Abstract
Chronic stress is a risk factor for numerous aging-related diseases and has been shown to shorten lifespan in humans and other social mammals. Yet, how life stress causes such a vast range of diseases is still largely unclear. In recent years, the impact of stress on health and aging has been increasingly associated with the dysregulation of the so-called hallmarks of aging. These are basic biological mechanisms that influence intrinsic cellular functions and whose alteration can lead to accelerated aging. Here, we review correlational and experimental literature (primarily focusing on evidence from humans and murine models) on the contribution of life stress – particularly stress derived from adverse social environments – to trigger hallmarks of aging including cellular senescence, sterile inflammation, telomere length, production of reactive oxygen species, DNA damage, and epigenetic changes. We also evaluate the validity of stress-induced senescence and accelerated aging as an etiopathological proposition. Finally, we highlight current gaps of knowledge and future directions for the field, and discuss perspectives for translational geroscience.
Keywords: geroscience, biological age, senescence, epigenetic, social stress, chronic stress
Graphical abstract
INTRODUCTION
Life stress – defined as the occurrence of unpredictable, uncontrollable and potentially/actually life-threatening conditions (Koolhaas et al., 2011) – is a major risk factor for a multitude of aging-related diseases, as shown by epidemiological, clinical, and preclinical studies. In particular, chronic stress confers increased risk and often earlier onset of aging-related diseases, which severely degrade quality of life and are the primary causes of death in older adults. For example, life stress increases risk for age-related diseases such as obesity, type 2 diabetes, cardiovascular disease, Alzheimer’s disease, vascular dementia, Parkinson’s disease, osteoarthritis and many others (Bonanni et al., 2018; Gerritsen et al., 2017; Hemmerle et al., 2012; Kivimäki et al., 2022; Kivimäki and Steptoe, 2018; McEwen, 1998; Mejfa et al., 2003; Snyder-Mackler et al., 2020).
In spite of the decades old proposition that chronic stress accelerates aging via global dysregulation of homeostatic mechanisms (Sapolsky et al., 1986; Selye, 1970), how life stress causes such a vast range of diseases is still largely unclear. The advent of geroscience provides a conceptual framework to interpret the impact of life stress on health and aging by addressing mechanisms that regulate the fundamental biological processes affecting aging at the cellular, organ and systemic level (Duque et al., 2023; Epel, 2020; Kennedy et al., 2014; Kivimäki et al., 2022; López-Otín et al., 2023; Lyons and Bartolomucci, 2020; Polsky et al., 2022; Sierra and Kohanski, 2023; Snyder-Mackler et al., 2020). For example, graying of the hair, a classical visual biomarker of aging and premature senescence, has recently been shown to be induced by stress (Rosenberg et al., 2021), with a potential role played by stress-induced hyperactivation of the sympathetic nervous system (SNS) (Zhang et al., 2020). Similarly, bone loss and osteoporosis increase with aging, and chronic social stress has been shown to affect cartilage-to-bone transition during bone growth and repair upon fracture in mice, via a mechanism requiring SNS activation (Tschaffon-Müller et al., 2023).
Here, we review the available correlational and experimental literature – primarily focusing our analysis on human data and preclinical murine models – on the contribution of stress to trigger select hallmarks of aging (Kennedy et al., 2014; López-Otín et al., 2023, 2013) including: i) cellular senescence and sterile inflammation; ii) telomere length; iii) production of reactive oxygen species; iv) DNA damage; v) epigenetic changes. Several excellent reviews discussed the role of some of the hallmarks of aging as potential biomarkers, or causal factors, of the link between life stress, aging-associated diseases, and accelerated biological aging (e.g. (Blackburn et al., 2015; Kivimäki et al., 2022; Palma-Gudiel et al., 2020; Polsky et al., 2022; Raffington and Belsky, 2022), but to the best of our knowledge, no previous review integrated animal models and human data in a translational geroscience perspective, as presented in this paper. Additionally, several important research papers have been published in the ~12 months preceding this publication, which have not been reviewed extensively elsewhere (e.g (Chiou et al., 2022; Dion et al., 2022; Kong et al., 2023; Obeng-Gyasi et al., 2023; Poganik et al., 2022; Razzoli et al., 2023; Rentscher et al., 2022; Tschaffon-Müller et al., 2023). The aims of this review are, i) to briefly summarize previous knowledge on the hallmarks of aging and their involvement in life stress (pointing readers to previous reviews, whenever possible), ii) to highlight recent findings in human studies and animal models of social determinants of health and aging (SDoHA), that enable the transition from correlational studies to causal inference, and finally iii) integrate those concepts within the framework of geroscience highlighting gaps of knowledge and future perspectives.
LIFE STRESS AND ACCELLERATION OF BIOLOGICAL AGING
A certain degree of physical, emotional, and social challenge is a constant feature of life, and all organisms must be capable of managing and adapting to perturbations to homeostasis. However, sometimes life challenges impose physiological demands outside an individual’s adaptive capacity. These challenges can be defined as stressors (Koolhaas et al., 2011). Over the years, the field has revised its nomenclature multiple times and there is not currently a universally accepted definition of stress (Aschbacher et al., 2013; Koolhaas et al., 2011; McEwen, 1998; Nederhof and Schmidt, 2012; Sapolsky et al., 2000). Here we will refer to the “stress revised” terminology, defining life stress as circumstances characterized by perception of/exposure to an uncontrollable and unpredictable hazard that can pose at least a perceived potential life threat and requires adaptations outside of an individual’s regulatory range (Koolhaas et al., 2011).
Aside from its definition, it is accepted that the physiological stress-response system includes activation of several neural and neuroendocrine axes such as the SNS, the sympatho-adrenal medullary (SAM) system and the hypothalamic pituitary adrenal (HPA) axis (Charmandari et al., 2005; Sapolsky et al., 2000; Ulrich-Lai and Herman, 2009). In response to a stressor, the SNS/SAM are rapidly activated mediating the classical “fight or flight response” characterized by increased heart rate, respiratory rate, glycolysis, glycogenolysis and lipolysis to sustain an acute metabolic demand to the body. Unlike the fast responding and rapidly deactivating responses mediated by the SNS, the HPA axis operates on a slower timescale. It’s activation originates from neurons in the paraventricular nucleus of the hypothalamus, that in response to perceived threat, release corticotropin releasing hormone (CRH) into the hypophysial portal system. This leads to release of pituitary adrenocorticotropic hormone (ACTH) into general circulation. In turn, this results in glucocorticoid (GC) secretion from the adrenal cortex. GCs activate several physiological responses while also exerting a negative feedback on the HPA axis. However, in cases of sustained stress, or during aging, GC hypersecretion can persist (Sapolsky et al., 1984; Spiga et al., 2014). The negative feedback system becomes impaired with age due in large part to downregulation of glucocorticoid receptors (GR) in the hippocampus and other critical nodes of the HPA axis (Polsky et al., 2022; Sapolsky et al., 1986). While this defines the main stress-related neuro-endocrine axes, additional endocrine mediators such as thyroid and growth hormone axes (Charmandari et al., 2005; Sapolsky et al., 2000; Tsigos and Chrousos, 2002), as well as adipose-derived IL-6 (Qing et al., 2020) and osteoclast-derived osteocalcin (Berger et al., 2019) are activated acutely and/or chronically during stress (Figure 1), which are also implicated in aging (Bonewald, 2019; Cappola et al., 2023; van den Beld et al., 2018).
Figure 1. Summary of the major stress-related axes, and stress-induced changes to select hallmarks of aging.
The major stress-related pathways include the hormones of the hypothalamus-pituitary-adrenocortical (HPA) axis: corticotrophin releasing factor (CRH), adrenocorticotropic hormone (ACTH), and cortisol (or corticosterone in rodents). The sympatho-adreno-medullary (SAM) axis secretes catecholamines, granins, and other hormones and neuropeptides. The neurotransmitters secreted by the sympathetic nervous system (SNS), include noradrenaline, ATP, neuropeptide Y (NPY), and other factors. The parasympathetic nervous system (PNS) releases neurotransmitters such as acetylcholine and other factors. Other neuroendocrine axes include the growth hormone (GH) axis [growth hormone releasing hormone (GHRH), GH, and insulin-like growth factor 1 (IGF1)], the thyroid axis [thyrotropin releasing hormone (TRH), thyroid stimulating hormone (TSH), triiodothyronine (T3), and thyroxine (T4)]. The adipose tissue (brown or white adipocytes) secretes factors such as interleukin-6 (IL-6), leptin, and others. Osteoclasts have been shown to secrete osteocalcin in response to stress. Evidence discussed in this review demonstrate that life stress promotes hallmarks of aging in several organs and systems. These include blood and other body fluids, circulating immune cells [peripheral blood mononuclear cells (PBMC)], central nervous system (CNS), thymus, lung, liver, gastric, muscle, and bone tissues. Stress-induced changes to hallmarks of aging have been primarily studied in animal models; studies in humans have been limited to PBMC and body fluids and are presented in italics.
Among the life stressors affecting body function and aging, stressors of social nature, aka “social stressors”, have singularly potent effects (Bartolomucci, 2007; Koolhaas et al., 2011; Sapolsky, 2005; Snyder-Mackler et al., 2020). Indeed, SDoHA - including adverse childhood experiences (ACE), low socioeconomic status (SES), loneliness, and caregiver stress - are increasingly regarded as modulators of the stress response, accelerators of biological age, and risk factors for aging-associated diseases (Kivimäki et al., 2022; Noren Hooten et al., 2022; Snyder-Mackler et al., 2020; Wang et al., 2023). For example, ACE cause particularly severe harm to both healthspan – the number of years lived without functional impairment – and lifespan (Kivimäki et al., 2022; Snyder-Mackler et al., 2020; Tung et al., 2016). Exposure to an increasing number of ACE elevates risk for chronic diseases in adulthood, including ischemic heart disease, cancer, and chronic lung disease (Kivimäki et al., 2022; Kivimäki and Steptoe, 2018; Lupien et al., 2009). Like ACE, low SES confers an immense burden of social stress and is associated with elevated mortality risk from nearly all causes (Elo, 2009; Marmot et al., 1991). The factors involved can include financial insecurity, uncertainty about future stability, and feelings of exclusion or marginalization due to occupational or economic status, in addition to limited or absent appropriate nutritional, educational and medical resources (Adler et al., 1994; Kivimäki et al., 2022; Marmot et al., 1991; Snyder-Mackler et al., 2020; Stringhini et al., 2017). Interestingly, the adverse effect of ACE on health and aging was recapitulated in some [baboons (Tung et al., 2016)] but not all [gorilla (Morrison et al., 2023)], non-human primates, suggesting that species-specific behavioral and socioecological factors can inform on biological resilience to ACE in humans.
While a number of factors contribute to the myriad negative health outcomes associated with low SES, material deprivation does not entirely explain this phenomenon (Richterman et al., 2023). Rather, it is widely hypothesized that chronic social stress and its physiological consequences at least partially underlie the shorter lifespan and numerous poor disease outcomes associated with low SES. Indeed, low SES is associated with higher basal levels of catecholamines and cortisol, as well as higher self-reported levels of stress (Cohen et al., 2006; Szanton et al., 2005). Supporting psychosocial stress as a principal intermediary of SES and poor health outcomes is a study showing that low SES individuals who still experience a high sense of control have health metrics comparable with individuals in higher income groups (Lachman and Weaver, 1998).
One cohort study followed >3,000 participants aged 6–18 years at baseline for over 30 years via repeated biomedical examinations (Kivimäki et al., 2018). The findings suggested that experiencing lower SES and its associated stressors accelerated age-related metabolic changes. At baseline, groups with high vs low SES stress did not differ in any metabolic traits, but by early adulthood the low SES group had notably increased blood lipid levels, decreased insulin sensitivity, and increased compensatory insulin secretion that was still insufficient to normalize circulating glucose concentrations. By middle age, low SES individuals were more likely to be obese, hypertensive, have a fatty liver, and have diabetes compared with those who consistently lived in high SES neighbourhoods (Kivimäki et al., 2018). Another study found that SES experiences from each life course period (childhood, early adulthood, and late adulthood) additively combine to predict the number of years an individual can expect to live with or without dementia (Cha et al., 2021). Childhood adversities alone have a profound effect on the likelihood of developing dementia. The risk of developing dementia for individuals experiencing two disadvantages and three disadvantages were found to respectively be 56%, and 87% higher than for those with no socioeconomic disadvantages during childhood. Low SES status during all three life periods shortened life expectancy for both men and women by 8.66 and 6.89 years, respectively. 18% of those years for men, or 17% for women were predicted to be lived with dementia, compared with 3% for men or 4% for women who experienced high SES throughout all life stages (Cha et al., 2021).
Although the relationship between low SES and poorer health/lifespan has long been documented, the biological mechanisms underlying this effect have been difficult to study (Kivimäki et al., 2022). Some important recent advancements have been made, e.g. the Environmental Risk Longitudinal Twin Study found that children raised in more socioeconomically disadvantaged neighbourhoods develop epigenetic DNA methylation patterns distinct from their more advantaged peers (Reuben et al., 2020). A Mendelian randomization study investigated causal associations of 19 modifiable socioeconomic, lifestyle, and cardiometabolic factors with DNA-based measures of biological age (Kong et al., 2023). In addition to known risk factors such as smoking, high education, and household income, operational measures of high SES, were found to be among the strongest protective factor for the DNA age of an individual (Kong et al., 2023).
Although Mendelian randomization studies are increasingly used to attempt to answer cause-effect relationships in human studies (Kivimäki et al., 2022), a more precise understanding of causal physiological underpinnings of the health risks conferred by low social status requires the use of animal models where randomization experiments are possible. This approach is supported by the existence of social gradients in health in virtually all social mammals (Snyder-Mackler et al., 2020). Consistent with human studies, in experimental animal models of social determinants of health, social stressors (e.g. chronic subordination stress) are particularly potent activators of stress responses compared with other types, such as physical (e.g. cold water stress, foot shock) or psychological (e.g. restraint stress) (Bartolomucci, 2007; Bartolomucci et al., 2009). In baboons, social rank as well as the strength/stability of an individual’s social bonds predict transcriptional profiling related to biological aging and survival (Chiou et al., 2022; Tung et al., 2016; Zipple et al., 2019) and experimental manipulation of social rank causes a lasting effect on immune functions (Snyder-Mackler et al., 2016). Similarly, subordinate status within a group of mice has been studied as a model for low SES in humans. Subordinate mice show persistent immunoendocrine impairments, reduced lifespan, elevation of cellular senescence markers and development of early-stage atherosclerotic lesions (W. Lee et al., 2022; Razzoli et al., 2018). Furthermore, new data demonstrate changes in methylation patterns linked to mouse genetic background and social rank, adding further credibility to social subordination as a relevant model of SDoHA (Razzoli et al., 2023).
