai;Zoltan Ungvari;Mónika Fekete;Andrea B Maier;Gabor Mikala;Hajnalka Andrikovics;Andrea Lehoczki. Genomic instability and genetic heterogeneity in aging: insights from clonal hematopoiesis (CHIP), monoclonal gammopathy (MGUS), and monoclonal B-cell lymphocytosis (MBL).. GeroScience(IF=5.4). 2025. PMID:39405013. DOI: 10.1007/s11357-024-01374-y.
  • [11] Matthew M Crane;Adam E Russell;Brent J Schafer;Ben W Blue;Riley Whalen;Jared Almazan;Mung Gi Hong;Bao Nguyen;Joslyn E Goings;Kenneth L Chen;Ryan Kelly;Matt Kaeberlein. DNA damage checkpoint activation impairs chromatin homeostasis and promotes mitotic catastrophe during aging.. eLife(IF=6.4). 2019. PMID:31714209. DOI: .
  • [12] Ricardo Afonso-Reis;Cristiana R Madeira;David V C Brito;Clévio Nóbrega. Insights Into Cockayne Syndrome Type B: What Underlies Its Pathogenesis?. Aging cell(IF=7.1). 2025. PMID:40536083. DOI: 10.1111/acel.70136.
  • [13] Tomas Schmauck-Medina;Adrian Molière;Sofie Lautrup;Jianying Zhang;Stefan Chlopicki;Helena Borland Madsen;Shuqin Cao;Casper Soendenbroe;Els Mansell;Mark Bitsch Vestergaard;Zhiquan Li;Yosef Shiloh;Patricia L Opresko;Jean-Marc Egly;Thomas Kirkwood;Eric Verdin;Vilhelm A Bohr;Lynne S Cox;Tinna Stevnsner;Lene Juel Rasmussen;Evandro F Fang. New hallmarks of ageing: a 2022 Copenhagen ageing meeting summary.. Aging(IF=3.9). 2022. PMID:36040386. DOI: 10.18632/aging.204248.
  • [14] Ji-Hye Oh;Hyo Jeong Lee;Wonkyung Kim;Da Eun Oh;Hong-Kyu Kim;Eun Hee Kim;Jaewon Choe;Ha Ra Jun;Chae Won Park;Young Gwang Kang;Chong Jai Kim;Chang Ohk Sung;Tae Won Kim. Whole genome sequencing reveals telomere associated genomic differences between healthy and unhealthy aging in a Korean population.. Biogerontology(IF=4.1). 2025. PMID:40844718. DOI: 10.1007/s10522-025-10310-2.
  • [15] Peter Vrtačnik;Lara G Merino;Santhilal Subhash;Hafdís T Helgadóttir;Matthieu Bardin;Fabiana Stefani;Depin Wang;Ping Chen;Irene Franco;Gwladys Revêchon;Maria Eriksson. Induced somatic mutation accumulation during skeletal muscle regeneration reduces muscle strength.. Nature aging(IF=19.4). 2025. PMID:40836125. DOI: 10.1038/s43587-025-00941-y.
  • [16] Lucía López-Gil;Amparo Pascual-Ahuir;Markus Proft. Genomic Instability and Epigenetic Changes during Aging.. International journal of molecular sciences(IF=4.9). 2023. PMID:37762580. DOI: 10.3390/ijms241814279.
  • [17] Christopher E Mason;Maria A Sierra;Henry J Feng;Susan M Bailey. Telomeres and aging: on and off the planet!. Biogerontology(IF=4.1). 2024. PMID:38581556. DOI: 10.1007/s10522-024-10098-7.
  • [18] Geraldine Aubert;Peter M Lansdorp. Telomeres and aging.. Physiological reviews(IF=28.7). 2008. PMID:18391173. DOI: 10.1152/physrev.00026.2007.
  • [19] Sarah E Wolf;Idan Shalev. The shelterin protein expansion of telomere dynamics: Linking early life adversity, life history, and the hallmarks of aging.. Neuroscience and biobehavioral reviews(IF=7.9). 2023. PMID:37268182. DOI: 10.1016/j.neubiorev.2023.105261.
