Appearance
This report is written by MaltSci based on the latest literature and research findings
What are the molecular mechanisms of aging?
Abstract
Aging is a complex biological phenomenon characterized by a progressive decline in physiological functions and increased susceptibility to age-related diseases. This review explores the molecular mechanisms underlying aging, emphasizing the interplay of genetic, environmental, and lifestyle factors. Key mechanisms include oxidative stress, telomere shortening, mitochondrial dysfunction, epigenetic changes, and chronic inflammation, which contribute to cellular dysfunction and the onset of age-related pathologies such as cardiovascular diseases, neurodegenerative disorders, and cancer. Oxidative stress arises from an imbalance between reactive oxygen species production and antioxidant defenses, leading to cumulative cellular damage. Telomere shortening, resulting from repeated cell divisions, triggers cellular senescence, while mitochondrial dysfunction impairs energy production and increases oxidative stress. Epigenetic alterations influence gene expression patterns associated with aging, and chronic inflammation exacerbates age-related decline through the senescence-associated secretory phenotype. Lifestyle factors, particularly dietary and physical activity interventions, modulate these aging pathways and offer potential strategies for promoting healthy aging. Emerging research areas, including senolytics, gene therapy, and the microbiome's role in aging, present exciting opportunities for innovative therapeutic approaches. This comprehensive review aims to synthesize existing literature and identify future research directions that could lead to effective interventions for age-related conditions, ultimately enhancing the health span of the aging population.
Outline
This report will discuss the following questions.
- 1 Introduction
- 2 Molecular Pathways of Aging
- 2.1 Oxidative Stress and Aging
- 2.2 Telomere Shortening and Cellular Senescence
- 2.3 Mitochondrial Dysfunction
- 2.4 Epigenetic Alterations
- 3 The Role of Inflammation in Aging
- 3.1 Chronic Inflammation and the Inflammaging Concept
- 3.2 Impact of Inflammatory Cytokines on Aging
- 4 Lifestyle Factors Influencing Aging
- 4.1 Diet and Nutritional Interventions
- 4.2 Physical Activity and its Molecular Impact
- 5 Emerging Research Areas
- 5.1 Senolytics and Senomorphics
- 5.2 Gene Therapy and Genetic Interventions
- 5.3 The Microbiome and Aging
- 6 Future Directions and Challenges
- 6.1 Translational Research in Aging
- 6.2 Ethical Considerations in Anti-Aging Therapies
- 7 Conclusion
1 Introduction
Aging is a complex biological phenomenon characterized by a progressive decline in physiological functions and an increased susceptibility to age-related diseases. This multifaceted process is not merely a consequence of time but is influenced by an intricate interplay of genetic, environmental, and lifestyle factors. As populations globally continue to age, understanding the molecular mechanisms that underlie aging has become a pressing scientific challenge. Research indicates that aging is associated with several hallmark features, including genomic instability, telomere shortening, mitochondrial dysfunction, epigenetic changes, and chronic inflammation [1][2]. These mechanisms contribute to the gradual deterioration of cellular function, ultimately leading to the onset of age-related pathologies such as cardiovascular diseases, neurodegenerative disorders, and cancer [3][4].
The significance of understanding the molecular underpinnings of aging cannot be overstated. Insights into these mechanisms hold the potential to inform the development of therapeutic interventions aimed at promoting healthy aging and extending lifespan. For instance, interventions targeting oxidative stress, a key contributor to cellular aging, have shown promise in preclinical models [1]. Furthermore, the exploration of natural compounds that modulate aging pathways offers a novel approach to enhance longevity and mitigate age-related diseases [2]. As the prevalence of age-associated diseases rises, elucidating the molecular pathways involved in aging is essential for advancing public health strategies and improving the quality of life for older adults.
Current research has made significant strides in identifying various molecular pathways involved in aging. These include oxidative stress, which results from an imbalance between reactive oxygen species (ROS) production and antioxidant defenses, leading to cellular damage [1]. Telomere shortening, a consequence of repeated cell division, plays a critical role in cellular senescence and the aging process [5]. Mitochondrial dysfunction is another crucial aspect, as mitochondria are not only the powerhouses of the cell but also significant sources of ROS [1]. Additionally, epigenetic alterations can influence gene expression patterns that contribute to aging, while chronic inflammation, often referred to as "inflammaging," exacerbates age-related decline [2][6].
This review is organized into several sections to comprehensively address the molecular mechanisms of aging. The first section delves into the key molecular pathways of aging, including oxidative stress, telomere shortening, mitochondrial dysfunction, and epigenetic changes. Following this, we explore the role of inflammation in aging, discussing the concept of inflammaging and the impact of inflammatory cytokines on the aging process. The subsequent section examines lifestyle factors influencing aging, particularly dietary and physical activity interventions that can modulate aging at the molecular level. Emerging research areas are also highlighted, including the potential of senolytics and senomorphics, gene therapy, and the microbiome's influence on aging. Finally, we address future directions and challenges in aging research, emphasizing the need for translational studies and ethical considerations in developing anti-aging therapies.
By synthesizing existing literature and identifying gaps in knowledge, this review aims to provide a thorough understanding of the molecular mechanisms of aging. Through this comprehensive approach, we hope to illuminate potential avenues for future research that could lead to innovative therapeutic strategies for age-related conditions, ultimately enhancing the health span of the aging population.
2 Molecular Pathways of Aging
2.1 Oxidative Stress and Aging
Aging is a complex biological process characterized by the gradual decline of physiological functions and an increased susceptibility to age-related diseases. One of the central mechanisms implicated in the aging process is oxidative stress, which arises from an imbalance between the production of reactive oxygen species (ROS) and the body's antioxidant defense systems. This imbalance leads to molecular damage, affecting various cellular components such as DNA, proteins, and lipids, ultimately contributing to the aging phenotype.
Oxidative stress is considered a fundamental mechanism driving aging due to its capacity to induce cumulative damage at the cellular level. This damage can lead to malfunctioning cellular machinery, affecting tissues and organs, and thereby accelerating the aging process (Mehdi et al. 2021). The evidence supporting the role of oxidative stress in aging includes observations that the accumulation of oxidative damage correlates with age-related decline in physiological functions and increased disease incidence (Kourtis & Tavernarakis 2011).
Several studies have highlighted the relationship between oxidative stress and specific aging-related pathologies. For instance, oxidative stress has been linked to cognitive decline, physical frailty, and other degenerative conditions associated with aging (Tan et al. 2018). Moreover, it has been observed that genetic and environmental factors can modulate the levels of oxidative stress, thereby influencing individual aging trajectories (Płóciniczak et al. 2025).
Molecularly, oxidative stress impacts various cellular pathways. The accumulation of oxidative damage to proteins can lead to loss of function, as oxidized proteins are preferentially degraded (Sohal 2002). Additionally, oxidative stress is known to activate stress response pathways, including those mediated by the p53 protein, which plays a dual role depending on the levels of oxidative stress—exhibiting antioxidant activities at low levels and pro-oxidative activities at high levels (Liu & Xu 2011). This context-dependent regulation of p53 highlights the intricate balance between oxidative stress and cellular responses that can either promote survival or lead to cell death.