EFFECT OF LIFE STRESS ON SELECTED HALLMARKS OF AGING
Accelerated biological aging is considered to reflect the cumulative progressive dysfunction and cellular damage compounded during lifespan. A set of cellular/physiological processes has been identified as age-promoting factors, and are collectively referred to as pillars/hallmarks of aging, including cellular senescence, telomere length, ROS and oxidative stress, genetic/epigenetic changes (Kennedy et al., 2014; López-Otín et al., 2023, 2013). While for clarity’s sake, this review describes some of these hallmarks individually in relation to stress exposure, it is crucial to note that a central feature of these pillars/hallmarks is their connectedness and interplay (Epel, 2020; Kennedy et al., 2014; López-Otín et al., 2023). This interconnection posits a conceptual challenge in that it is difficult to pinpoint individual physiological mechanisms responsible for age-associated damage. Many homeostatic and pathological processes occur concomitantly and/or consequentially during the life course, differentially affecting the pace of aging. Here, we will briefly review selected hallmarks/pillars of aging such as: i) telomere length, ii) cellular senescence and systemic inflammation, iii) production of reactive oxygen species, iv) DNA damage and v) epigenetic changes, and discuss evidence of their link to life stress (Figure 1). Because most/all of the hallmarks of aging are interconnected and multiple markers are often measured in a single research paper, the narrative will often create connection and overlaps while attempting to keep the sections separate. This review will primarily focus on in vivo studies where life stress can be investigated. However, cellular models exposed acutely/chronically to stress-related hormones such as glucocorticoids, offer promising mechanistic insights and will also briefly reviewed here. For examples, a recent study showed that chronic glucocorticoid exposure increases cellular energy expenditure (caused by a metabolic shift from glycolysis to mitochondrial oxidative phosphorylation), induced mitochondrial (mt)DNA instability, modulated senescence associated secretory phenotype (SASP), and accelerates cellular aging assessed using DNA methylation and telomere shortening (Bobba-Alves et al., 2023).
EFFECTS OF LIFE STRESS ON TELOMERES
Telomeres are repeated TTAGGG DNA sequences that cap the end of chromosomes, are subject to progressive reduction during replication and aging, and whose reduction is counterbalanced by the activity of telomerase (Blackburn et al., 2015; de Lange, 2009). Before telomeres are completely lost, they reach a critical length, termed the Hayflick limit, which triggers the DNA damage response (DDR) and activation of the tumor suppressor p53 resulting in either apoptosis, or exit from the cell cycle (Saretzki et al., 1999). In a given cell, the different chromosomes may have a range of telomere lengths; however, the stability and viability of the cell is defined by the shortest telomere present (Hemann et al., 2001). Numerous environmental factors, including life stress as discussed below, can affect telomere length maintenance (See also (Blackburn et al., 2015; Polsky et al., 2022) for additional review) (Table 1).
Table 1.
Effect of chronic life stress or neuroendcirne stress mediators on selected hallmarks of aging: effect on telomeres
Finding | Species | Population | Stress model/proxy | Tissue | Reference |
---|---|---|---|---|---|
| |||||
Shorter telomeres, lower telomerase activity, greater oxidative damage | Human | Pre-menopausal mothers | Reported stress levels | PBMCs | Epel et al., 2004 |
Shorter telomeres, higher cytokine production by PBMCs | Human | Men and women, avg age 65+/− 5 years | Caregivers of AD patients | PBMCs | Damjanovic et al., 2007 |
Shorter telomeres associated with stress and urinary catecholamine level | Human | Women with a sister with breast cancer, but not cancer themselves | Reported stress levels | Leukocytes | Parks et al., 2009 |
Shorter telomeres | Human | Female twins, avg age 46 | Low SES | White blood cells | Cherkas et al., 2006 |
No difference in telomere length | Human | Men and women, avg age 50+/− 5 years | Low SES | PBMCs | Adams et al., 2007 |
Shorter telomeres | Human | Men and women over 60; avg age 70 | Experience of childhood adverse events | Leukocytes | Schaakxs et al., 2016 |
Lower baseline telomerase activity; equivalent telomerase activity following exposure to acute stressor | Human | Women age 51–75; avg age 62 | Dementia caregivers reporting high stress levels; Trier Social Stress Test | PBMCs | Epel et al., 2010 |
decreased telomere | Mice | wild type young adult | chronic mild stress | liver | Xie et al. 2019 |
No change in telomere length | Mice | wild type 17 month old | Lifelong chronic psychosocial stress | liver, spleen, lung | Razzoli et al., 2018 |
Lower telomere length | Human | Sedentary postmenopausal women | Reported stress levels | Whole blood | Puterman et al., 2010 |
The first study to directly link life stress with one of the markers of biological aging was published in 2004 by Epel and coworkers (Epel et al., 2004). Participants were premenopausal mothers to either a healthy child or a chronically ill child. Telomere length and telomerase activity were measured in peripheral blood mononuclear cells (PBMCs) . Within the group of mothers with chronically ill children, there was a negative correlation between duration of caregiving and telomere length, independent of age. However, rather than the health status of the child, the degree of perceived stress reported by the mothers was a better predictor of telomere length and telomerase activity. Although the average age of the two groups was similar, telomeres in the higher stress group were shorter by an average of 550-bp, suggesting a difference of 9–17-years difference in biological age. The high stress group also had lower telomerase activity and higher levels of higher oxidative stress. This negative correlation between life stress level and telomere length has been replicated in many (Damjanovic et al., 2007; Parks et al., 2009) but not all studies (Rentscher et al., 2019). It has also been shown that urinary catecholamine content, indicative of chronic activation of one of the major stress neuroendocrine axes, is associated with shorter telomeres suggesting a possible causative link (Parks et al., 2009). Similar to life stress, low SES has been associated with lower telomere length in some (Cherkas et al., 2006) but not all studies (Adams et al., 2007). These differing findings may be influenced by the observation that in older adults, stress-associated differences in telomere length are more difficult to parse. In older age, other factors such as disease burden and disease survivorship account for greater variability (Schaakxs et al., 2016). Despite this, the negative effect of adverse childhood events can still be discerned in the telomere length of older adults (Schaakxs et al., 2016).
In a mouse model of lifelong chronic subordination stress, absolute telomere length in liver, lung and spleen of 17 month-old mice was not altered in spite of increased organ damage, increased markers of cellular senescence and shorter lifespan (Razzoli et al., 2018). Similarly, early life stress had no consequences on telomere length in hippocampi when mice were tested at 20 months of age (Kotah et al., 2021). Unlike early life stress and subordination, chronic mild stress in young adult mice led to a shortening of telomeres in the liver, in presence of increased oxidative damage (Xie et al., 2019). The tissue specificity of telomere length shortening due to chronic stress was also highlighted in a study of mice exposed to chronic immobilization stress, showing tissue-specific acceleration (Baek et al., 2019).
A dynamic relationship between psychological stress and telomerase activity has also emerged. High stress caregivers to a family member with dementia were found to have lower baseline telomerase activity; however, when exposed to an acute stressor they displayed increased telomerase activity equivalent to that of age-matched low stress controls (Epel et al., 2010). The increases in telomerase activity were associated with the magnitude of cortisol response to the stressor. However, another study found that caregivers to a dementia patient had higher basal telomerase activity, despite shorter telomeres (Damjanovic et al., 2007). Follow-up studies have looked for possible interventions for stress-induced telomere erosion. It was found that regular physical exercise can buffer the effect of stress-induced telomere shortening in post-menopausal women (Puterman et al., 2010).
EFFECTS OF LIFE STRESS ON CELLULAR SENESCENCE, SASP FACTORS AND INFLAMMATION
Cellular senescence is a complex phenomenon with yet undefined molecular definition (Gorgoulis et al., 2019; P. J. Lee et al., 2022), and implications for prevention of uncontrolled proliferation in cancer development, normal wound repair and tissue regeneration (Campisi and d’Adda di Fagagna, 2007; Childs et al., 2017; Gorgoulis et al., 2019). Senescent cells (SNC) accumulate with age, and in recent years have been demonstrated to actively drive numerous aging related diseases including atherosclerosis, cancer and Alzheimer’s disease (Bussian et al., 2018; Childs et al., 2017; Kirkland et al., 2017; Xu et al., 2018). Highlighting the complex and yet unresolved link between SNCs and diseases, clearance of senescent alveolar macrophages was recently shown to protect from early-stage lung tumorigenesis in mice (Prieto et al., 2023).
Cellular senescence is considered one of the fundamental aging process in which cells cease division and undergo a number of other distinctive phenotypic changes (Hayflick and Moorhead, 1961; McHugh and Gil, 2018). Throughout its lifespan, a cell is subjected to internal and external challenges leading to gradual accrual of damage. Simultaneously, as cells divide, they incrementally lose telomeric DNA, destabilizing the entire chromosomal structure. Both of these forms of damage make a cell susceptible to developing deleterious DNA mutations. Senescence programming prevents damaged cells from passing on damaged DNA to the next generation, making it a potent tumor suppressive mechanism. This process can be is advantageous in ensuring that an individual survives long enough to reproduce; however, it appears to be less advantageous to an aging organism. SNC become resistant to many apoptotic signals, contributing to their tendency to accumulate with age (Campisi and d’Adda di Fagagna, 2007). As these cells remain metabolically active and develop numerous phenotypic changes, their accrual can result in severe tissue dysfunction (Campisi and d’Adda di Fagagna, 2007; Childs et al., 2017). Apart from telomere erosion, multiple forms of cellular damage can induce senescence. Non-telomeric DNA damage, mitochondrial dysfunction, oxidative stress, chromatin disruption, and oncogene signaling are all capable of activating senescence programming. These cellular stresses can result from internal or external sources. Regardless of the initial stimulus, signaling converges on one of two pathways to halt progression through the cell cycle progression. There are two main tumor-suppressive pathways responsible for cell cycle arrest and senescence induction. These pathways are controlled by p16INK4a (p16)/ pRB (retinoblastoma protein), and p53 respectively. While both are capable of halting replication, the two pathways are not entirely equivalent. In addition to being activated by different stimuli, the p53 pathway, via the effector p21, induces escapable cell cycle arrest, while p16/pRB induction results in permanent cell cycle arrest (Childs et al., 2017). Shortened telomeres or other forms of DNA injury prompt a DDR, which activates the master gene regulator, p53. Depending on the nature and severity of the DNA damage, p53 activation can result in either cell death via apoptosis, or cellular senescence (Chen et al., 2000; Debacq-Chainiaux et al., 2005; Probin et al., 2006). The interplay between these alternative cell fates remains unclear. In conditions favoring senescence, p53 activates transcription of the cyclin-dependent kinase inhibitor p21 (Zhang et al., 1999). p21 prevents progression through the cell cycle by blocking the cyclin-dependent kinases responsible for moving the cell from G1 to S phase. If the damage is repaired and DDR signaling ceases, p21-induced arrest also ceases, and the now healthy cell is able to re-enter the cell cycle. If the DNA damage is not repaired, sustained arrest eventually causes induction of p16, resulting in permanent exit from the cell cycle (Dulić et al., 2000). p16 can also be activated independently in response to oncogenic Ras or BRAF signaling, although this process is incompletely understood (Campisi and d’Adda di Fagagna, 2007; Di Micco et al., 2006). Bias towards p16 vs p21 activation varies between cell types and species (Sturmlechner et al., 2021).
In addition to exit from the cell cycle, SNC are also characterized by: resistance to apoptosis, increased lysosomal volume and lysosomal β-galactosidase production (senescence-associated β-galactosidase; SA βGal), chromatin remodeling, enlarged and flattened morphology, and secretion of a set of factors termed the senescence associated secretory phenotype – SASP (Campisi and d’Adda di Fagagna, 2007; Childs et al., 2017; Gorgoulis et al., 2019). SASP composition varies between cell types, but in general includes a host of proinflammatory cytokines, chemokines, extracellular matrix molecules and even reactive oxygen species (Coppé et al., 2011). These molecules act as paracrine signals in a form of sterile inflammation; in this way, SNC can alter their tissue microenvironment and negatively affect the function of neighboring cells. While overexpression of p16 alone is sufficient to induce replicative arrest, DDR signaling is required for the SASP (Coppé et al., 2011; Rodier et al., 2009). It is believed that secretion of these proinflammatory molecules underlies most of the detrimental effects of SNC. A systemic rise in inflammatory mediators is a common feature of aging. This phenomenon, termed “inflammaging” is associated with elevated risk for cardiovascular disease as well as other chronic aging-related diseases (Ferrucci and Fabbri, 2018; Franceschi et al., 2018). The root causes of inflammaging remains poorly understood, though immune dysregulation and possibly SNC accumulation are likely critical factors (Ferrucci et al., 2010; Fulop et al., 2017; Yousefzadeh et al., 2021). Overall, increasing evidence suggests that accumulation of SNC may be a major driver of the functional decline associated with premature aging.
A growing body of literature suggests that in both humans and mouse models chronic stress might promote accumulation of SNC, SASP and sterile inflammation (See also (Polsky et al., 2022) for review) (Table 2). Life stress has been associated with increased p16INK4a in PBMC (Rentscher et al., 2019). Consistently, lifelong psychosocial stress in mice shortened lifespan and increased expression of senescence markers such as p16INK4a and p53 in several peripheral organs in subordinate individuals (Razzoli et al., 2018). Additionally, chronic exposure to a non-social psychological stressor (restraint stress) increased p16INK4a, p21, and SASP, and lowered DNA damage repair in severe combined immunodeficient (SCID) male mice (Rentscher et al., 2022). As discussed above, the vast majority of SASP factors contribute to a state of sterile local or systemic inflammation known as “inflammaging” (Ferrucci and Fabbri, 2018; Franceschi et al., 2018). Thus, studies measuring inflammation have the potential to capture signals coming from SNC, even if the SNC per se were not evaluated as a main source of those mediators. Likewise, aging immune cells can drive SNC and ageing of solid organs (Yousefzadeh et al., 2021) creating a circular loop resulting in increased sterile inflammation and organ damage. Several studies demonstrate that psychological stress promotes inflammation both in the brain, and in the periphery (Calcia et al., 2016; Frank et al., 2006; Rohleder, 2014). This finding has been replicated in both humans and laboratory rodents. For example, caregiver stress, low SES, work-related stress and childhood adversity are all associated with elevations in inflammatory markers (Rohleder, 2014). During an extended period of chronic stress, there are fluctuations in multiple cytokines. This was demonstrated in a study that subjected female mice to 6-hours of daily restraint stress for 28 days, monitoring serum cytokine levels throughout, and during 2 weeks of recovery (Voorhees et al., 2013). Circulating plasma levels of IL-6 were increased throughout the 4 weeks of restraint stress but returned to levels indistinguishable from controls by day 7 of recovery. Serum IL-10 was lower overall during the both the restraint and recovery phases. In the brain — specifically the cortex and hippocampus — IL-10 was reduced throughout the period of restraint and recovery as well. Chronic unpredictable mild stress increases circulating plasma levels of IL-6, IL-1β, TNF-α and IL-10 in rats (López-López et al., 2016). Changes in circulating cytokine levels are apparent after 20 days of stress and continued up until the conclusion of the experiment at 60 days. This preceded elevations in markers of oxidative damage to proteins and lipids in the liver.
Table 2.
