Next Article in Journal
ScFv-h3D6 Prevents Bapineuzumab-Induced Hemorrhagic Events in the APP23 Mouse Model of Alzheimer’s Disease
Previous Article in Journal
Serum PTH ≥ 40 pg/mL as a Marker of Bone Fragility and Vitamin D Deficiency in Periodontitis Patients: Biochemical, Densitometric and Genetic Evidence
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Exploring Chronic Pain, Immune Dysfunction and Lifestyle: A Focus on T Cell Exhaustion and Senescence

1
Pain in Motion Research Group (PAIN), Department of Physiotherapy, Human Physiology and Anatomy, Faculty of Physical Education and Physiotherapy, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussels, Belgium
2
Flanders Research Foundation-FWO, 1090 Brussels, Belgium
3
Department of Public Health and Primary Care, Centre for Environment & Health, KU Leuven, Kapucijnenvoer 35, 3000 Leuven, Belgium
4
Laboratory of Neuro-Aging & Viro-Immunotherapy, Center for Neurosciences, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussels, Belgium
5
Mucosal Immunology Lab, Institute of Biomedicine and Molecular Genetics (IBGM), University of Valladolid-CSIC, 47005 Valladolid, Spain
6
Unit of Physiotherapy, Department of Health and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, SE-405 30 Gothenburg, Sweden
7
PijnPraxis.be Practice for Pain Management, 3970 Leopoldsburg, Belgium
*
Author to whom correspondence should be addressed.
Biomolecules 2025, 15(11), 1601; https://doi.org/10.3390/biom15111601 (registering DOI)
Submission received: 2 October 2025 / Revised: 12 November 2025 / Accepted: 13 November 2025 / Published: 15 November 2025
(This article belongs to the Section Molecular Biology)

Abstract

Chronic pain conditions are debilitating and have an enormous impact on quality of life, yet underlying biological mechanisms remain poorly understood, hindering the development of diagnostic tools and effective treatments. Emerging evidence suggests a role for immune dysfunction in chronic pain. Among the various forms of immune dysfunction, T cell exhaustion and senescence, well-characterized in cancer and chronic infections, remain largely unexplored in chronic pain research. At the same time, lifestyle factors such as sleep, stress, physical activity, and diet are increasingly recognized as modulators of both pain and immune function. This review explores the potential interplay between these behavioural factors, immune exhaustion/senescence, and chronic pain. Critical gaps in current knowledge are identified, and future directions are outlined to clarify immune dysfunction and the influence of lifestyle factors in chronic pain conditions.

1. Linking Chronic Pain to Immune Dysfunction and Lifestyle Factors

Pain is the most common reason why individuals seek medical care and represents a significant burden on healthcare systems worldwide [1]. Chronic pain, defined as pain persisting for more than three months, is a multifaceted and debilitating condition affecting 11–40% of the population worldwide, with a higher prevalence in women [1,2,3]. Notably, three of the top four causes of disability (back pain, musculoskeletal disorders, and neck pain) are classified as chronic pain conditions [4]. Despite its profound impact on individuals, society, and the economy, chronic pain still lacks objective biomarkers for diagnosis [1]. This absence stems from a limited understanding of its underlying biological mechanisms, leaving diagnosis relying on subjective self-reporting [1,5]. The lack of biomarkers poses a barrier to the development of effective diagnostic tools and treatments for chronic pain, highlighting an urgent unmet medical need.
Mounting evidence supports the involvement of the immune system in chronic pain, with immune cells and nociceptors engaging in a dynamic, bidirectional network that modulates pain states [6,7]. Neuroimmune interactions are tightly regulated by both the central and peripheral nervous systems, for instance through stress-related pathways such as the hypothalamic–pituitary–adrenal (HPA) axis, which can influence immune cell activity and pain perception [8]. Despite evidence suggesting a role for immune dysfunction in chronic pain, focused research into immune states remains limited. Many studies are limited by small sample sizes and non-specific analyses of immune markers, leaving key questions unanswered [9]. Nonetheless, these findings are valuable when interpreted in the context of specific immune processes such as immune exhaustion and senescence, two distinct and fundamental states of the immune system. Exhaustion and senescence are particularly relevant in the context of chronic conditions. Unlike acute dysfunctional immune states, they represent long-term alterations of immune function [10]. Both processes are well-established contributors to pathologies such as cancer, chronic viral infections, and autoimmunity, where they have greatly advanced the understanding of immune dysfunction and therapeutic strategies [10,11,12]. In chronic pain, however, their contribution is remarkably underexplored.
Chronic pain is influenced by multiple lifestyle factors, amongst which are sleep, psychological stress, physical activity, and diet [13]. Each of these factors has independently been linked to both pain outcomes and immune regulation, which suggests an interesting interplay between chronic pain, lifestyle factors, and the immune system [14,15,16]. Dysregulation in these domains, such as sleep deprivation, chronic stress, and physical inactivity, can promote systemic low-grade inflammation and alter cytokine profiles [14,15,16]. These immune changes may, in turn, contribute to chronic pain through mechanisms such as neuroinflammation and central sensitization [6,17]. Investigating the influence of lifestyle factors may therefore offer valuable insights into the underlying biological contributors of chronic pain, particularly regarding immune-related mechanisms.
In this review, the potential interplay between the four main lifestyle factors (i.e., sleep, psychological stress, physical activity, and diet), immune exhaustion/senescence, and chronic pain is explored. Critical knowledge gaps are highlighted and directions for future research into immune processes and modifiable lifestyle factors in chronic pain conditions are proposed.

2. Immune Exhaustion and Senescence

The immune system plays a central role in defending the body against pathogens and injury through interactions between physical barriers, immune cells, and signalling molecules [18]. Immune dysfunction, however, is linked to a wide range of diseases, including cancer, autoimmune disorders, and neurodegenerative conditions [18,19,20]. Evidence also suggests a central role for immune dysfunction in chronic pain, where immune cells and nociceptors engage in bidirectional communication. Activated nociceptors release inflammatory mediators that further stimulate immune cells, sustaining a continuous feedback loop [7]. Moreover, immune signalling is implicated in the transition from acute to chronic pain, with T cells playing both protective and pain-promoting roles [9].
The activity of T cells is tightly regulated by stimulatory and inhibitory receptors on their surface, whose expression changes dynamically during differentiation [10]. Both CD4+ and CD8+ T cells interact with antigen-presenting cells (APCs) via these receptors, recognizing antigens presented on major histocompatibility complex (MHC) molecules [21]. Ligand binding modulates downstream signalling pathways, controlling cell activation and function [21]. The level of receptor expression thus finetunes the immune response and is used to identify specific T cell subsets, such as CD4+ and CD8+ T cells, and immune states. Among these, T cell exhaustion and T cell senescence are two immune states that share phenotypic similarities, including reduced functionality and altered expression of receptors, but differ fundamentally in their underlying triggers, molecular features, and reversibility.
Immune exhaustion refers to a progressive, yet potentially reversible, decline in T cell functionality, primarily affecting effector T cells [10]. Different from the regulatory suppression of T cells, the exhausted state is triggered by persistent antigenic overstimulation and prolonged immune activation, such as during cancer and chronic viral infections [10,11]. Exhausted T cells exhibit transcriptional and epigenetic changes, the gradual loss of effector function, a distinct cytokine profile, metabolic dysregulation, and overexpression of inhibitory receptors [22]. These include, but are not restricted to, programmed cell death protein 1 (PD-1), cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4), B- and T-lymphocyte attenuator (BTLA), T-cell immunoglobulin and mucin domain 3 (Tim-3), T-cell immunoreceptor with Ig and ITIM domains (TIGIT), and lymphocyte-activation gene 3 (LAG-3) [22,23]. These receptors interact with their corresponding ligand(s) on APCs, such as PD-1 with programmed death-ligand 1 (PD-L1) and Tim-3 with Galectin-9 (Gal-9), thereby affecting T cell function [22,24].
At the transcriptional level, exhaustion is associated with dysregulation of key transcription factors involved in T cell differentiation, such as T-box expressed in T cells (T-bet), Eomesodermin (Eomes), and T cell factor 1 (TCF-1) [22,25]. Functionally, exhausted T cells show impaired production of cytotoxic effector proteins such as granzyme B and perforin, and reduced polyfunctional cytokine production of interleukin 2 (IL-2), interferon gamma (IFN-γ), and tumour necrosis factor alpha (TNF-α) [22,23,26]. Increased levels of IL-10 have also been implicated in promoting T cell exhaustion [23]. Importantly, exhaustion can be reversed, as evidenced by the remarkable clinical success of immune checkpoint inhibitors (ICIs) targeting PD-1 or CTLA-4, which reverse T cell exhaustion and are now established therapies for several cancers [27,28,29,30,31].
By contrast, immune senescence represents an irreversible state of cell cycle arrest; typically triggered by ageing or cellular stress [10,32]. While senescence plays beneficial roles in processes such as wound healing and tumour suppression, chronic senescence can lead to pathological outcomes [33]. Senescent immune cells exhibit telomere shortening and loss of telomerase activity, distinct transcriptional and epigenetic changes, metabolic dysregulation, apoptosis resistance, and a pro-inflammatory phenotype known as the senescence-associated secretory phenotype (SASP) [33,34,35,36]. SASP involves the release of signalling molecules, including IL-6, IL-10, TNF-α, and IFN-γ [36,37,38].
Senescent cells are further characterized by overexpression of surface markers, including Killer cell lectin-like receptor G1 (KLRG-1), Tim-3, TIGIT, and the terminal differentiation marker cluster of differentiation 57 (CD57), while having reduced CD27 and CD28 expression [10,22,36]. Unlike exhausted T cells, these cells have typically reached a terminal differentiation state and exhibit a reduced naive to memory ratio [36]. Functionally, senescent T cells show decreased granzyme B and perforin production, alongside a more pro-inflammatory secretory profile, with increased activity of cyclic GMP-AMP synthase (cGAS), nuclear factor kappa B (NF-κB), and cyclin-dependent kinase inhibitor 2A (p16INK4a), evoking SASP [34,38,39]. This shift toward increased cytokine-mediated signalling may contribute to sustained inflammation and sensitization, mechanisms which are increasingly recognized as relevant to chronic pain pathophysiology [8,9]. Immune senescence is irreversible; however, senolytic drugs have been developed to selectively target and remove senescent cells [40]. An overview of these properties, including the similarities and differences between T cell exhaustion and senescence, is summarized in Table 1.
Despite major advances in our understanding of immune exhaustion and senescence in the context of cancer and infectious diseases, where they have paved the way for therapy breakthroughs such as ICIs, their contribution to chronic pain remains largely unexplored [27,28,31]. This knowledge gap is striking and underscores the urgent need for in-depth studies focusing on T cell phenotypes in chronic pain populations.

3. Role of Lifestyle Factors in Immune Exhaustion and Senescence

Sleep, stress, physical activity, and diet are considered the four main pillars of health and are essential for proper immune function [14,15,16]. When these lifestyle factors are not well-maintained or dysregulated, immune dysfunction can occur. Because they affect both immune function and chronic pain, exploring lifestyle factors could provide critical insights into how T cell exhaustion and senescence arise or persist in chronic pain. Current knowledge on the influence of sleep, psychological stress, physical activity, and diet, interpreted in the context of T cell exhaustion and senescence, is summarized in Figure 1. The next sections explore this information in more detail.