Antioxidants, both endogenous and exogenous, are critical in mitigating oxidative stress and its associated damage. They function by scavenging free radicals, thereby reducing oxidative damage and promoting healthy aging (Warraich et al. 2020). The modulation of antioxidant defense mechanisms has been proposed as a potential therapeutic strategy to delay the aging process and improve healthspan, although the evidence regarding the effectiveness of such interventions remains mixed (Iakovou & Kourti 2022).
In summary, the molecular mechanisms of aging are significantly influenced by oxidative stress, which contributes to cellular and molecular damage over time. The interplay between oxidative stress, cellular responses, and genetic factors shapes the aging process, making it a critical area of study for developing interventions aimed at promoting healthy aging and mitigating age-related diseases. Understanding these complex relationships is essential for identifying potential biomarkers of aging and devising personalized strategies to enhance longevity and healthspan (Mehdi et al. 2023; Płóciniczak et al. 2025).
2.2 Telomere Shortening and Cellular Senescence
Aging is a complex biological process characterized by a progressive decline in the functional capacity of tissues and organs, ultimately leading to increased mortality. Central to this process is the phenomenon of telomere shortening, which plays a critical role in cellular senescence and contributes to various age-related diseases.
Telomeres are repetitive DNA sequences located at the ends of linear eukaryotic chromosomes, serving to protect chromosome ends from degradation and prevent illegitimate recombination. As cells undergo division, telomeres shorten due to the end-replication problem inherent in DNA replication. This shortening leads to cellular senescence, a state where cells lose their ability to proliferate, thereby contributing to tissue degeneration and dysfunction. The progressive loss of telomeres is closely linked to the aging process, with telomere attrition being a fundamental mechanism driving cellular aging (Zhu et al., 2019; Xi et al., 2013) [5][7].
Cellular senescence, defined as an irreversible arrest of the cell cycle, is influenced by various factors, including oxidative stress, DNA damage, inflammation, and telomere shortening. When telomeres reach a critically short length, they trigger DNA damage responses that activate cell cycle checkpoints, leading to senescence. This process serves as a protective mechanism against genomic instability and cancer but also limits the regenerative capacity of tissues, thus exacerbating age-related decline (Fyhrquist et al., 2013; von Figura et al., 2009) [8][9].
The interplay between telomere shortening and mitochondrial dysfunction further complicates the aging process. Mitochondria, as the metabolic centers of cells, are significantly affected by the loss of telomere integrity. Dysfunctional mitochondria contribute to increased oxidative stress, which in turn accelerates telomere shortening. This crosstalk between telomeres and mitochondrial metabolism highlights the multifaceted nature of cellular aging and underscores the importance of maintaining mitochondrial health in the context of aging (Gao et al., 2022) [10].
Moreover, telomere shortening is implicated in various age-related diseases, including cardiovascular diseases, neurodegeneration, and cancer. The accumulation of critically short telomeres is proposed as a primary molecular cause of these conditions, with telomere maintenance gene mutations being associated with specific pathologies termed telomeropathies (Martínez & Blasco, 2018) [11]. The senescence-associated secretory phenotype (SASP) of senescent cells can promote chronic inflammation and tissue remodeling, contributing to the pathogenesis of age-related diseases (Hornsby, 2011) [12].
In summary, the molecular mechanisms of aging, particularly through telomere shortening and subsequent cellular senescence, involve a complex interplay of genetic, metabolic, and environmental factors. Understanding these pathways provides insights into potential therapeutic strategies aimed at mitigating the effects of aging and enhancing healthspan.
2.3 Mitochondrial Dysfunction
Aging is a complex biological process characterized by the progressive decline of physiological functions, which is closely associated with various molecular mechanisms, notably mitochondrial dysfunction. Mitochondria play a central role in cellular energy production and regulation of metabolic processes, and their dysfunction has been implicated in the aging process and age-related diseases.
Mitochondrial dysfunction contributes to aging through several interconnected pathways. Firstly, as individuals age, there is an accumulation of dysfunctional mitochondria, which leads to decreased efficiency in oxidative phosphorylation and an increase in the production of reactive oxygen species (ROS) [13]. This imbalance in ROS production and clearance results in oxidative stress, causing damage to cellular components, including lipids, proteins, and DNA [1]. Such oxidative damage is a key factor in the aging phenotype, contributing to cellular senescence and the development of age-related diseases [14].
Moreover, mitochondrial dysfunction affects the cellular signaling pathways involved in aging. It has been observed that mitochondrial signaling components, such as mitochondrial DNA, ROS, and mitochondrial metabolites, play critical roles in immune system activation and chronic inflammation, a phenomenon termed "inflammageing" [15]. This low-grade inflammation further exacerbates age-related pathologies, including neurodegenerative diseases and sarcopenia [15].
Another significant aspect of mitochondrial dysfunction in aging is the disruption of mitochondrial quality control mechanisms, including mitochondrial biogenesis, dynamics, and mitophagy. With aging, the efficiency of these processes declines, leading to an accumulation of damaged mitochondria, which in turn contributes to cellular dysfunction [13].
Mitochondrial dynamics, including fission and fusion, are essential for maintaining mitochondrial function and integrity. Disruption in these dynamics has been linked to age-related decline in cellular homeostasis [14]. For instance, the proteins involved in mitochondrial fission (such as DRP1) and fusion (such as MFN1 and OPA1) are crucial for adapting to cellular stress and maintaining mitochondrial function. Aging is associated with altered expression and activity of these proteins, leading to impaired mitochondrial morphology and function [13].
In addition to these pathways, metabolic reprogramming due to mitochondrial dysfunction has been identified as a contributor to aging. Mitochondria influence metabolic pathways, and their dysfunction can lead to altered energy metabolism, further promoting age-related diseases [16]. The sirtuin family of proteins, which are involved in the regulation of mitochondrial homeostasis, also plays a significant role in the aging process. They modulate various cellular pathways that influence mitochondrial function and cellular senescence [16].
Furthermore, the senescence-associated secretory phenotype (SASP), characterized by the secretion of pro-inflammatory cytokines and other factors by senescent cells, is influenced by mitochondrial dysfunction. This secretion contributes to the aging process by promoting a pro-inflammatory environment, which can exacerbate tissue degeneration [16].
In summary, mitochondrial dysfunction is a pivotal mechanism in the aging process, influencing various pathways including oxidative stress, inflammatory responses, metabolic reprogramming, and cellular senescence. Understanding these interconnected pathways provides valuable insights into potential therapeutic strategies aimed at mitigating the effects of aging and associated diseases by targeting mitochondrial health and function [17].
2.4 Epigenetic Alterations
Aging is a multifaceted biological process characterized by progressive decline in tissue and organ function, which is closely linked to a series of molecular alterations, particularly epigenetic changes. Epigenetic mechanisms play a pivotal role in regulating gene expression without altering the underlying DNA sequence, thereby influencing various cellular processes associated with aging.