Effect of chronic life stress or neuroendcirne stress mediators on selected hallmarks of aging: SNCs, SASP and sterile inflammation
Finding | Species | Population | Stress model/proxy | Tissue | Reference |
---|---|---|---|---|---|
| |||||
Elevated proinflammatory cytokines | Rats | wildtype | Chronic unpredictable mild stress; 20–60 days | Blood plasma | Lopez-Lopez et al., 2016 |
Oxidative damage to proteins and lipids | Rats | wildtype | Chronic unpredictable mild stress; 20–60 days | Liver | Lopez-Lopez et al., 2016 |
Elevated Il-6, decreased IL-10 | Mice | Female wildtype | Restraint; 6h/day for 28 days | Blood plasma | Voorhees et al., 2013 |
Decreased IL-10 | Mice | Female wildtype | Restraint; 6h/day for 28 days | hippocampus, cortex | Voorhees et al., 2013 |
Microglial activation | Mice | Male wildtype | Restraint; 2 30 min sessions per day; 14 days | Brain | Tynan et al.,2010 |
Microglial activation and production of proinflammatory cytokines | Mice | Male wildtype | Repeated social defeat; 2h per day for 6 days | Brain | Wohleb et al., 2011 |
Elevated proinflammatory cytokines, iNOS Elevated | Mice | Male wildtype | Repeated social defeat; 2h per day for 6 days | Brain | Wohleb et al., 2012 |
proinflammatory cytokines, invasion by peripheral immune cells | Mice | Male wildtype | Repeated social defeat; 2h per day for 6 days | Brain | Wohleb et al., 2013 |
Elevated proinflammatory immune cell activity | Human | Young adults | Low SES | PBMCs | Powell et al., 2013 |
Elevated proinflammatory immune cell activity | Mice | Male wildtype | Repeated social defeat; 2h per day for 6 days | PBMCs | Powell et al., 2013 |
Elevated proinflammatory immune cell activity | Human | Medical residents | Reported stress levels | PBMCs | Heidt et al., 2014 |
Increased p16 or p53 in various organs; | Mice | wild type | chronic psychosocial stress | liver, lung, spleen | Razzoli et al., 2018 |
Increased p16 | Human | adults | chronic stress | PBMC | Rentscher et al., 2019 |
Neuroinflammation is another well-established consequence of chronic stress and increased inflammation is a common phenotype of normal brain aging across species (Frank et al., 2006). Several stressors promote microglial activation, as measured by Iba1 activity (Calcia et al., 2016). In particular, a very strong effect has been found in area CA1 of the hippocampus (Bian et al., 2012). This increase in microglial activation leads to the secretion of cytokines, chemokines and other SASP factors facilitating the development of anxiety-like behavior (Tynan et al., 2010; Wohleb et al., 2013). Stress exposure also promotes production of proinflammatory monocytes in the bone marrow, which are released into general circulation and can migrate and localize in the brain in addition to other organs (Heidt et al., 2014; Wohleb et al., 2013) thereby secreting SASP factors. Finally, a less explored area in this field is the neuro-immune modulation of bone structure during homeostasis and upon trauma. Mice lacking tyrosine hydroxylase, the rate limiting enzyme for the synthesis of norepinephrine in myeloid cells are protected from chronic psychosocial stress-induced disturbance of bone growth and healing (Tschaffon-Müller et al., 2023), a finding matched by knockout of β2-adrenoceptor in chondrocyte.
EFFECT OF LIFE STRESS ON ROS AND DNA DAMAGE
The free radical theory of aging, first proposed in the 1950s, posits that aging is the result of accumulated damage created by reactive oxygen species (ROS) (Harman, 2009). Though it has now been shown that oxidative damage does not alone drive aging, it is certainly one of the mechanisms contributing to the aging process. ROS, and the damage that they wreak are a major cause of cellular senescence. ROS and oxidative damage increase with age, and increased production of ROS is associated with shortened lifespan (Kirkwood and Kowald, 2012; Stadtman and Levine, 2000). ROS are byproducts of normal cellular metabolism, are present at low levels during homeostasis (Boonstra and Post, 2004) and serve as signaling molecules in optimally functioning cells. ROS are also capable of modifications to DNA. The most frequent form of oxidative damage to DNA is 8-hydroxylation of guanine (Poulsen et al., 2014). In some cases, ROS-induced damage causes rupture to the DNA sugar-phosphate backbone resulting in single or double strand breaks (DSB), which are the most dangerous form of DNA damage. Attempts to repair DSBs can cause loss or translocation of chromosomal segments causing tumorigenesis or replicative senescence. Specialized protein kinases sense the DSB, causing local phosphorylation of the histone H2AX (called γ-H2AX in its phosphorylated form), a classical marker of SNC and a signaling cascade culminating in activation of the master regulator p53. p53 induces cell cycle arrest by activating transcription of p21, as described above in the cellular senescence section.
Studies over the last 30 years have demonstrated that psychological stress, both acute and chronic, can result in the formation of ROS (See also (Salim, 2016; Sharifi-Rad et al., 2020) for additional review) (Table 3). Measures of oxidative damage were obtained in lymphocytes from undergraduate students immediately prior to an exam, and on a “non-stress” occasion for comparison (Sivonová et al., 2004) and found increased DNA damage, greater lipid susceptibility to oxidation, and decreased antioxidant levels in plasma on the exam day. In another study on undergraduate students, exam stress increased saliva levels of the antioxidant catalase and sialic acid, and decreased levels of oxidized proteins (Tsuber et al., 2014). The increase in catalase was greater in female participants than in male, indicative of greater intrinsic antioxidant defense. This could represent a compensatory mechanism to combat elevated ROS. Exposure to the Trier Social Stress Test (Kirschbaum et al., 1993) – a classical test in which participants perform a public speaking task and then attempt mental arithmetic – elevates saliva levels of several miRNAs (Wiegand et al., 2018) including miR-21, directly inhibits superoxide dismutase (SOD), resulting in impaired ROS degradation.
Table 3.
Effect of chronic life stress or neuroendcirne stress mediators on selected hallmarks of aging: ROS and DNA damage
Finding | Species | Population | Stress model/proxy | Tissue | Reference |
---|---|---|---|---|---|
| |||||
Greater DNA damage (assessed by comet assay), higher lipid peroxidation, decreased antioxidant levels | Human | University students | Taking an exam | Leukocytes, plasma | Sivonova et al., 2004 |
Greater protein and lipid oxidative damage; lower antioxidant level | Human | Female university students | Taking an exam | Blood serum | Nakhaee et al., 2013 |
Lower antioxidant levels; wack sperm | Human | Male university students | Taking an exam | Semen | Eskiocak et al., 2005 |
Higher inhibitors of antioxidants | Human | University students | Trier Social Stress Test | Saliva | Wiegand et al., 2018 |
Increased antioxidant levels | Human | University students | Taking an exam | Saliva | Tsuber et al., 2014 |
Greater oxidative stress | Human | Female adolescents | Adverse childhood events | Urine | Horn et al., 2019 |
Greater lipid peroxidation, IL-6 | Human | Children | Low SES | Blood serum | Mansur et al., 2016 |
Greater oxidative stress | Human | Hypertensive patients | High trait anxiety | PBMCs, blood plasma | Yasunari et al., 2006 |
Greater lipid peroxidation | Rat | wildtype | Acute air jet stress | Plasma | D’Angelo et al., 2010 |
Single 5-hour communication box stress | Adachi et al., 1993 | ||||
No effect | Rat | wildtype | Liver | ||
Increased oxidative damage; returns to control levels within 1 hour | Rat | wildtype | 3–4 exposures to 5-hour communication box stress | Liver | Adachi et al., 1993 |
Increased oxidative damage, HSP70 decreased antioxidant activity, | Rat | wildtype | Communication box stress exposure; 1h/day for 3 or 5 weeks | Masseter muscle | Li, Zhang et al., 2011 |
Increased lipid peroxidation, DNA fragmentation, decreased cytochrome C | Rat | wildtype | Water immersion stress; acute; 90min | Gastric/Intestinal mucosa | Bagchi et al., 1999 |
Increased lipid peroxidation, DNA fragmentation, decreased cytochrome C | Rat | wildtype | Water immersion stress; 15min/day for 15 days | Gastric/Intestinal mucosa | Bagchi et al., 1999 |
Elevated ROS, elevated NO, increased lipid peroxidation, protein carbonylation; reduced antioxidants | Mice | wildtype | Restraint; 5h/day, 14 days | hippocampus, cortex, cerebellum | Son et al., 2016 |
Increased oxidative DNA damage, reactive nitrogen species | Rat | wildtype | Social isolation; 14 days | Nucleus accumbens, hypothalamus | Colaianna et al., 2013 |
Increased oxidative DNA damage, reactive nitrogen species Increased | Rat | wildtype | Social isolation; 28 days | prefrontal cortex | Colaianna et al., 2013 |
oxidative DNA damage, reactive nitrogen species | Rat | wildtype | Social isolation; 49 days | adrenal glands | Colaianna et al., 2013 |
Increased oxidative DNA damage | Mice | SAMP-10 Human | confrontational housing | cerebral cortex | Unno et al., 2011 |
Increased ROS, RNS, DNA damage, impaired DNA repair | Cell culture | Breast cancer lines MDA-MB-231, MCF-7 Human | Cortisol (15, 30, or 90min) | cell culture | Flaherty et al., 2017 |
Increased ROS, RNS, DNA damage, impaired DNA repair | Cell culture | Breast cancer lines MDA-MB-231, MCF-7 | Cortisol (15, 30, or 90min) | cell culture | Flaherty et al., 2017 |
Increased iNOS | Mice | females injected in mammary fat pad with human breast cancer cells | Restraint; 5h/day, 14 days | Mammary tumors | Flaherty et al., 2017 |
Elevated DNA damage, impaired DNA repair, upregulated cell cycle inhibitor CDC25A | Cell culture | Murine 3T3 cells | Cortisol (30 min) | cell culture | Flint et al., 2007 |
Elevated DNA damage, impaired DNA repair, upregulated cell cycle inhibitor CDC25A | Cell culture | Murine 3T3 cells | Norepinephrine (30 min) | cell culture | Flint et al., 2007 |
Elevated DNA damage, upregulated cell cycle inhibitor CDC25A | Cell culture | Murine 3T3 cells | Epinephrine (30 min) | cell culture | Flint et al., 2007 |
Poorer DNA repair | Human | Nonpsychotic psychiatric patients avg age 32 | Reported distress (Minnesota Multiphasic Personality Index) | Lymphocytes | Kiecolt-Glaser et al., 1985 |
Increased DNA damage | Mice | wildtype males | Isoproterenol | Thymus | Hara et al., 2011 |
Increased DNA damage | Cell culture | Human U2OS, Murine MEF | Isoproterenol | cell culture | Hara et al., 2011 |
The stress increase in ROS suggests that compensatory mechanisms to combat acute stress-induced ROS can be insufficient in the long term under certain conditions. Adolescents who experience multiple ACE showed increased urinary levels of the oxidative stress markers F2-isoprostanes (Horn et al., 2019). Socioeconomic disadvantage is associated with elevated plasma serum thiobarbituric acid-reactive substance as well as IL-6 (Mansur et al., 2016). Finally, hypertensive patients with high trait anxiety have higher plasma levels of norepinephrine, which correlates with increased ROS in mononuclear immune cells (Yasunari et al., 2006).
Rodent studies have recapitulated the stress-induced ROS phenomenon observed in humans and have shed greater light on the mechanisms underlying this effect. A 5-hour exposure to the communication box stress paradigm did not affect rat liver levels of OH8dG – a product of hydroxyl radical induced DNA damage (Adachi et al., 1993). The induced increase returned to levels equivalent to that of controls 1-hour after the stress exposure. Thus, it appears that neutralization of ROS is possible after several repeated stress exposures. However, after a second, third or fourth exposure, stress-exposed rats had significantly higher levels of OH8dG in liver nuclear DNA. Another experiment further elucidated the time course in which stress-induced ROS recovery can be compromised. This experiment employed the same communication box stress paradigm on rats in a chronic study design. The rats were exposed for 1-hour per day for 1, 3 or 5 weeks. Researchers subsequently found an increase in lipid peroxidation marker MDA content in the masseter muscles after 3 and 5 weeks but not 1 week of stress (Li et al., 2011). Concurrently, following 3 or 5 weeks of stress, they found a decrease in the activity of antioxidant enzymes SOD, glutathione peroxidase, and catalase in the masseter muscles. In rats exposed for 5 weeks there was also an elevation in the cellular stress-sensitive protein HSP70. Together, these studies suggest that psychological stress can induce an imbalance favoring greater ROS and resulting in oxidative damage, despite upregulation of the protective HSP70. Psychological stress is capable of increasing ROS in numerous tissue types, though it seems to have a particularly profound effect on the brain. 14 days of non-social psychological restraint stress resulted in higher levels of ROS in the hippocampus and cerebral cortex, and higher levels of nitric oxide in the hippocampus and cerebellum in mice (Son et al., 2016). It also induced greater levels of lipid peroxidation marker MDA in all three brain regions and protein peroxidation marker protein carbonyl in the hippocampus and cortex. In contrast it reduced levels of the antioxidant enzymes SOD and glutathione reductase-transferase.
In rats, 2 weeks of social isolation are sufficient to increase mRNA expression of NADPH oxidase (NOX2), an enzyme that catalyzes formation of superoxide anion O2, in the nucleus accumbens and hypothalamus (Colaianna et al., 2013). This is associated with an increase in 8-OHdG in both regions, and an increase in nitrotyrosine – a marker of reactive RNS— in the hypothalamus. After 4 weeks, elevations of NOX2 and 8-OHdG in the prefrontal cortex can be detected as well. After 7 weeks, these markers are also elevated in the adrenal glands. The majority of 8-OHdG and nitrotyrosine positive cells in the hypothalamus were neurons. In the prefrontal cortex, the NOX2 subunit p47phox was increased specifically in pyramidal neurons, not in GABAergic neurons, astrocytes, or microglia (Schiavone et al., 2012). Alterations in behavior, increase in glutamate levels and loss of parvalbumin were detectable after 4 weeks of social isolation.
In the accelerated senescence SAMP-10 mouse model, confrontational housing — a model for psychosocial stress — increased levels of the DNA damage marker 8-oxo-dG can be found in the cerebral cortex (Unno et al., 2011). This was also associated with accelerated cognitive decline and shorter lifespan.
The mechanisms underlying stress-induced ROS overproduction have been further illuminated using molecular mediators of the stress response using cell culture models. Acute exposure to cortisol or norepinephrine was sufficient to increase levels of ROS and RNS in breast cancer cells, which was paralleled by increase in γ-H2AX foci, indicative of DNA damage and senescence, an effect mediated by their respective receptor antagonists (Flaherty et al., 2017). In vitro and in vivo studies using endocrine mediators of the stress response indicate that stress may impair the repair of damaged DNA. Short term (<30 minute) in vitro exposure of murine 3T3-L1 cells to cortisol or norepinephrine not only increase DNA damage on its own, but also interfered with DNA repair in cells exposed to UV radiation (Flint et al., 2007). Epinephrine exposure produced a similar degree of DNA damage but did not impair DNA repair. All three stress-related hormones induced upregulation of DNA damage sensors Chk1 and Chk2, and the proto-oncogene CDC25A, which is involved in cell cycle delay following DNA damage. Consistently, 24-hour exposure to epinephrine or norepinephrine increases DNA damage, in the murine 3T3-L1 cells, an effect that was blocked by pre-treatment with the β-adrenergic receptor antagonist propranolol (Flint et al., 2013). This damage is functionally relevant, as cells treated with either catecholamine undergo greater malignant transformation in-vitro and become tumors more rapidly in-vivo when implanted in mice (Flint et al., 2013). These findings in murine systems can be extended to other cell types and species. In human breast cancer cells, DNA repair following exposure to H2O2 was impaired in the presence of cortisol (Flaherty et al., 2017). Furthermore, lymphocytes sampled from participants who scored highly for distress on the Minnesota Multiphasic Personality Inventory exhibited poorer DNA repair 5 hours after X-irradiation (Damjanovic et al., 2007; Kiecolt-Glaser et al., 1985). Consistent with in vitro work, mice treated with the β-adrenergic receptor agonist isoproterenol accumulated DNA damage in the thymus, as quantified by γ-H2AX (Hara et al., 2011). Isoproterenol treatment also decreased levels of the DNA damage responder, and senescence regulator p53. These findings were replicated in vitro in several cell lines and was further demonstrated to be mediated specifically by β2-adrenergic receptor (β2-AR) signaling. The authors suggest that stress-induced sympathetic nervous system activity, promotes DNA damage both directly, and indirectly via suppression of p53-mediated DNA repair (Hara et al., 2011). This may consequently result in worse DNA damage, and a lower likelihood of successful repair ultimately leading to permanent senescence.