  • [20] Xiao Dong;Shixiang Sun;Lei Zhang;Seungsoo Kim;Zhidong Tu;Cristina Montagna;Alexander Y Maslov;Yousin Suh;Tao Wang;Judith Campisi;Jan Vijg. Age-related telomere attrition causes aberrant gene expression in sub-telomeric regions.. Aging cell(IF=7.1). 2021. PMID:34018656. DOI: 10.1111/acel.13357.
  • [21] Yukun Zhu;Xuewen Liu;Xuelu Ding;Fei Wang;Xin Geng. Telomere and its role in the aging pathways: telomere shortening, cell senescence and mitochondria dysfunction.. Biogerontology(IF=4.1). 2019. PMID:30229407. DOI: 10.1007/s10522-018-9769-1.
  • [22] Bartu Eren Güneşliol;Esen Karaca;Duygu Ağagündüz;Zeynep Alanur Acar. Association of physical activity and nutrition with telomere length, a marker of cellular aging: A comprehensive review.. Critical reviews in food science and nutrition(IF=8.8). 2023. PMID:34553645. DOI: 10.1080/10408398.2021.1952402.
  • [23] Xi-Yuen Kuan;Nurul Syahira Ahmad Fauzi;Khuen Yen Ng;Athirah Bakhtiar. Exploring the Causal Relationship Between Telomere Biology and Alzheimer's Disease.. Molecular neurobiology(IF=4.3). 2023. PMID:37046137. DOI: 10.1007/s12035-023-03337-4.
  • [24] Stylianos Daios;Antonia Anogeianaki;Georgia Kaiafa;Anastasia Kontana;Stavroula Veneti;Christiana Gogou;Eleni Karlafti;Dimitrios Pilalas;Ilias Kanellos;Christos Savopoulos. Telomere Length as a Marker of Biological Aging: A Critical Review of Recent Literature.. Current medicinal chemistry(IF=3.5). 2022. PMID:35838223. DOI: 10.2174/0929867329666220713123750.
  • [25] Daniel Baylis;Georgia Ntani;Mark H Edwards;Holly E Syddall;David B Bartlett;Elaine M Dennison;Carmen Martin-Ruiz;Thomas von Zglinicki;Diana Kuh;Janet M Lord;Avan Aihie Sayer;Cyrus Cooper. Inflammation, telomere length, and grip strength: a 10-year longitudinal study.. Calcified tissue international(IF=3.2). 2014. PMID:24858709. DOI: 10.1007/s00223-014-9862-7.
  • [26] Qiaofeng Ye;Abner T Apsley;Laura Etzel;Waylon J Hastings;John T Kozlosky;Cade Walker;Sarah E Wolf;Idan Shalev. Telomere length and chronological age across the human lifespan: A systematic review and meta-analysis of 414 study samples including 743,019 individuals.. Ageing research reviews(IF=12.4). 2023. PMID:37567392. DOI: 10.1016/j.arr.2023.102031.
  • [27] Marlies Schellnegger;Elisabeth Hofmann;Martina Carnieletto;Lars-Peter Kamolz. Unlocking longevity: the role of telomeres and its targeting interventions.. Frontiers in aging(IF=4.3). 2024. PMID:38333665. DOI: 10.3389/fragi.2024.1339317.
  • [28] Pan Liao;Bo Yan;Conglin Wang;Ping Lei. Telomeres: Dysfunction, Maintenance, Aging and Cancer.. Aging and disease(IF=6.9). 2023. PMID:38270117. DOI: 10.14336/AD.2023.1128.
  • [29] Yifei Feng;Yan Lu. The nuclear-mitochondrial crosstalk in aging: From mechanisms to therapeutics.. Free radical biology & medicine(IF=8.2). 2025. PMID:40086490. DOI: 10.1016/j.freeradbiomed.2025.03.012.