3.1. Sleep and T Cell Dysregulation

When interpreting current evidence of sleep and its effects on immune function in the context of T cell exhaustion and senescence, most studies remain limited and are restricted to either surface receptors analyses or functional assessments. Nevertheless, their insights are valuable. For instance, sleep fragmentation is correlated to an increase in late differentiated, CD8+CD28 T cells, a hallmark of senescent T cells [41]. Yet, in mice, sleep fragmentation does not significantly alter PD-1 expression on CD4+ T cells, suggesting that sleep may not drive T cell exhaustion [42].
Sleep does not affect the number of granzyme B or perforin expressing cells [43]. However, sleep restriction has been shown to suppress T-bet expression, a key transcription factor regulating T cell differentiation and function [44]. This downregulation was accompanied by reduced IFN-γ levels and granzyme B expression [44]. Additionally, poor sleep quality is linked to elevated IL-6 production [45]. This effect is likely driven by activation of NF-κB, which regulates pro-inflammatory genes and was found to be increased in immune cells during sleep loss [44,46,47,48]. On the other hand, sleep deprivation also upregulates senescence-related genes such as p16INK4a, which normalize after successful remission of insomnia, a sleep-related disorder that is highly prevalent in the chronic pain population [47,49]. Most evidence on cytokine changes remains inconsistent, with studies reporting increased, decreased and unaffected cytokines such as IL-2, TNF-α, and IFN-γ [15].
Telomere shortening in relation to sleep has been examined in multiple studies, many of which indicate a correlation between poor sleep quality or duration and shorter telomere length (TL) in immune cells [50,51,52,53,54,55,56,57,58,59,60,61,62]. Prather et al. showed this association specifically in CD4+ and CD8+ T cells [52]. Interestingly, a recent study reported a genetic component for short sleep and its association with decreased TL [59]. However, not all studies confirm this relationship, with several reports of no significant association between sleep quality or duration and TL in immune cells [51,52,53,56,63,64,65]. Zgheib et al. linked sleep difficulties to shorter telomeres, although sleep duration itself was not significantly related [65]. Age plays an important role, as evidenced by Cribbet et al. who found an association between sleep and TL in older adults, but not in younger adults [53]. Other studies report sex-specific effects; Liang et al. in women under 50 years old, and Jackowska et al. in older men but not in women [54,55]. Several studies indicate that insomnia is associated with shorter telomeres, although results depend on age and are not always significant [66,67,68]. Conversely, one study found no relation between insomnia and TL [64]. For a more comprehensive overview of current knowledge on the link between sleep and TL, readers are referred to the review by Barragán et al. [69].

3.2. Stress and T Cell Dysregulation

Stress and stress-related disorders are linked to fewer naive and more terminally differentiated CD4+ and CD8+ T cells [70,71,72,73,74]. When looking at the expression of T cell surface markers, stress has been associated with upregulation of PD-1 and CD57 and downregulation of CTLA-4 and CD28 on CD4+ T cells, as well as increased KLRG-1, PD-1, and CD57, yet reduced CD28, expression on CD8+ T cells [70,75,76,77]. Duggal et al. strengthened these claims by combining T cell phenotyping with functional assays, demonstrating significant results between stress groups [75]. They also studied the effect of physical and psychological stress on T cells, showing that experiencing both stressors results in a higher senescent T cell phenotype than experiencing only one, indicating a link between stress and senescence [75]. Although conducted in a cancer context, one study showed that stress hormones modulate T cell phenotypes in humans and mice, influencing cytokine-mediated signalling and regulation of cell surface receptors, further supporting a potential link between stress and immune dysfunction [78]. Psychological stress is related to lower granzyme B and IFN-γ, as well as higher IL-10 production [79,80]. However, others report contradictory results by demonstrating elevated or conflicting levels of TNF-α, IFN-γ, IL-6, granzyme B, and perforin [43,75,80,81,82].
Ornish et al. demonstrated that lifestyle interventions, including stress management, physical activity, and dietary changes, are associated with longer TL, as measured by quantitative PCR (qPCR) [83]. This aligns with other studies linking stress to shorter telomeres [80,84,85,86,87]. Notably, while perceived stress is generally associated with shorter TL, this effect is not observed among individuals who are more physically active, suggesting a buffering effect of exercise [85]. Not all studies are consistent: one study reported no direct link between perceived stress and TL [52]. However, stress did influence the sleep–telomere relationship, with poor sleep linked to short telomeres in individuals with high perceived stress [52]. Rentscher et al. reported a link between stress and increased mRNA expression of p16INK4a, though no direct association with TL in leukocytes was observed [88]. Genetic susceptibility may further explain these associations, as individuals with the GABRA6 TT genotype, leading to increased stress susceptibility, have CD8+CD28+ T cells with shorter TL [89].

3.3. Physical Activity and T Cell Dysregulation

Exercise has been linked to lower expression of exhaustion and senescence markers (PD-1, KLRG-1, and CD57) and higher levels of CD28, granzyme B, and perforin [43,90,91,92,93]. Du et al. further showed that genes related to granzyme B, perforin, IL-2, and T-bet are upregulated in peripheral blood mononuclear cells (PBMCs) after acute exercise [94]. However, increased KLRG-1 and CD57 expression after exercise has also been demonstrated, while others found no effect on KLRG-1 expression on CD4+ T cells or the CD28CD57+ phenotype [95,96,97,98]. Despite these inconsistencies, physically active individuals display a more favourable T cell phenotype than sedentary ones, suggesting that exercise has a moderating effect [95,96].
Physical activity has been linked to shifts in T cell composition, including a reduced naive to memory ratio, as well as conflicting findings on TNF-α, IFN-γ, and IL-2 production [90,93,96,99,100]. Exercise also reduces cGAS, p16INK4a, and other senescent-related proteins, potentially having a diminishing effect on immune senescence [90].
Most studies report a positive effect between physical activity and TL, with exercise being linked to longer TL [50,63,101,102,103,104,105,106,107,108,109,110]. Moreover, physical activity may also attenuate the effect of stress on TL, as their link has been described in sedentary individuals but not in those who are physically active [85]. However, evidence remains conflicting, as some studies indicate no effect of physical activity on TL [111,112,113].

3.4. Diet and T Cell Dysregulation

Human studies investigating the link between diet and T cell function are extremely limited, but preliminary data suggests that healthy nutrition correlates with a higher number of granzyme B and perforin expressing immune cells, showing a buffering effect of a healthy diet on immune dysfunction [43]. Animal studies could offer insights but remain scarce and highlight the need for more comprehensive research. In mice, a high-fat diet increased PD-1 expression on CD8+ T cells and reduced levels of granzyme B [114]. Caloric restriction in monkeys leads to an increase in naive T cells, decrease in memory T cells, and decrease in TNF-α and IFN-γ levels [115]. Another study, however, links caloric restriction to elevated IFN-γ protein and gene expression, without effect on IL-6 and IL-10 levels [116].
Research consistently shows that diet influences TL [117]. Consumption of foods such as nuts, vegetables, fruits, and coffee is linked to immune cells with longer telomeres [111,113,117,118,119,120]. On the other hand, the Western diet, characterized by higher fat and meat intake, has been associated with shorter TL [113,117,121].

3.5. Lifestyle Factors, Immune Exhaustion and Immune Senescence

There is growing evidence suggesting that poor sleep quality and duration are associated with altered immune function. However, when interpreted in the context of T cell exhaustion, current knowledge remains limited and inconsistent. While some studies report shorter telomeres and increased inflammatory signalling suggestive of immune senescence, others fail to confirm these in T cells specifically. Findings on cytokine profiles are particularly variable, highlighting the complexity of the relationship between sleep and T cell dysfunction. Importantly, assessing sleep is challenging as it can be subjective (e.g., using questionnaires) or objective (e.g., via polysomnography), complicating direct comparison between studies. In addition, sleep is closely related to other lifestyle factors such as psychological stress, with poor sleep reducing stress resilience and high stress levels leading to sleep disturbance.
Similarly, when investigating psychological stress and its influence on T cell differentiation and signalling, no clear conclusion on T cell exhaustion can be drawn. While stress has been associated with changes in exhaustion markers, such as altered cytokine levels (e.g., TNF-α, IFN-γ, and IL-10) and expression of T cell inhibitory receptors, these findings are limited and inconsistent across studies, which underscores the need for in-depth, targeted research. Substantial evidence links psychological stress to immune senescence, such as shorter telomeres, altered receptor expression, and elevated pro-inflammatory cytokines. However, as observed for sleep, evidence regarding cytokine levels is inconsistent, limiting direct comparison between studies. Future work should therefore combine both phenotypic markers and functional assays to better define the impact of stress on immune exhaustion and senescence, while considering other confounding lifestyle factors such as sleep, physical activity, and diet.
Similar to sleep and stress, physical activity is a broad and heterogeneous concept. An important distinction should be made between studies investigating acute exercise effects (following a single bout of exercise) and long-term training effects (resulting from regular exercise over weeks or months). This difference, along with factors such as timing of measurements, participants’ age, and type or intensity of exercise, all significantly influence outcomes. Through changes in stress hormones and energy metabolism, physical activity influences T cell activity and may therefore modulate exhaustion, but further research should investigate true T cell functionality to clarify its effect and resolve current inconsistencies [122]. Mounting evidence suggests that exercise attenuates the senescent phenotype, as it has been inversely associated with signs of senescence. In 2021, Barragán et al. and Mathot et al., respectively, reviewed how physical activity might modulate TL and other senescent markers; for more information readers are referred to their articles [69,123]. Current evidence suggests that long-term physical activity decreases T cell senescence in older adults.
Human studies on diet and immune regulation in the context of T cell differentiation, exhaustion, and senescence remain extremely limited. A healthy diet is associated with longer telomeres and higher frequencies of granzyme B and perforin-expressing immune cells, yet a clear conclusion on exhaustion or senescence cannot be drawn. Findings from animal studies further suggest that diet may influence immune senescence, but results remain inconsistent and are based on a limited number of models. Notably, metabolic diseases such as obesity and type 2 diabetes are linked to elevated pro-inflammatory cytokines and an accumulation of CD28CD57+ T cells, reflecting immune senescence [124,125,126]. This aligns with the framework of this review, suggesting that dietary factors can shape T cell function and promote a chronic pro-inflammatory phenotype. However, the current lack of comprehensive experimental and human data highlights a key research gap, underscoring the need for well-controlled studies to clarify the causal effects of dietary effects on T cell exhaustion and senescence.
Overall, the interplay between lifestyle factors, immune exhaustion or senescence, and chronic pain is complex and characterized by conflicting findings. Current studies often investigate single variables and rarely control for confounding factors such as sleep, stress, physical activity, or diet simultaneously. For now, these associations should therefore be interpreted as correlative rather than causal. Future research integrating multiple lifestyle factors with in-depth immune phenotyping is essential to clarify causal relationships and their potential contribution to chronic pain.

4. Immune Exhaustion and Senescence in Chronic Pain Conditions

Despite substantial evidence implicating the involvement of the immune system in chronic pain, research into specific immune processes, such as exhaustion and senescence, remains limited [7,9]. Exploring T cell exhaustion and senescence in chronic pain conditions may therefore shed light on previously overlooked disease mechanisms.

4.1. Primary Chronic Pain Conditions

Chronic primary pain is defined as pain lasting for more than three months that is associated with significant functional disability and/or emotional distress, without being better explained by another condition [127]. Disorders such as myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS), fibromyalgia (FM), and chronic widespread pain (CWP) fall within this category and are characterized by persistent pain without a clear understanding of their underlying etiology and pathophysiology. In this context, T cell exhaustion and senescence may therefore not only arise as consequences of disease but could also contribute to the development or maintenance of chronic pain.
The potential link between ME/CFS and immune exhaustion or senescence has previously been reviewed [128]. This chronic disorder is often confused with macrophagic myofasciitis (MM), characterized by widespread pain and severe fatigue, or with long coronavirus disease (Long COVID). In MM, decreased TNF-α levels have been reported and interpreted as a reflection of an exhausted immune system as seen in ME/CFS [129]. Patients with Long COVID and ME/CFS symptoms exhibit elevated expression of exhaustion markers, including PD-1, Tim-3, TIGIT, and Gal-9 [130,131]. Saito et al. found that Tim-3+ cells correlate with impaired TNF-α and IFN-γ production, whereas PD-1+ cells are associated with elevated cytokines levels [130].
Several studies have studied cytokine levels in FM patients, with increased, decreased, and unchanged results for TNF-α, IFN-γ, IL-2, IL-6, and IL-10 [132,133,134,135,136,137,138,139,140,141,142]. However, these cytokines were measured in plasma or serum without confirmation by functional assays, such as in vitro T cell stimulation, limiting conclusions about T cell functionality. Wallace et al. investigated cytokine production in PBMCs, reporting no significant differences for TNF-α, IFN-γ, IL-2, and IL-10, and an increase for IL-6 [143]. In serum, elevated IL-6 levels correlate to FM symptoms and thus chronic pain, which may be suggestive of immune senescence [142].
A genomic study of patients with CWP identified differentially methylated regions in immune signalling pathways, including PD-1 and T cell exhaustion [144]. While preliminary, these findings suggest a potential role for immune exhaustion and highlight the need for more focused research.