Epigenetic alterations encompass a range of modifications, including DNA methylation, histone modifications, chromatin remodeling, and the regulation of non-coding RNAs. These modifications are crucial for maintaining genomic stability and proper gene expression. As organisms age, these epigenetic marks can become dysregulated, leading to aberrant gene expression patterns that contribute to the aging process and age-related diseases.
One of the primary epigenetic changes observed during aging is DNA methylation, which involves the addition of methyl groups to cytosine residues in DNA. This process can lead to the silencing of genes essential for maintaining cellular functions. Studies have demonstrated that age-related changes in DNA methylation can serve as biomarkers for biological age, highlighting their potential in longevity research and age-associated disorder diagnostics [18].
Histone modifications, another critical aspect of epigenetic regulation, include the addition or removal of chemical groups to histone proteins, which can alter chromatin structure and accessibility. These modifications influence transcriptional activity and can result in changes to cellular plasticity, particularly in neuronal contexts, where they impact memory and cognitive functions [19]. As individuals age, the patterns of histone modifications can become disrupted, contributing to the decline in synaptic plasticity and memory formation [19].
Moreover, chromatin remodeling, which refers to the dynamic restructuring of chromatin, is also affected by aging. The loss of histones and alterations in nucleosome occupancy have been identified as key processes that exacerbate genomic instability and contribute to the aging phenotype [20]. The emergence of noncanonical histone variants and disruptions in higher-order chromatin architecture are indicative of the complex interplay between histone dynamics and aging [20].
The relationship between epigenetic alterations and aging is further complicated by the impact of environmental factors. Lifestyle choices, such as diet and exercise, have been shown to influence epigenetic marks, suggesting that interventions targeting epigenetic mechanisms may offer potential strategies for delaying aging and mitigating age-related diseases [21]. For instance, caloric restriction has been recognized as an effective intervention that induces beneficial epigenetic changes, thereby extending lifespan in various model organisms [22].
In conclusion, the molecular mechanisms of aging, particularly through the lens of epigenetic alterations, reveal a complex network of interactions that govern cellular aging processes. Understanding these mechanisms not only enhances our comprehension of aging but also opens avenues for therapeutic interventions aimed at promoting healthy aging and addressing age-associated diseases. The reversible nature of epigenetic modifications presents promising opportunities for developing strategies to counteract the effects of aging and improve healthspan [23].
3 The Role of Inflammation in Aging
3.1 Chronic Inflammation and the Inflammaging Concept
Aging is increasingly recognized as a complex biological process characterized by a progressive decline in physiological functions, which predisposes individuals to a variety of chronic diseases. One of the critical components of this process is chronic inflammation, often referred to as "inflammaging." This term describes a state of low-grade, chronic inflammation that occurs in the absence of overt infection and is associated with aging. The underlying molecular mechanisms of aging, particularly concerning inflammation, involve several interconnected pathways and cellular processes.
Chronic inflammation in aging is primarily driven by the activation of the innate immune system, which can be triggered by various factors, including cellular debris, damaged organelles, and pro-inflammatory cytokines. The accumulation of such stimuli leads to the activation of pattern recognition receptors and inflammasomes, such as the NLRP3 inflammasome, which play a significant role in the inflammatory response. This chronic activation results in the secretion of pro-inflammatory mediators like TNF-alpha, IL-1beta, and IL-6, contributing to a sustained inflammatory state that can exacerbate age-related diseases [24][25].
Furthermore, aging is associated with cellular senescence, a process where cells lose their ability to divide and function effectively. Senescent cells secrete a variety of inflammatory factors, collectively known as the senescence-associated secretory phenotype (SASP), which further perpetuates inflammation and contributes to the deterioration of tissue homeostasis [15]. The interplay between cellular senescence and chronic inflammation creates a vicious cycle, where inflammation promotes senescence, and senescent cells, in turn, amplify inflammation [26].
Mitochondrial dysfunction is another key mechanism implicated in inflammaging. Aging is characterized by a decline in mitochondrial function, leading to increased production of reactive oxygen species (ROS) and altered mitochondrial signaling. This dysfunction not only contributes to oxidative stress but also enhances the production of pro-inflammatory cytokines, thereby sustaining the chronic inflammatory state associated with aging [15][27].
Moreover, the gut microbiota plays a pivotal role in the aging process and its associated inflammation. Changes in gut microbiota composition with age can lead to dysbiosis, which is linked to increased intestinal permeability and systemic inflammation. The release of microbial products can trigger inflammatory responses, further linking gut health to systemic inflammatory processes and aging [28][29].
In summary, the molecular mechanisms of aging, particularly through the lens of chronic inflammation and the concept of inflammaging, involve a multifaceted interplay of immune activation, cellular senescence, mitochondrial dysfunction, and gut microbiota alterations. Understanding these interconnected pathways is crucial for developing potential therapeutic interventions aimed at mitigating the effects of aging and promoting healthy longevity [27][30].
3.2 Impact of Inflammatory Cytokines on Aging
Aging is a complex biological process characterized by a progressive decline in physiological functions, and it is increasingly recognized that inflammation plays a crucial role in this process. The phenomenon known as "inflammaging" describes the chronic, low-grade inflammation that accompanies aging, which is linked to various age-related diseases and the overall decline in health status. Inflammatory cytokines are central to this process, mediating intercellular communication and influencing immune responses.
One of the primary mechanisms by which inflammation affects aging is through the dysregulation of cytokine production. Cytokines are small glycoproteins secreted mainly by immune cells, and they exert both pro-inflammatory and anti-inflammatory effects. With aging, there is often a shift towards a pro-inflammatory cytokine profile, which has been associated with various age-related diseases such as cardiovascular diseases, neurodegenerative disorders, and metabolic syndromes. This shift is characterized by elevated levels of cytokines such as interleukin (IL)-6, tumor necrosis factor-alpha (TNF-α), and IL-1, which contribute to tissue damage and the progression of age-related pathologies (Michaud et al., 2013; Rea et al., 2018).
The role of mitochondria in aging and inflammation has also been highlighted. Mitochondrial dysfunction, a hallmark of aging, contributes to the production of reactive oxygen species (ROS) and the release of pro-inflammatory cytokines, further perpetuating the inflammatory state (Kalykaki et al., 2024). Mitochondrial signaling components, including mitochondrial DNA and metabolites, play a significant role in regulating immune responses and the secretion of cytokines. This dysregulation can lead to an exacerbation of chronic inflammation and contribute to cellular senescence, a state where cells lose their ability to divide and function properly (Poulios et al., 2025).
Moreover, cytokine dysregulation is linked to immunosenescence, the age-related decline in immune function. As individuals age, the immune system becomes less capable of effectively responding to pathogens and repairing tissues, leading to an increased susceptibility to infections and diseases. This decline is accompanied by a persistent inflammatory state, where the body is unable to resolve inflammation effectively, resulting in tissue damage and accelerated aging (Chaudhary et al., 2025).