  • [30] Carola Ingrid Weidner;Qiong Lin;Carmen Maike Koch;Lewin Eisele;Fabian Beier;Patrick Ziegler;Dirk Olaf Bauerschlag;Karl-Heinz Jöckel;Raimund Erbel;Thomas Walter Mühleisen;Martin Zenke;Tim Henrik Brümmendorf;Wolfgang Wagner. Aging of blood can be tracked by DNA methylation changes at just three CpG sites.. Genome biology(IF=9.4). 2014. PMID:24490752. DOI: 10.1186/gb-2014-15-2-r24.
  • [31] Meaghan J Jones;Sarah J Goodman;Michael S Kobor. DNA methylation and healthy human aging.. Aging cell(IF=7.1). 2015. PMID:25913071. DOI: 10.1111/acel.12349.
  • [32] Archana Unnikrishnan;Willard M Freeman;Jordan Jackson;Jonathan D Wren;Hunter Porter;Arlan Richardson. The role of DNA methylation in epigenetics of aging.. Pharmacology & therapeutics(IF=12.5). 2019. PMID:30419258. DOI: 10.1016/j.pharmthera.2018.11.001.
  • [33] Kang Wang;Huicong Liu;Qinchao Hu;Lingna Wang;Jiaqing Liu;Zikai Zheng;Weiqi Zhang;Jie Ren;Fangfang Zhu;Guang-Hui Liu. Epigenetic regulation of aging: implications for interventions of aging and diseases.. Signal transduction and targeted therapy(IF=52.7). 2022. PMID:36336680. DOI: 10.1038/s41392-022-01211-8.
  • [34] Mehran Izadi;Nariman Sadri;Amirhossein Abdi;Sahar Serajian;Dorsa Jalalei;Safa Tahmasebi. Epigenetic biomarkers in aging and longevity: Current and future application.. Life sciences(IF=5.1). 2024. PMID:38879158. DOI: 10.1016/j.lfs.2024.122842.
  • [35] Beatriz Pereira;Francisca P Correia;Inês A Alves;Margarida Costa;Mariana Gameiro;Ana P Martins;Jorge A Saraiva. Epigenetic reprogramming as a key to reverse ageing and increase longevity.. Ageing research reviews(IF=12.4). 2024. PMID:38272265. DOI: 10.1016/j.arr.2024.102204.
  • [36] Alice E Kane;David A Sinclair. Epigenetic changes during aging and their reprogramming potential.. Critical reviews in biochemistry and molecular biology(IF=6.4). 2019. PMID:30822165. DOI: 10.1080/10409238.2019.1570075.
  • [37] Annamaria la Torre;Filomena Lo Vecchio;Antonio Greco. Epigenetic Mechanisms of Aging and Aging-Associated Diseases.. Cells(IF=5.2). 2023. PMID:37190071. DOI: 10.3390/cells12081163.
  • [38] Xin Liu;Jing Feng;Madi Guo;Chen Chen;Tong Zhao;Xiuxiu Sun;Yong Zhang. Resetting the aging clock through epigenetic reprogramming: Insights from natural products.. Pharmacology & therapeutics(IF=12.5). 2025. PMID:40221101. DOI: 10.1016/j.pharmthera.2025.108850.
  • [39] Antero Salminen;Kai Kaarniranta;Anu Kauppinen. Insulin/IGF-1 signaling promotes immunosuppression via the STAT3 pathway: impact on the aging process and age-related diseases.. Inflammation research : official journal of the European Histamine Research Society ... et al.. 2021. PMID:34476533. DOI: 10.1007/s00011-021-01498-3.
  • [40] Marielisa Rincon;Radhika Muzumdar;Gil Atzmon;Nir Barzilai. The paradox of the insulin/IGF-1 signaling pathway in longevity.. Mechanisms of ageing and development(IF=5.1). 2004. PMID:15272501. DOI: 10.1016/j.mad.2004.03.006.
  • [41] Kylie Hesp;Geert Smant;Jan E Kammenga. Caenorhabditis elegans DAF-16/FOXO transcription factor and its mammalian homologs associate with age-related disease.. Experimental gerontology(IF=4.3). 2015. PMID:26363351. DOI: .