4.2. Secondary Chronic Pain Conditions

In several disorders, chronic pain arises as a consequence of an underlying disease process, as is seen in cancer or rheumatoid arthritis (RA). In these contexts, specific pathophysiological mechanisms drive the development of chronic pain, and immune alterations such as T cell exhaustion and senescence might reflect disease progression or chronicity rather than act as primary drivers of chronic pain.
Genetic data suggests a possible link between exhaustion and RA, as T-bet gene polymorphisms have been associated with RA susceptibility [145]. Patients with RA also exhibit features of immune senescence, including telomere shortening, the CD28CD57+ phenotype in CD4+ T cells, and elevated levels of TNF-α and IFN-γ [146,147,148,149]. While elevated levels of TNF-α and IFN-γ primarily reflect systemic inflammation, they may also indicate a role for senescent T cells in RA, as further described by Chalan et al. [150]. However, it remains unclear whether such immune changes directly contribute to chronic pain in RA.
In osteoarthritis (OA), a condition causing chronic pain in joints, decreased IL-10 levels have been linked to reduced Tim-3 expression, suggesting that T cell exhaustion is unlikely a prominent feature in this population [151]. However, pain-associated differently methylated regions in knee OA were enriched for genes involved in the PD-1 immunotherapy pathway, suggesting a potential role for T cell exhaustion [152]. Given the limited number of studies, interpreting these results in the context of immune exhaustion may be misleading.
Chronic osteomyelitis, a persistent bone infection accompanied by pain, has been associated with increased LAG-3 expression on T cells and reduced IFN-γ production, suggestive of T cell exhaustion [153]. In contrast, PD-1 and CD57 expression remained unchanged, and Tim-3 expression was reduced on T follicular helper cells [153,154]. Interestingly, B and NK cells of these patients show significant increases in the expression of exhaustion markers [154]. Moreover, a lack of alteration in CD57 expression suggests that T cell senescence is unlikely in people with osteomyelitis, although no strong conclusion can be drawn.
In chronic neuropathic pain, CD4+ T cells exhibit increased CD27 expression, contradicting a senescent phenotype [155]. Still, upregulation of NF-κB in T cells, and not in other immune cell types, alongside a reduction in CD8+ naive and an increase in CD4+ memory T cells, may reflect some sign of senescence [155]. Postherpetic neuralgia (PHN), a neuropathic pain condition following herpes zoster infection, has been linked to signs of T cell exhaustion. Patients with PHN show reduced CD28 expression in both CD4+ and CD8+ T cells, alongside increased levels of TNF-α, PD-1, CD57, and granzyme B [156]. Notably, T-bet is upregulated in patients’ CD4+ T cells but downregulated in their CD8+ T cells [156]. Furthermore, BTLA expression is positively correlated with pain scores, supporting a role for exhaustion in PHN [156].
Individuals suffering from sickle cell disease (SCD) have elevated levels of soluble exhaustion markers, including PD-1, BTLA, Tim-3, CTLA-4, and LAG-3, some of which highly correlate with pain [157]. However, elevated CD27 and CD28 levels in these patients contradict immune senescence [157]. It is important to note that pain in SCD is driven by well-established mechanisms such as inflammation and ischemia, which may overshadow contributions from T cell exhaustion or senescence [158]. Nonetheless, as there are currently no other SCD studies evaluating exhaustion and senescence, further research is warranted to clarify their potential role.
Taken together, these findings suggest that immune exhaustion features may occur in a variety of chronic pain conditions, both primary and secondary, although patterns remain condition-specific and inconsistent. While features of immune senescence are present in diseases like RA, they are not consistently observed across all chronic pain conditions. To clarify the role of immune exhaustion in chronic pain, standardized investigations of T cell phenotypes and cytokine profiles are needed, ideally in studies that carefully monitor or control for lifestyle factors known to affect immune states. Ultimately, future research should aim to clarify whether specific immune states, such as T cell exhaustion and senescence, contribute to chronic pain pathophysiology, and to what extent the established perpetuating role of an unhealthy lifestyle relates to these states in chronic pain. Additionally, while T cell subset-specific changes have been described in some chronic pain conditions, these are not always investigated in detail across all studies, making it difficult to draw in-depth conclusions on the role of subsets in T cell exhaustion or senescence. Integrating comprehensive subset analyses in future studies could provide a more complete understanding of immune dysregulation in chronic pain.

4.3. Potential Treatments

ICIs and senolytics, established therapies against exhaustion and senescence, respectively, illustrate how targeting dysfunctional immune states can lead to therapeutic breakthroughs. While their use is well-characterized in oncology, their application in chronic pain research is still in its very early stages. ICIs, drugs that target ligand-receptors interactions associated with exhaustion, modulate downstream signalling pathways and ultimately restore effective T cell function [40]. In the context of chronic pain, evidence is scarce. For instance, a recent study in cancer patients reported that PD-1/PD-L1 blockade on macrophages enhances chemotherapy-induced neuropathic pain [159]. This effect is likely driven by increased inflammation following checkpoint inhibition rather than direct reactivation of exhausted T cells, underscoring the complex interactions between immune checkpoints, immune cell function, and pain modulation. These findings highlight the need for dedicated research to investigate the role of T cell exhaustion, and its potential blockade, in chronic pain modulation. Additionally, the use of other ICIs, such as those targeting CTLA-4, should be studied, as preliminary evidence suggests PD-1-based ICIs may not be beneficial for chronic pain patients [160].
Senolytic drugs, on the other hand, do not reverse the senescent phenotype but instead cause selective cell death of senescent cells [161]. Recently, Mannarino et al. showed that senolytics diminish chronic low back pain in mice, although their study was not focused on T cells [162]. In line with this, the use of senolytics reduced neuropathic pain in mice and rat models [163,164,165]. Interestingly, senolytic drugs have already been tested in OA patients up until phase II clinical trials, moderately reducing associated pain levels [166,167]. However, senescence is an established feature in the chondrocytes of these patients, whereas its contribution to T cell function is not well understood.

5. Conclusions

Despite valuable insights discussed in this review, current studies on lifestyle factors and chronic pain are limited by incomplete T cell profiling and selective analysis of cytokines. Rather than focusing on a few selected markers, broader immune assessments using advanced techniques such as high-dimensional flow cytometry and single-cell RNA sequencing are needed. Moreover, the interplay between lifestyle factors and immune exhaustion or senescence remains complex, as most studies investigate these parameters in isolation and rarely account for potential confounders. Integrative, multi-variable approaches are therefore essential to unravel their complex interplay. In chronic pain research focusing on immunity, systematically incorporating lifestyle variables such as sleep, psychological stress, physical activity, and diet could provide important insights, as all these factors have been associated with both chronic pain and immune function, and may therefore act as modulators.
To achieve this, future studies should adopt more concrete and standardized approaches. Currently, the absence of standardized definitions and uniform objective measurements (e.g., receptor expression, cytokine profiles, and TL) complicates direct comparison across studies. Heterogeneity in study populations (e.g., age, sex, and comorbidities) and methodological approaches further complicate interpretation of current knowledge. Future studies should apply robust methodological designs, such as randomized cross-over designs, to examine causal interference. Importantly, interaction effects, such as how sleep, stress, physical activity, and diet may influence each other, TL, or ageing of the immune system, should always be considered. Finally, objective measurements should be combined with validated pain questionnaires to distinguish between immune alterations linked to disease mechanisms and those specifically associated with the experience of chronic pain, as these do not necessarily overlap.
Alongside methodological improvements, therapeutic implications should also be considered. Clarifying the therapeutic potential of ICIs and senolytics in chronic pain conditions represents an important future direction but requires careful characterization of T cell involvement before translation to patients.

Author Contributions

Conceptualization, J.N. and A.P.; methodology, Y.B., J.N., and A.P.; investigation, Y.B.; recourses, Y.B., J.N., and A.P.; writing—original draft preparation, Y.B.; writing—review and editing, J.H., A.W., H.-Y.X., J.V.C., T.L., J.L.A., S.C.-S., J.N., and A.P.; visualization, Y.B.; supervision, J.N. and A.P. All authors have read and agreed to the published version of the manuscript.

Funding

Andrea Polli, Jolien Hendrix and Huan-Yu Xiong are Fonds Wetenschappelijk Onderzoek (FWO) fellowship holders. Jente Van Campenhout is funded by ME Research UK (SCIO charity number SCO36942). This work is furthermore supported by the Strategic Research Program SRP90 (‘Pain Never Sleeps: Unravelling the Sleep-Pain Interaction in Patients with Chronic Pain’), funded by the research council of the Vrije Universiteit Brussel, Brussels, Belgium.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study.

Acknowledgments

The authors gratefully acknowledge the valuable input received by David Bernardo Ordiz, which helped improve this manuscript.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

APCsAntigen-presenting cells
BTLAB- and T-lymphocyte attenuator
CD27/28/57Cluster of differentiation 27/28/57
cGASCyclic GMP-AMP synthase
COVIDCoronavirus disease
CTLA-4Cytotoxic T-lymphocyte-associated antigen 4
CWPChronic widespread pain
EomesEomesodermin
FMFibromyalgia
Gal-9Galectin-9
HPA axisHypothalamic–pituitary–adrenal axis
ICIsImmune checkpoint inhibitors
IFN-γInterferon gamma
IL-2/6/10Interleukin 2/6/10
KLRG-1Killer cell lectin-like receptor G1
LAG-3Lymphocyte-activation gene 3
Long COVIDLong Coronavirus disease
MHCMajor histocompatibility complex
MMMacrophagic myofasciitis
ME/CFSMyalgic encephalomyelitis/chronic fatigue syndrome
NF-κBNuclear factor kappa B
NK cellNatural killer cell
OAOsteoarthritis
PBMCsPeripheral blood mononuclear cells
PD-L1Programmed Death-Ligand 1
PD-1Programmed cell death protein 1
PHNPostherpetic neuralgia
p16INK4aCyclin-dependent kinase inhibitor 2A
qPCRQuantitative polymerase chain reaction
RARheumatoid arthritis
SASPSenescence-associated secretory phenotype
SCDSickle cell disease
T-betT-box expressed in T cells
TCF-1T cell factor 1
TIGITT-cell immunoreceptor with Ig and ITIM domains
Tim-3T-cell immunoglobulin and mucin domain 3
TLTelomere length
TNF-αTumour necrosis factor alpha