This substantial body of work provides convincing evidence that life stress or stress-related hormones causes an increase in ROS production, and that this results in oxidative damage to DNA in numerous cell types/organs. Stress-induced ROS overproduction directly results in DNA damage — one of the primary triggers for cellular senescence. Multiple stress hormones appear to induce DNA damage via increased ROS production, as well as impair DNA repair. However, stress relevant studies using murine models are limited in this area and conclusions obtained using in vitro system remain to be confirmed in vivo.
EFFECT OF LIFE STRESS ON EPIGENETIC REGULATION
The hallmarks of aging discussed in the previous sections are implicated in shaping the progression of biological aging, the gradual process that leads to a functional decline over the lifetime and that represents a major risk factor for age-related diseases. Biological age is not necessarily reflective of an individual’s chronological age because it encodes exposures to disease, lifestyle changes, environmental factors, stress, etc., that result in acceleration or deceleration of the pace of aging and of the linked propensity to develop pathologies (Zhang and Gladyshev, 2020). Biological age can be estimated using various approaches including epigenetic biomarkers – specifically DNA methylation (DNAm) changes. DNAm consists of the addition of a methyl group to a CpG dinulceotide in most cases associated with transcriptional repression via its effect on chromatin accessibility. DNAm occurs mostly following predictable patterns, initially established for organismal development and later modulated for maintenance, resulting in gain of DNAm at gene promoters and loss of general DNAm during aging (Singer, 2019). Similarly, epigenetic alterations to DNA and chromatin play a crucial role in mediating the induction and maintenance of cellular senescence, though processes such as heterochromatin loss and instability, histone acetylation and methylation, the action of Polycomb-group proteins (Crouch et al., 2022). Like in aging, DNAm patterns in senescent cells consist of a general state of hypomethylation (Cruickshanks et al., 2013). While sharing considerable overlap with age-associated methylation patterns, the ones associated with senescence also present differences at specific CpG sites, most commonly at HOX genes and genes mediating cellular differentiation (Bork et al., 2010). Altogether, alterations in DNAm and associated enzymes during aging and senescence significantly contribute to cellular dysfunction, facilitate a chronic inflammation state via SASP activation and may mediate certain age-related pathologies.
These changes in methylation patterns occupy a major field in geroscience. The predictability of these changes enable the application of machine learning techniques to develop age predictors – termed epigenetic clocks (Horvath and Raj, 2018). Since their advent (Horvath clock, (Horvath, 2013); Hannum clock, (Hannum et al., 2013)), a number of additional epigenetic clocks have been developed incorporating DNAm levels at sites correlating with morbidity and mortality and therefore better associated with measures of physiological dysregulation PhenoAge, (Levine et al., 2018); GrimAge, (Lu et al., 2019); DunedinPoAm, (Belsky et al., 2020)).
Increasing evidence links psychosocial stress with accelerated epigenetic aging (See also (Palma-Gudiel et al., 2020) for additional review) (Table 4). Social stress is believed to become biologically encoded through epigenetic modifications of genes such as the glucocorticoid receptor, CRH and FKBP5 genes (Cunliffe, 2016; Zannas et al., 2019). Stress related alterations in neuronal activity have also been linked to changes in DNAm (Fitzgerald et al., 2021), possibly due to numerous CpG sites they include are in the glucocorticoid response elements (Horvath et al., 2013). The administration of exogenous glucocorticoids has been shown to affect DNAm and transcription in many of the genes neighboring CpG of the Horvath’s clock (Zannas et al., 2020). The DunedinPACE biomarker of aging represents the proof of concept that the pace of aging can be measured from the DNAm levels of the most reliable CpG sites assessed at one timepoint jointly with biomarkers of physiological integrity (Belsky et al., 2022). The landmark Dunedin studies reflect at a cohort of over 1000 individuals followed longitudinally, determined biological age through indicators of organ system integrity recorded in multiple occasions and a single time point DNAm and estimated a faster aging pace in young adults with a history of ACE (Belsky et al., 2022, 2020).
Table 4.
Effect of chronic life stress or neuroendcirne stress mediators on selected hallmarks of aging: Epigenetic changes
Finding | Species | Population | Stress model/proxy | Tissue | Reference |
---|---|---|---|---|---|
| |||||
Resilience dependent Grim Age acceleration | Human | adults | Perceived stress score | Whole blood | Bergquist et al., 2022 |
No association with DNA methylaton age acceleration | Human | older adults | Perceived stress score | Whole blood | Vetter et al., 2022 |
Increased epigenetic aging via DNA methylation | Human | adolescents | economic trajectories during the Great Recession | blood leukocytes | Chen et al., 2016 |
Increased epigenetic aging via DNA methylation | Human | adults | Low SES | Whole blood | Fiorito et al., 2017 |
Increased epigenetic aging via DNA methylation | Human | adults | Low SES | Whole blood | Simons et al., 2017 |
Older epigenetic age in high ranking individuals | baboons | adults | high rank | Whole blood | Anderson et al, 2022 |
Increased DNAm in low rank individuals | mice | adult/old | low rank | liver | Razzoli et al., 2023 |
Childhood trauma has been linked by several studies to epigenetic age acceleration similarly to long term shift work, sleep deprivation and obesity (Lim et al., 2022). Interestingly, the effect of stress on epigenetic aging depends on the individual coping style and resilience, with multiple studies showing an inverse correlation between acceleration of epigenetic age due to stress exposure and coping ability in individuals exposed to earlier trauma (Bergquist et al., 2022; Galow and Peleg, 2022; Vetter et al., 2022).
Human epigenetic clocks are based on DNAm levels obtained mostly with micro-array assay, while the development of array-based aging clock for other species has been somewhat trailing behind, due to the lack of availability of BeadChips platforms for non-human species. Nevertheless, new studies now show that in social species individuals experiencing high levels of glucocorticoids and upregulated inflammation related pathways also show acceleration of biological aging based on telomere length as well as epigenetic clocks (Anderson et al., 2021). Wild baboons have been shown to pay the elevated energetic cost of maintaining high ranking social position by accruing an older epigenetic age than lower ranking individuals (Anderson et al., 2021), thus attributing to social determinants as the source of the variance in biological aging observed in natural populations. Murine epigenetic clocks have been developed based on sequencing-based methods as tissue-specific (i.e., liver (Wang et al., 2017), blood (Petkovich et al., 2017), brain (Liu et al., 2021); or multi-tissue (Kerepesi et al., 2021; Stubbs et al., 2017). Among the manipulations that have been found to affect epigenetic age in mice so far are, caloric restriction (Cole et al., 2017), dietary rapamycin treatment (Wang et al., 2017) or high fat diet feeding (Stubbs et al., 2017). The association between stress and epigenetic changes has been recently reviewed, particularly regarding their impact on rodent behavior (Dion et al., 2022). However, only a recent study to date has specifically examined the relationship between life stress, DNAm, epigenetic clocks and lifespan in a mouse model of chronic psychosocial stress (Razzoli et al., 2023). This study identified epigenetic changes (DNAm) in the liver of male mice, which are in line with increased risk conferred by genetic background (Sv129Ev > C57BL/6J > CD1) and social rank (low rank > high rank) on healthspan and lifespan. In this study, mouse epigenetic age was found to be strain dependent, while no link was found to the mouse social standing, likely due to the inadequacy of the available epigenetic clocks based on mouse DNAm array data. This limited knowledge on how life stress impacts epigenetic clock, as well as the unclear interaction of stress, epigenetic clocks, biological age and cellular senescence is likely to be rectified as research and technology advances. Learning from the evolution undergone by the human epigenetic clock, it’s desirable that future mouse studies will be designed taking into consideration measures stress experiences, DNAm and physiological phenotypes in the same population. Acquisition of this knowledge will be facilitated by accessibility of reliable experimental and computational tools and by conscious experimental design.
KNOWLEDGE GAP AND FUTURE DIRECTIONS
Given the degree of novelty the field of geroscience is infused of in terms of conceptual, computational, biological and technological innovations (Duque et al., 2023; Epel, 2020; Kennedy et al., 2014; Sierra and Kohanski, 2023), it is not surprising that knowledge gaps of are still numerous. Amongst the more urgent needs, studies are required in which social experiences, biological age measures and stress phenotypes are measured in the same population to elucidate the mechanistic link between social experience and system function. In line with this, more animal studies should examine the effect of varying type/timing of social experience on biomarkers of aging and stress phenotype, both in the laboratory as well as in wild settings.
While DNAm is one of the most accurate and reliable biomarkers of aging and disease, our understanding of it is still in its infancy. Particularly whether changes in DNAm is causal to aging remains to be shown (Simpson and Chandra, 2021). Similarly, despite the role of SNC in aging and disease risk, there is sparse information about their cellular identity and features as well as about the implications of their heterogeneity in human tissues. None of the current SNC biomarkers are specific or unique to SNC (P. J. Lee et al., 2022), calling for the need of developing multiple targeted and unbiased approaches to identify these cells. Overall, these efforts should culminate in the identification of sensitive and specific gerodiagnostics – biomarkers to quantify the various hallmarks of aging. – and in the design of interventions; this would allow the elevation of biological age by stress to become a truly quantifiable and actionable therapeutic target (Chaib et al., 2022; Kirkland et al., 2017).
Lastly, a significant gap of knowledge concerns the mechanism through which life stress affects multiple hallmarks of aging, impairs multi-organ physiology, causes comorbid diseases, and overall affects life expectancy. Figure 1 describes the major stress-related neuro-endocrine systems as well as the consequences of life stress on selected hallmarks of aging at the organ level in humans and animal models. However, the specific link between one or more of those stress-related axes, and a given change in these hallmarks is still unclear, and largely unexplored. Cellular models have been used to tackle this question by testing the effects of “stress hormones” such as cortisol on induction of hallmarks of aging (Bobba-Alves et al., 2023; Laberge et al., 2012; Zannas et al., 2020). Limited studies have also tested the role of such mediators (catecholamines) on grey hair formation and stem cell exhaustion (Zhang et al., 2020). Yet contrasting findings have been obtained in those studies ranging from induction to inhibition of SNC, SASP and telomere shortening, suggesting that the in vivo “neuro-symphony of stress” mediators (Joëls and Baram, 2009) can interact in complex manners during the life course to impact fundamental biological mechanisms affecting organ function and aging. In essence, it appears that the use of conventional reductionist approaches of decomposing organisms into unitary tracts manipulated one at a time by using loss/gain of function of single genes/factors can be ineffective to tackle the fundamental question of how life stress accelerates aging. To use Jacob’s words “Clearly, an understanding of the simple is necessary to understand the more complex, but whether it is sufficient is questionable” (Jacob, 1977). An alternative holistic approach focused on the organism as a unit, regulated by multiple – sometime opposing – physiological control mechanisms should be preferred (Stambler, 2017). These physiological control mechanisms should be ideally investigated simultaneously to determine how the organism adapts or succumbs to the strain posed by multiple environmental challenges (including social interactions). However, such a construct remains to be defined in terms of experimental design and feasibility. Artificial intelligence algorithms and innovative bioengineering approaches can ultimately benefit the use of holistic approaches by sharing the potential to address multiple functions in the same organism over its lifespan as well as in large enough populations to contrast background noise and intrinsic inter-individual variability.
The field of geroscience attempts to identify fundamental processes affecting aging in multiple organs and in multiple dimensions, aiming at developing therapies to extend the healthy lifespan. Classical examples of interventions improving healthy-lifespan, and/or extending lifespan in some animal studies, include caloric restriction and exercise (Balasubramanian et al., 2017; Huffman et al., 2016; Waziry et al., 2023). One of the major innovations concerns the promise offered by drug treatments with anti-aging effect such as rapamycin, metformin and others (Partridge et al., 2020) as well as systemic, or targeted clearance of SNC (senolytics) or inhibition of the effects exerted by SASP (senomorphics) to offset multi-morbidities of aging and compress pathologies overall benefiting healthy aging (Childs et al., 2017; Kirkland et al., 2017; P. J. Lee et al., 2022). To the best of our knowledge, no study has been conducted in which human subjects or animal models exposed to life stress – characterized by impaired healthspan, increased biological age, and reduced lifespan - were randomized to receive therapeutic interventions proven to exert a beneficial effect in stress-free populations.
Intriguingly, in parallel to the extensive literature discussed above showing that life stress increase biological aging in humans and mice, a recent study showed that surgery, pregnancy or viral infection increase epigenetic age in humans, albeit reversibly upon recovery (Poganik et al., 2022). Although pregnancy, surgery, and viral infections cannot be defined as stressors per se [based on the definition used in this paper (Koolhaas et al., 2011) and by others (Epel et al., 2018)], they share some similarity with life stressors, including elevated HPA axis, and often catecholamines, suggesting that life stress-induced acceleration of biological aging could be sensitive to interventions increasing resilience and/or inducing reversal.
Additionally, genetic reprogramming of aging-induced DNA methylation patterns has recently been proposed to “rejuvenate” organ functions and hallmarks of aging in mice models (Lu et al., 2020; Poganik et al., 2022; Yang et al., 2023). Can clearance of stress-induced SNC or targeted/global reprogramming of stress-induced DNA hypo/hyper methylation be sufficient to oppose the multi-morbidities imposed by life stress? No empiric answer is available yet.
CONCLUSION
Chronic stress poses a significant public health threat and is a likely major source of health disparities. It is associated with elevated risk for chronic aging related diseases, which are the primary causes of disability and premature death in older adults. Psychosocial stress is a particularly potent form of stress and is a key contributor to the numerous adverse health outcomes associated with ACE and low SES. Chronic stress has long been theorized to accelerate the aging process. In this Review, we gave an overview of the scientific literature that implicates a link between psychological stress and several pillars/hallmarks of aging including cellular senescence and DNAm. Stress has been demonstrated to promote numerous senescence triggers, including telomere erosion, oxidative damage, and DNA damage. Moreover, stress is associated with systemic inflammation, which can both promote senescence-inducing cellular damage, and be a consequence of SNC accumulation. Overall, available studies suggest that life stress can lead to a vast array of aging-related diseases at least in part by activating cellular senescence and other biological processes associated with premature aging. Therefore, we postulate that these senescence/aging pathways should be considered as an intrinsic biological manifestation of chronic stress-induced perturbations. More attention should be focused on how endocrine/neural mediators of the stress response affect aging-associated processes and how buffering those negative effects could be used therapeutically to reduce the pathological burden of stress on life expectancy.
Highlights.
Chronic stress is a major risk factor for a multitude of age-related diseases.
Hallmarks of aging are basic biological mechanisms that can lead to accelerated aging.
Stress has been increasingly associated with the dysregulation of the hallmarks of aging.
We review evidence on the contribution of life stress on cellular senescence, sterile inflammation, telomere length, production of reactive oxygen species, DNA damage, and epigenetic changes.
We evaluate stress-induced senescence and accelerated aging as valid etiopathological proposition.
Acknowledgments
FINANCIAL SUPPORT
A.B. is supported by NIH/NIA R61 AG078520, NIH/NIA R24 AG065172, MN Partnership for Biotechnology and Molecular Genomic #18.4, NEXTGENERATIONEU (NGEU), funded by the Ministry of University and Research (MUR), National Recovery and Resilience Plan (NRRP), project MNESYS (PE0000006) - A Multiscale integrated approach to the study of the nervous system in health and disease (DN. 1553 11.10.2022).
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
BIBLIOGRAPHY
- Adachi S, Kawamura K, Takemoto K, 1993. Oxidative damage of nuclear DNA in liver of rats exposed to psychological stress. Cancer Res 53, 4153–4155. [PubMed] [Google Scholar]
- Adams J, Martin-Ruiz C, Pearce MS, White M, Parker L, von Zglinicki T, 2007. No association between socio-economic status and white blood cell telomere length. Aging Cell 6, 125–128. 10.1111/j.1474-9726.2006.00258.x [DOI] [PubMed] [Google Scholar]
- Adler NE, Boyce T, Chesney M. a, Cohen S, Folkman S, Kahn RL, Syme SL, 1994. Socioeconomic status and health. The challenge of the gradient. The American psychologist 49, 15–24. 10.1037/0003-066X.49.1.15 [DOI] [PubMed] [Google Scholar]
- Anderson JA, Johnston RA, Lea AJ, Campos FA, Voyles TN, Akinyi MY, Alberts SC, Archie EA, Tung J, 2021. High social status males experience accelerated epigenetic aging in wild baboons. Elife 10, e66128. 10.7554/eLife.66128 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Aschbacher K, O’Donovan A, Wolkowitz OM, Dhabhar FS, Su Y, Epel E, 2013. Good stress, bad stress and oxidative stress: insights from anticipatory cortisol reactivity. Psychoneuroendocrinology 38, 1698–708. 10.1016/j.psyneuen.2013.02.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Baek JH, Son H, Jeong Y-H, Park SW, Kim HJ, 2019. Chronological Aging Standard Curves of Telomere Length and Mitochondrial DNA Copy Number in Twelve Tissues of C57BL/6 Male Mouse. Cells 8, 247. 10.3390/cells8030247 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Balasubramanian P, Howell PR, Anderson RM, 2017. Aging and Caloric Restriction Research: A Biological Perspective With Translational Potential. EBioMedicine 21, 37–44. 10.1016/j.ebiom.2017.06.015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bartolomucci A, 2007. Social stress, immune functions and disease in rodents. Frontiers in Neuroendocrinology 28, 28–49. 10.1016/j.yfrne.2007.02.001 [DOI] [PubMed] [Google Scholar]
- Bartolomucci A, Fuchs E, Koolhaas JM, Ohl F, 2009. Acute and chronic social defeat: Stress protocols and behavioral testing, in: Mood and Anxiety Related Phenotypes in Mice, Neuromethods. Humana Press, pp. 261–275. 10.1007/978-1-60761-303-9_14 [DOI] [Google Scholar]
- Belsky DW, Caspi A, Arseneault L, Baccarelli A, Corcoran D, Gao X, Hannon E, Harrington HL, Rasmussen LJH, Houts R, Huffman K, Kraus WE, Kwon D, Mill J, Pieper CF, Prinz J, Poulton R, Schwartz J, Sugden K, Vokonas P, Williams BS, Moffitt TE, 2020. Quantification of the pace of biological aging in humans through a blood test, the DunedinPoAm DNA methylation algorithm. eLife 9, 1–56. 10.7554/ELIFE.54870 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Belsky DW, Caspi A, Corcoran DL, Sugden K, Poulton R, Arseneault L, Baccarelli A, Chamarti K, Gao X, Hannon E, Harrington HL, Houts R, Kothari M, Kwon D, Mill J, Schwartz J, Vokonas P, Wang C, Williams BS, Moffitt TE, 2022. DunedinPACE, a DNA methylation biomarker of the pace of aging. Elife 11, e73420. 10.7554/eLife.73420 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Berger JM, Singh P, Khrimian L, Morgan DA, Chowdhury S, Arteaga-Solis E, Horvath TL, Domingos AI, Marsland AL, Kumar Yadav V, Rahmouni K, Gao X-B, Karsenty G, 2019. Mediation of the Acute Stress Response by the Skeleton. Cell Metabolism. 10.1016/j.cmet.2019.08.012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bergquist SH, Wang D, Smith AK, Roberts DL, Moore MA, 2022. Hormetic association between perceived stress and human epigenetic aging based on resilience capacity. Biogerontology 23, 615–627. 10.1007/s10522-022-09985-8 [DOI] [PubMed] [Google Scholar]
- Bian Y, Pan Z, Hou Z, Huang C, Li W, Zhao B, 2012. Learning, memory, and glial cell changes following recovery from chronic unpredictable stress. Brain Research Bulletin 88, 471–476. 10.1016/j.brainresbull.2012.04.008 [DOI] [PubMed] [Google Scholar]
- Blackburn EH, Epel ES, Lin J, 2015. Human telomere biology: A contributory and interactive factor in aging, disease risks, and protection. Science 350, 1193–1198. 10.1126/science.aab3389 [DOI] [PubMed] [Google Scholar]
- Bobba-Alves N, Sturm G, Lin J, Ware SA, Karan KR, Monzel AS, Bris C, Procaccio V, Lenaers G, Higgins-Chen A, Levine M, Horvath S, Santhanam BS, Kaufman BA, Hirano M, Epel E, Picard M, 2023. Cellular Allostatic Load is linked to Increased Energy Expenditure and Accelerated Biological Aging. Psychoneuroendocrinology 106322. 10.1016/j.psyneuen.2023.106322 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bonanni L, Franciotti R, Martinotti G, Vellante F, Flacco ME, Di Giannantonio M, Thomas A, Onofrj M, 2018. Post Traumatic Stress Disorder Heralding the Onset of Semantic Frontotemporal Dementia. Journal of Alzheimer’s disease : JAD 63, 203–215. 10.3233/JAD-171134 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bonewald L, 2019. Use it or lose it to age: A review of bone and muscle communication. Bone 120, 212–218. 10.1016/j.bone.2018.11.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Boonstra J, Post JA, 2004. Molecular events associated with reactive oxygen species and cell cycle progression in mammalian cells. Gene 337, 1–13. 10.1016/j.gene.2004.04.032 [DOI] [PubMed] [Google Scholar]
- Bork S, Pfister S, Witt H, Horn P, Korn B, Ho AD, Wagner W, 2010. DNA methylation pattern changes upon long-term culture and aging of human mesenchymal stromal cells. Aging Cell 9, 54–63. 10.1111/j.1474-9726.2009.00535.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bussian TJ, Aziz A, Meyer CF, Swenson BL, van Deursen JM, Baker DJ, 2018. Clearance of senescent glial cells prevents tau-dependent pathology and cognitive decline. Nature 562, 578–582. 10.1038/s41586-018-0543-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- Calcia MA, Bonsall DR, Bloomfield PS, Selvaraj S, Barichello T, Howes OD, 2016. Stress and neuroinflammation: A systematic review of the effects of stress on microglia and the implications for mental illness. Psychopharmacology 233, 1637–1650. 10.1007/s00213-016-4218-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Campisi J, d’Adda di Fagagna F, 2007. Cellular senescence: when bad things happen to good cells. Nature Reviews Molecular Cell Biology 8, 729–740. 10.1038/nrm2233 [DOI] [PubMed] [Google Scholar]
- Cappola AR, Auchus RJ, El-Hajj Fuleihan G, Handelsman DJ, Kalyani RR, McClung M, Stuenkel CA, Thorner MO, Verbalis JG, 2023. Hormones and Aging: An Endocrine Society Scientific Statement. The Journal of Clinical Endocrinology & Metabolism dgad225. 10.1210/clinem/dgad225 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cha H, Farina MP, Hayward MD, 2021. Socioeconomic status across the life course and dementia-status life expectancy among older Americans. SSM Popul Health 15, 100921. 10.1016/j.ssmph.2021.100921 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chaib S, Tchkonia T, Kirkland JL, 2022. Cellular senescence and senolytics: the path to the clinic. Nat Med 28, 1556–1568. 10.1038/s41591-022-01923-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- Charmandari E, Tsigos C, Chrousos G, 2005. Endocrinology of the stress response. Annual Review of Physiology 67, 259–284. 10.1146/annurev.physiol.67.040403.120816 [DOI] [PubMed] [Google Scholar]
- Cherkas LF, Aviv A, Valdes AM, Hunkin JL, Gardner JP, Surdulescu GL, Kimura M, Spector TD, 2006. The effects of social status on biological aging as measured by white-blood-cell telomere length. Aging Cell 5, 361–365. 10.1111/j.1474-9726.2006.00222.x [DOI] [PubMed] [Google Scholar]
- Childs BG, Gluscevic M, Baker DJ, Laberge RM, Marquess D, Dananberg J, Van Deursen JM, 2017. Senescent cells: An emerging target for diseases of ageing. Nature Reviews Drug Discovery 16, 718–735. 10.1038/nrd.2017.116 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chiou KL, DeCasien AR, Rees KP, Testard C, Spurrell CH, Gogate AA, Pliner HA, Tremblay S, Mercer A, Whalen CJ, Negrón-Del Valle JE, Janiak MC, Bauman Surratt SE, González O, Compo NR, Stock MK, Ruiz-Lambides AV, Martínez MI, Cayo Biobank Research Unit, Wilson MA, Melin AD, Antón SC, Walker CS, Sallet J, Newbern JM, Starita LM, Shendure J, Higham JP, Brent LJN, Montague MJ, Platt ML, Snyder-Mackler N, 2022. Multiregion transcriptomic profiling of the primate brain reveals signatures of aging and the social environment. Nat Neurosci 25, 1714–1723. 10.1038/s41593-022-01197-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cohen S, Doyle WJ, Baum A, 2006. Socioeconomic status is associated with stress hormones. Psychosom Med 68, 414–420. 10.1097/01.psy.0000221236.37158.b9 [DOI] [PubMed] [Google Scholar]
- Colaianna M, Schiavone S, Zotti M, Tucci P, Morgese MG, Bäckdahl L, Holmdahl R, Krause K-H, Cuomo V, Trabace L, 2013. Neuroendocrine profile in a rat model of psychosocial stress: relation to oxidative stress. Antioxid Redox Signal 18, 1385–1399. 10.1089/ars.2012.4569 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cole JJ, Robertson NA, Rather MI, Thomson JP, McBryan T, Sproul D, Wang T, Brock C, Clark W, Ideker T, Meehan RR, Miller RA, Brown-Borg HM, Adams PD, 2017. Diverse interventions that extend mouse lifespan suppress shared age-associated epigenetic changes at critical gene regulatory regions. Genome Biol 18, 58. 10.1186/s13059-017-1185-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Coppé J-P, Rodier F, Patil CK, Freund A, Desprez P-Y, Campisi J, 2011. Tumor suppressor and aging biomarker p16(INK4a) induces cellular senescence without the associated inflammatory secretory phenotype. J Biol Chem 286, 36396–36403. 10.1074/jbc.M111.257071 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Crouch J, Shvedova M, Thanapaul RJRS, Botchkarev V, Roh D, 2022. Epigenetic Regulation of Cellular Senescence. Cells 11, 672. 10.3390/cells11040672 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cruickshanks HA, McBryan T, Nelson DM, VanderKraats ND, Shah PP, van Tuyn J, Singh Rai T, Brock C, Donahue G, Dunican DS, Drotar ME, Meehan RR, Edwards JR, Berger SL, Adams PD, 2013. Senescent cells harbour features of the cancer epigenome. Nat Cell Biol 15, 1495–1506. 10.1038/ncb2879 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cunliffe VT, 2016. The epigenetic impacts of social stress: how does social adversity become biologically embedded? Epigenomics 8, 1653–1669. 10.2217/epi-2016-0075 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Damjanovic AK, Yang Y, Glaser R, Kiecolt-Glaser JK, Nguyen H, Laskowski B, Zou Y, Beversdorf DQ, Weng N, 2007. Accelerated telomere erosion is associated with a declining immune function of caregivers of Alzheimer’s disease patients. J Immunol 179, 4249–4254. 10.4049/jimmunol.179.6.4249 [DOI] [PMC free article] [PubMed] [Google Scholar]
- de Lange T, 2009. How telomeres solve the end-protection problem. Science 326, 948–952. 10.1126/science.1170633 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Di Micco R, Fumagalli M, Cicalese A, Piccinin S, Gasparini P, Luise C, Schurra C, Garre’ M, Nuciforo PG, Bensimon A, Maestro R, Pelicci PG, d’Adda di Fagagna F, 2006. Oncogene-induced senescence is a DNA damage response triggered by DNA hyper-replication. Nature 444, 638–642. 10.1038/nature05327 [DOI] [PubMed] [Google Scholar]
- Dion A, Muñoz PT, Franklin TB, 2022. Epigenetic mechanisms impacted by chronic stress across the rodent lifespan. Neurobiol Stress 17, 100434. 10.1016/j.ynstr.2022.100434 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dulić V, Beney GE, Frebourg G, Drullinger LF, Stein GH, 2000. Uncoupling between phenotypic senescence and cell cycle arrest in aging p21-deficient fibroblasts. Mol Cell Biol 20, 6741–6754. 10.1128/MCB.20.18.6741-6754.2000 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Duque G, Lipsitz LA, Ferrucci L, Addie S, Carrington-Lawrence S, Kohanski R, 2023. Geroscience for the Next Chapter of Medicine. J Gerontol A Biol Sci Med Sci 78, 791–792. 10.1093/gerona/glad083 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Elo IT, 2009. Social Class Differentials in Health and Mortality: Patterns and Explanations in Comparative Perspective. Annual Review of Sociology 35, 553–572. 10.1146/annurev-soc-070308-115929 [DOI] [Google Scholar]