  • [42] Kanako Iwasaki;Benjamin Lalani;Jiho Kahng;Priscila Carapeto;Stephanie Sanjines;Francesko Hela;Cristian Abarca;Tadataka Tsuji;Justin Darcy;Andrzej Bartke;Yu-Hua Tseng;Rohit N Kulkarni;Cristina Aguayo-Mazzucato. Decreased IGF1R attenuates senescence and improves function in pancreatic β-cells.. Frontiers in endocrinology(IF=4.6). 2023. PMID:37441495. DOI: 10.3389/fendo.2023.1203534.
  • [43] Edio Maldonado;Sebastián Morales-Pison;Fabiola Urbina;Aldo Solari. Aging Hallmarks and the Role of Oxidative Stress.. Antioxidants (Basel, Switzerland)(IF=6.6). 2023. PMID:36978899. DOI: 10.3390/antiox12030651.
  • [44] Jordan J Baechle;Nan Chen;Priya Makhijani;Shawn Winer;David Furman;Daniel A Winer. Chronic inflammation and the hallmarks of aging.. Molecular metabolism(IF=6.6). 2023. PMID:37329949. DOI: 10.1016/j.molmet.2023.101755.
  • [45] Celine E Riera;Andrew Dillin. Tipping the metabolic scales towards increased longevity in mammals.. Nature cell biology(IF=19.1). 2015. PMID:25720959. DOI: 10.1038/ncb3107.
  • [46] Elangbam Tomtheelnganbee;Puja Sah;R Sharma. Mitochondrial function and nutrient sensing pathways in ageing: enhancing longevity through dietary interventions.. Biogerontology(IF=4.1). 2022. PMID:35842501. DOI: 10.1007/s10522-022-09978-7.
  • [47] Camilla Barbero Mazzucca;Giuseppe Cappellano;Annalisa Chiocchetti. Nutrition, Immunity and Aging: Current Scenario and Future Perspectives in Neurodegenerative Diseases.. CNS & neurological disorders drug targets(IF=3.0). 2024. PMID:37138438. DOI: 10.2174/1871527322666230502123255.
  • [48] Aygul Dagbasi;Adrian Holliday;Bernadette Carroll;Chiara de Lucia;Oliver M Shannon;John Mathers;John McLaughlin;Chloe French;Aylin Hanyaloglu;Sorrel Burden;Douglas Morrison;Viktor Korolchuk;Gary Frost. White Paper on Nutrition Sensing and Ageing.. Nutrition bulletin(IF=3.6). 2025. PMID:40635164. DOI: 10.1111/nbu.70020.
  • [49] Aygul Dagbasi;Amy Fuller;Aylin C Hanyaloglu;Bernadette Carroll;John McLaughlin;Gary Frost;Adrian Holliday. The role of nutrient sensing dysregulation in anorexia of ageing: The little we know and the much we don't.. Appetite(IF=3.8). 2024. PMID:39423861. DOI: 10.1016/j.appet.2024.107718.
  • [50] Jose Ignacio Escrig-Larena;Sandra Delgado-Pulido;María Mittelbrunn. Mitochondria during T cell aging.. Seminars in immunology(IF=7.8). 2023. PMID:37473558. DOI: 10.1016/j.smim.2023.101808.
  • [51] Zhejun Ji;Guang-Hui Liu;Jing Qu. Mitochondrial sirtuins, key regulators of aging.. Life medicine(IF=6.0). 2025. PMID:40799515. DOI: 10.1093/lifemedi/lnaf019.
  • [52] Indumathi Somasundaram;Samatha M Jain;Marcel Blot-Chabaud;Surajit Pathak;Antara Banerjee;Sonali Rawat;Neeta Raj Sharma;Asim K Duttaroy. Mitochondrial dysfunction and its association with age-related disorders.. Frontiers in physiology(IF=3.4). 2024. PMID:39015222. DOI: 10.3389/fphys.2024.1384966.