References

  1. Cohen, S.P.; Vase, L.; Hooten, W.M. Chronic pain: An update on burden, best practices, and new advances. Lancet 2021, 397, 2082–2097. [Google Scholar] [CrossRef]
  2. Treede, R.-D.; Rief, W.; Barke, A.; Aziz, Q.; Bennett, M.I.; Benoliel, R.; Cohen, M.; Evers, S.; Finnerup, N.B.; First, M.B.; et al. Chronic pain as a symptom or a disease: The IASP Classification of Chronic Pain for the International Classification of Diseases (ICD-11). PAIN 2019, 160, 19–27. [Google Scholar] [CrossRef]
  3. Dahlhamer, J.; Lucas, J.; Zelaya, C.; Nahin, R.; Mackey, S.; DeBar, L.; Kerns, R.; Von Korff, M.; Porter, L.; Helmick, C. Prevalence of Chronic Pain and High-Impact Chronic Pain Among Adults—United States, 2016. Morb. Mortal. Wkly. Rep. 2018, 67, 1001–1006. [Google Scholar] [CrossRef]
  4. Murray, C.J.L.; Abraham, J.; Ali, M.K.; Alvarado, M.; Atkinson, C.; Baddour, L.M.; Bartels, D.H.; Benjamin, E.J.; Bhalla, K.; Birbeck, G.; et al. The State of US health, 1990-2010: Burden of diseases, injuries, and risk factors. JAMA 2013, 310, 591–608. [Google Scholar] [CrossRef]
  5. Polli, A.; Nijs, J.; Thienpont, B. Epigenetics as the molecular substrate of multimodal lifestyle approaches for patients with persistent pain. Braz. J. Phys. Ther. 2024, 29, 101170. [Google Scholar] [CrossRef] [PubMed]
  6. Ji, R.R.; Nackley, A.; Huh, Y.; Terrando, N.; Maixner, W. Neuroinflammation and Central Sensitization in Chronic and Widespread Pain. Anesthesiology 2018, 129, 343–366. [Google Scholar] [CrossRef]
  7. Ji, R.R.; Chamessian, A.; Zhang, Y.Q. Pain regulation by non-neuronal cells and inflammation. Science 2016, 354, 572–577. [Google Scholar] [CrossRef]
  8. Sawicki, C.M.; Humeidan, M.L.; Sheridan, J.F. Neuroimmune Interactions in Pain and Stress: An Interdisciplinary Approach. Neuroscientist 2021, 27, 113–128. [Google Scholar] [CrossRef]
  9. Laumet, G.; Ma, J.; Robison, A.J.; Kumari, S.; Heijnen, C.J.; Kavelaars, A. T Cells as an Emerging Target for Chronic Pain Therapy. Front. Mol. Neurosci. 2019, 12, 216. [Google Scholar] [CrossRef] [PubMed]
  10. Akbar, A.N.; Henson, S.M. Are senescence and exhaustion intertwined or unrelated processes that compromise immunity? Nat. Rev. Immunol. 2011, 11, 289–295. [Google Scholar] [CrossRef] [PubMed]
  11. McLane, L.M.; Abdel-Hakeem, M.S.; Wherry, E.J. CD8 T Cell Exhaustion During Chronic Viral Infection and Cancer. Annu. Rev. Immunol. 2019, 37, 457–495. [Google Scholar] [CrossRef]
  12. Ray, D.; Yung, R. Immune senescence, epigenetics and autoimmunity. Clin. Immunol. 2018, 196, 59–63. [Google Scholar] [CrossRef] [PubMed]
  13. Mills, S.E.E.; Nicolson, K.P.; Smith, B.H. Chronic pain: A review of its epidemiology and associated factors in population-based studies. Br. J. Anaesth. 2019, 123, e273–e283. [Google Scholar] [CrossRef]
  14. Weyh, C.; Krüger, K.; Strasser, B. Physical Activity and Diet Shape the Immune System during Aging. Nutrients 2020, 12, 622. [Google Scholar] [CrossRef] [PubMed]
  15. Besedovsky, L.; Lange, T.; Born, J. Sleep and immune function. Pflugers Arch. 2012, 463, 121–137. [Google Scholar] [CrossRef]
  16. Raza, M.L. The stress-immune system axis: Exploring the interplay between stress and immunity. Prog. Brain Res. 2025, 291, 289–317. [Google Scholar] [CrossRef] [PubMed]
  17. Zhang, J.M.; An, J. Cytokines, inflammation, and pain. Int. Anesthesiol. Clin. 2007, 45, 27–37. [Google Scholar] [CrossRef]
  18. Chaplin, D.D. Overview of the immune response. J. Allergy Clin. Immunol. 2010, 125 (Suppl. S2), S3–S23. [Google Scholar] [CrossRef]
  19. Abbott, M.; Ustoyev, Y. Cancer and the Immune System: The History and Background of Immunotherapy. Semin. Oncol. Nurs. 2019, 35, 150923. [Google Scholar] [CrossRef]
  20. DeMaio, A.; Mehrotra, S.; Sambamurti, K.; Husain, S. The role of the adaptive immune system and T cell dysfunction in neurodegenerative diseases. J. Neuroinflammation 2022, 19, 251. [Google Scholar] [CrossRef]
  21. Chen, L.; Flies, D.B. Molecular mechanisms of T cell co-stimulation and co-inhibition. Nat. Rev. Immunol. 2013, 13, 227–242. [Google Scholar] [CrossRef]
  22. Zhao, Y.; Shao, Q.; Peng, G. Exhaustion and senescence: Two crucial dysfunctional states of T cells in the tumor microenvironment. Cell Mol. Immunol. 2020, 17, 27–35. [Google Scholar] [CrossRef] [PubMed]
  23. Wherry, E.J. T cell exhaustion. Nat. Immunol. 2011, 12, 492–499. [Google Scholar] [CrossRef]
  24. Okoye, I.; Xu, L.; Motamedi, M.; Parashar, P.; Walker, J.W.; Elahi, S. Galectin-9 expression defines exhausted T cells and impaired cytotoxic NK cells in patients with virus-associated solid tumors. J. Immunother. Cancer 2020, 8, e001849. [Google Scholar] [CrossRef]
  25. Zhang, J.; Lyu, T.; Cao, Y.; Feng, H. Role of TCF-1 in differentiation, exhaustion, and memory of CD8(+) T cells: A review. FASEB J. 2021, 35, e21549. [Google Scholar] [CrossRef]
  26. Jiang, Y.; Li, Y.; Zhu, B. T-cell exhaustion in the tumor microenvironment. Cell Death Dis. 2015, 6, e1792. [Google Scholar] [CrossRef] [PubMed]
  27. Ribas, A.; Wolchok, J.D. Cancer immunotherapy using checkpoint blockade. Science 2018, 359, 1350–1355. [Google Scholar] [CrossRef]
  28. Sharma, P.; Allison, J.P. The future of immune checkpoint therapy. Science 2015, 348, 56–61. [Google Scholar] [CrossRef] [PubMed]
  29. Leach, D.R.; Krummel, M.F.; Allison, J.P. Enhancement of antitumor immunity by CTLA-4 blockade. Science 1996, 271, 1734–1736. [Google Scholar] [CrossRef]
  30. Freeman, G.J.; Long, A.J.; Iwai, Y.; Bourque, K.; Chernova, T.; Nishimura, H.; Fitz, L.J.; Malenkovich, N.; Okazaki, T.; Byrne, M.C.; et al. Engagement of the PD-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation. J. Exp. Med. 2000, 192, 1027–1034. [Google Scholar] [CrossRef]
  31. Tabana, Y.; Moon, T.C.; Siraki, A.; Elahi, S.; Barakat, K. Reversing T-cell exhaustion in immunotherapy: A review on current approaches and limitations. Expert. Opin. Ther. Targets 2021, 25, 347–363. [Google Scholar] [CrossRef]
  32. Campisi, J. Aging, cellular senescence, and cancer. Annu. Rev. Physiol. 2013, 75, 685–705. [Google Scholar] [CrossRef]
  33. Adams, P.D. Healing and hurting: Molecular mechanisms, functions, and pathologies of cellular senescence. Mol. Cell 2009, 36, 2–14. [Google Scholar] [CrossRef] [PubMed]
  34. He, S.; Sharpless, N.E. Senescence in Health and Disease. Cell 2017, 169, 1000–1011. [Google Scholar] [CrossRef]
  35. Campisi, J.; d’Adda di Fagagna, F. Cellular senescence: When bad things happen to good cells. Nat. Rev. Mol. Cell Biol. 2007, 8, 729–740. [Google Scholar] [CrossRef] [PubMed]
  36. Liu, Z.; Liang, Q.; Ren, Y.; Guo, C.; Ge, X.; Wang, L.; Cheng, Q.; Luo, P.; Zhang, Y.; Han, X. Immunosenescence: Molecular mechanisms and diseases. Signal Transduct. Target. Ther. 2023, 8, 200. [Google Scholar] [CrossRef] [PubMed]
  37. Coppé, J.P.; Desprez, P.Y.; Krtolica, A.; Campisi, J. The senescence-associated secretory phenotype: The dark side of tumor suppression. Annu. Rev. Pathol. 2010, 5, 99–118. [Google Scholar] [CrossRef]
  38. Jin, P.; Duan, X.; Li, L.; Zhou, P.; Zou, C.G.; Xie, K. Cellular senescence in cancer: Molecular mechanisms and therapeutic targets. MedComm 2024, 5, e542. [Google Scholar] [CrossRef]
  39. Yang, H.; Wang, H.; Ren, J.; Chen, Q.; Chen, Z.J. cGAS is essential for cellular senescence. Proc. Natl. Acad. Sci. USA 2017, 114, E4612–E4620. [Google Scholar] [CrossRef]
  40. Naimi, A.; Mohammed, R.N.; Raji, A.; Chupradit, S.; Yumashev, A.V.; Suksatan, W.; Shalaby, M.N.; Thangavelu, L.; Kamrava, S.; Shomali, N.; et al. Tumor immunotherapies by immune checkpoint inhibitors (ICIs); the pros and cons. Cell Commun. Signal 2022, 20, 44. [Google Scholar] [CrossRef]
  41. Carroll, J.E.; Irwin, M.R.; Levine, M.; Seeman, T.E.; Absher, D.; Assimes, T.; Horvath, S. Epigenetic Aging and Immune Senescence in Women With Insomnia Symptoms: Findings From the Women’s Health Initiative Study. Biol. Psychiatry 2017, 81, 136–144. [Google Scholar] [CrossRef]
  42. Cubillos-Zapata, C.; Almendros, I.; Díaz-García, E.; Toledano, V.; Casitas, R.; Galera, R.; López-Collazo, E.; Farre, R.; Gozal, D.; García-Rio, F. Differential effect of intermittent hypoxia and sleep fragmentation on PD-1/PD-L1 upregulation. Sleep 2020, 43, zsz285. [Google Scholar] [CrossRef]
  43. Li, Q.; Morimoto, K.; Nakadai, A.; Qu, T.; Matsushima, H.; Katsumata, M.; Shimizu, T.; Inagaki, H.; Hirata, Y.; Hirata, K.; et al. Healthy lifestyles are associated with higher levels of perforin, granulysin and granzymes A/B-expressing cells in peripheral blood lymphocytes. Prev. Med. 2007, 44, 117–123. [Google Scholar] [CrossRef]
  44. Aho, V.; Ollila, H.M.; Rantanen, V.; Kronholm, E.; Surakka, I.; van Leeuwen, W.M.; Lehto, M.; Matikainen, S.; Ripatti, S.; Härmä, M.; et al. Partial sleep restriction activates immune response-related gene expression pathways: Experimental and epidemiological studies in humans. PLoS ONE 2013, 8, e77184. [Google Scholar] [CrossRef]
  45. Prather, A.A.; Marsland, A.L.; Hall, M.; Neumann, S.A.; Muldoon, M.F.; Manuck, S.B. Normative variation in self-reported sleep quality and sleep debt is associated with stimulated pro-inflammatory cytokine production. Biol. Psychol. 2009, 82, 12–17. [Google Scholar] [CrossRef]
  46. Irwin, M.R.; Wang, M.; Ribeiro, D.; Cho, H.J.; Olmstead, R.; Breen, E.C.; Martinez-Maza, O.; Cole, S. Sleep loss activates cellular inflammatory signaling. Biol. Psychiatry 2008, 64, 538–540. [Google Scholar] [CrossRef] [PubMed]
  47. Carroll, J.E.; Cole, S.W.; Seeman, T.E.; Breen, E.C.; Witarama, T.; Arevalo, J.M.G.; Ma, J.; Irwin, M.R. Partial sleep deprivation activates the DNA damage response (DDR) and the senescence-associated secretory phenotype (SASP) in aged adult humans. Brain Behav. Immun. 2016, 51, 223–229. [Google Scholar] [CrossRef] [PubMed]
  48. Irwin, M.R.; Wang, M.; Campomayor, C.O.; Collado-Hidalgo, A.; Cole, S. Sleep deprivation and activation of morning levels of cellular and genomic markers of inflammation. Arch. Intern. Med. 2006, 166, 1756–1762. [Google Scholar] [CrossRef]
  49. Carroll, J.E.; Olmstead, R.; Cole, S.W.; Breen, E.C.; Arevalo, J.M.; Irwin, M.R. Remission of insomnia in older adults treated with cognitive behavioral therapy for insomnia (CBT-I) reduces p16(INK4a) gene expression in peripheral blood: Secondary outcome analysis from a randomized clinical trial. Geroscience 2023, 45, 2325–2335. [Google Scholar] [CrossRef]
  50. Puterman, E.; Lin, J.; Krauss, J.; Blackburn, E.H.; Epel, E.S. Determinants of telomere attrition over 1 year in healthy older women: Stress and health behaviors matter. Mol. Psychiatry 2015, 20, 529–535. [Google Scholar] [CrossRef] [PubMed]
  51. Prather, A.A.; Puterman, E.; Lin, J.; O’Donovan, A.; Krauss, J.; Tomiyama, A.J.; Epel, E.S.; Blackburn, E.H. Shorter leukocyte telomere length in midlife women with poor sleep quality. J. Aging Res. 2011, 2011, 721390. [Google Scholar] [CrossRef]
  52. Prather, A.A.; Gurfein, B.; Moran, P.; Daubenmier, J.; Acree, M.; Bacchetti, P.; Sinclair, E.; Lin, J.; Blackburn, E.; Hecht, F.M.; et al. Tired telomeres: Poor global sleep quality, perceived stress, and telomere length in immune cell subsets in obese men and women. Brain Behav. Immun. 2015, 47, 155–162. [Google Scholar] [CrossRef]
  53. Cribbet, M.R.; Carlisle, M.; Cawthon, R.M.; Uchino, B.N.; Williams, P.G.; Smith, T.W.; Gunn, H.E.; Light, K.C. Cellular aging and restorative processes: Subjective sleep quality and duration moderate the association between age and telomere length in a sample of middle-aged and older adults. Sleep 2014, 37, 65–70. [Google Scholar] [CrossRef] [PubMed]