- Epel ES, 2020. The geroscience agenda: Toxic stress, hormetic stress, and the rate of aging. Ageing Research Reviews 63, 101167–101167. 10.1016/j.arr.2020.101167 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Epel ES, Blackburn EH, Lin J, Dhabhar FS, Adler NE, Morrow JD, Cawthon RM, 2004. Accelerated telomere shortening in response to life stress. Proceedings of the National Academy of Sciences of the United States of America 101, 17312–5. 10.1073/pnas.0407162101 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Epel ES, Crosswell AD, Mayer SE, Prather AA, Slavich GM, Puterman E, Mendes WB, 2018. More than a feeling: A unified view of stress measurement for population science. Front Neuroendocrinol 49, 146–169. 10.1016/j.yfrne.2018.03.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Epel ES, Lin J, Dhabhar FS, Wolkowitz OM, Puterman E, Karan L, Blackburn EH, 2010. Dynamics of telomerase activity in response to acute psychological stress. Brain Behav Immun 24, 531–539. 10.1016/j.bbi.2009.11.018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ferrucci L, Fabbri E, 2018. Inflammageing: chronic inflammation in ageing, cardiovascular disease, and frailty. Nat Rev Cardiol 15, 505–522. 10.1038/s41569-018-0064-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ferrucci L, Semba RD, Guralnik JM, Ershler WB, Bandinelli S, Patel KV, Sun K, Woodman RC, Andrews NC, Cotter RJ, Ganz T, Nemeth E, Longo DL, 2010. Proinflammatory state, hepcidin, and anemia in older persons. Blood 115, 3810–3816. 10.1182/blood-2009-02-201087 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fitzgerald E, Sinton MC, Wernig-Zorc S, Morton NM, Holmes MC, Boardman JP, Drake AJ, 2021. Altered hypothalamic DNA methylation and stress-induced hyperactivity following early life stress. Epigenetics Chromatin 14, 31. 10.1186/s13072-021-00405-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Flaherty RL, Owen M, Fagan-Murphy A, Intabli H, Healy D, Patel A, Allen MC, Patel BA, Flint MS, 2017. Glucocorticoids induce production of reactive oxygen species/reactive nitrogen species and DNA damage through an iNOS mediated pathway in breast cancer. Breast Cancer Res 19, 35. 10.1186/s13058-017-0823-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Flint MS, Baum A, Chambers WH, Jenkins FJ, 2007. Induction of DNA damage, alteration of DNA repair and transcriptional activation by stress hormones. Psychoneuroendocrinology 32, 470–479. 10.1016/j.psyneuen.2007.02.013 [DOI] [PubMed] [Google Scholar]
- Flint MS, Baum A, Episcopo B, Knickelbein KZ, Liegey Dougall AJ, Chambers WH, Jenkins FJ, 2013. Chronic exposure to stress hormones promotes transformation and tumorigenicity of 3T3 mouse fibroblasts. Stress 16, 114–121. 10.3109/10253890.2012.686075 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Franceschi C, Garagnani P, Parini P, Giuliani C, Santoro A, 2018. Inflammaging: a new immune–metabolic viewpoint for age-related diseases. Nature Reviews Endocrinology 14, 576–590. 10.1038/s41574-018-0059-4 [DOI] [PubMed] [Google Scholar]
- Frank MG, Barrientos RM, Biedenkapp JC, Rudy JW, Watkins LR, Maier SF, 2006. mRNA up-regulation of MHC II and pivotal pro-inflammatory genes in normal brain aging. Neurobiology of Aging 27, 717–722. 10.1016/j.neurobiolaging.2005.03.013 [DOI] [PubMed] [Google Scholar]
- Fulop T, Larbi A, Dupuis G, Le Page A, Frost EH, Cohen AA, Witkowski JM, Franceschi C, 2017. Immunosenescence and Inflamm-Aging As Two Sides of the Same Coin: Friends or Foes? Front Immunol 8, 1960. 10.3389/fimmu.2017.01960 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Galow A-M, Peleg S, 2022. How to Slow down the Ticking Clock: Age-Associated Epigenetic Alterations and Related Interventions to Extend Life Span. Cells 11, 468. 10.3390/cells11030468 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gerritsen L, Wang HX, Reynolds CA, Fratiglioni L, Gatz M, Pedersen NL, 2017. Influence of Negative Life Events and Widowhood on Risk for Dementia. American Journal of Geriatric Psychiatry 25, 766–778. 10.1016/j.jagp.2017.02.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gorgoulis V, Adams PD, Alimonti A, Bennett DC, Bischof O, Bishop C, Campisi J, Collado M, Evangelou K, Ferbeyre G, Gil J, Hara E, Krizhanovsky V, Jurk D, Maier AB, Narita M, Niedernhofer L, Passos JF, Robbins PD, Schmitt CA, Sedivy J, Vougas K, von Zglinicki T, Zhou D, Serrano M, Demaria M, 2019. Cellular Senescence: Defining a Path Forward. Cell 179, 813–827. 10.1016/j.cell.2019.10.005 [DOI] [PubMed] [Google Scholar]
- Hannum G, Guinney J, Zhao L, Zhang L, Hughes G, Sadda S, Klotzle B, Bibikova M, Fan J-B, Gao Y, Deconde R, Chen M, Rajapakse I, Friend S, Ideker T, Zhang K, 2013. Genome-wide methylation profiles reveal quantitative views of human aging rates. Mol Cell 49, 359–367. 10.1016/j.molcel.2012.10.016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hara MR, Kovacs JJ, Whalen EJ, Rajagopal S, Strachan RT, Grant W, Towers AJ, Williams B, Lam CM, Xiao K, Shenoy SK, Gregory SG, Ahn S, Duckett DR, Lefkowitz RJ, 2011. A stress response pathway regulates DNA damage through β2-adrenoreceptors and β-arrestin-1. Nature 477, 349–353. 10.1038/nature10368 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Harman D, 2009. Origin and evolution of the free radical theory of aging: a brief personal history, 1954–2009. Biogerontology 10, 773–781. 10.1007/s10522-009-9234-2 [DOI] [PubMed] [Google Scholar]
- Hayflick L, Moorhead PS, 1961. The serial cultivation of human diploid cell strains. Experimental Cell Research 25, 585–621. 10.1016/0014-4827(61)90192-6 [DOI] [PubMed] [Google Scholar]
- Heidt T, Sager HB, Courties G, Dutta P, Iwamoto Y, Zaltsman A, Von Zur Muhlen C, Bode C, Fricchione GL, Denninger J, Lin CP, Vinegoni C, Libby P, Swirski FK, Weissleder R, Nahrendorf M, 2014. Chronic variable stress activates hematopoietic stem cells. Nature Medicine 20, 754–758. 10.1038/nm.3589 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hemann MT, Strong MA, Hao LY, Greider CW, 2001. The shortest telomere, not average telomere length, is critical for cell viability and chromosome stability. Cell 107, 67–77. 10.1016/s0092-8674(01)00504-9 [DOI] [PubMed] [Google Scholar]
- Hemmerle AM, Herman JP, Seroogy KB, 2012. Stress, depression and Parkinson’s disease. Experimental Neurology 233, 79–86. 10.1016/J.EXPNEUROL.2011.09.035 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Horn SR, Leve LD, Levitt P, Fisher PA, 2019. Childhood adversity, mental health, and oxidative stress: A pilot study. PLoS One 14, e0215085. 10.1371/journal.pone.0215085 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Horvath S, 2013. DNA methylation age of human tissues and cell types. Genome Biology 14, 1–20. 10.1186/gb-2013-14-10-r115 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Horvath S, Raj K, 2018. DNA methylation-based biomarkers and the epigenetic clock theory of ageing. Nat Rev Genet 19, 371–384. 10.1038/s41576-018-0004-3 [DOI] [PubMed] [Google Scholar]
- Huffman DM, Schafer MJ, LeBrasseur NK, 2016. Energetic interventions for healthspan and resiliency with aging. Exp Gerontol 86, 73–83. 10.1016/j.exger.2016.05.012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jacob F, 1977. Evolution and tinkering. Science 196, 1161–1166. 10.1126/science.860134 [DOI] [PubMed] [Google Scholar]
- Joëls M, Baram TZ, 2009. The neuro-symphony of stress. Nat Rev Neurosci 10, 459–466. 10.1038/nrn2632 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kennedy BK, Berger SL, Brunet A, Campisi J, Cuervo AM, Epel ES, Franceschi C, Lithgow GJ, Morimoto RI, Pessin JE, Rando TA, Richardson A, Schadt EE, Wyss-Coray T, Sierra F, 2014. Geroscience: Linking aging to chronic disease. Cell 159, 709–713. 10.1016/j.cell.2014.10.039 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kerepesi C, Zhang B, Lee S-G, Trapp A, Gladyshev VN, 2021. Epigenetic clocks reveal a rejuvenation event during embryogenesis followed by aging. Sci Adv 7, eabg6082. 10.1126/sciadv.abg6082 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kiecolt-Glaser JK, Stephens RE, Lipetz PD, Speicher CE, Glaser R, 1985. Distress and DNA repair in human lymphocytes. J Behav Med 8, 311–320. 10.1007/BF00848366 [DOI] [PubMed] [Google Scholar]
- Kirkland JL, Tchkonia T, Zhu Y, Niedernhofer LJ, Robbins PD, 2017. The Clinical Potential of Senolytic Drugs. Journal of the American Geriatrics Society 65, 2297–2301. 10.1111/jgs.14969 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kirkwood TBL, Kowald A, 2012. The free-radical theory of ageing--older, wiser and still alive: modelling positional effects of the primary targets of ROS reveals new support. Bioessays 34, 692–700. 10.1002/bies.201200014 [DOI] [PubMed] [Google Scholar]
- Kirschbaum C, Pirke K-M, Hellhammer DH, 1993. The ‘Trier Social Stress Test’ – A Tool for Investigating Psychobiological Stress Responses in a Laboratory Setting. NPS 28, 76–81. 10.1159/000119004 [DOI] [PubMed] [Google Scholar]
- Kivimäki M, Bartolomucci A, Kawachi I, 2022. The multiple roles of life stress in metabolic disorders. Nature Reviews Endocrinology 2022 1–18. 10.1038/s41574-022-00746-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kivimäki M, Steptoe A, 2018. Effects of stress on the development and progression of cardiovascular disease. Nature Reviews Cardiology 15, 215–229. 10.1038/nrcardio.2017.189 [DOI] [PubMed] [Google Scholar]
- Kivimäki M, Vahtera J, Tabák AG, Halonen JI, Vineis P, Pentti J, Pahkala K, Rovio S, Viikari J, Kähönen M, Juonala M, Ferrie JE, Stringhini S, Raitakari OT, 2018. Neighbourhood socioeconomic disadvantage, risk factors, and diabetes from childhood to middle age in the Young Finns Study: a cohort study. Lancet Public Health 3, e365–e373. 10.1016/S2468-2667(18)30111-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kong L, Ye C, Wang Y, Hou T, Zheng J, Zhao Z, Li M, Xu Y, Lu J, Chen Y, Xu M, Wang W, Ning G, Bi Y, Wang T, 2023. Genetic Evidence for Causal Effects of Socioeconomic, Lifestyle, and Cardiometabolic Factors on Epigenetic-Age Acceleration. The Journals of Gerontology: Series A glad 078. 10.1093/gerona/glad078 [DOI] [PubMed] [Google Scholar]
- Koolhaas JM, Bartolomucci A, Buwalda B, de Boer SF, Fl?gge G, Korte SM, Meerlo P, Murison R, Olivier B, Palanza P, Richter-Levin G, Sgoifo A, Steimer T, Stiedl O, van Dijk G, W?hr M, Fuchs E, 2011. Stress revisited: A critical evaluation of the stress concept. Neuroscience and Biobehavioral Reviews 35. 10.1016/j.neubiorev.2011.02.003 [DOI] [PubMed] [Google Scholar]
- Kotah JM, Hoeijmakers L, Nutma E, Lucassen PJ, Korosi A, 2021. Early-life stress does not alter spatial memory performance, hippocampal neurogenesis, neuroinflammation, or telomere length in 20-month-old male mice. Neurobiol Stress 15, 100379. 10.1016/j.ynstr.2021.100379 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Laberge R-M, Zhou L, Sarantos MR, Rodier F, Freund A, de Keizer PLJ, Liu S, Demaria M, Cong Y-S, Kapahi P, Desprez P-Y, Hughes RE, Campisi J, 2012. Glucocorticoids suppress selected components of the senescence-associated secretory phenotype. Aging Cell 11, 569–578. 10.1111/j.1474-9726.2012.00818.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lachman ME, Weaver SL, 1998. The sense of control as a moderator of social class differences in health and well-being. J Pers Soc Psychol 74, 763–773. 10.1037//0022-3514.74.3.763 [DOI] [PubMed] [Google Scholar]
- Lee PJ, Benz CC, Blood P, Börner K, Campisi J, Chen F, Daldrup-Link H, De Jager P, Ding L, Duncan FE, Eickelberg O, Fan R, Finkel T, Furman D, Garovic V, Gehlenborg N, Glass C, Heckenbach I, Joseph Z-B, Katiyar P, Kim S-J, Königshoff M, Kuchel GA, Lee H, Lee JH, Ma J, Ma Q, Melov S, Metis K, Mora AL, Musi N, Neretti N, Passos JF, Rahman I, Rivera-Mulia JC, Robson P, Rojas M, Roy AL, Scheibye-Knudsen M, Schilling B, Shi P, Silverstein JC, Suryadevara V, Xie J, Wang J, Wong AI, Niedernhofer LJ, Wang S, Anvari H, Balough J, Benz C, Bons J, Brenerman B, Evans W, Gerencser A, Gregory H, Hansen M, Justice J, Kapahi P, Murad N, O’Broin A, Pavone ME, Powell M, Scott G, Shanes E, Shankaran M, Verdin E, Winer D, Wu F, Adams A, Blood PD, Bueckle A, Cao-Berg I, Chen H, Davis M, Filus S, Hao Y, Hartman A, Hasanaj E, Helfer J, Herr B, Joseph ZB, Molla G, Mou G, Puerto J, Quardokus EM, Ropelewski AJ, Ruffalo M, Satija R, Schwenk M, Scibek R, Shirey W, Sibilla M, Welling J, Yuan Z, Bonneau R, Christiano A, Izar B, Menon V, Owens DM, Phatnani H, Smith C, Suh Y, Teich AF, Bekker V, Chan C, Coutavas E, Hartwig MG, Ji Z, Nixon AB, Dou Z, Rajagopal J, Slavov N, Holmes D, Jurk D, Kirkland JL, Lagnado A, Tchkonia T, Abraham K, Dibattista A, Fridell Y-W, Howcroft TK, Jhappan C, Montes VP, Prabhudas M, Resat H, Taylor V, Kumar M, Suryadevara V, Cigarroa F, Cohn R, Cortes TM, Courtois E, Chuang J, Davé M, Domanskyi S, Enninga EAL, Eryilmaz GN, Espinoza SE, Gelfond J, Kirkland J, Kuchel GA, Kuo C-L, Lehman JS, Aguayo-Mazzucato C, Meves A, Rani M, Sanders S, Thibodeau A, Tullius SG, Ucar D, White B, Wu Q, Xu Ming, Yamaguchi S, Assarzadegan N, Cho C-S, Hwang I, Hwang Y, Xi J, Adeyi OA, Aliferis CF, Bartolomucci A, Dong X, DuFresne-To MJ, Ikramuddin S, Johnson SG, Nelson AC, Niedernhofer LJ, Revelo XS, Trevilla-Garcia C, Sedivy JM, Thompson EL, Robbins PD, Wang J, Aird KM, Alder JK, Beaulieu D, Bueno M, Calyeca J, Chamucero-Millaris JA, Chan SY, Chung D, Corbett A, Gorbunova V, Gowdy KM, Gurkar A, Horowitz JC, Hu Q, Kaur G, Khaliullin TO, Lafyatis R, Lanna S, Li D, Ma A, Morris A, Muthumalage TM, Peters V, Pryhuber GS, Reader BF, Rosas L, Sembrat JC, Shaikh S, Shi H, Stacey SD, Croix C St., Wang C, Wang Q, Watts A, Gu L, Lin Y, Rabinovitch PS, Sweetwyne MT, Artyomov MN, Ballentine SJ, Chheda MG, Davies SR, DiPersio JF, Fields RC, Fitzpatrick JAJ, Fulton RS, Imai S, Jain S, Ju T, Kushnir VM, Link DC, Ben Major M, Oh ST, Rapp D, Rettig MP, Stewart SA, Veis DJ, Vij KR, Wendl MC, Wyczalkowski MA, Craft JE, Enninful A, Farzad N, Gershkovich P, Halene S, Kluger Y, VanOudenhove J, Xu Mina, Yang J, Yang M, 2022. NIH SenNet Consortium to map senescent cells throughout the human lifespan to understand physiological health. Nature Aging 2022 1–11. 10.1038/s43587-022-00326-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee W, Milewski TM, Dwortz MF, Young RL, Gaudet AD, Fonken LK, Champagne FA, Curley JP, 2022. Distinct immune and transcriptomic profiles in dominant versus subordinate males in mouse social hierarchies. Brain, behavior, and immunity 103, 130–144. 10.1016/J.BBI.2022.04.015 [DOI] [PubMed] [Google Scholar]