  • [53] Angie K Torres;Claudia Jara;Jesús Llanquinao;Matías Lira;Daniela Cortés-Díaz;Cheril Tapia-Rojas. Mitochondrial Bioenergetics, Redox Balance, and Calcium Homeostasis Dysfunction with Defective Ultrastructure and Quality Control in the Hippocampus of Aged Female C57BL/6J Mice.. International journal of molecular sciences(IF=4.9). 2023. PMID:36982549. DOI: 10.3390/ijms24065476.
  • [54] Lloye M Dillon;Adriana P Rebelo;Carlos T Moraes. The role of PGC-1 coactivators in aging skeletal muscle and heart.. IUBMB life(IF=3.2). 2012. PMID:22279035. DOI: 10.1002/iub.608.
  • [55] David Sebastián;Manuel Palacín;Antonio Zorzano. Mitochondrial Dynamics: Coupling Mitochondrial Fitness with Healthy Aging.. Trends in molecular medicine(IF=13.8). 2017. PMID:28188102. DOI: 10.1016/j.molmed.2017.01.003.
  • [56] Lanlan Zhang;Jianlong Wu;Ziguan Zhu;Yuchen He;Renpeng Fang. Mitochondrion: A bridge linking aging and degenerative diseases.. Life sciences(IF=5.1). 2023. PMID:37030614. DOI: 10.1016/j.lfs.2023.121666.
  • [57] Caio M P F Batalha;Anibal Eugênio Vercesi;Nadja C Souza-Pinto. The Many Roles Mitochondria Play in Mammalian Aging.. Antioxidants & redox signaling(IF=6.1). 2022. PMID:34715739. DOI: 10.1089/ars.2021.0074.
  • [58] Guo Chen;Guido Kroemer;Oliver Kepp. Mitophagy: An Emerging Role in Aging and Age-Associated Diseases.. Frontiers in cell and developmental biology(IF=4.3). 2020. PMID:32274386. DOI: 10.3389/fcell.2020.00200.
  • [59] Claudia Morganti;Keisuke Ito. Mitochondrial Contributions to Hematopoietic Stem Cell Aging.. International journal of molecular sciences(IF=4.9). 2021. PMID:34681777. DOI: 10.3390/ijms222011117.
  • [60] Peter J McGuire. Mitochondrial Dysfunction and the Aging Immune System.. Biology(IF=3.5). 2019. PMID:31083529. DOI: 10.3390/biology8020026.
  • [61] Pei Wei;Xiaoyan Zhang;Chi Yan;Siyu Sun;Zhigang Chen;Fei Lin. Mitochondrial dysfunction and aging: multidimensional mechanisms and therapeutic strategies.. Biogerontology(IF=4.1). 2025. PMID:40634825. DOI: 10.1007/s10522-025-10273-4.
  • [62] Yu Sun;Qingfeng Li;James L Kirkland. Targeting senescent cells for a healthier longevity: the roadmap for an era of global aging.. Life medicine(IF=6.0). 2022. PMID:36699942. DOI: 10.1093/lifemedi/lnac030.
  • [63] Toby Chin;Xin Er Lee;Pei Yi Ng;Yaelim Lee;Oliver Dreesen. The role of cellular senescence in skin aging and age-related skin pathologies.. Frontiers in physiology(IF=3.4). 2023. PMID:38074322. DOI: 10.3389/fphys.2023.1297637.
  • [64] Darya Bulbiankova;Rocío Díaz-Puertas;Francisco Javier Álvarez-Martínez;María Herranz-López;Enrique Barrajón-Catalán;Vicente Micol. Hallmarks and Biomarkers of Skin Senescence: An Updated Review of Skin Senotherapeutics.. Antioxidants (Basel, Switzerland)(IF=6.6). 2023. PMID:36830002. DOI: 10.3390/antiox12020444.
  • [65] Margherita Protasoni;Manuel Serrano. Targeting Mitochondria to Control Ageing and Senescence.. Pharmaceutics(IF=5.5). 2023. PMID:36839673. DOI: 10.3390/pharmaceutics15020352.