  54. Liang, G.; Schernhammer, E.; Qi, L.; Gao, X.; De Vivo, I.; Han, J. Associations between rotating night shifts, sleep duration, and telomere length in women. PLoS ONE 2011, 6, e23462. [Google Scholar] [CrossRef]
  55. Jackowska, M.; Hamer, M.; Carvalho, L.A.; Erusalimsky, J.D.; Butcher, L.; Steptoe, A. Short sleep duration is associated with shorter telomere length in healthy men: Findings from the Whitehall II cohort study. PLoS ONE 2012, 7, e47292. [Google Scholar] [CrossRef]
  56. Grieshober, L.; Wactawski-Wende, J.; Hageman Blair, R.; Mu, L.; Liu, J.; Nie, J.; Carty, C.L.; Hale, L.; Kroenke, C.H.; LaCroix, A.Z.; et al. A Cross-Sectional Analysis of Telomere Length and Sleep in the Women’s Health Initiative. Am. J. Epidemiol. 2019, 188, 1616–1626. [Google Scholar] [CrossRef]
  57. Révész, D.; Milaneschi, Y.; Terpstra, E.M.; Penninx, B.W. Baseline biopsychosocial determinants of telomere length and 6-year attrition rate. Psychoneuroendocrinology 2016, 67, 153–162. [Google Scholar] [CrossRef]
  58. Tempaku, P.F.; Mazzotti, D.R.; Hirotsu, C.; Andersen, M.L.; Xavier, G.; Maurya, P.K.; Rizzo, L.B.; Brietzke, E.; Belangero, S.I.; Bittencourt, L.; et al. The effect of the severity of obstructive sleep apnea syndrome on telomere length. Oncotarget 2016, 7, 69216–69224. [Google Scholar] [CrossRef]
  59. Hu, J.; Lu, J.; Lu, Q.; Weng, W.; Guan, Z.; Wang, Z. Mendelian randomization and colocalization analyses reveal an association between short sleep duration or morning chronotype and altered leukocyte telomere length. Commun. Biol. 2023, 6, 1014. [Google Scholar] [CrossRef] [PubMed]
  60. James, S.; McLanahan, S.; Brooks-Gunn, J.; Mitchell, C.; Schneper, L.; Wagner, B.; Notterman, D.A. Sleep Duration and Telomere Length in Children. J. Pediatr. 2017, 187, 247–252.e241. [Google Scholar] [CrossRef] [PubMed]
  61. Carroll, J.E.; Irwin, M.R.; Seeman, T.E.; Diez-Roux, A.V.; Prather, A.A.; Olmstead, R.; Epel, E.; Lin, J.; Redline, S. Obstructive sleep apnea, nighttime arousals, and leukocyte telomere length: The Multi-Ethnic Study of Atherosclerosis. Sleep 2019, 42, zsz089. [Google Scholar] [CrossRef] [PubMed]
  62. Kwon, A.M.; Baik, I.; Thomas, R.J.; Shin, C. The association between leukocyte telomere lengths and sleep instability based on cardiopulmonary coupling analysis. Sleep Breath. 2015, 19, 963–968. [Google Scholar] [CrossRef] [PubMed]
  63. Puterman, E.; Epel, E.S.; Lin, J.; Blackburn, E.H.; Gross, J.J.; Whooley, M.A.; Cohen, B.E. Multisystem resiliency moderates the major depression-telomere length association: Findings from the Heart and Soul Study. Brain Behav. Immun. 2013, 33, 65–73. [Google Scholar] [CrossRef]
  64. Wynchank, D.; Bijlenga, D.; Penninx, B.W.; Lamers, F.; Beekman, A.T.; Kooij, J.J.S.; Verhoeven, J.E. Delayed sleep-onset and biological age: Late sleep-onset is associated with shorter telomere length. Sleep 2019, 42, zsz139. [Google Scholar] [CrossRef]
  65. Zgheib, N.K.; Sleiman, F.; Nasreddine, L.; Nasrallah, M.; Nakhoul, N.; Isma’eel, H.; Tamim, H. Short Telomere Length is Associated with Aging, Central Obesity, Poor Sleep and Hypertension in Lebanese Individuals. Aging Dis. 2018, 9, 77–89. [Google Scholar] [CrossRef]
  66. Tempaku, P.; Hirotsu, C.; Mazzotti, D.; Xavier, G.; Maurya, P.; Brietzke, E.; Belangero, S.; Poyares, D.; Bittencourt, L.; Tufik, S. Long Sleep Duration, Insomnia, and Insomnia With Short Objective Sleep Duration Are Independently Associated With Short Telomere Length. J. Clin. Sleep. Med. 2018, 14, 2037–2045. [Google Scholar] [CrossRef]
  67. Garland, S.N.; Palmer, C.; Donelson, M.; Gehrman, P.; Johnson, F.B.; Mao, J.J. A nested case-controlled comparison of telomere length and psychological functioning in breast cancer survivors with and without insomnia symptoms. Rejuvenation Res. 2014, 17, 453–457. [Google Scholar] [CrossRef]
  68. Carroll, J.E.; Esquivel, S.; Goldberg, A.; Seeman, T.E.; Effros, R.B.; Dock, J.; Olmstead, R.; Breen, E.C.; Irwin, M.R. Insomnia and Telomere Length in Older Adults. Sleep 2016, 39, 559–564. [Google Scholar] [CrossRef]
  69. Barragán, R.; Ortega-Azorín, C.; Sorlí, J.V.; Asensio, E.M.; Coltell, O.; St-Onge, M.P.; Portolés, O.; Corella, D. Effect of Physical Activity, Smoking, and Sleep on Telomere Length: A Systematic Review of Observational and Intervention Studies. J. Clin. Med. 2021, 11, 76. [Google Scholar] [CrossRef] [PubMed]
  70. Freier, E.; Weber, C.S.; Nowottne, U.; Horn, C.; Bartels, K.; Meyer, S.; Hildebrandt, Y.; Luetkens, T.; Cao, Y.; Pabst, C.; et al. Decrease of CD4(+)FOXP3(+) T regulatory cells in the peripheral blood of human subjects undergoing a mental stressor. Psychoneuroendocrinology 2010, 35, 663–673. [Google Scholar] [CrossRef]
  71. Bosch, J.A.; Fischer, J.E.; Fischer, J.C. Psychologically adverse work conditions are associated with CD8+ T cell differentiation indicative of immunesenescence. Brain Behav. Immun. 2009, 23, 527–534. [Google Scholar] [CrossRef] [PubMed]
  72. Wang, J.; Shao, Y.; Deng, X.; Du, J. Causal Relationship Between Post-Traumatic Stress Disorder and Immune Cell Traits: A Mendelian Randomization Study. Brain Behav. 2024, 14, e70073. [Google Scholar] [CrossRef]
  73. Maydych, V.; Claus, M.; Dychus, N.; Ebel, M.; Damaschke, J.; Diestel, S.; Wolf, O.T.; Kleinsorge, T.; Watzl, C. Impact of chronic and acute academic stress on lymphocyte subsets and monocyte function. PLoS ONE 2017, 12, e0188108. [Google Scholar] [CrossRef] [PubMed]
  74. Klopack, E.T.; Crimmins, E.M.; Cole, S.W.; Seeman, T.E.; Carroll, J.E. Social stressors associated with age-related T lymphocyte percentages in older US adults: Evidence from the US Health and Retirement Study. Proc. Natl. Acad. Sci. USA 2022, 119, e2202780119. [Google Scholar] [CrossRef] [PubMed]
  75. Duggal, N.A.; Upton, J.; Phillips, A.C.; Hampson, P.; Lord, J.M. Depressive symptoms post hip fracture in older adults are associated with phenotypic and functional alterations in T cells. Immun. Ageing 2014, 11, 25. [Google Scholar] [CrossRef]
  76. Reed, R.G.; Presnell, S.R.; Al-Attar, A.; Lutz, C.T.; Segerstrom, S.C. Perceived stress, cytomegalovirus titers, and late-differentiated T and NK cells: Between-, within-person associations in a longitudinal study of older adults. Brain Behav. Immun. 2019, 80, 266–274. [Google Scholar] [CrossRef]
  77. Amati, M.; Tomasetti, M.; Ciuccarelli, M.; Mariotti, L.; Tarquini, L.M.; Bracci, M.; Baldassari, M.; Balducci, C.; Alleva, R.; Borghi, B.; et al. Relationship of job satisfaction, psychological distress and stress-related biological parameters among healthy nurses: A longitudinal study. J. Occup. Health 2010, 52, 31–38. [Google Scholar] [CrossRef]
  78. Aquino-Acevedo, A.N.; Knochenhauer, H.; Castillo-Ocampo, Y.; Ortiz-León, M.; Rivera-López, Y.A.; Morales-López, C.; Cruz-Robles, M.E.; Hernández-Cordero, E.R.; Russell, S.; Whitaker, R.; et al. Stress hormones are associated with inflammatory cytokines and attenuation of T-cell function in the ascites from patients with high grade serous ovarian cancer. Brain Behav. Immun. Health 2022, 26, 100558. [Google Scholar] [CrossRef]
  79. Xiong, Y.; Wang, Z.; Young, M.R.I. Reduced Expression of Immune Mediators by T-Cell Subpopulations of Combat-Exposed Veterans With Post-Traumatic Stress Disorder. Front. Psychiatry 2019, 10, 693. [Google Scholar] [CrossRef]
  80. Damjanovic, A.K.; Yang, Y.; Glaser, R.; Kiecolt-Glaser, J.K.; Nguyen, H.; Laskowski, B.; Zou, Y.; Beversdorf, D.Q.; Weng, N.P. Accelerated telomere erosion is associated with a declining immune function of caregivers of Alzheimer’s disease patients. J. Immunol. 2007, 179, 4249–4254. [Google Scholar] [CrossRef]
  81. von Känel, R.; Hepp, U.; Kraemer, B.; Traber, R.; Keel, M.; Mica, L.; Schnyder, U. Evidence for low-grade systemic proinflammatory activity in patients with posttraumatic stress disorder. J. Psychiatr. Res. 2007, 41, 744–752. [Google Scholar] [CrossRef] [PubMed]
  82. Atanackovic, D.; Schnee, B.; Schuch, G.; Faltz, C.; Schulze, J.; Weber, C.S.; Schafhausen, P.; Bartels, K.; Bokemeyer, C.; Brunner-Weinzierl, M.C.; et al. Acute psychological stress alerts the adaptive immune response: Stress-induced mobilization of effector T cells. J. Neuroimmunol. 2006, 176, 141–152. [Google Scholar] [CrossRef]
  83. Ornish, D.; Lin, J.; Chan, J.M.; Epel, E.; Kemp, C.; Weidner, G.; Marlin, R.; Frenda, S.J.; Magbanua, M.J.M.; Daubenmier, J.; et al. Effect of comprehensive lifestyle changes on telomerase activity and telomere length in men with biopsy-proven low-risk prostate cancer: 5-year follow-up of a descriptive pilot study. Lancet Oncol. 2013, 14, 1112–1120. [Google Scholar] [CrossRef]
  84. Epel, E.S.; Blackburn, E.H.; Lin, J.; Dhabhar, F.S.; Adler, N.E.; Morrow, J.D.; Cawthon, R.M. Accelerated telomere shortening in response to life stress. Proc. Natl. Acad. Sci. USA 2004, 101, 17312–17315. [Google Scholar] [CrossRef]
  85. Puterman, E.; Lin, J.; Blackburn, E.; O’Donovan, A.; Adler, N.; Epel, E. The power of exercise: Buffering the effect of chronic stress on telomere length. PLoS ONE 2010, 5, e10837. [Google Scholar] [CrossRef] [PubMed]
  86. Parks, C.G.; Miller, D.B.; McCanlies, E.C.; Cawthon, R.M.; Andrew, M.E.; DeRoo, L.A.; Sandler, D.P. Telomere length, current perceived stress, and urinary stress hormones in women. Cancer Epidemiol. Biomarkers Prev. 2009, 18, 551–560. [Google Scholar] [CrossRef] [PubMed]
  87. Schaakxs, R.; Wielaard, I.; Verhoeven, J.E.; Beekman, A.T.; Penninx, B.W.; Comijs, H.C. Early and recent psychosocial stress and telomere length in older adults. Int. Psychogeriatr. 2016, 28, 405–413. [Google Scholar] [CrossRef]
  88. Rentscher, K.E.; Carroll, J.E.; Repetti, R.L.; Cole, S.W.; Reynolds, B.M.; Robles, T.F. Chronic stress exposure and daily stress appraisals relate to biological aging marker p16(INK4a). Psychoneuroendocrinology 2019, 102, 139–148. [Google Scholar] [CrossRef]
  89. Martino, P.; Perez-Alarcón, M.; Deconinck, L.; De Raedt, R.; Vanderhasselt, M.A.; Kozusznik, M.W.; Kooy, F.; Hidalgo, V.; Venero, C.; Salvador, A.; et al. Stress and telomere length in leukocytes: Investigating the role of GABRA6 gene polymorphism and cortisol. Psychoneuroendocrinology 2025, 173, 107358. [Google Scholar] [CrossRef]
  90. Englund, D.A.; Sakamoto, A.E.; Fritsche, C.M.; Heeren, A.A.; Zhang, X.; Kotajarvi, B.R.; Lecy, D.R.; Yousefzadeh, M.J.; Schafer, M.J.; White, T.A.; et al. Exercise reduces circulating biomarkers of cellular senescence in humans. Aging Cell 2021, 20, e13415. [Google Scholar] [CrossRef]
  91. Cao Dinh, H.; Bautmans, I.; Beyer, I.; Onyema, O.O.; Liberman, K.; De Dobbeleer, L.; Renmans, W.; Vander Meeren, S.; Jochmans, K.; Delaere, A.; et al. Six weeks of strength endurance training decreases circulating senescence-prone T-lymphocytes in cytomegalovirus seropositive but not seronegative older women. Immun. Ageing 2019, 16, 17. [Google Scholar] [CrossRef]
  92. Minuzzi, L.G.; Rama, L.; Chupel, M.U.; Rosado, F.; Dos Santos, J.V.; Simpson, R.; Martinho, A.; Paiva, A.; Teixeira, A.M. Effects of lifelong training on senescence and mobilization of T lymphocytes in response to acute exercise. Exerc. Immunol. Rev. 2018, 24, 72–84. [Google Scholar]
  93. Spielmann, G.; McFarlin, B.K.; O’Connor, D.P.; Smith, P.J.; Pircher, H.; Simpson, R.J. Aerobic fitness is associated with lower proportions of senescent blood T-cells in man. Brain Behav. Immun. 2011, 25, 1521–1529. [Google Scholar] [CrossRef] [PubMed]