The relationship between nutrition and inflammation also plays a significant role in aging. Malnutrition and imbalances in nutrient intake can exacerbate the inflammatory state, contributing to the decline in health observed in the elderly. For instance, specific cytokines have been shown to modulate appetite and nutrient sensing, indicating that the inflammatory status of an individual can influence their nutritional health and, consequently, their aging trajectory (Coperchini et al., 2025).
In summary, the molecular mechanisms of aging are significantly influenced by inflammation and the dysregulation of cytokines. The chronic inflammatory state associated with aging, characterized by an imbalance of pro-inflammatory and anti-inflammatory cytokines, contributes to the pathogenesis of age-related diseases and the overall decline in physiological functions. Understanding these mechanisms offers potential therapeutic avenues for mitigating the effects of aging and promoting healthier aging trajectories through anti-inflammatory strategies and nutritional interventions.
4 Lifestyle Factors Influencing Aging
4.1 Diet and Nutritional Interventions
Aging is a multifactorial biological process characterized by various molecular mechanisms that contribute to cellular senescence, oxidative stress, inflammation, and genomic instability. These processes are influenced significantly by lifestyle factors, particularly diet and nutritional interventions, which have been shown to modulate aging pathways and impact healthspan.
One of the primary mechanisms of aging involves oxidative stress, which results from an imbalance between reactive oxygen species (ROS) production and the body's ability to detoxify these reactive products. Dietary restriction has been identified as a potent modulator of oxidative stress, with studies indicating that reduced caloric intake can extend lifespan by decreasing oxidative damage [31]. The free radical theory of aging posits that oxidative damage accumulates over time, leading to age-related functional decline [31].
In addition to oxidative stress, chronic inflammation plays a critical role in aging. The concept of "inflammaging" refers to the low-grade, chronic inflammation that often accompanies aging and contributes to the development of age-related diseases. Nutritional components, such as polyphenols found in various foods, exhibit anti-inflammatory properties and may help mitigate these effects [32].
Cellular senescence, a state in which cells lose the ability to divide and function properly, is another crucial aspect of aging. It is associated with various pathologies, including neurodegenerative diseases and cardiovascular disorders. Nutrition has been shown to influence cellular senescence through various pathways, including the modulation of key metabolic pathways and the removal of senescent cells [33]. For instance, certain bioactive nutrients can selectively target these pathways, promoting healthier aging [34].
The role of the gut microbiota in aging is increasingly recognized, as it can influence systemic inflammation and metabolic health. Dietary patterns, such as the Mediterranean diet, have been associated with beneficial effects on gut microbiota diversity and composition, which in turn can modulate aging-related processes [35]. This diet, rich in fruits, vegetables, whole grains, and healthy fats, supports the growth of beneficial microbes that produce short-chain fatty acids, which have protective effects against inflammation and metabolic disorders [36].
Moreover, caloric restriction and the intake of specific bioactive compounds, such as resveratrol and curcumin, have been linked to the activation of longevity pathways, including sirtuins and AMPK. These compounds can enhance autophagy, a process that clears damaged cellular components, thus promoting cellular health and longevity [2].
The interplay between diet and aging is complex, as genetic and environmental factors also influence how dietary components affect aging. For example, variations in insulin/IGF-1 signaling and other metabolic pathways can mediate the impact of nutrients on lifespan and health [37]. Therefore, understanding these molecular mechanisms and the effects of nutritional interventions can provide insights into strategies for promoting healthy aging and delaying age-related diseases.
In conclusion, the molecular mechanisms of aging are influenced by a variety of factors, with diet playing a significant role in modulating these processes. Nutritional interventions can target oxidative stress, inflammation, cellular senescence, and gut microbiota composition, ultimately contributing to healthier aging and improved quality of life in the elderly population. Future research should continue to explore these interactions to develop effective dietary strategies for aging populations.
4.2 Physical Activity and its Molecular Impact
Aging is characterized by a progressive decline in physiological function and an accumulation of molecular damage across various biological systems. The molecular mechanisms underlying aging are complex and multifaceted, involving genetic, epigenetic, and environmental factors. Key mechanisms include oxidative stress, inflammation, telomere attrition, mitochondrial dysfunction, and cellular senescence.
Oxidative stress arises from the accumulation of reactive oxygen species (ROS), which can damage cellular components such as DNA, proteins, and lipids, leading to functional impairments. Inflammation is another hallmark of aging, where chronic low-grade inflammation can disrupt homeostasis and contribute to the development of age-related diseases. Telomere shortening, which occurs with each cell division, limits the replicative capacity of cells and is associated with cellular senescence, a state where cells lose their ability to proliferate and function properly. Mitochondrial dysfunction, characterized by reduced energy production and increased ROS generation, further exacerbates the aging process by impairing cellular metabolism and promoting apoptosis.
Recent research highlights the significant role of lifestyle factors, particularly physical activity, in modulating these molecular mechanisms of aging. Regular physical exercise has been shown to improve mitochondrial function, enhance antioxidant defenses, and reduce markers of inflammation. For instance, physical activity increases the expression of neurotrophic factors and promotes neuroplasticity, which can counteract cognitive decline associated with aging (Kaliman et al. 2011). Additionally, exercise has been linked to changes in epigenetic markers, including DNA methylation and histone modifications, that can influence gene expression related to aging and longevity (Li et al. 2011).
Moreover, physical activity can enhance the body’s ability to manage oxidative stress by upregulating antioxidant enzymes and reducing the accumulation of ROS (Pangrazzi & Meryk 2024). This interplay between exercise and molecular mechanisms underscores the potential of lifestyle interventions in mitigating the effects of aging and promoting healthspan.
In summary, the molecular mechanisms of aging involve a complex interplay of oxidative stress, inflammation, telomere attrition, and mitochondrial dysfunction. Lifestyle factors, particularly physical activity, play a crucial role in influencing these mechanisms, thereby offering potential strategies for promoting healthy aging and longevity. Integrating regular exercise into daily routines may help counteract the detrimental effects of aging at the molecular level, contributing to improved health outcomes in older adults.
5 Emerging Research Areas
5.1 Senolytics and Senomorphics
Aging is a multifaceted biological process characterized by progressive physiological decline and increased susceptibility to various diseases. One of the primary mechanisms underlying aging is cellular senescence, which involves a stable exit from the cell cycle in response to cellular damage and stress. This phenomenon leads to the accumulation of senescent cells (SnCs) that secrete a range of pro-inflammatory molecules, collectively known as the senescence-associated secretory phenotype (SASP). The SASP not only disrupts tissue homeostasis but also promotes a state of chronic inflammation, contributing to age-related pathologies such as neurodegenerative diseases, cancer, and metabolic disorders[38][39].
The molecular regulation of cellular senescence is primarily mediated through pathways such as p53/p21 and p16INK4a/pRb. These pathways are crucial for inducing cell cycle arrest and play protective roles against cancer; however, their prolonged activation can lead to detrimental effects, including the promotion of tissue dysfunction and inflammation[40].