- Levine ME, Lu AT, Quach A, Chen BH, Assimes TL, Bandinelli S, Hou L, Baccarelli AA, Stewart JD, Li Y, Whitsel EA, Wilson JG, Reiner1 AP, Aviv1 A, Lohman K, Liu Y, Ferrucci L, Horvath S, 2018. An epigenetic biomarker of aging for lifespan and healthspan. Aging 10, 573–591. 10.18632/aging.101414 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li Q, Zhang M, Chen Y-J, Wang Y-J, Huang F, Liu J, 2011. Oxidative damage and HSP70 expression in masseter muscle induced by psychological stress in rats. Physiol Behav 104, 365–372. 10.1016/j.physbeh.2011.04.012 [DOI] [PubMed] [Google Scholar]
- Lim S, Nzegwu D, Wright ML, 2022. The Impact of Psychosocial Stress from Life Trauma and Racial Discrimination on Epigenetic Aging-A Systematic Review. Biol Res Nurs 24, 202–215. 10.1177/10998004211060561 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu H, Zhou J, Tian W, Luo C, Bartlett A, Aldridge A, Lucero J, Osteen JK, Nery JR, Chen H, Rivkin A, Castanon RG, Clock B, Li YE, Hou X, Poirion OB, Preissl S, Pinto-Duarte A, O’Connor C, Boggeman L, Fitzpatrick C, Nunn M, Mukamel EA, Zhang Z, Callaway EM, Ren B, Dixon JR, Behrens MM, Ecker JR, 2021. DNA methylation atlas of the mouse brain at single-cell resolution. Nature 598, 120–128. 10.1038/s41586-020-03182-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- López-López AL, Jaime HB, Escobar Villanueva MDC, Padilla MB, Palacios GV, Aguilar FJA, 2016. Chronic unpredictable mild stress generates oxidative stress and systemic inflammation in rats. Physiol Behav 161, 15–23. 10.1016/j.physbeh.2016.03.017 [DOI] [PubMed] [Google Scholar]
- López-Otín C, Blasco MA, Partridge L, Serrano M, Kroemer G, 2023. Hallmarks of aging: An expanding universe. Cell 0. 10.1016/j.cell.2022.11.001 [DOI] [PubMed] [Google Scholar]
- López-Otín C, Blasco MA, Partridge L, Serrano M, Kroemer G, 2013. The hallmarks of aging. Cell 153, 1194–1194. 10.1016/j.cell.2013.05.039 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lu AT, Quach A, Wilson JG, Reiner AP, Aviv A, Raj K, Hou L, Baccarelli AA, Li Y, Stewart JD, Whitsel EA, Assimes TL, Ferrucci L, Horvath S, 2019. DNA methylation GrimAge strongly predicts lifespan and healthspan. Aging (Albany NY) 11, 303–327. 10.18632/aging.101684 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lu Y, Brommer B, Tian X, Krishnan A, Meer M, Wang C, Vera DL, Zeng Q, Yu D, Bonkowski MS, Yang J-H, Zhou S, Hoffmann EM, Karg MM, Schultz MB, Kane AE, Davidsohn N, Korobkina E, Chwalek K, Rajman LA, Church GM, Hochedlinger K, Gladyshev VN, Horvath S, Levine ME, Gregory-Ksander MS, Ksander BR, He Z, Sinclair DA, 2020. Reprogramming to recover youthful epigenetic information and restore vision. Nature 588, 124–129. 10.1038/s41586-020-2975-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lupien SJ, McEwen BS, Gunnar MR, Heim C, 2009. Effects of stress throughout the lifespan on the brain, behaviour and cognition. Nature Reviews Neuroscience 10, 434–445. 10.1038/nrn2639 [DOI] [PubMed] [Google Scholar]
- Lyons CE, Bartolomucci A, 2020. Stress and Alzheimer’s disease: A senescence link? Neuroscience and Biobehavioral Reviews 115, 285–298. 10.1016/j.neubiorev.2020.05.010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mansur RB, Cunha GR, Asevedo E, Zugman A, Zeni-Graiff M, Rios AC, Sethi S, Maurya PK, Levandowski ML, Gadelha A, Pan PM, Stertz L, Belangero SI, Kauer-Sant’ Anna M, Teixeira AL, Mari JJ, Rohde LA, Miguel EC, McIntyre RS, Grassi-Oliveira R, Bressan RA, Brietzke E, 2016. Socioeconomic Disadvantage Moderates the Association between Peripheral Biomarkers and Childhood Psychopathology. PLoS One 11, e0160455. 10.1371/journal.pone.0160455 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Marmot MG, Stansfeld S, Patel C, North F, Head J, White I, Brunner E, Feeney A, Marmot MG, Smith GD, 1991. Health inequalities among British civil servants: the Whitehall II study. The Lancet 337, 1387–1393. 10.1016/0140-6736(91)93068-K [DOI] [PubMed] [Google Scholar]
- McEwen BS, 1998. Protective and Damaging Effects of Stress Mediators. New England Journal of Medicine 338, 171–179. 10.1056/NEJM199801153380307 [DOI] [PubMed] [Google Scholar]
- McHugh D, Gil J, 2018. Senescence and aging: Causes, consequences, and therapeutic avenues. The Journal of Cell Biology 217, 65–77. 10.1083/jcb.201708092 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mejfa S, Giraldo M, Pineda D, Ardila A, Lopera Francisco, Mejia S, Giraldo M, Pineda D, Lopera F, 2003. Nongenetic Factors as Modifiers of the Age of Onset of Familial Alzheimer’s Disease. International Psychogeriah-ics 15, 337–349. [DOI] [PubMed] [Google Scholar]
- Morrison RE, Eckardt W, Stoinski TS, Rosenbaum S, 2023. Cumulative early-life adversity does not predict reduced adult longevity in wild gorillas. Curr Biol 33, 2307–2314.e4. 10.1016/j.cub.2023.04.051 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nederhof E, Schmidt MV, 2012. Mismatch or cumulative stress: Toward an integrated hypothesis of programming effects. Physiology & Behavior 106, 691–700. 10.1016/j.physbeh.2011.12.008 [DOI] [PubMed] [Google Scholar]
- Noren Hooten N, Pacheco NL, Smith JT, Evans MK, 2022. The accelerated aging phenotype: The role of race and social determinants of health on aging. Ageing research reviews 73. 10.1016/J.ARR.2021.101536 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Obeng-Gyasi S, Elsaid MI, Lu Y, Chen J, Carson WE, Ballinger TJ, Andersen BL, 2023. Association of Allostatic Load With All-Cause Mortality in Patients With Breast Cancer. JAMA Network Open 6, e2313989. 10.1001/jamanetworkopen.2023.13989 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Palma-Gudiel H, Fañanás L, Horvath S, Zannas AS, 2020. Psychosocial stress and epigenetic aging. Int Rev Neurobiol 150, 107–128. 10.1016/bs.irn.2019.10.020 [DOI] [PubMed] [Google Scholar]
- Parks CG, Miller DB, McCanlies EC, Cawthon RM, Andrew ME, DeRoo LA, Sandler DP, 2009. Telomere length, current perceived stress, and urinary stress hormones in women. Cancer Epidemiol Biomarkers Prev 18, 551–560. 10.1158/1055-9965.EPI-08-0614 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Partridge L, Fuentealba M, Kennedy BK, 2020. The quest to slow ageing through drug discovery. Nat Rev Drug Discov 19, 513–532. 10.1038/s41573-020-0067-7 [DOI] [PubMed] [Google Scholar]
- Petkovich DA, Podolskiy DI, Lobanov AV, Lee S-G, Miller RA, Gladyshev VN, 2017. Using DNA Methylation Profiling to Evaluate Biological Age and Longevity Interventions. Cell Metab 25, 954–960.e6. 10.1016/j.cmet.2017.03.016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Poganik JR, Zhang B, Baht GS, Kerepesi C, Yim SH, Lu AT, Haghani A, Gong T, Hedman AM, Andolf E, Pershagen G, Almqvist C, White JP, Horvath S, Gladyshev VN, 2022. Biological age is increased by stress and restored upon recovery. 10.1101/2022.05.04.490686 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Polsky LR, Rentscher KE, Carroll JE, 2022. Stress-induced biological aging: A review and guide for research priorities. Brain, Behavior, and Immunity 104, 97–109. 10.1016/J.BBI.2022.05.016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Poulsen HE, Nadal LL, Broedbaek K, Nielsen PE, Weimann A, 2014. Detection and interpretation of 8-oxodG and 8-oxoGua in urine, plasma and cerebrospinal fluid. Biochim Biophys Acta 1840, 801–808. 10.1016/j.bbagen.2013.06.009 [DOI] [PubMed] [Google Scholar]
- Prieto LI, Sturmlechner I, Graves SI, Zhang C, Goplen NP, Yi ES, Sun J, Li H, Baker DJ, 2023. Senescent alveolar macrophages promote early-stage lung tumorigenesis. Cancer Cell S1535–6108(23)00171-X. 10.1016/j.ccell.2023.05.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Puterman E, Lin J, Blackburn E, O’Donovan A, Adler N, Epel E, 2010. The power of exercise: buffering the effect of chronic stress on telomere length. PLoS One 5, e10837. 10.1371/journal.pone.0010837 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Qing H, Desrouleaux R, Israni-Winger K, Mineur YS, Fogelman N, Zhang C, Rashed S, Palm NW, Sinha R, Picciotto MR, Perry RJ, Wang A, 2020. Origin and Function of Stress-Induced IL-6 in Murine Models. Cell 182, 372–387.e14. 10.1016/j.cell.2020.05.054 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Raffington L, Belsky DW, 2022. Integrating DNA Methylation Measures of Biological Aging into Social Determinants of Health Research. Curr Environ Health Rep 9, 196–210. 10.1007/s40572-022-00338-8 [DOI] [PubMed] [Google Scholar]
- Razzoli M, Nyuyki-Dufe K, Chen BH, Bartolomucci A, 2023. Contextual modifiers of healthspan, lifespan, and epigenome in mice under chronic social stress. Proc Natl Acad Sci U S A 120, e2211755120. 10.1073/pnas.2211755120 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Razzoli M, Nyuyki-Dufe K, Gurney A, Erickson C, McCallum J, Spielman N, Marzullo M, Patricelli J, Kurata M, Pope EA, Touma C, Palme R, Largaespada DA, Allison DB, Bartolomucci A, 2018. Social stress shortens lifespan in mice. Aging Cell 17. 10.1111/acel.12778 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rentscher KE, Carroll JE, Polsky LR, Lamkin DM, 2022. Chronic stress increases transcriptomic indicators of biological aging in mouse bone marrow leukocytes. Brain Behav Immun Health 22, 100461. 10.1016/j.bbih.2022.100461 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rentscher KE, Carroll JE, Repetti RL, Cole SW, Reynolds BM, Robles TF, 2019. Chronic stress exposure and daily stress appraisals relate to biological aging marker p16 INK4a. Psychoneuroendocrinology 102, 139–148. 10.1016/j.psyneuen.2018.12.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Reuben A, Sugden K, Arseneault L, Corcoran DL, Danese A, Fisher HL, Moffitt TE, Newbury JB, Odgers C, Prinz J, Rasmussen LJH, Williams B, Mill J, Caspi A, 2020. Association of Neighborhood Disadvantage in Childhood With DNA Methylation in Young Adulthood. JAMA Netw Open 3, e206095. 10.1001/jamanetworkopen.2020.6095 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Richterman A, Millien C, Bair EF, Jerome G, Suffrin JCD, Behrman JR, Thirumurthy H, 2023. The effects of cash transfers on adult and child mortality in low- and middle-income countries. Nature 618, 575–582. 10.1038/s41586-023-06116-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rodier F, Coppé J-P, Patil CK, Hoeijmakers WAM, Muñoz DP, Raza SR, Freund A, Campeau E, Davalos AR, Campisi J, 2009. Persistent DNA damage signalling triggers senescence-associated inflammatory cytokine secretion. Nat Cell Biol 11, 973–979. 10.1038/ncb1909 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rohleder N, 2014. Stimulation of systemic low-grade inflammation by psychosocial stress. Psychosom Med 76, 181–189. 10.1097/PSY.0000000000000049 [DOI] [PubMed] [Google Scholar]
- Rosenberg AM, Rausser S, Ren J, Mosharov EV, Sturm G, Ogden RT, Patel P, Kumar Soni R, Lacefield C, Tobin DJ, Paus R, Picard M, 2021. Quantitative mapping of human hair greying and reversal in relation to life stress. Elife 10, e67437. 10.7554/eLife.67437 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Salim S, 2016. Oxidative stress: a potential link between emotional wellbeing and immune response. Curr Opin Pharmacol 29, 70–76. 10.1016/j.coph.2016.06.006 [DOI] [PubMed] [Google Scholar]
- Sapolsky RM, 2005. The Influence of Social Hierarchy on Primate Health. Science 308, 648–652. 10.1126/science.1106477 [DOI] [PubMed] [Google Scholar]
- Sapolsky RM, Krey LC, McEwen BS, 1986. The Neuroendocrinology of Stress and Aging: The Glucocorticoid Cascade Hypothesis*. Endocrine Reviews 7, 284–301. 10.1210/edrv-7-3-284 [DOI] [PubMed] [Google Scholar]
- Sapolsky RM, Krey LC, McEwen BS, 1984. Glucocorticoid-sensitive hippocampal neurons are involved in terminating the adrenocortical stress response. Proceedings of the National Academy of Sciences of the United States of America 81, 6174–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sapolsky RM, Romero LM, Munck AU, 2000. How Do Glucocorticoids Influence Stress Responses? Integrating Permissive, Suppressive, Stimulatory, and Preparative Actions 1. Endocrine Reviews 21, 55–89. 10.1210/edrv.21.1.0389 [DOI] [PubMed] [Google Scholar]
- Saretzki G, Sitte N, Merkel U, Wurm RE, von Zglinicki T, 1999. Telomere shortening triggers a p53-dependent cell cycle arrest via accumulation of G-rich single stranded DNA fragments. Oncogene 18, 5148–5158. 10.1038/sj.onc.1202898 [DOI] [PubMed] [Google Scholar]
- Schaakxs R, Wielaard I, Verhoeven JE, Beekman ATF, Penninx BWJH, Comijs HC, 2016. Early and recent psychosocial stress and telomere length in older adults. Int Psychogeriatr 28, 405–413. 10.1017/S1041610215001155 [DOI] [PubMed] [Google Scholar]
- Schiavone S, Jaquet V, Sorce S, Dubois-Dauphin M, Hultqvist M, Bäckdahl L, Holmdahl R, Colaianna M, Cuomo V, Trabace L, Krause K-H, 2012. NADPH oxidase elevations in pyramidal neurons drive psychosocial stress-induced neuropathology. Transl Psychiatry 2, e111. 10.1038/tp.2012.36 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Selye H, 1970. Stress and aging. J Am Geriatr Soc 18, 669–680. 10.1111/j.1532-5415.1970.tb02813.x [DOI] [PubMed] [Google Scholar]
- Sharifi-Rad M, Anil Kumar NV, Zucca P, Varoni EM, Dini L, Panzarini E, Rajkovic J, Tsouh Fokou PV, Azzini E, Peluso I, Prakash Mishra A, Nigam M, El Rayess Y, Beyrouthy ME, Polito L, Iriti M, Martins N, Martorell M, Docea AO, Setzer WN, Calina D, Cho WC, Sharifi-Rad J, 2020. Lifestyle, Oxidative Stress, and Antioxidants: Back and Forth in the Pathophysiology of Chronic Diseases. Front Physiol 11, 694. 10.3389/fphys.2020.00694 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sierra F, Kohanski RA, 2023. The Role of the National Institute on Aging in the Development of the Field of Geroscience. Cold Spring Harb Perspect Med; a041211. 10.1101/cshperspect.a041211 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Simpson DJ, Chandra T, 2021. Epigenetic age prediction. Aging Cell 20, e13452. 10.1111/acel.13452 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Singer BD, 2019. A Practical Guide to the Measurement and Analysis of DNA Methylation. Am J Respir Cell Mol Biol 61, 417–428. 10.1165/rcmb.2019-0150TR [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sivonová M, Zitnanová I, Hlincíková L, Skodácek I, Trebatická J, Duracková Z, 2004. Oxidative stress in university students during examinations. Stress 7, 183–188. 10.1080/10253890400012685 [DOI] [PubMed] [Google Scholar]
- Snyder-Mackler N, Burger JR, Gaydosh L, Belsky DW, Noppert GA, Campos FA, Bartolomucci A, Yang YC, Aiello AE, O’Rand A, Harris KM, Shively CA, Alberts SC, Tung J, 2020. Social determinants of health and survival in humans and other animals. Science 368, eaax9553–eaax9553. 10.1126/science.aax9553 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Snyder-Mackler N, Sanz J, Kohn JN, Brinkworth JF, Morrow S, Shaver AO, Grenier JC, Pique-Regi R, Johnson ZP, Wilson ME, Barreiro LB, Tung J, 2016. Social status alters immune regulation and response to infection in macaques. Science 354, 1041–1045. 10.1126/science.aah3580 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Son S-W, Lee J-S, Kim H-G, Kim D-W, Ahn Y-C, Son C-G, 2016. Testosterone depletion increases the susceptibility of brain tissue to oxidative damage in a restraint stress mouse model. J Neurochem 136, 106–117. 10.1111/jnc.13371 [DOI] [PubMed] [Google Scholar]
- Spiga F, Walker JJ, Terry JR, Lightman SL, 2014. HPA Axis-Rhythms, in: Comprehensive Physiology. John Wiley & Sons, Inc., Hoboken, pp. 1273–1298. 10.1002/cphy.c140003 [DOI] [PubMed] [Google Scholar]
- Stadtman ER, Levine RL, 2000. Protein oxidation. Ann N Y Acad Sci 899, 191–208. 10.1111/j.1749-6632.2000.tb06187.x [DOI] [PubMed] [Google Scholar]
- Stambler I, 2017. Longevity Promotion: Multidisciplinary Perspectives. Lulu Press, Inc. [Google Scholar]
- Stringhini S, Carmeli C, Jokela M, Avendaño M, Muennig P, Guida F, Ricceri F, d’Errico A, Barros H, Bochud M, Chadeau-Hyam M, Clavel-Chapelon F, Costa G, Delpierre C, Fraga S, Goldberg M, Giles GG, Krogh V, Kelly-Irving M, Layte R, Lasserre AM, Marmot MG, Preisig M, Shipley MJ, Vollenweider P, Zins M, Kawachi I, Steptoe A, Mackenbach JP, Vineis P, Kivimäki M, Alenius H, Avendano M, Barros H, Bochud M, Carmeli C, Carra L, Castagné R, Chadeau-Hyam M, Clavel-Chapelon F, Costa G, Courtin E, Delpierre C, D’Errico A, Dugué P-A, Elliott P, Fraga S, Gares V, Giles G, Goldberg M, Greco D, Hodge A, Irving MK, Karisola P, Kivimäki M, Krogh V, Lang T, Layte R, Lepage B, Mackenbach J, Marmot M, McCrory C, Milne R, Muennig P, Nusselder W, Panico S, Petrovic D, Polidoro S, Preisig M, Raitakari O, Ribeiro AI, Ribeiro AI, Ricceri F, Robinson O, Valverde JR, Sacerdote C, Satolli R, Severi G, Shipley MJ, Stringhini S, Tumino R, Vineis P, Vollenweider P, Zins M, 2017. Socioeconomic status and the 25 × 25 risk factors as determinants of premature mortality: a multicohort study and meta-analysis of 1·7 million men and women. The Lancet 389, 1229–1237. 10.1016/S0140-6736(16)32380-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stubbs TM, Bonder MJ, Stark A-K, Krueger F, von Meyenn F, Stegle O, Reik W, 2017. Multi-tissue DNA methylation age predictor in mouse. Genome Biology 2017 18:1 18, 1–14. 10.1186/S13059-017-1203-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sturmlechner I, Zhang C, Sine CC, van Deursen E-J, Jeganathan KB, Hamada N, Grasic J, Friedman D, Stutchman JT, Can I, Hamada M, Lim DY, Lee J-H, Ordog T, Laberge R-M, Shapiro V, Baker DJ, Li H, van Deursen JM, 2021. p21 produces a bioactive secretome that places stressed cells under immunosurveillance. Science 374, eabb3420. 10.1126/science.abb3420 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Szanton SL, Gill JM, Allen JK, 2005. Allostatic load: A mechanism of socioeconomic health disparities? Biological Research for Nursing 7, 7–15. 10.1177/1099800405278216 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tschaffon-Müller MEA, Kempter E, Steppe L, Kupfer S, Kuhn MR, Gebhard F, Pankratz C, Kalbitz M, Schütze K, Gündel H, Kaleck N, Strauß G, Vacher J, Ichinose H, Weimer K, Ignatius A, Haffner-Luntzer M, Reber SO, 2023. Neutrophil-derived catecholamines mediate negative stress effects on bone. Nat Commun 14, 3262. 10.1038/s41467-023-38616-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tsigos C, Chrousos GP, 2002. Hypothalamic-pituitary-adrenal axis, neuroendocrine factors and stress. J Psychosom Res 53, 865–871. 10.1016/s0022-3999(02)00429-4 [DOI] [PubMed] [Google Scholar]
- Tsuber V, Kadamov Y, Tarasenko L, 2014. Activation of antioxidant defenses in whole saliva by psychosocial stress is more manifested in young women than in young men. PLoS One 9, e115048. 10.1371/journal.pone.0115048 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tung J, Archie EA, Altmann J, Alberts SC, 2016. Cumulative early life adversity predicts longevity in wild baboons. Nature Communications 7, 11181–11181. 10.1038/ncomms11181 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tynan RJ, Naicker S, Hinwood M, Nalivaiko E, Buller KM, Pow DV, Day TA, Walker FR, 2010. Chronic stress alters the density and morphology of microglia in a subset of stress-responsive brain regions. Brain, Behavior, and Immunity 24, 1058–1068. 10.1016/j.bbi.2010.02.001 [DOI] [PubMed] [Google Scholar]
- Ulrich-Lai YM, Herman JP, 2009. Neural regulation of endocrine and autonomic stress responses. Nature reviews. Neuroscience 10, 397–409. 10.1038/nrn2647 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Unno K, Fujitani K, Takamori N, Takabayashi F, Maeda K-I, Miyazaki H, Tanida N, Iguchi K, Shimoi K, Hoshino M, 2011. Theanine intake improves the shortened lifespan, cognitive dysfunction and behavioural depression that are induced by chronic psychosocial stress in mice. Free Radic Res 45, 966–974. 10.3109/10715762.2011.566869 [DOI] [PubMed] [Google Scholar]
- van den Beld AW, Kaufman J-M, Zillikens MC, Lamberts SW, Egan JM, van der Lely AJ, 2018. The physiology of endocrine systems with ageing. Lancet Diabetes Endocrinol 6, 647–658. 10.1016/S2213-8587(18)30026-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vetter VM, Drewelies J, Sommerer Y, Kalies CH, Regitz-Zagrosek V, Bertram L, Gerstorf D, Demuth I, 2022. Epigenetic aging and perceived psychological stress in old age. Transl Psychiatry 12, 410. 10.1038/s41398-022-02181-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Voorhees JL, Tarr AJ, Wohleb ES, Godbout JP, Mo X, Sheridan JF, Eubank TD, Marsh CB, 2013. Prolonged restraint stress increases IL-6, reduces IL-10, and causes persistent depressive-like behavior that is reversed by recombinant IL-10. PLoS One 8, e58488. 10.1371/journal.pone.0058488 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang F, Gao Y, Han Z, Yu Y, Long Z, Jiang X, Wu Y, Pei B, Cao Y, Ye J, Wang M, Zhao Y, 2023. A systematic review and meta-analysis of 90 cohort studies of social isolation, loneliness and mortality. Nat Hum Behav. 10.1038/s41562-023-01617-6 [DOI] [PubMed] [Google Scholar]
- Wang T, Tsui B, Kreisberg JF, Robertson NA, Gross AM, Yu MK, Carter H, Brown-Borg HM, Adams PD, Ideker T, 2017. Epigenetic aging signatures in mice livers are slowed by dwarfism, calorie restriction and rapamycin treatment. Genome Biol 18, 57. 10.1186/s13059-017-1186-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Waziry R, Ryan CP, Corcoran DL, Huffman KM, Kobor MS, Kothari M, Graf GH, Kraus VB, Kraus WE, Lin DTS, Pieper CF, Ramaker ME, Bhapkar M, Das SK, Ferrucci L, Hastings WJ, Kebbe M, Parker DC, Racette SB, Shalev I, Schilling B, Belsky DW, 2023. Effect of long-term caloric restriction on DNA methylation measures of biological aging in healthy adults from the CALERIE trial. Nat Aging 3, 248–257. 10.1038/s43587-022-00357-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wiegand C, Heusser P, Klinger C, Cysarz D, Büssing A, Ostermann T, Savelsbergh A, 2018. Stress-associated changes in salivary microRNAs can be detected in response to the Trier Social Stress Test: An exploratory study. Sci Rep 8, 7112. 10.1038/s41598-018-25554-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wohleb ES, Powell ND, Godbout JP, Sheridan JF, 2013. Stress-induced recruitment of bone marrow-derived monocytes to the brain promotes anxiety-like behavior. Journal of Neuroscience 33, 13820–13833. 10.1523/JNEUROSCI.1671-13.2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xie X, Shen Q, Cao L, Chen Y, Ma L, Xiao Q, Yu C, Fu Z, 2019. Depression caused by long-term stress regulates premature aging and is possibly associated with disruption of circadian rhythms in mice. Physiol Behav 199, 100–110. 10.1016/j.physbeh.2018.11.011 [DOI] [PubMed] [Google Scholar]
- Xu M, Pirtskhalava T, Farr JN, Weigand BM, Palmer AK, Weivoda MM, Inman CL, Ogrodnik MB, Hachfeld CM, Fraser DG, Onken JL, Johnson KO, Verzosa GC, Langhi LGP, Weigl M, Giorgadze N, LeBrasseur NK, Miller JD, Jurk D, Singh RJ, Allison DB, Ejima K, Hubbard GB, Ikeno Y, Cubro H, Garovic VD, Hou X, Weroha SJ, Robbins PD, Niedernhofer LJ, Khosla S, Tchkonia T, Kirkland JL, 2018. Senolytics improve physical function and increase lifespan in old age. Nature medicine 24, 1246–1256. 10.1038/s41591-018-0092-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yang J-H, Hayano M, Griffin PT, Amorim JA, Bonkowski MS, Apostolides JK, Salfati EL, Blanchette M, Munding EM, Bhakta M, Chew YC, Guo W, Yang X, Maybury-Lewis S, Tian X, Ross JM, Coppotelli G, Meer MV, Rogers-Hammond R, Vera DL, Lu YR, Pippin JW, Creswell ML, Dou Z, Xu C, Mitchell SJ, Das A, O’Connell BL, Thakur S, Kane AE, Su Q, Mohri Y, Nishimura EK, Schaevitz L, Garg N, Balta A-M, Rego MA, Gregory-Ksander M, Jakobs TC, Zhong L, Wakimoto H, Andari JE, Grimm D, Mostoslavsky R, Wagers AJ, Tsubota K, Bonasera SJ, Palmeira CM, Seidman JG, Seidman CE, Wolf NS, Kreiling JA, Sedivy JM, Murphy GF, Green RE, Garcia BA, Berger SL, Oberdoerffer P, Shankland SJ, Gladyshev VN, Ksander BR, Pfenning AR, Rajman LA, Sinclair DA, 2023. Loss of epigenetic information as a cause of mammalian aging. Cell 0. 10.1016/j.cell.2022.12.027 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yasunari K, Matsui T, Maeda K, Nakamura M, Watanabe T, Kiriike N, 2006. Anxiety-induced plasma norepinephrine augmentation increases reactive oxygen species formation by monocytes in essential hypertension. Am J Hypertens 19, 573–578. 10.1016/j.amjhyper.2005.10.027 [DOI] [PubMed] [Google Scholar]
- Yousefzadeh MJ, Flores RR, Zhu Y, Schmiechen ZC, Brooks RW, Trussoni CE, Cui Y, Angelini L, Lee KA, McGowan SJ, Burrack AL, Wang D, Dong Q, Lu A, Sano T, O’Kelly RD, McGuckian CA, Kato JI, Bank MP, Wade EA, Pillai SPS, Klug J, Ladiges WC, Burd CE, Lewis SE, LaRusso NF, Vo NV, Wang Y, Kelley EE, Huard J, Stromnes IM, Robbins PD, Niedernhofer LJ, 2021. An aged immune system drives senescence and ageing of solid organs. Nature 594, 100–105. 10.1038/s41586-021-03547-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zannas AS, Jia M, Hafner K, Baumert J, Wiechmann T, Pape JC, Arloth J, Ködel M, Martinelli S, Roitman M, Röh S, Haehle A, Emeny RT, Iurato S, Carrillo-Roa T, Lahti J, Räikkönen K, Eriksson JG, Drake AJ, Waldenberger M, Wahl S, Kunze S, Lucae S, Bradley B, Gieger C, Hausch F, Smith AK, Ressler KJ, Müller-Myhsok B, Ladwig KH, Rein T, Gassen NC, Binder EB, 2019. Epigenetic upregulation of FKBP5 by aging and stress contributes to NF-κB-driven inflammation and cardiovascular risk. Proceedings of the National Academy of Sciences of the United States of America 166, 11370–11379. 10.1073/PNAS.1816847116/-/DCSUPPLEMENTAL [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zannas AS, Kosyk O, Leung CS, 2020. Prolonged Glucocorticoid Exposure Does Not Accelerate Telomere Shortening in Cultured Human Fibroblasts. Genes (Basel) 11, 1425. 10.3390/genes11121425 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang B, Gladyshev VN, 2020. How can aging be reversed? Exploring rejuvenation from a damage-based perspective. Advanced Genetics 1, e10025. 10.1002/ggn2.10025 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang B, Ma S, Rachmin I, He M, Baral P, Choi S, Gonçalves WA, Shwartz Y, Fast EM, Su Y, Zon LI, Regev A, Buenrostro JD, Cunha TM, Chiu IM, Fisher DE, Hsu YC, 2020. Hyperactivation of sympathetic nerves drives depletion of melanocyte stem cells. Nature 577, 676–681. 10.1038/s41586-020-1935-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang Y, Fujita N, Tsuruo T, 1999. Caspase-mediated cleavage of p21Waf1/Cip1 converts cancer cells from growth arrest to undergoing apoptosis. Oncogene 18, 1131–1138. 10.1038/sj.onc.1202426 [DOI] [PubMed] [Google Scholar]
- Zipple MN, Archie EA, Tung J, Altmann J, Alberts SC, 2019. Intergenerational effects of early adversity on survival in wild baboons. eLife 8. 10.7554/eLife.47433 [DOI] [PMC free article] [PubMed] [Google Scholar]