  94. Du, L.J.; Song, Y.H.; Tang, L.X. Effect of acute exercise on gene expression in peripheral blood mononuclear cells of puberty children. Sci. Rep. 2024, 14, 27977. [Google Scholar] [CrossRef]
  95. Despeghel, M.; Reichel, T.; Zander, J.; Krüger, K.; Weyh, C. Effects of a 6 Week Low-Dose Combined Resistance and Endurance Training on T Cells and Systemic Inflammation in the Elderly. Cells 2021, 10, 843. [Google Scholar] [CrossRef]
  96. Duggal, N.A.; Pollock, R.D.; Lazarus, N.R.; Harridge, S.; Lord, J.M. Major features of immunesenescence, including reduced thymic output, are ameliorated by high levels of physical activity in adulthood. Aging Cell 2018, 17, e12750. [Google Scholar] [CrossRef]
  97. Minuzzi, L.G.; Rama, L.; Bishop, N.C.; Rosado, F.; Martinho, A.; Paiva, A.; Teixeira, A.M. Lifelong training improves anti-inflammatory environment and maintains the number of regulatory T cells in masters athletes. Eur. J. Appl. Physiol. 2017, 117, 1131–1140. [Google Scholar] [CrossRef]
  98. Simpson, R.J.; Cosgrove, C.; Ingram, L.A.; Florida-James, G.D.; Whyte, G.P.; Pircher, H.; Guy, K. Senescent T-lymphocytes are mobilised into the peripheral blood compartment in young and older humans after exhaustive exercise. Brain Behav. Immun. 2008, 22, 544–551. [Google Scholar] [CrossRef] [PubMed]
  99. Shinkai, S.; Kohno, H.; Kimura, K.; Komura, T.; Asai, H.; Inai, R.; Oka, K.; Kurokawa, Y.; Shephard, R. Physical activity and immune senescence in men. Med. Sci. Sports Exerc. 1995, 27, 1516–1526. [Google Scholar] [CrossRef]
  100. Tylutka, A.; Morawin, B.; Gramacki, A.; Zembron-Lacny, A. Lifestyle exercise attenuates immunosenescence; flow cytometry analysis. BMC Geriatr. 2021, 21, 200. [Google Scholar] [CrossRef] [PubMed]
  101. Cherkas, L.F.; Hunkin, J.L.; Kato, B.S.; Richards, J.B.; Gardner, J.P.; Surdulescu, G.L.; Kimura, M.; Lu, X.; Spector, T.D.; Aviv, A. The association between physical activity in leisure time and leukocyte telomere length. Arch. Intern. Med. 2008, 168, 154–158. [Google Scholar] [CrossRef]
  102. Dankel, S.J.; Loenneke, J.P.; Loprinzi, P.D. The impact of overweight/obesity duration and physical activity on telomere length: An application of the WATCH paradigm. Obes. Res. Clin. Pract. 2017, 11, 247–252. [Google Scholar] [CrossRef] [PubMed]
  103. Du, M.; Prescott, J.; Kraft, P.; Han, J.; Giovannucci, E.; Hankinson, S.E.; De Vivo, I. Physical activity, sedentary behavior, and leukocyte telomere length in women. Am. J. Epidemiol. 2012, 175, 414–422. [Google Scholar] [CrossRef]
  104. Fretts, A.M.; Mete, M.; Howard, B.V.; Best, L.G.; Siscovick, D.S.; Eilat-Adar, S.; Zhao, J. Physical activity and telomere length in American Indians: The Strong Heart Study. Eur. J. Epidemiol. 2018, 33, 497–500. [Google Scholar] [CrossRef]
  105. Kim, J.H.; Ko, J.H.; Lee, D.C.; Lim, I.; Bang, H. Habitual physical exercise has beneficial effects on telomere length in postmenopausal women. Menopause 2012, 19, 1109–1115. [Google Scholar] [CrossRef] [PubMed]
  106. Latifovic, L.; Peacock, S.D.; Massey, T.E.; King, W.D. The Influence of Alcohol Consumption, Cigarette Smoking, and Physical Activity on Leukocyte Telomere Length. Cancer Epidemiol. Biomarkers Prev. 2016, 25, 374–380. [Google Scholar] [CrossRef]
  107. Saßenroth, D.; Meyer, A.; Salewsky, B.; Kroh, M.; Norman, K.; Steinhagen-Thiessen, E.; Demuth, I. Sports and Exercise at Different Ages and Leukocyte Telomere Length in Later Life--Data from the Berlin Aging Study II (BASE-II). PLoS ONE 2015, 10, e0142131. [Google Scholar] [CrossRef]
  108. Shadyab, A.H.; LaMonte, M.J.; Kooperberg, C.; Reiner, A.P.; Carty, C.L.; Manini, T.M.; Hou, L.; Di, C.; Macera, C.A.; Gallo, L.C.; et al. Leisure-time physical activity and leukocyte telomere length among older women. Exp. Gerontol. 2017, 95, 141–147. [Google Scholar] [CrossRef]
  109. Tucker, L.A. Physical activity and telomere length in U.S. men and women: An NHANES investigation. Prev. Med. 2017, 100, 145–151. [Google Scholar] [CrossRef] [PubMed]
  110. LaRocca, T.J.; Seals, D.R.; Pierce, G.L. Leukocyte telomere length is preserved with aging in endurance exercise-trained adults and related to maximal aerobic capacity. Mech. Ageing Dev. 2010, 131, 165–167. [Google Scholar] [CrossRef]
  111. Cassidy, A.; De Vivo, I.; Liu, Y.; Han, J.; Prescott, J.; Hunter, D.J.; Rimm, E.B. Associations between diet, lifestyle factors, and telomere length in women. Am. J. Clin. Nutr. 2010, 91, 1273–1280. [Google Scholar] [CrossRef]
  112. von Känel, R.; Bruwer, E.J.; Hamer, M.; de Ridder, J.H.; Malan, L. Association between objectively measured physical activity, chronic stress and leukocyte telomere length. J. Sports Med. Phys. Fitness 2017, 57, 1349–1358. [Google Scholar] [CrossRef]
  113. Tiainen, A.M.; Männistö, S.; Blomstedt, P.A.; Moltchanova, E.; Perälä, M.M.; Kaartinen, N.E.; Kajantie, E.; Kananen, L.; Hovatta, I.; Eriksson, J.G. Leukocyte telomere length and its relation to food and nutrient intake in an elderly population. Eur. J. Clin. Nutr. 2012, 66, 1290–1294. [Google Scholar] [CrossRef]
  114. Kado, T.; Nawaz, A.; Takikawa, A.; Usui, I.; Tobe, K. Linkage of CD8(+) T cell exhaustion with high-fat diet-induced tumourigenesis. Sci. Rep. 2019, 9, 12284. [Google Scholar] [CrossRef]
  115. Messaoudi, I.; Warner, J.; Fischer, M.; Park, B.; Hill, B.; Mattison, J.; Lane, M.A.; Roth, G.S.; Ingram, D.K.; Picker, L.J.; et al. Delay of T cell senescence by caloric restriction in aged long-lived nonhuman primates. Proc. Natl. Acad. Sci. USA 2006, 103, 19448–19453. [Google Scholar] [CrossRef] [PubMed]
  116. Mascarucci, P.; Taub, D.; Saccani, S.; Paloma, M.A.; Dawson, H.; Roth, G.S.; Lane, M.A.; Ingram, D.K. Cytokine responses in young and old rhesus monkeys: Effect of caloric restriction. J. Interferon Cytokine Res. 2002, 22, 565–571. [Google Scholar] [CrossRef]
  117. Gürel, S.; Pak, E.N.; Tek, N.A. Aging Processes Are Affected by Energy Balance: Focused on the Effects of Nutrition and Physical Activity on Telomere Length. Curr. Nutr. Rep. 2024, 13, 264–279. [Google Scholar] [CrossRef] [PubMed]
  118. Lee, J.Y.; Jun, N.R.; Yoon, D.; Shin, C.; Baik, I. Association between dietary patterns in the remote past and telomere length. Eur. J. Clin. Nutr. 2015, 69, 1048–1052. [Google Scholar] [CrossRef]
  119. Zhou, M.; Zhu, L.; Cui, X.; Feng, L.; Zhao, X.; He, S.; Ping, F.; Li, W.; Li, Y. Influence of diet on leukocyte telomere length, markers of inflammation and oxidative stress in individuals with varied glucose tolerance: A Chinese population study. Nutr. J. 2016, 15, 39. [Google Scholar] [CrossRef] [PubMed]
  120. Liu, J.J.; Crous-Bou, M.; Giovannucci, E.; De Vivo, I. Coffee Consumption Is Positively Associated with Longer Leukocyte Telomere Length in the Nurses’ Health Study. J. Nutr. 2016, 146, 1373–1378. [Google Scholar] [CrossRef]
  121. Chang, X.; Dorajoo, R.; Sun, Y.; Wang, L.; Ong, C.N.; Liu, J.; Khor, C.C.; Yuan, J.M.; Koh, W.P.; Friedlander, Y.; et al. Effect of plasma polyunsaturated fatty acid levels on leukocyte telomere lengths in the Singaporean Chinese population. Nutr. J. 2020, 19, 119. [Google Scholar] [CrossRef] [PubMed]
  122. Pedersen, B.K.; Hoffman-Goetz, L. Exercise and the immune system: Regulation, integration, and adaptation. Physiol. Rev. 2000, 80, 1055–1081. [Google Scholar] [CrossRef] [PubMed]
  123. Mathot, E.; Liberman, K.; Cao Dinh, H.; Njemini, R.; Bautmans, I. Systematic review on the effects of physical exercise on cellular immunosenescence-related markers—An update. Exp. Gerontol. 2021, 149, 111318. [Google Scholar] [CrossRef]
  124. Yi, H.S.; Kim, S.Y.; Kim, J.T.; Lee, Y.S.; Moon, J.S.; Kim, M.; Kang, Y.E.; Joung, K.H.; Lee, J.H.; Kim, H.J.; et al. T-cell senescence contributes to abnormal glucose homeostasis in humans and mice. Cell Death Dis. 2019, 10, 249. [Google Scholar] [CrossRef]
  125. Valentine, Y.; Nikolajczyk, B.S. T cells in obesity-associated inflammation: The devil is in the details. Immunol. Rev. 2024, 324, 25–41. [Google Scholar] [CrossRef]
  126. Shirakawa, K.; Sano, M. T Cell Immunosenescence in Aging, Obesity, and Cardiovascular Disease. Cells 2021, 10, 2435. [Google Scholar] [CrossRef] [PubMed]
  127. Nicholas, M.; Vlaeyen, J.W.S.; Rief, W.; Barke, A.; Aziz, Q.; Benoliel, R.; Cohen, M.; Evers, S.; Giamberardino, M.A.; Goebel, A.; et al. The IASP classification of chronic pain for ICD-11: Chronic primary pain. Pain 2019, 160, 28–37. [Google Scholar] [CrossRef]
  128. Van Campenhout, J.; Buntinx, Y.; Xiong, H.Y.; Wyns, A.; Polli, A.; Nijs, J.; Aerts, J.L.; Laeremans, T.; Hendrix, J. Unravelling the Connection Between Energy Metabolism and Immune Senescence/Exhaustion in Patients with Myalgic Encephalomyelitis/Chronic Fatigue Syndrome. Biomolecules 2025, 15, 357. [Google Scholar] [CrossRef]
  129. Masson, J.D.; Badran, G.; Gherardi, R.K.; Authier, F.J.; Crépeaux, G. Widespread Myalgia and Chronic Fatigue: Phagocytes from Macrophagic Myofasciitis Patients Exposed to Aluminum Oxyhydroxide-Adjuvanted Vaccine Exhibit Specific Inflammatory, Autophagic, and Mitochondrial Responses. Toxics 2024, 12, 491. [Google Scholar] [CrossRef]
  130. Saito, S.; Shahbaz, S.; Osman, M.; Redmond, D.; Bozorgmehr, N.; Rosychuk, R.J.; Lam, G.; Sligl, W.; Cohen Tervaert, J.W.; Elahi, S. Diverse immunological dysregulation, chronic inflammation, and impaired erythropoiesis in long COVID patients with chronic fatigue syndrome. J. Autoimmun. 2024, 147, 103267. [Google Scholar] [CrossRef]
  131. Klein, J.; Wood, J.; Jaycox, J.R.; Dhodapkar, R.M.; Lu, P.; Gehlhausen, J.R.; Tabachnikova, A.; Greene, K.; Tabacof, L.; Malik, A.A.; et al. Distinguishing features of long COVID identified through immune profiling. Nature 2023, 623, 139–148. [Google Scholar] [CrossRef]
  132. Bote, M.E.; García, J.J.; Hinchado, M.D.; Ortega, E. Inflammatory/stress feedback dysregulation in women with fibromyalgia. Neuroimmunomodulation 2012, 19, 343–351. [Google Scholar] [CrossRef]
  133. Hernandez, M.E.; Becerril, E.; Perez, M.; Leff, P.; Anton, B.; Estrada, S.; Estrada, I.; Sarasa, M.; Serrano, E.; Pavon, L. Proinflammatory cytokine levels in fibromyalgia patients are independent of body mass index. BMC Res. Notes 2010, 3, 156. [Google Scholar] [CrossRef]
  134. Bazzichi, L.; Rossi, A.; Massimetti, G.; Giannaccini, G.; Giuliano, T.; De Feo, F.; Ciapparelli, A.; Dell’Osso, L.; Bombardieri, S. Cytokine patterns in fibromyalgia and their correlation with clinical manifestations. Clin. Exp. Rheumatol. 2007, 25, 225–230. [Google Scholar]
  135. Ranzolin, A.; Duarte, A.L.; Bredemeier, M.; da Costa Neto, C.A.; Ascoli, B.M.; Wollenhaupt-Aguiar, B.; Kapczinski, F.; Xavier, R.M. Evaluation of cytokines, oxidative stress markers and brain-derived neurotrophic factor in patients with fibromyalgia—A controlled cross-sectional study. Cytokine 2016, 84, 25–28. [Google Scholar] [CrossRef] [PubMed]
  136. Pernambuco, A.P.; Schetino, L.P.; Alvim, C.C.; Murad, C.M.; Viana, R.S.; Carvalho, L.S.; Reis, D. Increased levels of IL-17A in patients with fibromyalgia. Clin. Exp. Rheumatol. 2013, 31 (Suppl. S79), S60–S63. [Google Scholar] [PubMed]
  137. Ernberg, M.; Christidis, N.; Ghafouri, B.; Bileviciute-Ljungar, I.; Löfgren, M.; Bjersing, J.; Palstam, A.; Larsson, A.; Mannerkorpi, K.; Gerdle, B.; et al. Plasma Cytokine Levels in Fibromyalgia and Their Response to 15 Weeks of Progressive Resistance Exercise or Relaxation Therapy. Mediators Inflamm. 2018, 2018, 3985154. [Google Scholar] [CrossRef] [PubMed]