  • [66] Antoneta Granic;Karen Suetterlin;Tea Shavlakadze;Miranda D Grounds;Avan A Sayer. Hallmarks of ageing in human skeletal muscle and implications for understanding the pathophysiology of sarcopenia in women and men.. Clinical science (London, England : 1979)(IF=7.7). 2023. PMID:37986616. DOI: 10.1042/CS20230319.
  • [67] Teow J Phua. Hallmarks of aging: middle-aging hypovascularity, tissue perfusion and nitric oxide perspective on healthspan.. Frontiers in aging(IF=4.3). 2024. PMID:39839443. DOI: 10.3389/fragi.2024.1526230.
  • [68] Yajuan Zhang;Yaoyao Jiang;Xiaoyue Yang;Yumei Huang;An Pan;Yunfei Liao. Adipose tissue senescence: Biological changes, hallmarks and therapeutic approaches.. Mechanisms of ageing and development(IF=5.1). 2024. PMID:39265709. DOI: 10.1016/j.mad.2024.111988.
  • [69] Derrick J Rossi;David Bryder;Jun Seita;Andre Nussenzweig;Jan Hoeijmakers;Irving L Weissman. Deficiencies in DNA damage repair limit the function of haematopoietic stem cells with age.. Nature(IF=48.5). 2007. PMID:17554309. DOI: 10.1038/nature05862.
  • [70] Jeyan Jayarajan;Michael D Milsom. The role of the stem cell epigenome in normal aging and rejuvenative therapy.. Human molecular genetics(IF=3.2). 2020. PMID:32744315. DOI: 10.1093/hmg/ddaa167.
  • [71] Miren Revuelta;Ander Matheu. Autophagy in stem cell aging.. Aging cell(IF=7.1). 2017. PMID:28782921. DOI: 10.1111/acel.12655.
  • [72] Monica Boyle;Chihunt Wong;Michael Rocha;D Leanne Jones. Decline in self-renewal factors contributes to aging of the stem cell niche in the Drosophila testis.. Cell stem cell(IF=20.4). 2007. PMID:18371382. DOI: 10.1016/j.stem.2007.08.002.
  • [73] Jason Doles;Mekayla Storer;Luca Cozzuto;Guglielmo Roma;William M Keyes. Age-associated inflammation inhibits epidermal stem cell function.. Genes & development(IF=7.7). 2012. PMID:22972935. DOI: 10.1101/gad.192294.112.
  • [74] Feng Chen;Yingxia Liu;Nai-Kei Wong;Jia Xiao;Kwok-Fai So. Oxidative Stress in Stem Cell Aging.. Cell transplantation(IF=3.2). 2017. PMID:29113471. DOI: 10.1177/0963689717735407.
  • [75] Thomas A Rando;Anne Brunet;Margaret A Goodell. Hallmarks of stem cell aging.. Cell stem cell(IF=20.4). 2025. PMID:40562035. DOI: 10.1016/j.stem.2025.06.004.
  • [76] Jéssica M Vilas;Carmen Carneiro;Sabela Da Silva-Álvarez;Alba Ferreirós;Patricia González;María Gómez;Sagrario Ortega;Manuel Serrano;Tomás García-Caballero;Miguel González-Barcia;Anxo Vidal;Manuel Collado. Adult Sox2+ stem cell exhaustion in mice results in cellular senescence and premature aging.. Aging cell(IF=7.1). 2018. PMID:30129215. DOI: 10.1111/acel.12834.
  • [77] Jitendra Kumar Chaudhary;Ajay Kumar Danga;Anita Kumari;Akshay Bhardwaj;Pramod C Rath. Role of stem cells in ageing and age-related diseases.. Mechanisms of ageing and development(IF=5.1). 2025. PMID:40324541. DOI: 10.1016/j.mad.2025.112069.

MaltSci Intelligent Research Services

Search for more papers on MaltSci.com

hallmarks of aging · genomic instability · mitochondrial dysfunction · cellular senescence · chronic inflammation


© 2025 MaltSci