In response to the detrimental effects of senescent cells, emerging therapeutic strategies have focused on targeting these cells through two main approaches: senolytics and senomorphics. Senolytics are compounds that selectively induce apoptosis in senescent cells, thereby reducing their numbers and alleviating the associated inflammatory burden. Conversely, senomorphics aim to modulate the SASP and other deleterious aspects of the senescent phenotype without necessarily eliminating the senescent cells themselves[41][42].
Recent research highlights the potential of various natural and synthetic compounds as senolytics, including tyrosine kinase inhibitors, BCL-2 family inhibitors, and natural polyphenols. Additionally, senomorphics such as mTOR inhibitors and epigenetic modulators have shown promise in mitigating the harmful effects of the SASP[41][43].
Furthermore, the biological heterogeneity of senescent cells poses challenges in developing specific biomarkers for their identification and targeting. This heterogeneity is influenced by factors such as cell type, tissue context, and the temporal dynamics of the SASP, which can exert both pro-inflammatory and immunosuppressive effects[41][42].
Overall, the dual approach of employing senolytics and senomorphics represents a promising strategy in the field of anti-aging interventions. By targeting cellular senescence and its associated pathways, these therapies hold the potential to improve health outcomes and enhance the quality of life as organisms age[40][41].
5.2 Gene Therapy and Genetic Interventions
Aging is a complex biological process characterized by a progressive decline in physiological functions and increased vulnerability to diseases. Various molecular mechanisms have been identified that contribute to aging, which include telomere shortening, genomic instability, mitochondrial dysfunction, oxidative stress, and cellular senescence.
Telomeres, the protective caps at the ends of chromosomes, play a crucial role in cellular aging. As cells divide, telomeres shorten, which eventually leads to cellular senescence and mitochondrial dysfunction. This shortening of telomeres is linked to the decline in cellular function and contributes to the aging phenotype, as described by Zhu et al. (2019) in their review on telomeres and aging pathways[5].
Genomic instability is another significant factor in aging. It encompasses various mutational alterations in the genome, including point mutations, chromosomal aberrations, and loss of telomeric repeats. This instability increases with age and is believed to play a causative role in the aging process, as indicated by Bürkle (2002) who discusses the implications of genomic instability in aging[44].
Mitochondrial dysfunction is also a key mechanism associated with aging. Mitochondria are the primary source of reactive oxygen species (ROS), and as aging progresses, there is a decline in ATP production alongside an increase in ROS, leading to oxidative stress. This oxidative stress causes damage to cellular components, which further accelerates the aging process[1].
Oxidative stress is characterized by an imbalance between ROS production and the cell's antioxidant defenses. Elevated ROS levels can damage DNA, lipids, and proteins, contributing to the aging phenotype. Research has shown that oxidative stress plays a significant role in age-related diseases, including neurodegenerative disorders[45].
Cellular senescence, the process by which cells lose the ability to divide and function properly, is another hallmark of aging. Senescent cells can secrete pro-inflammatory factors that contribute to tissue dysfunction and promote age-related diseases. This phenomenon is associated with various stressors, including oxidative stress and telomere shortening[6].
Moreover, aging is influenced by lifestyle factors, which can modulate these molecular pathways. For instance, lifestyle modifications such as exercise and caloric restriction have been shown to have anti-aging effects by improving metabolic health and reducing the risk of age-related diseases[46].
Emerging research areas, particularly gene therapy and genetic interventions, are exploring ways to manipulate these molecular mechanisms to extend healthspan and lifespan. For example, targeting pathways associated with oxidative stress and inflammation may offer new therapeutic avenues to mitigate the effects of aging and prevent age-related diseases[2].
In summary, the molecular mechanisms of aging are multifaceted and interconnected, involving telomere attrition, genomic instability, mitochondrial dysfunction, oxidative stress, and cellular senescence. Understanding these processes is crucial for developing interventions aimed at promoting healthy aging and extending lifespan.
5.3 The Microbiome and Aging
Aging is a multifaceted biological process characterized by the gradual decline in physiological functions and increased susceptibility to various diseases. Recent studies have increasingly highlighted the significant role of the microbiome in influencing aging processes and associated molecular mechanisms.
The gut microbiota undergoes notable changes with aging, including alterations in microbial composition and diversity, which can lead to dysbiosis. This dysbiosis has been linked to various age-related diseases, such as cardiovascular, neurodegenerative, and metabolic disorders. For instance, as described by Gyriki et al. (2025), dysbiosis can significantly impact health by altering immune responses and promoting systemic inflammation, a phenomenon often referred to as "inflammaging" [47].
Molecularly, the interactions between the host and microbiota can modulate key metabolic pathways. Best et al. (2025) demonstrated that aging is associated with a pronounced reduction in metabolic activity within the microbiome, which coincides with decreased beneficial interactions among bacterial species. This reduction in microbial metabolic function is critical, as it is linked to the downregulation of essential host pathways, particularly those involved in nucleotide metabolism. Such pathways are vital for maintaining intestinal barrier integrity, cellular replication, and overall homeostasis [48].
Moreover, the metabolites produced by gut microbiota play crucial roles in signaling pathways that regulate aging and longevity. Zhou et al. (2021) reviewed how specific metabolites act as signaling molecules that can actively influence longevity and health outcomes. This suggests that the metabolites derived from the microbiome are integral to understanding the molecular mechanisms of aging [49].
The microbiota-gut-brain axis also emerges as a critical area of research in understanding aging-related behavioral changes. Jing et al. (2024) highlighted how age-induced dysbiosis can lead to alterations in the production of short-chain fatty acids (SCFAs), which are essential for maintaining gut-brain communication. Changes in SCFA levels are associated with neuroinflammation and cognitive decline, linking gut microbiota composition directly to neurological health in aging [50].
Additionally, interventions targeting the microbiome, such as probiotics and dietary modifications, show promise in mitigating age-related declines. Ramasinghe et al. (2025) emphasized the potential of specific dietary patterns, like the Mediterranean diet, in enhancing microbial diversity and function, thereby promoting healthier aging outcomes [36].
In conclusion, the molecular mechanisms of aging are intricately linked to the dynamics of the microbiome. Changes in microbial composition, the metabolites produced, and the resulting effects on systemic inflammation and metabolic pathways collectively contribute to the aging process. Ongoing research continues to unravel these complex interactions, paving the way for novel therapeutic strategies aimed at promoting healthy aging through microbiome modulation.
6 Future Directions and Challenges
6.1 Translational Research in Aging
Aging is a multifaceted biological process characterized by a progressive decline in cellular and physiological functions, which is accompanied by a series of molecular alterations. Several molecular mechanisms have been identified as pivotal contributors to the aging process, including genomic instability, loss of telomere function, epigenetic changes, increased cellular senescence, depletion of the stem cell pool, altered intercellular communication, and loss of protein homeostasis [3].
One significant area of focus is the role of the translation machinery in aging. Alterations in the ribosome and protein biosynthesis are central to understanding how aging affects cellular function. The ribosome, a conserved RNA-protein complex, is crucial for accurate polypeptide synthesis and co-translational protein folding. Age-related changes in the translation apparatus can influence the rate and selectivity of protein synthesis, which has implications for cellular senescence and overall organismal aging [3].
In a comprehensive study analyzing the proteomic and metabolomic profiles of ten mouse organs across different life stages, it was found that aging is associated with significant molecular dynamics, including alterations in protein expression and metabolite levels. For instance, 18 proteins exhibited consistent age-related differential expression across multiple organs, with functional enrichment analysis indicating that the humoral immune response is a primary driver of these changes [51]. Furthermore, metabolites such as NAD+, inosine, and hypoxanthine showed significant expression changes during aging, suggesting that metabolic pathways are also crucial in the aging process [51].
Another important molecular mechanism involves the high mobility group Box 1 protein (HMGB1), which plays diverse roles in cellular homeostasis and is associated with aging and age-related diseases. HMGB1 acts as a DNA chaperone and regulator of gene transcription in the nucleus, while also being involved in autophagy and inflammation in the cytoplasm. Its expression levels change with age, indicating its potential as a universal biomarker for aging [6].
Moreover, the relationship between aging and translational control is underscored by the integrated stress response and pathways mediated by mechanistic target of rapamycin (mTOR) and elongation factor 2 kinase. These pathways are critical for maintaining cellular homeostasis and respond to environmental stresses, thereby influencing the aging process [52].
As the field of aging research continues to evolve, future directions should focus on elucidating the interconnectedness of these molecular mechanisms. There is a pressing need for translational research that bridges the gap between basic science and clinical applications, particularly in understanding how these molecular changes can be targeted to promote healthy aging and mitigate age-related diseases [[pmid:41063300],[pmid:39427887]]. Developing novel therapeutic strategies that can address the underlying molecular alterations associated with aging will be essential for improving geriatric health outcomes.
6.2 Ethical Considerations in Anti-Aging Therapies
Aging is a complex biological process characterized by a progressive decline in physiological functions and an increased susceptibility to age-related diseases. At the molecular level, aging is marked by various alterations that affect cellular integrity and functionality. The primary molecular mechanisms associated with aging include oxidative stress, chronic inflammation, genomic instability, mitochondrial dysfunction, and cellular senescence.
Oxidative stress results from an imbalance between the production of reactive oxygen species (ROS) and the body's ability to detoxify these harmful compounds. This imbalance can lead to damage of cellular components, including lipids, proteins, and DNA, ultimately contributing to aging and age-related diseases (Ahmed 2025). Chronic inflammation, often referred to as "inflammaging," is another hallmark of aging. It is characterized by the persistent activation of pro-inflammatory pathways, such as the NF-κB signaling pathway, which can exacerbate tissue damage and promote the development of various age-related pathologies (Chung et al. 2009; Młynarska et al. 2025).
Genomic instability, resulting from factors such as telomere shortening and DNA damage, plays a crucial role in the aging process. As cells divide, telomeres—the protective caps at the ends of chromosomes—shorten, leading to cellular senescence and loss of regenerative capacity (Torre et al. 2023). Mitochondrial dysfunction is also a significant contributor to aging, as it impairs energy production and increases ROS generation, further exacerbating oxidative stress (Kang 2020).
Cellular senescence, a state in which cells lose their ability to divide and function, is driven by various stressors, including oxidative damage and telomere shortening. Senescent cells secrete a range of pro-inflammatory cytokines, known as the senescence-associated secretory phenotype (SASP), which can affect neighboring cells and promote tissue degeneration (Lee et al. 2021).
Future directions in aging research focus on identifying potential therapeutic targets to mitigate these molecular mechanisms. Strategies include pharmacological interventions, lifestyle modifications, and dietary approaches aimed at enhancing autophagy, reducing oxidative stress, and combating inflammation (Li et al. 2025; Ahmed 2025). Natural compounds, such as polyphenols and other phytochemicals, have shown promise in modulating these aging hallmarks, offering a potential avenue for developing anti-aging therapies (Zhuang et al. 2024).
Ethical considerations surrounding anti-aging therapies are critical, particularly as the pursuit of longevity raises questions about accessibility, societal implications, and the potential for exacerbating existing inequalities. The commercialization of anti-aging products and the ethical implications of extending lifespan must be carefully examined to ensure that such interventions promote overall health and well-being rather than merely prolonging life without quality.
In conclusion, the molecular mechanisms of aging encompass a range of interrelated processes, including oxidative stress, inflammation, genomic instability, mitochondrial dysfunction, and cellular senescence. Addressing these mechanisms through innovative therapeutic strategies holds promise for improving healthspan and mitigating age-related diseases, while also necessitating a thoughtful approach to the ethical challenges that arise in the field of anti-aging research.
7 Conclusion
The review highlights the intricate molecular mechanisms of aging, including oxidative stress, telomere shortening, mitochondrial dysfunction, epigenetic alterations, and chronic inflammation, which collectively contribute to the decline in physiological functions and increased susceptibility to age-related diseases. Each mechanism is interlinked, suggesting that a holistic understanding of aging is crucial for developing effective therapeutic strategies. Current research has made significant strides in identifying these pathways, yet gaps remain in translating this knowledge into clinical practice. Future research should focus on innovative interventions, such as senolytics, gene therapy, and dietary modifications, while also addressing the ethical considerations associated with anti-aging therapies. By advancing our understanding of the molecular underpinnings of aging, we can enhance healthspan and improve the quality of life for aging populations.
References
- [1] 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.
- [2] Mohamed Ahmed. Targeting aging pathways with natural compounds: a review of curcumin, epigallocatechin gallate, thymoquinone, and resveratrol.. Immunity & ageing : I & A(IF=5.6). 2025. PMID:40611111. DOI: 10.1186/s12979-025-00522-y.
- [3] Yulia Gonskikh;Norbert Polacek. Alterations of the translation apparatus during aging and stress response.. Mechanisms of ageing and development(IF=5.1). 2017. PMID:28414025. DOI: 10.1016/j.mad.2017.04.003.
- [4] Juanli Zhao;Zhenjie Han;Li Ding;Ping Wang;Xiutang He;Li Lin. The molecular mechanism of aging and the role in neurodegenerative diseases.. Heliyon(IF=3.6). 2024. PMID:38312598. DOI: 10.1016/j.heliyon.2024.e24751.
- [5] 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.
- [6] Elena Ruggieri;Erika Di Domenico;Andrea Giacomo Locatelli;Flavio Isopo;Sarah Damanti;Rebecca De Lorenzo;Enrico Milan;Giovanna Musco;Patrizia Rovere-Querini;Simone Cenci;Emilie Vénéreau. HMGB1, an evolving pleiotropic protein critical for cellular and tissue homeostasis: Role in aging and age-related diseases.. Ageing research reviews(IF=12.4). 2024. PMID:39427887. DOI: 10.1016/j.arr.2024.102550.
- [7] Huanjiu Xi;Changyong Li;Fu Ren;Hailong Zhang;Luping Zhang. Telomere, aging and age-related diseases.. Aging clinical and experimental research(IF=3.4). 2013. PMID:23739898. DOI: 10.1007/s40520-013-0021-1.
- [8] Frej Fyhrquist;Outi Saijonmaa;Timo Strandberg. The roles of senescence and telomere shortening in cardiovascular disease.. Nature reviews. Cardiology(IF=44.2). 2013. PMID:23478256. DOI: 10.1038/nrcardio.2013.30.
- [9] Guido von Figura;Daniel Hartmann;Zhangfa Song;Karl Lenhard Rudolph. Role of telomere dysfunction in aging and its detection by biomarkers.. Journal of molecular medicine (Berlin, Germany)(IF=4.2). 2009. PMID:19669107. DOI: 10.1007/s00109-009-0509-5.
- [10] Xingyu Gao;Xiao Yu;Chang Zhang;Yiming Wang;Yanan Sun;Hui Sun;Haiying Zhang;Yingai Shi;Xu He. Telomeres and Mitochondrial Metabolism: Implications for Cellular Senescence and Age-related Diseases.. Stem cell reviews and reports(IF=4.2). 2022. PMID:35460064. DOI: 10.1007/s12015-022-10370-8.
- [11] Paula Martínez;Maria A Blasco. Heart-Breaking Telomeres.. Circulation research(IF=16.2). 2018. PMID:30355079. DOI: 10.1161/CIRCRESAHA.118.312202.
- [12] Peter J Hornsby. Cellular aging and cancer.. Critical reviews in oncology/hematology(IF=5.6). 2011. PMID:20705476. DOI: 10.1016/j.critrevonc.2010.07.011.
- [13] Anna Panfilova;Tatiana Zubareva;Ekaterina Mironova;Gianluigi Mazzoccoli;Maria Greta Pia Marasco;Sofya Balazovskaia;Peter Yablonsky;Igor Kvetnoy. Mitochondrial proteins as biomarkers of cellular senescence and age-associated diseases.. Aging(IF=3.9). 2025. PMID:40856658. DOI: 10.18632/aging.206305.
- [14] Corina T Madreiter-Sokolowski;Ursula Hiden;Jelena Krstic;Katrin Panzitt;Martin Wagner;Christian Enzinger;Michael Khalil;Mahmoud Abdellatif;Ernst Malle;Tobias Madl;Elena Osto;Markus Schosserer;Christoph J Binder;Andrea Olschewski. Targeting organ-specific mitochondrial dysfunction to improve biological aging.. Pharmacology & therapeutics(IF=12.5). 2024. PMID:39179117. DOI: 10.1016/j.pharmthera.2024.108710.
- [15] Maria Kalykaki;Teresa Rubio-Tomás;Nektarios Tavernarakis. The role of mitochondria in cytokine and chemokine signalling during ageing.. Mechanisms of ageing and development(IF=5.1). 2024. PMID:39307464. DOI: 10.1016/j.mad.2024.111993.
- [16] 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.
- [17] 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.
- [18] Anne-Marie Galow;Shahaf Peleg. How to Slow down the Ticking Clock: Age-Associated Epigenetic Alterations and Related Interventions to Extend Life Span.. Cells(IF=5.2). 2022. PMID:35159278. DOI: 10.3390/cells11030468.
- [19] Marsha R Penner;Tania L Roth;Carol A Barnes;J David Sweatt. An epigenetic hypothesis of aging-related cognitive dysfunction.. Frontiers in aging neuroscience(IF=4.5). 2010. PMID:20552047. DOI: 10.3389/fnagi.2010.00009.
- [20] Sushil Kumar Dubey;Rashmi Dubey;Mark Ellsworth Kleinman. Unraveling Histone Loss in Aging and Senescence.. Cells(IF=5.2). 2024. PMID:38391933. DOI: 10.3390/cells13040320.
- [21] 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.
- [22] Yuanyuan Li;Michael Daniel;Trygve O Tollefsbol. Epigenetic regulation of caloric restriction in aging.. BMC medicine(IF=8.3). 2011. PMID:21867551. DOI: 10.1186/1741-7015-9-98.
- [23] 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.
- [24] Hae Young Chung;Matteo Cesari;Stephen Anton;Emanuele Marzetti;Silvia Giovannini;Arnold Young Seo;Christy Carter;Byung Pal Yu;Christiaan Leeuwenburgh. Molecular inflammation: underpinnings of aging and age-related diseases.. Ageing research reviews(IF=12.4). 2009. PMID:18692159. DOI: 10.1016/j.arr.2008.07.002.
- [25] Ádám Mészáros;Kinga Molnár;Bernát Nógrádi;Zsófia Hernádi;Ádám Nyúl-Tóth;Imola Wilhelm;István A Krizbai. Neurovascular Inflammaging in Health and Disease.. Cells(IF=5.2). 2020. PMID:32635451. DOI: 10.3390/cells9071614.
- [26] Xia Li;Chentao Li;Wanying Zhang;Yanan Wang;Pengxu Qian;He Huang. Inflammation and aging: signaling pathways and intervention therapies.. Signal transduction and targeted therapy(IF=52.7). 2023. PMID:37291105. DOI: 10.1038/s41392-023-01502-8.
- [27] Amit Singh;Shepherd H Schurman;Arsun Bektas;Mary Kaileh;Roshni Roy;David M Wilson;Ranjan Sen;Luigi Ferrucci. Aging and Inflammation.. Cold Spring Harbor perspectives in medicine(IF=10.1). 2024. PMID:38052484. DOI: 10.1101/cshperspect.a041197.
- [28] Mario Caldarelli;Pierluigi Rio;Andrea Marrone;Vincenzo Giambra;Antonio Gasbarrini;Giovanni Gambassi;Rossella Cianci. Inflammaging: The Next Challenge-Exploring the Role of Gut Microbiota, Environmental Factors, and Sex Differences.. Biomedicines(IF=3.9). 2024. PMID:39200181. DOI: 10.3390/biomedicines12081716.
- [29] Claudio Franceschi;Paolo Garagnani;Paolo Parini;Cristina Giuliani;Aurelia Santoro. Inflammaging: a new immune-metabolic viewpoint for age-related diseases.. Nature reviews. Endocrinology(IF=40.0). 2018. PMID:30046148. DOI: 10.1038/s41574-018-0059-4.
- [30] 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.
- [31] B P Yu. Aging and oxidative stress: modulation by dietary restriction.. Free radical biology & medicine(IF=8.2). 1996. PMID:8891668. DOI: 10.1016/0891-5849(96)00162-1.
- [32] Hong Zhuang;Xiaoliang Zhang;Sijia Wu;Pang Yong;Haiyang Yan. Opportunities and challenges of foodborne polyphenols applied to anti-aging health foods.. Food science and biotechnology(IF=3.1). 2024. PMID:39493397. DOI: 10.1007/s10068-024-01686-x.
- [33] Bhawna Diwan;Rohit Sharma. Nutritional components as mitigators of cellular senescence in organismal aging: a comprehensive review.. Food science and biotechnology(IF=3.1). 2022. PMID:35756719. DOI: 10.1007/s10068-022-01114-y.
- [34] Tania Rescigno;Luigina Micolucci;Mario F Tecce;Anna Capasso. Bioactive Nutrients and Nutrigenomics in Age-Related Diseases.. Molecules (Basel, Switzerland)(IF=4.6). 2017. PMID:28075340. DOI: 10.3390/molecules22010105.
- [35] Maria Carmen Andreo-López;Victoria Contreras-Bolívar;Manuel Muñoz-Torres;Beatriz García-Fontana;Cristina García-Fontana. Influence of the Mediterranean Diet on Healthy Aging.. International journal of molecular sciences(IF=4.9). 2023. PMID:36901921. DOI: 10.3390/ijms24054491.
- [36] Chapa Ramasinghe;Matteo Bordiga;Baojun Xu. A Comprehensive Review of the Triangular Relationship Among Diet, Gut Microbiota, and Aging.. International journal of molecular sciences(IF=4.9). 2025. PMID:41009354. DOI: 10.3390/ijms26188785.
- [37] Serena Dato;Dina Bellizzi;Giuseppina Rose;Giuseppe Passarino. The impact of nutrients on the aging rate: A complex interaction of demographic, environmental and genetic factors.. Mechanisms of ageing and development(IF=5.1). 2016. PMID:26876763. DOI: .
- [38] Masayoshi Suda;Karl H Paul;Utkarsh Tripathi;Tohru Minamino;Tamara Tchkonia;James L Kirkland. Targeting Cell Senescence and Senolytics: Novel Interventions for Age-Related Endocrine Dysfunction.. Endocrine reviews(IF=22.0). 2024. PMID:38500373. DOI: 10.1210/endrev/bnae010.
- [39] Di Wu;Qinzheng Xu;Shuang Wu;Jiachen Tan;Faheem Ahmed Khan;Nuruliarizki Shinta Pandupuspitasari;Chunjie Huang. Pursuing healthy aging and longevity through natural product and stem cell-driven rejuvenation.. Biochemical pharmacology(IF=5.6). 2025. PMID:40998022. DOI: 10.1016/j.bcp.2025.117359.
- [40] Hubert Klepacki;Krystyna Kowalczuk;Natalia Łepkowska;Justyna Magdalena Hermanowicz. Molecular Regulation of SASP in Cellular Senescence: Therapeutic Implications and Translational Challenges.. Cells(IF=5.2). 2025. PMID:40643463. DOI: 10.3390/cells14130942.
- [41] Timur Saliev;Prim B Singh. Targeting Senescence: A Review of Senolytics and Senomorphics in Anti-Aging Interventions.. Biomolecules(IF=4.8). 2025. PMID:40563501. DOI: 10.3390/biom15060860.
- [42] Liyao Zheng;Shipei He;Hong Wang;Jinling Li;Yuanyuan Liu;Sijia Liu. Targeting Cellular Senescence in Aging and Age-Related Diseases: Challenges, Considerations, and the Emerging Role of Senolytic and Senomorphic Therapies.. Aging and disease(IF=6.9). 2024. PMID:38421832. DOI: 10.14336/AD.2024.0206.
- [43] Sofia M Lagoumtzi;Niki Chondrogianni. Senolytics and senomorphics: Natural and synthetic therapeutics in the treatment of aging and chronic diseases.. Free radical biology & medicine(IF=8.2). 2021. PMID:33989756. DOI: 10.1016/j.freeradbiomed.2021.05.003.
- [44] Alexander Bürkle. In memoriam Bernard Strehler--genomic instability in ageing: a persistent challenge.. Mechanisms of ageing and development(IF=5.1). 2002. PMID:12044938. DOI: 10.1016/s0047-6374(02)00027-1.
- [45] Paul D Ray;Bo-Wen Huang;Yoshiaki Tsuji. Reactive oxygen species (ROS) homeostasis and redox regulation in cellular signaling.. Cellular signalling(IF=3.7). 2012. PMID:22286106. DOI: 10.1016/j.cellsig.2012.01.008.
- [46] Qiao Li;Heng Zhang;Nanyin Xiao;Guangyu Liang;Yan Lin;Xiao Yang;Jiankun Yang;Zonghao Qian;Yangguang Fu;Cuntai Zhang;Anding Liu. Aging and Lifestyle Modifications for Preventing Aging-Related Diseases.. FASEB journal : official publication of the Federation of American Societies for Experimental Biology(IF=4.2). 2025. PMID:40293686. DOI: 10.1096/fj.202402797RR.
- [47] Despoina Gyriki;Christos G Nikolaidis;Eugenia Bezirtzoglou;Chrysa Voidarou;Elisavet Stavropoulou;Christina Tsigalou. The gut microbiota and aging: interactions, implications, and interventions.. Frontiers in aging(IF=4.3). 2025. PMID:40438731. DOI: 10.3389/fragi.2025.1452917.
- [48] Lena Best;Thomas Dost;Daniela Esser;Stefano Flor;Andy Mercado Gamarra;Madlen Haase;A Samer Kadibalban;Georgios Marinos;Alesia Walker;Johannes Zimmermann;Rowena Simon;Silvio Schmidt;Jan Taubenheim;Sven Künzel;Robert Häsler;Sören Franzenburg;Marco Groth;Silvio Waschina;Philip Rosenstiel;Felix Sommer;Otto W Witte;Philippe Schmitt-Kopplin;John F Baines;Christiane Frahm;Christoph Kaleta. Metabolic modelling reveals the aging-associated decline of host-microbiome metabolic interactions in mice.. Nature microbiology(IF=19.4). 2025. PMID:40140706. DOI: 10.1038/s41564-025-01959-z.
- [49] Yue Zhou;Guo Hu;Meng C Wang. Host and microbiota metabolic signals in aging and longevity.. Nature chemical biology(IF=13.7). 2021. PMID:34552221. DOI: 10.1038/s41589-021-00837-z.
- [50] Yingli Jing;Qiuying Wang;Fan Bai;Zihan Li;Yan Li;Weijin Liu;Yitong Yan;Shuangyue Zhang;Chen Gao;Yan Yu. Role of microbiota-gut-brain axis in natural aging-related alterations in behavior.. Frontiers in neuroscience(IF=3.2). 2024. PMID:38699678. DOI: 10.3389/fnins.2024.1362239.
- [51] Qingwen Wang;Zhixiao Xu;Xinwen Ding;Aiting Wang;Sunfengda Song;Shuang Zhang;Youming Chen;Yi Ding;Lai Jiang;Xianting Ding. Ten mouse organs proteome and metabolome atlas from adult to aging.. Genome medicine(IF=11.2). 2025. PMID:41063300. DOI: 10.1186/s13073-025-01535-4.
- [52] Katrina Woodward;Nikolay E Shirokikh. Translational control in cell ageing: an update.. Biochemical Society transactions(IF=4.3). 2021. PMID:34913471. DOI: 10.1042/BST20210844.
MaltSci Intelligent Research Services
Search for more papers on MaltSci.com
Oxidative Stress · Telomere Shortening · Mitochondrial Dysfunction · Epigenetic Changes · Chronic Inflammation
© 2025 MaltSci