  138. Topal, G.; Donmez, A.; Doğan, B.S.; Kucur, M.; Cengiz, D.T.; Berkoz, F.B.; Erdogan, N. Asymmetric dimethylarginine (ADMA) levels are increased in patients with fibromyalgia: Correlation with tumor necrosis factor-α (TNF-α) and 8-iso-prostaglandin F(2α) (8-iso-PGF(2α)). Clin. Biochem. 2011, 44, 364–367. [Google Scholar] [CrossRef]
  139. Tsilioni, I.; Russell, I.J.; Stewart, J.M.; Gleason, R.M.; Theoharides, T.C. Neuropeptides CRH, SP, HK-1, and Inflammatory Cytokines IL-6 and TNF Are Increased in Serum of Patients with Fibromyalgia Syndrome, Implicating Mast Cells. J. Pharmacol. Exp. Ther. 2016, 356, 664–672. [Google Scholar] [CrossRef]
  140. Wang, H.; Moser, M.; Schiltenwolf, M.; Buchner, M. Circulating cytokine levels compared to pain in patients with fibromyalgia—A prospective longitudinal study over 6 months. J. Rheumatol. 2008, 35, 1366–1370. [Google Scholar]
  141. Ghizal, F.; Das, S.K.; Verma, N.; Mahdi, A.A. Evaluating relationship in cytokines level, Fibromyalgia Impact Questionnaire and Body Mass Index in women with Fibromyalgia syndrome. J. Back. Musculoskelet. Rehabil. 2016, 29, 145–149. [Google Scholar] [CrossRef]
  142. Mendieta, D.; De la Cruz-Aguilera, D.L.; Barrera-Villalpando, M.I.; Becerril-Villanueva, E.; Arreola, R.; Hernández-Ferreira, E.; Pérez-Tapia, S.M.; Pérez-Sánchez, G.; Garcés-Alvarez, M.E.; Aguirre-Cruz, L.; et al. IL-8 and IL-6 primarily mediate the inflammatory response in fibromyalgia patients. J. Neuroimmunol. 2016, 290, 22–25. [Google Scholar] [CrossRef]
  143. Wallace, D.J.; Linker-Israeli, M.; Hallegua, D.; Silverman, S.; Silver, D.; Weisman, M.H. Cytokines play an aetiopathogenetic role in fibromyalgia: A hypothesis and pilot study. Rheumatology 2001, 40, 743–749. [Google Scholar] [CrossRef]
  144. Montesino-Goicolea, S.; Sinha, P.; Huo, Z.; Rani, A.; Foster, T.C.; Cruz-Almeida, Y. Enrichment of genomic pathways based on differential DNA methylation profiles associated with chronic musculoskeletal pain in older adults: An exploratory study. Mol. Pain. 2020, 16, 1744806920966902. [Google Scholar] [CrossRef]
  145. Chae, S.C.; Shim, S.C.; Chung, H.T. Association of TBX21 polymorphisms in a Korean population with rheumatoid arthritis. Exp. Mol. Med. 2009, 41, 33–41. [Google Scholar] [CrossRef] [PubMed]
  146. Koetz, K.; Bryl, E.; Spickschen, K.; O’Fallon, W.M.; Goronzy, J.J.; Weyand, C.M. T cell homeostasis in patients with rheumatoid arthritis. Proc. Natl. Acad. Sci. USA 2000, 97, 9203–9208. [Google Scholar] [CrossRef]
  147. Schönland, S.O.; Lopez, C.; Widmann, T.; Zimmer, J.; Bryl, E.; Goronzy, J.J.; Weyand, C.M. Premature telomeric loss in rheumatoid arthritis is genetically determined and involves both myeloid and lymphoid cell lineages. Proc. Natl. Acad. Sci. USA 2003, 100, 13471–13476. [Google Scholar] [CrossRef]
  148. Pedersen-Lane, J.H.; Zurier, R.B.; Lawrence, D.A. Analysis of the thiol status of peripheral blood leukocytes in rheumatoid arthritis patients. J. Leukoc. Biol. 2007, 81, 934–941. [Google Scholar] [CrossRef] [PubMed]
  149. Fasth, A.E.; Cao, D.; van Vollenhoven, R.; Trollmo, C.; Malmström, V. CD28nullCD4+ T cells—characterization of an effector memory T-cell population in patients with rheumatoid arthritis. Scand. J. Immunol. 2004, 60, 199–208. [Google Scholar] [CrossRef] [PubMed]
  150. Chalan, P.; van den Berg, A.; Kroesen, B.J.; Brouwer, L.; Boots, A. Rheumatoid Arthritis, Immunosenescence and the Hallmarks of Aging. Curr. Aging Sci. 2015, 8, 131–146. [Google Scholar] [CrossRef]
  151. Li, S.; Wan, J.; Anderson, W.; Sun, H.; Zhang, H.; Peng, X.; Yu, Z.; Wang, T.; Yan, X.; Smith, W. Downregulation of IL-10 secretion by Treg cells in osteoarthritis is associated with a reduction in Tim-3 expression. Biomed. Pharmacother. 2016, 79, 159–165. [Google Scholar] [CrossRef]
  152. Montesino-Goicolea, S.; Meng, L.; Rani, A.; Huo, Z.; Foster, T.C.; Fillingim, R.B.; Cruz-Almeida, Y. Enrichment of genomic pathways based on differential DNA methylation profiles associated with knee osteoarthritis pain. Neurobiol. Pain. 2022, 12, 100107. [Google Scholar] [CrossRef]
  153. Wang, Y.; Wang, J.; Meng, J.; Jiang, H.; Zhao, J.; Qian, H.; Chen, T. Epigenetic Modification Mediates the Increase of LAG-3(+) T Cells in Chronic Osteomyelitis. Inflammation 2017, 40, 414–421. [Google Scholar] [CrossRef] [PubMed]
  154. Surendar, J.; Hackenberg, R.K.; Schmitt-Sánchez, F.; Ossendorff, R.; Welle, K.; Stoffel-Wagner, B.; Sage, P.T.; Burger, C.; Wirtz, D.C.; Strauss, A.C.; et al. Osteomyelitis is associated with increased anti-inflammatory response and immune exhaustion. Front. Immunol. 2024, 15, 1396592. [Google Scholar] [CrossRef] [PubMed]
  155. O’Brien, J.A.; McGuire, H.M.; Shinko, D.; Fazekas de St Groth, B.; Russo, M.A.; Bailey, D.; Santarelli, D.M.; Wynne, K.; Austin, P.J. T lymphocyte and monocyte subsets are dysregulated in type 1 diabetes patients with peripheral neuropathic pain. Brain Behav. Immun. Health 2021, 15, 100283. [Google Scholar] [CrossRef] [PubMed]
  156. Peng, Q.; Guo, X.; Luo, Y.; Wang, G.; Zhong, L.; Zhu, J.; Li, Y.; Zeng, X.; Feng, Z. Dynamic Immune Landscape and VZV-Specific T Cell Responses in Patients With Herpes Zoster and Postherpetic Neuralgia. Front. Immunol. 2022, 13, 887892. [Google Scholar] [CrossRef]
  157. Li, W.; Pucka, A.Q.; Houran, L.; Huang, X.; Debats, C.; Reyes, B.A.; O’Brien, A.R.; Yu, Q.; Wang, Y. Soluble immune checkpoints are dysregulated in patients with sickle cell disease and correlate with inflammatory mediators, autoantibodies, immune cell profiles, and clinical outcomes. medRxiv 2025. [Google Scholar] [CrossRef]
  158. Brandow, A.M.; Liem, R.I. Advances in the diagnosis and treatment of sickle cell disease. J. Hematol. Oncol. 2022, 15, 20. [Google Scholar] [CrossRef]
  159. Wanderley, C.W.S.; Maganin, A.G.M.; Adjafre, B.; Mendes, A.S.; Silva, C.E.A.; Quadros, A.U.; Luiz, J.P.M.; Silva, C.M.S.; Silva, N.R.; Oliveira, F.F.B.; et al. PD-1/PD-L1 Inhibition Enhances Chemotherapy-Induced Neuropathic Pain by Suppressing Neuroimmune Antinociceptive Signaling. Cancer Immunol. Res. 2022, 10, 1299–1308. [Google Scholar] [CrossRef]
  160. Deng, D.; Zhang, T.; Ma, L.; Zhao, W.; Huang, S.; Wang, K.; Shu, S.; Chen, X. PD-L1/PD-1 pathway: A potential neuroimmune target for pain relief. Cell Biosci. 2024, 14, 51. [Google Scholar] [CrossRef]
  161. Chaib, S.; Tchkonia, T.; Kirkland, J.L. Cellular senescence and senolytics: The path to the clinic. Nat. Med. 2022, 28, 1556–1568. [Google Scholar] [CrossRef]
  162. Mannarino, M.; Cherif, H.; Ghazizadeh, S.; Martinez, O.W.; Sheng, K.; Cousineau, E.; Lee, S.; Millecamps, M.; Gao, C.; Gilbert, A.; et al. Senolytic treatment for low back pain. Sci. Adv. 2025, 11, eadr1719. [Google Scholar] [CrossRef]
  163. Techameena, P.; Feng, X.; Zhang, K.; Hadjab, S. The single-cell transcriptomic atlas iPain identifies senescence of nociceptors as a therapeutical target for chronic pain treatment. Nat. Commun. 2024, 15, 8585. [Google Scholar] [CrossRef]
  164. Du, J.; Cheng, N.; Deng, Y.; Xiang, P.; Liang, J.; Zhang, Z.; Hei, Z.; Li, X. Astrocyte senescence-like response related to peripheral nerve injury-induced neuropathic pain. Cell Mol. Biol. Lett. 2023, 28, 65. [Google Scholar] [CrossRef]
  165. Borgonetti, V.; Galeotti, N. Rosmarinic Acid Reduces Microglia Senescence: A Novel Therapeutic Approach for the Management of Neuropathic Pain Symptoms. Biomedicines 2022, 10, 1468. [Google Scholar] [CrossRef] [PubMed]
  166. Jeon, O.H.; Kim, C.; Laberge, R.M.; Demaria, M.; Rathod, S.; Vasserot, A.P.; Chung, J.W.; Kim, D.H.; Poon, Y.; David, N.; et al. Local clearance of senescent cells attenuates the development of post-traumatic osteoarthritis and creates a pro-regenerative environment. Nat. Med. 2017, 23, 775–781. [Google Scholar] [CrossRef] [PubMed]
  167. Ansari, M.M.; Ghosh, M.; Lee, D.S.; Son, Y.O. Senolytic therapeutics: An emerging treatment modality for osteoarthritis. Ageing Res. Rev. 2024, 96, 102275. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Associations between lifestyle factors and immune dysfunction interpreted in the context of T cell exhaustion and senescence. By looking at receptor expression, telomere length, effector proteins, and signalling molecules, signs of T cell exhaustion and senescence in human cohorts are summarized for sleep disturbances, psychological stress, physical activity, and healthy diet. Abbreviations: CD28/57 = cluster of differentiation 28/57; cGAS = cyclic GMP-AMP synthase; KLRG-1 = Killer cell lectin-like receptor G1; NF-κB = nuclear factor kappa B; PD-1 = programmed cell death protein 1; p16INK4a = cyclin-dependent kinase inhibitor 2A; TL = telomere length; = increased; = decreased. Created in BioRender. Buntinx, Y. (2025) https://BioRender.com/hiv36in (accessed on 12 November 2025).
Figure 1. Associations between lifestyle factors and immune dysfunction interpreted in the context of T cell exhaustion and senescence. By looking at receptor expression, telomere length, effector proteins, and signalling molecules, signs of T cell exhaustion and senescence in human cohorts are summarized for sleep disturbances, psychological stress, physical activity, and healthy diet. Abbreviations: CD28/57 = cluster of differentiation 28/57; cGAS = cyclic GMP-AMP synthase; KLRG-1 = Killer cell lectin-like receptor G1; NF-κB = nuclear factor kappa B; PD-1 = programmed cell death protein 1; p16INK4a = cyclin-dependent kinase inhibitor 2A; TL = telomere length; = increased; = decreased. Created in BioRender. Buntinx, Y. (2025) https://BioRender.com/hiv36in (accessed on 12 November 2025).
Biomolecules 15 01601 g001
Table 1. Key characteristics of T cell exhaustion and senescence.
Table 1. Key characteristics of T cell exhaustion and senescence.
T Cell ExhaustionT Cell Senescence
TriggerPersistent antigenic stimulationAgeing, cellular stress
Inhibitory receptors ⇑ PD-1, CTLA-4, BTLA, Tim-3, TIGIT, Gal-9, LAG-3 ⇑ CD57, KLRG-1, Tim-3, TIGIT
Stimulatory receptors-⇓ CD27, CD28
Effector proteins, chemokines, and signalling molecules ⇓ granzyme B, perforinSASP
⇓ granzyme B, perforin
⇑ cGAS, NF-κB, p16INK4a
Cytokines ⇓ TNF-α, IFN-γ, IL-2
⇑ IL-10
⇑ TNF-α, IFN-γ, IL-6, IL-10
Transcription factorsT-bet, Eomes, TCF-1-
Telomere length-
Reversible?YesNo
Abbreviations: BTLA = B- and T-lymphocyte attenuator; CD27 = cluster of differentiation 27; CD28 = cluster of differentiation 28; CD57 = cluster of differentiation 57; cGAS = cyclic GMP-AMP synthase; CTLA-4 = cytotoxic T-lymphocyte-associated antigen 4; Eomes = Eomesodermin; Gal-9 = galectin-9; IFN-γ = interferon gamma; IL-2 = interleukin 2; IL-6 = interleukin 6; IL-10 = interleukin 10; KLRG-1; Killer cell lectin-like receptor G1; LAG-3 = lymphocyte-activation gene 3; NF-κB = nuclear factor kappa B; PD-1 = programmed cell death protein 1; p16INK4a = cyclin-dependent kinase inhibitor 2A; SASP = senescence-associated secretory phenotype; T-bet = T-box expressed in T cells; TCF-1 = T cell factor 1; TIGIT = T-cell immunoreceptor with Ig and ITIM domains; Tim-3 = T-cell immunoglobulin and mucin domain 3; TNF-α = tumour necrosis factor alpha; ⇑ = increased; ⇓ = decreased.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Buntinx, Y.; Hendrix, J.; Wyns, A.; Van Campenhout, J.; Xiong, H.-Y.; Laeremans, T.; Cuesta-Sancho, S.; Aerts, J.L.; Nijs, J.; Polli, A. Exploring Chronic Pain, Immune Dysfunction and Lifestyle: A Focus on T Cell Exhaustion and Senescence. Biomolecules 2025, 15, 1601. https://doi.org/10.3390/biom15111601

AMA Style

Buntinx Y, Hendrix J, Wyns A, Van Campenhout J, Xiong H-Y, Laeremans T, Cuesta-Sancho S, Aerts JL, Nijs J, Polli A. Exploring Chronic Pain, Immune Dysfunction and Lifestyle: A Focus on T Cell Exhaustion and Senescence. Biomolecules. 2025; 15(11):1601. https://doi.org/10.3390/biom15111601

Chicago/Turabian Style

Buntinx, Yanthe, Jolien Hendrix, Arne Wyns, Jente Van Campenhout, Huan-Yu Xiong, Thessa Laeremans, Sara Cuesta-Sancho, Joeri L. Aerts, Jo Nijs, and Andrea Polli. 2025. "Exploring Chronic Pain, Immune Dysfunction and Lifestyle: A Focus on T Cell Exhaustion and Senescence" Biomolecules 15, no. 11: 1601. https://doi.org/10.3390/biom15111601

APA Style

Buntinx, Y., Hendrix, J., Wyns, A., Van Campenhout, J., Xiong, H.-Y., Laeremans, T., Cuesta-Sancho, S., Aerts, J. L., Nijs, J., & Polli, A. (2025). Exploring Chronic Pain, Immune Dysfunction and Lifestyle: A Focus on T Cell Exhaustion and Senescence. Biomolecules, 15(11), 1601. https://doi.org/10.3390/biom15111601

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop