Back to Journals » Clinical Interventions in Aging » Volume 20

T Cell Aging: An Important Target for Perioperative Immunomodulation

Authors Lan H, Xu S, Li H , Guo R , Feng Z, Wang Y 

Received 25 January 2025

Accepted for publication 19 April 2025

Published 1 May 2025 Volume 2025:20 Pages 537—557

DOI https://doi.org/10.2147/CIA.S519438

Checked for plagiarism Yes

Review by Single anonymous peer review

Peer reviewer comments 2

Editor who approved publication: Prof. Dr. Nandu Goswami



Haoning Lan, Songchao Xu, Huili Li, Ruijuan Guo, Zhong Feng, Yun Wang

Department of Anaesthesiology, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, People’s Republic of China

Correspondence: Yun Wang, Department of Anesthesiology, Beijing Friendship Hospital, Capital Medical University, No. 95, Yong’ an Road, Beijing, 100050, People’s Republic of China, Tel +86-10-63133069, Email [email protected]

Abstract: Although T cells are crucially involved in maintaining immune function, their roles change with age. Furthermore, T cell aging has a unique onset and progression mechanism and several clinical indicators have been developed to detect it. Moreover, perioperative pain and stressful stimuli could affect the body’s immune status, influencing patients’ recovery. This article examines how preoperative and intraoperative complications influence T cell aging. These factors include conditions such as hypertension, diabetes, acute respiratory distress syndrome, hypoxemia, depression, pain, obesity, neurologic diseases, tumors, autoimmune diseases, as well as aspects like anesthetic modalities, types of surgery, and medications. This analysis could help identify groups at a high risk of perioperative T cell aging. For example, elderly cancer patients with multiple chronic diseases may be the most affected by T cell aging. We also discuss the effects of T cell aging on postoperative phenomena such as neurological dysfunction and recovery quality. Based on insights from this discussion, we deduced that prehabilitation, pharmacological treatment, and adoptive neuro-immunotherapy could modulate T cell aging in the perioperative period, thus improving clinical prognosis.

Keywords: perioperative period, T cell aging, immunomodulation, CD57, terminal effector memory T cells, senescence-associated secretory phenotype

Invasive perioperative procedures often trigger an inflammatory response in patients, stimulating immune regulatory mechanisms in their bodies. Patients’ immune function gradually declines as they age due to the degeneration of immune organs, chronic infections and inflammatory senescence, mitochondrial dysfunction, protein homeostasis imbalance, and epigenetic alterations.1 In addition to negatively impacting vital organ function in patients, imbalanced perioperative immune regulation could increase the incidence of postoperative infections and tumor metastasis recurrences.2

According to reports, appropriate anesthetic drugs and perioperative adjuvants could help improve innate and adaptive immune function. For instance, sevoflurane attenuated neutrophil recruitment and phagocytosis, whereas propofol exerted enhanced protective effects on neutrophil function. Furthermore, Non-Steroidal Anti-Inflammatory Drugs (NSAIDs), commonly used perioperative analgesics, upregulated Interleukin (IL)-10 (which has immunosuppressive properties) and downregulated IL-12 (which has anti-angiogenic properties).3 However, due to a lack of appropriate immunomodulatory indicators, clinicians can only administer drugs to modulate perioperative immune function based on clinical experience. Although IL-6, calcitonin, C-Reactive Protein (CRP), absolute leukocyte value, and T cell ratio have been established as common immunomodulation targets, infections and drugs easily interfere with them; hence they cannot be employed to accurately quantify the immune reserve function. In other words, they are not optimal indicators of perioperative immunomodulation, a phenomenon that necessitates additional research. In addition to mediating adaptive immunity, T cells are also an important component of both humoral and innate immunity. T cells play a role in maintaining the cell pool, frequently processing endogenous signals and exogenous stimuli, reaching a new steady state, and preserving memory of previous antigen encounters.4 Furthermore, the indicators of T cell could effectively reflect patients’ immune reserve function. With increasing age, factors such as immune organ damage, chronic infection, and inflammatory senescence pose significant challenges to the intricate and delicate T cell system. Epidemiological studies have shown that the immune system in elderly individuals is weakened. For example, for the 2017/2018 flu vaccine season, the clinical effectiveness of the flu vaccine in preventing influenza was 68% in children, 30–40% in middle-aged adults, and only 17% in the elderly.5,6 Although the overall immune capacity of the elderly population is diminished, there is considerable heterogeneity in immune function. Currently, there is a research gap in identifying elderly individuals with compromised immune function during the perioperative period and quantifying their residual immune capacity. T cell aging represents a promising approach to addressing this issue. First, as mentioned earlier, T cell function is an important indicator of immune function. Second, chronic diseases frequently observed in the elderly population are risk factors for T cell aging; thus, patients with multiple chronic conditions or tumors during the perioperative period are at high risk of immune decline. Third, evaluating T cell aging markers requires only peripheral blood extraction, making it highly practical for clinical applications. Fourth, T cell aging markers are quantifiable metrics that facilitate the assessment of immune function. Consequently, T cell aging is a vital indicator of perioperative immunity. Many prior investigations into T cell aging have relied on rodent models, but substantial disparities exist in immune patterns between rodents and humans. For example, in young adult rats, the majority of T cells originate from the thymus.7,8 In contrast, for a 20-year-old male, less than 20% of T cells are derived from the thymus, and for a 50-year-old male, this proportion decreases to less than 1%.9 Additionally, the lifespan of immature T cells in mice is limited to 6–11 weeks, whereas in humans, it extends to several years.8 Therefore, this article focuses on clinical evidence related to the perioperative period while appropriately integrating findings from animal research. This article reviews the mechanisms, phenotypes, and clinical detection indexes of T cell aging. It also summarizes the preoperative and intraoperative factors that could help identify people at high risk of T cell aging. Furthermore, it discusses the impacts of T cell aging in the postoperative period and outlines the potential for modulating T cell aging in the perioperative period, to improve clinical prognosis.

Mechanisms of T Cell Aging

Unique T immune-related factors [immune organ damage (thymic degeneration and peripheral homeostasis disorders), long-term infections, and inflammatory senescence] and universal senescence-associated factors [genomic instability, epigenetic alterations, mitochondrial dysfunction and Oxidative Stress (OS), and protein homeostasis imbalance] are some of the influencing factors of T cell aging (Figure 1).

Figure 1 Mechanisms and Phenotypes of T Cell Aging.

Abbreviations: TCR, T Cell Receptor; CD, Cluster of Differentiation.

Immune Organ Damage and T Cell Aging

The thymus, the central immune organ, is responsible for T cell differentiation, development, selection, and maturation. It also secretes cytokines and hormones, thus regulating T cell immune function in peripheral lymphoid organs. Notably, thymus atrophies and thymocytes are always downregulated post-puberty. Thymic degeneration could reduce the output of thymic naïve T cells,10 leading to a decrease in the number of peripheral naïve T cells and the diversity of T Cell Receptors (TCRs). This phenomenon implies that when the ability to fight against external pathogens is weakened, memory T cells will be upregulated as a means of compensating for the immune defense function.11 Furthermore, autoreactive T cell upregulation following negative selection disruption could imply an increased risk of autoimmune diseases.12 Besides the central immune organs, peripheral immune organs are also crucially involved in T cell aging. For instance, fibroblast reticulocytes in secondary lymphoid organs produce IL-7, which regulates T cell immunity. Additionally, lymph node-derived IL-7 deficiency somewhat mediated T cell apoptosis and depletion associated with lymph node fibrosis in individuals affected with the Human Immunodeficiency Virus (HIV) or the yellow fever virus.8 Moreover, the combined analysis of chromatin accessibility and transcriptome suggested that silencing of the IL7R gene and the IL-7 signaling pathway could serve as potential biomarkers of CD8+ T cell aging.13 Therefore, impaired pathways or structural alterations in secondary lymphoid organs could also affect T cell aging.

Chronic Infection, Inflammatory Senescence, and T Cell Aging

Cytomegalovirus (CMV) infection is a key pathogenic driver of CD8+ T cell aging.14 According to research, in the elderly, CMV-specific T cells account for a significant proportion of CD8+ T cells.15 Furthermore, giant cell-associated latent infections could trigger a dramatic expansion of T cell clones, seizing ecological niches and biasing the TCR repertoire, thus diminishing protection against other antigens.16 Additionally, with continued antigenic exposure and immune cell activation, Hepatitis B and C viruses, as well as HIV, could cause chronic infections. Through mechanisms such as topoisomerase inhibition or phosphatidylinositol 3-kinase/protein kinase B/mammalian target of rapamycin 1 (mTORC1) signaling activation, chronic infections could lead to T cell aging-related manifestations such as DNA damage, telomere truncation, and metabolic abnormalities.17,18

Inflammatory senescence, a low-grade chronic inflammatory state that develops with age, has been associated with various age-related defects, such as increased intestinal permeability, chronic infections, metabolic stress, and senescent cell accumulation.19 Multiple studies have established that inflammatory stimuli could induce naïve T cell differentiation. For instance, inflammatory factors IL-15 and IL-4 induced naïve T cells to differentiate into virtual memory T cells, which exhibited phenotypes comparable to those of antigen-induced memory T cells and exerted immune functions, such as bystander immunity.20 It is also noteworthy that inflammation-induced T cell differentiation is diffuse. Various inflammatory factors (such as IL-6, IL-8, and TNF) can induce CD8+ granzyme K+ T cells, and granzyme K could lead to the senescence of peripheral cells such as fibroblasts.21 Such inflammatory environments may also suppress the responsiveness of T cells to cytokines and other stimuli.22 Notably, the secretion of inflammatory cytokines [such as IL-1β, IL-6, and Tumor Necrosis Factor (TNF)], and chemokines (such as CCL2 and IL-8) by senescent T cells could further exacerbate inflammatory senescence.19

Mitochondrial Dysfunction, OS, and T Cell Aging

Low doses of rotenone could suppress the Respiratory Chain Complex I (NADH Oxidoreductase) and upregulate Terminal Effector Memory T Cells (TEMRA Cells),23 thus inhibiting mitochondrial function. This phenomenon highlights the potential direct effects of mitochondrial dysfunction on the proportion of senescent T cell subsets. The characteristic features of age-related mitochondrial dysfunction include mitochondrial DNA mutations, decreased respiratory capacity, decreased mitochondrial membrane potential, and increased Reactive Oxygen Species (ROS) production.24 Besides inducing progressive changes in mitochondrial morphology, activity, and signaling function, these factors could also cause telomere wear and reduced enzyme repair activity, leading to decreased telomere length in T cells.25 Apart from decreased telomere length, massive inflammatory factor secretion (a process mitochondria significantly mediate) is the other key hallmark of senescent T cells. In aged T cells, the altered membrane permeability of damaged mitochondria could promote the release of mitochondrial damage-associated molecular patterns, such as mitochondrial DNA, cardiolipin, or second mitochondria-derived activator of caspase. Mitochondrial DNA and cardiolipin could further activate inflammatory pathways such as the cyclic guanosine monophosphate -adenosine monophosphate synthase - stimulator of interferon gene pathway or the assembly of inflammasomes, promoting the release of Interferon Type I (IFN-I) molecules, TNF, IL-18, and IL-1β. On the other hand, second mitochondria-derived activator of caspase could switch the nonclassical nuclear factor-kappaB (NF-κB) pathway to the classical NF-κB pathway.26 Notably, the genetic deletion of Mpc1 (a vital subunit of the mitochondrial pyruvate carrier) could intensify aerobic glycolysis and reduce Oxidative Phosphorylation (OPHOS), affecting thymocyte differentiation and ultimately promoting the expansion of hyperinflammatory T cells.27

Protein Imbalance and T Cell Aging

Proteasome and autophagy primarily mediate the degradation of misfolded or senescent proteins, and defects in either process could lead to T cell aging. Notably, elderly subjects exhibited calpain and tripeptidyl peptidase II downregulation in the proteasome pathway. Furthermore, protease function impairment, either by protease inhibitors or the specific knockdown of the mouse T cell Proteasome Subunit Non-ATPase-dependent Regulatory Granule 13 (PRG-13), increased in aged CD4+ T cells in mice.28 Moreover, defects in the tripeptidyl peptidase II gene (Evan’s syndrome) have been associated with premature CD8+ T cell immunosenescence,29 a phenomenon that supports the tight link between proteasome activity and T cell aging. Furthermore, the autophagy blockade in the autophagy pathway could lead to the accumulation of depolarized mitochondria in CD8+ T cells, limiting tissue-resident function.30 Additionally, during senescence, mitochondrial dysfunction could persist due to autophagy impairment in CD4+ T lymphocytes.31

Genetic and Epigenetic Alterations and T Cell Aging

Chromatin accessibility, DNA methylation levels, and microRNAs have all been established to influence the onset of T cell aging. According to research, chromatin accessibility is significantly altered during CD4+ and CD8+ T cell aging, resulting in the disruption of the IL-7R signaling pathway.13 Furthermore, double-strand break repair nuclease meiotic recombination 11 homolog A (MRE11A) downregulation directly affects mitotic heterochromatin unraveling and leads to telomere damage, as well as increased levels of the aging markers Cyclin-Dependent Kinase Inhibitor 1 (CDKN1) and CDKN2A.32 Notably, histone acetylation has also been established to influence chromatin accessibility. At the single-cell level, aged T cells exhibited increased histone acetylation heterogeneity, implying that altered chromatin accessibility could lead to functional and differentiation diversity in aged T cells.33 It is also noteworthy that DNA methylation has recently gained increasing attention. Senescent CD8+ TEMRA cells exhibited significantly altered DNA methylation levels, Furthermore, the methylation levels of immune senescence-related +6 off methylation sites correlated positively with the degree of immune senescence.34 Finally, microRNAs were found to bind to messenger RNA molecules, inhibiting their translation or promoting their degradation, thus making them crucial regulators of immune function. Notably, miR-181a and miR-155 have been specifically highlighted as hot spots in the study of T cell aging. Specific deletion of miR-181a from mature T lymphocytes in a mouse model resulted in multiple defects comparable to human T cell aging, including upregulation of the negative regulator Dual-Specificity Phosphatase 6 (DUSP6)- Silent Mating Type Information Regulation 2 homolog 1 (SITR1) and impairment of antiviral CD8+ T cell responses.35 On the other hand, miR-155 was linked with an age-related upregulation of Toll-Like Receptor 5 (TLR5), leading to an enhanced inflammatory response in aged T cells.36

T Cell Aging Phenotype

In the bone marrow, Bone Marrow Pluripotent Hematopoietic Stem Cells (BM-HSCs) differentiate into lymphoid progenitor cells (also known as lymphoblasts), which enter the thymus through blood circulation and mature into initial T cells. The T cells then enter peripheral lymphoid organs and differentiate into effector T cells and memory T cells after the immune response. Notably, T cell aging entails dysfunction and subpopulation imbalance at the individual and overall cell levels respectively, with the former referred to as “qualitative aging” and the latter as “quantitative aging”. Notably, the two aging mechanisms work synergistically to affect immune function in individuals (Figure 1).

Qualitative T Cell Aging

Qualitative aging, a manifestation of cellular senescence at an individual cellular level, is often evidenced by T cell senescence. Senescence manifestations in T cells include low telomerase activity, short telomeres, DNA damage, mitochondrial dysfunction, enhanced classical senescence hallmarks (eg, β-galactosidase), increased secretion of pro-inflammatory factors (eg, TNF and IFN -γ), decreased cell proliferation, and heightened cytotoxicity.37 Furthermore, compared to immune cells, aged T cells exhibit unique senescence characteristics, including loss of TCR co-stimulatory receptors [eg, Cluster of Differentiation 27 (CD27) and CD28] and upregulation of terminal differentiation markers [CD57 and Killer Cell Lectin-like Receptor Subfamily G1 (KLRG1), Table 1].38

Table 1 Changes in CD Markers, Functions, Affected Signaling Pathways, and Transcription Factors of T Cells at Different Stages

Various T cell subpopulations differ in senescence sensitivity. For instance, CD8+ T cells are more sensitive to senescence than CD4+ T cells. Furthermore, CD8+ terminally differentiated TEMRAs accumulate faster than CD4+ cells. This phenomenon could be attributed to CD8+ TEMRA cells being more susceptible to mitochondrial decline,23 lower homeostatic naïve cell proliferation rates,46 increased chromatin accessibility alterations,13 and higher Tribbles Homolog 2 (TRIB2) abundance.47 Furthermore, regulatory T cells (Tregs) are more severely senescent than effector T cells. Notably, DNA Damage-Binding Protein 1 (DDB1) and Cullin4- Associated Factor 1 (CAF1) are essential for Treg senescence inhibition via Glutathione-S-Transferase P (GSTP1)-regulated ROS production.48

Although T cell senescence is closely linked to T cell exhaustion, the two phenomena are distinct. Whereas senescent T cells undergo an irreversible cell cycle arrest, the reduced proliferative capacity of exhausted T cells is a reversible functional state disorder.49 The expression profiles of the two phenomena are even more different. While senescent T cells exhibit CD28 and CD27 downregulation and CD57 and inflammatory factor upregulation, exhausted T cells show higher levels of inhibitory receptors [eg, Programmed Death 1 (PD-1), Cytotoxic T Lymphocyte-Associated Antigen 4 (CTLA4), T-cell immunoglobulin, and T Cell Membrane Protein 3(TIM3)] and lower levels of inflammatory factors (eg, IL-2 and TNF).34,38

Reduced T cell plasticity is the other important marker of T cell senescence. Besides their great proliferative potential and plasticity, young naïve CD4+ T cells could differentiate into different lineages relatively freely. On the other hand, elderly naïve CD4+ T cells have a specific differentiation tendency. Elderly naïve CD4+ T cells exhibit Transforming Growth Factor-β Receptor 3 (TGF-βR3) upregulation; hence, their responsiveness to TGF-β increases with age, which, in turn, favors Th9 differentiation. Furthermore, aging naïve CD4+ T cells respond to TCR stimulation, leading to Basic Leucine Zipper ATF-Like 2 (BATF2) and Interferon Regulatory Factor 4 (IRF4) upregulation and downregulation of inhibitors of DNA binding 3 and B Cell Lymphoma 6 (BCL6), all of which promote Th9 differentiation.50 Aged naïve CD4+ T cells also exhibit CD39 upregulation, which inhibits follicular helper T cell production.51 Furthermore, due to an increase in miR-21 levels, aged naïve CD4+ T cells are more likely to differentiate into inflammatory effector cells rather than follicular helper and memory T cells.52 In addition to the differentiation capacity of naïve cells, T cell plasticity also entails an alteration in the functional efficiency of T cells. In aged mice, Tregs do not suppress conventional T cell function as efficiently as young Tregs.48 Additionally, follicular helper T cells (T follicular helper) are inefficient in aged mice due to pathway defects.53

Quantitative T Cell Aging

One of the most striking phenotypes of quantitative T cell aging is an imbalance in the ratio of naïve T cells to memory T cells. On the one hand, naïve T cell pool maintenance depends on thymic output. Thymic degeneration, a phenomenon that occurs post-puberty, could lead to a decrease in the output of naïve T cells, resulting in a leaky naïve T cell pool.4 On the other hand, naïve T cell pool maintenance also depends on homeostatic peripheral naïve T cell proliferation. Notably, naïve T-cell pool maintenance mechanisms differ significantly between humans and mice, with steady-state peripheral naïve T cell proliferation and thymic output dominating in humans and mice, respectively.46 Furthermore, continuous antigenic stimulation could induce the conversion of naïve T cells into memory T cells; hence, the proportion of naïve T cells might gradually decrease with age. Besides antigenic stimulation, the peripheral lymphatic environment also influences the proportion of naïve T cells.

Perfect T cell immunity necessitates not just a sufficient number of T cells but also a diverse TCR pool to respond to new antigens. Both cohort and cross-sectional studies reported a decrease in TCR pool diversity with age54 and a high degree of convergence in CD4+ TCRs in the elderly.55 These phenomena could be attributed to several factors. First, naïve T cells with neo-specific TCRs originate from the thymus and thymic degeneration correlates with aging. As a result, the decrease in the output of naïve T cells correlates with neo-specific TCRs from the thymus, potentially leading to decreased TCR diversity. Second, long-term chronic infections (such as CMV and influenza virus infections) could result in a disproportionate expansion of specific T cells, altering the proportion of cells with different TCR populations.54 Third, due to the loss of wingless/β-catenin signaling, the human CD4+ stem cell memory T lymphocyte pool might get depleted with age.56 Reduced TCR diversity in naïve T cells is not entirely random and TCRs with a higher affinity for both self- and non-self-peptide-MHC complexes are more readily eliminated, probably because they are more readily converted to the “memory” phenotype.55

T Cell Aging Indicators Commonly Used in Clinical Trials

Flow cytometry is often used in clinical research to categorize T cell subpopulations. Moreover, this approach is often used to quantify the degree of T cell aging in patients, primarily through two methods. First, based on the C-C motif Chemokine Receptor 7 (CCR 7) and CD45RA, T cells are classified into naïve cells, central memory cells, effector memory T cells (TEM), and TEMRA cells. In this method, patients are often evaluated for T cell aging based on changes in the ratio of the four types of cells or by focusing specifically on the ratio of TEMRA cells. Notably, cytokines such as CD31, CD103, and Protein Tyrosine Kinase 7 (PTK7) are sometimes added to naïve cells to assess thymic function.40 Second, T-cell activation markers (CD28, CD38, and Human Leukocyte Antigen (HLA)-DR, among others) and T-cell senescence markers (CD57) are examined. Earlier clinical studies often employed CD28 or CD57+ T cells alone to define T cell aging, but in recent years, T cell aging is mostly defined using CD28 CD57+ T cells.57 To ensure experimental reliability, these two cell groups are often evaluated simultaneously. Nonetheless, some differences have been noted between the two methods, with the former and latter tending to reflect “T cell quantitative aging” and “T-cell qualitative aging”, respectively. There is a also difference in the reversibility of the two, with the cell cycles of TEMRA and CD28 CD57+ T cells being reversible and irreversible, respectively. There were also differences in the phenotypes of the two methods. The stress-inducible protein sestrins in TEMRA provide a scaffold for Mitogen-Activated Protein Kinase (MAPK) pathway activation, enhancing the downstream activation of extracellular signal-regulated kinase, MAPK, p38, and c-Jun N-Kinase (JNK), while inhibiting mTORC1. For CD28 CD57+ T cells, mTORC1, NF-κB, p38, and CCAAT/Enhancer Binding Protein β (C/EBPβ) are upregulated due to mitochondrial dysfunction and other factors, which mediate the differentiation of the senescence-associated secretory phenotype.58 Besides cell sorting, cytokines are sometimes employed in testing the functional level of T cells. Specifically, they help in assessing the telomere length, telomerase reverse transcriptase expression in T cells, or cellular senescence markers, such as p16INK4a and p21CIP1/Waf1.59 For the rigor of the test, CMV expression is always tested more often to further exclude the effect of chronic infections on T cell aging.

Preoperative and Intraoperative Factors and T Cell Aging

Preoperative [hypertension, Diabetes, Acute Respiratory Distress Syndrome (ARDS), hypoxemia, depression, pain, obesity, tumors, and autoimmune diseases] and intraoperative (anesthesia methods, surgery types, and medications) factors can affect mitochondrial function and create an inflammatory environment, increasing the risk of T cell aging. Although the evaluation of these factors could help identify patient groups at high risk of T-cell senescence and allow for prompt intervention to avoid a poor prognosis, there are no clinical prediction models for quantifying the risk of T cell aging more accurately (Figure 2).

Figure 2 Preoperative and Intraoperative Factors Increasing the Risk of T Cell Aging.

Abbreviations: CD, Cluster of Differentiation; CCR, C-C motif Chemokine Receptor; ROS, Reactive Oxygen Species; PD-1, Programmed Death 1; CMV, Cytomegalovirus; TEMRA, Terminal Effector Memory T Cells; MS, Multiple Sclerosis; KLRG1, Killer Cell Lectin-like Receptor Subfamily G1; AD, Alzheimer’s Disease; CBF, Cerebrospinal Fluid; PD, Parkinson’s Disease; IFN, Interferon; TNF, Tumor Necrosis Factor; RA, Rheumatoid arthritis; SLE, Systemic lupus erythematosus; IBD, Inflammatory bowel disease; TEM, Effector memory T cells; AAD, Acute Aortic Dissection.

Hypertension and T Cell Aging

Hypertension is among the most common comorbidities in the perioperative period. According to research, T-cell-specific AT1 receptor deficiency can exacerbate angiotensin II–induced renal injury, increasing the accumulation of pro-inflammatory Th1 cells and upregulating IFN -c and TNFα in the kidney.60 In hypertensive patients, T cells, especially CD8+ T cells, showed senescence-associated features such as CD28 CD57+, compared to their peers.61 Additionally, both cellular assays and mouse models demonstrated that T cell aging accelerated Ang II–induced cardiovascular and renal fibrosis and promoted the expression of inflammatory factors and superoxide production in the target organs.62 These findings suggest a tight link between T cell aging and both hypertension and damage to related organs. Moreover, although mice lacking T and B cells were not protected from Ang II–induced vascular abnormalities in a previous study, direct transfer of T cells restored vascular dysfunction.63 Furthermore, compared to young T cells, conditioned cultures of aged T cells stimulated inflammatory factor expression and OS in Ang II-treated renal epithelial cells, although these effects were attenuated after IFN -c neutralizing antibody treatment.62 Based on these research insights, targeting specific T cells could be a therapeutic avenue for managing elderly hypertension with comorbid end-organ dysfunction.

Diabetes and T Cell Aging

Diabetes is the other comorbidity common in the perioperative period. In previous research, insulin-resistant participants showed significantly more CD28 CD57+ senescent T cell subsets in CD4+ and CD8+ T cells than those with lower resistance values, as assessed by the Insulin Resistance (IR) homeostasis model. Furthermore, the abundance of senescent CD4+ and CD8+ T cells and the IR index correlated positively with the severity of liver fibrosis.64 Type 2 Diabetes (T2D) patients often have higher amounts of circulating aged T cells, a phenomenon that correlates with higher levels of systemic inflammation. This inflammatory milieu promotes the expression of a unique set of CCRs on aged T cells, particularly CCR2.65 Another clinical study reported that mitochondria from CD8+ T cells from T2D patients exhibited a higher oxidative capacity and mitochondrial ROS levels compared to age-matched controls. Furthermore, despite increased fatty acid uptake, T2D CD8+ TEMRA cells exhibited impaired fatty acid oxidation, lipid droplet accumulation, and decreased AMP-Activated Protein Kinase (AMPK) activity.66 These findings support the tight link between diabetes and T cell aging, with the latter being specifically linked with alterations in inflammatory responses and OS levels. Moreover, another clinical study involving pre-diabetic patients suggested that exercise stimulates naïve T cell production and mobilization, with differentiated TEMRA cells lost through apoptosis in the process.67 Additionally, the Rosuvastatin+ezetimibe combination therapy downregulated aged CD8+ T cells and increased the ratio of naïve to memory CD8+ T cells in diabetic patients. Notably, this treatment’s mechanism of action was not associated with improved lipid or glycated hemoglobin levels.68 Therefore, exercise, diet, and pharmacotherapy may be leveraged to prevent diabetes-associated T cell aging.

ARDS and T Cell Aging

Longitudinal studies involving patients with Corona Virus Disease 2019 (Covid-19)-related ARDS revealed that patients had a 2-3-fold increase in blood effector T cell expression and that most effector T-cells expressed PD-1 (associated with failure) and CD57 (associated with senescence). Furthermore, the patients showed a surge of pro-inflammatory cytokines, waves of Th1 and Th2 activation, and a 15-fold increase in γδ T-cell and proliferating NK-cell populations.69 It is also noteworthy that COVID-19-associated immune aging has been linked strongly with CMV infection. Compared to the healthy population, a previous study showed that patients with mild or asymptomatic COVID-19 in the unvaccinated and CMV-antibody-positive populations showed a significant upregulation of CD28 CD57+ Chemokine C-X3-C-Motif Ligand 1 (CX3CR1) in CD4+, CD8+, and γδ T-cell subsets. Notably, this variability was not observed in the CMV antibody-negative or vaccinated populations,70 probably because these two groups exhibited relatively weak immune activation and immune aging; hence, the small sample study could not yield positive results.

Hypoxemia and T Cell Aging

Short-term hypoxemia has been established to promote T cell aging. In previous research, the ratio of senescent (CD28 CD57+) and activated (CD62L CD11a+) cells in peripheral blood increased significantly after intense exercise (up to maximal oxygen uptake). Furthermore, lymphocyte surface thiols, intracellular total glutathione levels, and reduced glutathione all decreased. There was also an enhanced peroxide-mediated mitochondrial membrane potential reduction, cysteine protease 3/8/9 activation, Poly-ADP ribose polymerase cleavage, and phosphatidylserine exposure. Notably, these phenomena were not observed after moderate-intensity exercise (up to 50% of maximal oxygen uptake) but were observed after moderate exercise in a hypoxic environment, becoming even more pronounced with decreasing oxygen concentration.71 These findings collectively suggest that the hypoxic state increases the entry of senescent/activated forms of lymphocytes into peripheral blood while enhancing OS-induced lymphocyte apoptosis by decreasing cellular antioxidant levels during exercise. In another study, exercise to the point of volitional fatigue was followed by a 42.4% and 45.9% increase in senescent CD4+ and CD8+ T lymphocytes, respectively. Furthermore, males and less-exercised populations showed more pronounced levels of exercise-induced acute T cell aging.72 These findings suggest that during the perioperative period, especially in the anesthesia induction and maintenance phases, special attention should be paid to monitoring the blood gases of patient groups at a high risk of hypoxemia, including males, patients with airway difficulties, Chronic Obstructive Pulmonary Disease (COPD) patients, and debilitated patients, to reduce hypoxia-induced T cell aging and avoid related adverse outcomes.

Depression and Pain and T Cell Aging

According to reports, T cells are crucially involved in psychiatric regulation. In a previous study, compared to the healthy population, patients with major depression showed a higher proportion of TEMRA cells. Furthermore, patients with major depression showed a lower proportion of naïve T cells compared to the normal population. This phenomenon was more pronounced in the CMV-positive cohort, highlighting the potential link between depression and a more pronounced T cell aging.73 Similarly, in an Irritable Bowel Syndrome (IBS) patient cohort, the depressed group showed a significantly lower number of circulating T cells than non-depressed patients, implying that the co-morbid state of pain and depression had a more detrimental effect on immune status than the disease alone.74 The finding also suggested that patients with poor chronic pain control or psychiatric disorders such as depression were potential candidates for T cell aging, and that extra attention should be paid to such groups during the perioperative period.

Obesity and T Cell Aging

A high-fat diet will promote the accumulation of aging T cells (CD153+ PD-1+ CD44hi CD4+ T cells), and this subset will increase visceral adipose tissue inflammation by producing osteopontin.75 Obesity has been established to promote T cell senescence via several mechanisms. First, chronic stimulation by leptin and free fatty acids in the blood, repetitive antigenic stimulation, stress response, and hypoxia could all directly induce CD4+ T cell exhaustion.76 Second, obesity promotes thymus enlargement through growth factors such as the Insulin-like Growth Factor (IGF); however, this is only temporary as the thymus decline intensifies after 6–7 months, ultimately inducing T cell aging.77 Third, obesity induces OS and inflammation, leading to telomere shortening and ultimately T cell aging.78 Finally, obesity could trigger widespread gene expression changes in multiple organs79 and DNA methylation changes in blood leukocytes,80 leading to immune dysfunction and T cell aging. Notably, metabolic syndrome could further accelerate T cell aging in obese individuals. In previous research, obese patients with metabolic syndrome exhibited increased CD8+ T cell differentiation and shorter CD4+ T cell telomere length compared to those without metabolic syndrome.81 It is also noteworthy that surgical interventions could temporarily reverse accelerated T cell aging in cases of metabolic syndrome. Following bariatric surgery, the increased thymic output and accelerated T cell differentiation trend in obese patients would be suppressed, with patients with metabolic syndrome experiencing a more pronounced effect of the surgical intervention.81,82 This phenomenon suggests that the risky events associated with T cell aging in obese patients could be surgically prevented during the perioperative period, especially in those with metabolic syndrome.

Neurologic Diseases and T Cell Aging

In previous research, patients with multiple sclerosis exhibited features of T cell aging such as accelerated thymic degeneration and upregulation of CD4+ CD28 cells.83,84 Moreover, clinical studies demonstrated that senescence and systemic elevation of late memory T and B lymphocytes were some of the key manifestations in individuals with faster amyotrophic lateral sclerosis progression and medullary involvement.85

Notably, T cell aging has also been strongly associated with Alzheimer’s Disease (AD). In previous research, the percentage of naïve CD4+ T cells in the peripheral blood of AD patients decreased, whereas that of KLRG1-expressing terminally differentiated memory CD4+ T cells increased.86 Furthermore, another study revealed that compared to the normal population, AD patients exhibited higher levels of CD8+ TEMRA cells in the Cerebrospinal Fluid and blood.87 Additionally, AD patients showed a significantly shorter telomere length of peripheral blood T cells than the non-Alzheimer’s population. Moreover, the telomere length was found to correlate negatively with the serum TNF-α level, the proportion of CD8+ CD28 T cells, and the level of apoptosis.88 These findings collectively highlight the crucial involvement of T cell aging in various neurodegenerative diseases. It is also noteworthy that AD patients and the elderly often show the upregulation of A-β-specific T-cells.89 In another study, these cells penetrated the brain parenchyma and contributed to AD development in mice.90

There have also been some links between T cell aging and Parkinson’s Disease (PD). In a previous study, compared to the normal population, PD patients showed significantly lower levels of peripheral blood CD8+ TEMRA and CD8+ CD28 CD57+ T cells, significantly higher CD8+ CD28 expression, and lower expression levels of the cellular senescence marker p16. Furthermore, the two groups showed no significant differences in T cell telomere length, Telomerase Reverse Transcriptase (TERT) activity, or thymic migration.57,59 In another study, PD differed significantly from most of the aforementioned neurologic disorders regarding the state of T cell aging, a phenomenon attributable to the overreaction of the more active, less senescent T cells to PD-associated antigens, such as α-synuclein.91

Tumor and T Cell Aging

Numerous clinical trials have shown that the tumor microenvironment influences the distribution of T cell subsets, by increasing the proportions of CD8+ CD28 T cells and CD8+ CD57+ T cells in the blood circulation of patients with head and neck cancer, lung cancer, pleural mesothelioma, and lymphoma.92 This suggests that the tumor microenvironment may modulate the T cell aging. Meanwhile, the number of typical T-cell senescent subpopulations (CD8+ CD28 CD57+ T cells) in the circulation of lung cancer was reported to be highly elevated relative to levels in the normal population, and the proportion of T cell aging subpopulations in patients with the advanced stage was significantly higher compared with that of patients with early stage.93 Similarly, the expression of KLRG1 on CD4+ T cells increases with tumor progression in patients with renal clear cell carcinoma,94 further demonstrating that the tumor microenvironment influences T cell aging. Meanwhile, studies have demonstrated that chemotherapy increases the degree of T cell aging in tumor patients.95 Collectively, the aforementioned clinical trials have shown that the proportion of T cell aging is significantly higher in tumor patients, especially those with advanced tumors and following chemotherapy treatment. Therefore, researchers should formulate interventions during the perioperative period thereby improve T cell aging and prevent perioperative adverse events, ensuring good prognostic outcomes.

Autoimmune Diseases and T Cell Aging

The commonly reported autoimmune diseases include rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), and inflammatory bowel disease (IBD). A previous study demonstrated that the percentage of CD4+CD28 T cells was increased in the peripheral blood of RA patients, whereas CD4+CD28 T cells in the peripheral blood of RA patients generated more IFN-γ and TNF compared to regular CD4+CD28 T cells.96 Elsewhere, the CD4+CD28 T cells were not detected in the synovial fluid of RA patients, but KLRG1+ T cells (a typical T-cell senescence phenotype) were recorded in the synovial fluid of RA patients and spondylarthritis patients. This subset of T cells produces large amounts of IFN-γ and TNF-α.96,97 In patients with SLE, the number of CD4+CD28 T cells (a typical T-cell senescence phenotype) was significantly increased and the percentage of senescent T cells correlated significantly with SLE activity and disease severity.98 Similarly, high proportions of CD4+ CD28 T cells and CD8+ CD28 T cells were detected in patients with IBD, and the ratio of CD4+ CD28 T cells to CD8+ CD28 T cells serve as an important indicator of IBD severity.99 These studies provide evidence that autoimmune diseases influence T cell aging, and that the progression of autoimmune diseases will may influence T cell aging, necessitating careful preoperative operation.

Anesthesia Modality and T Cell Aging

Several clinical studies have shown that patients with colorectal cancer administered with a general anesthesia combined with epidural anesthesia compared to general anesthesia exhibit positive changes in NK cells, CD4+ cells, and CD4+/CD8+ ratio.100 This indicates that the mode of anesthesia has a directly effect on immune function. It can also alter the inflammatory milieu and trigger mitochondrial dysfunction. To prevent inflammation, general anesthesia combined with epidural anesthesia positively regulated IL-6, IL-8, IL-10, and TNF-α in patients with colorectal cancer.101,102 Modification to the inflammatory environment is accompanied by changes in the expression profile of healthy mesenchymal stem cells and genes influencing the survival of lymphocytes.103 Senescent mesenchymal stem cells that produce inflammatory factors impair the function and clonogenicity of young hematopoietic stem cells.104 It has been reported that general anesthesia lead to mitochondrial dysfunction by altering mitochondrial morphology and distribution, increasing mitochondrial ROS production, mitochondria-mediated regulation of cell death pathways, and mitochondria’s ability to clear misfolded proteins.105 As previously mentioned, mitochondrial dysfunction may promote T cell aging either directly or indirectly by inducing excessive production of ROS, affecting telomerase wear, and excessive inflammatory expansion. This demonstrates that mitochondria also serve as a mediator between anesthesia and T cell aging.

Type of Surgery and Medication and T Cell Aging

Studies have demonstrated potential association between the type of surgery and T cell aging. First, T cell aging is strongly associated with thymic hypoplasia, and preclinical surgical operations that remove the thymus may induce T cell aging, and hence, clinicians should pay attention to patients with a history of this surgery, and perform a long-term postoperative follow-up.106 In other studies, it was observed that sex steroid hormones mediate thymic degeneration by driving steroid signaling in thymic epithelial cells,107 and thus, additional attention should be paid to patients receiving long-term hormone therapy preoperatively. Second, patients with acute Stanford type-A aortic dissection exhibited reduced proportion of CD4+ naïve T and increased proportion of CD4+ TEMs on admission compared to healthy volunteers,108 suggesting that T cell aging may occur in all patients undergoing critical surgery and that additional clinical studies are needed to confirm the existence of T cell aging in patients undergoing other types of critical surgeries. As mentioned earlier, tumors is a high-risk factor for T cell aging, therefore, patients undergoing tumor-related surgery need to be monitored for potential T cell aging.

T Cell Aging and Postoperative Factors

T cell aging leads to poor prognosis by inducing perioperative neurologic dysfunction and impairing postoperative recovery. Therefore, additional clinical studies are needed to further explore the association of T cell aging with the remaining postoperative factors (Figure 3).

Figure 3 Postoperative Factors Associated with T Cell Aging.

Abbreviations: CD, Cluster of Differentiation; TEM, Effector memory T cells; KLRG1, Killer Cell Lectin-like Receptor Subfamily G1; PD-1, Programmed Death 1.

T Cell Aging and Postoperative Neurologic Dysfunction

Accumulating studies have demonstrated a link between T cell aging and postoperative cognitive dysfunction. In a model of neurocognitive impairment induced by internal fixation of tibial fracture in aged mice, circulating Tregs were increased accompanied by elevation of plasma inflammatory factor levels. Infusion of Tregs from young mice partially restored blood-brain barrier damage and alleviated postoperative cognitive dysfunction in aged mice. Compared with Tregs from young mice, differentially expressed genes were enriched in the TNF signaling pathway and the cytokine-cytokine receptor interaction pathway,109 implying that T cell aging may lead to postoperative cognitive dysfunction. T cell aging influences postoperative cognitive dysfunction via several mechanisms. First, a higher proportion of senescent cells in the brain increases the development of postoperative cognitive dysfunction. Aging CD8+ T cells present with features of elevated natural killer cell lectin-like receptor subfamily C member 1 and PD-1 and decreased perforin binding, which may potentially contribute to senescent cell escape.110–112 Senescent CD8+ cells are not only less capable of clearing senescent cells but also promoting the senescence of other brain cells via granzyme K and TNF-α, which may exacerbate the accumulation of senescent cells in the brain.21,113 Collectively, these findings indicate that long-term T cell aging increases the proportion of senescent cells in the brain, which in turn enhances the risk of postoperative cognitive dysfunction. Secondly, T cell aging can directly kill neural cells and senescent CD8+ T cells in the brain have also been shown to directly promote axonal and myelin degeneration via granzyme B and IFNγ-mediated microglia activation,114,115 and CD4+ CX3CR1+ T cells and CD8+ CX3CR1+ T cells. In addition, they are close to cleaved cysteinyl aspartate specific proteinase-3+ oligodendrocytes,116,117 indicating that aging T cells may directly lead to oligodendrocyte death and demyelination. Therefore, aged T cells kill neuronal cells and trigger degenerative brain changes enhancing the occurrence of postoperative delirium. Thirdly, inflammatory response may mediate the processes leading to T cell aging which impairs postoperative cognitive dysfunction. Brain inflammation has been associated with increased expression of the chemokine CX3C motif ligand 1 in the cerebrospinal fluid,116 and therefore, CD4+ CD28 T cells highly expressing CX3CR1 migrate to sites of inflammation in response to chemokine signals. Aged CD4+ T cells acquire an extreme pro-inflammatory phenotype, which upregulates IL17, IL9, IL1-β, IFN -γ, and the chemokine CX3CR1, while also downregulating CCR7.118 IL-17 triggers cognitive and synaptic deficits and damages the cerebral blood barrier.119,120 As a classic inflammatory mediator, IL1-β regulates neuronal survival and growth, and neuroplasticity.121 The levels of IL-9 were found to be negatively correlated with inflammatory activity, serving as an indicator of neurodegenerative and progressive disorders.122 Finally, in 5xFAD transgenic mice, CCR7 deletion caused deleterious neurovascular and microglia activation, thereby reducing lymphatic in-flow and cognitive deficits.123

T cell aging has been linked to occurrence of postoperative delirium. Firstly, senescent T cells are often accompanied by mitochondrial dysfunction in peripheral platelets, which serves as a predictor of postoperative delirium in elderly patients.124 Application of WS635, a mitochondrial function-enhancing agent, or transferring B cells attenuates anesthesia- and surgery-induced delirium-like alterations via the transmicrotubule-associated protein tau-PT217 pathway.125 These findings demonstrate that mitochondrial dysfunction is a potential mediator of T cell aging and postoperative delirium, making it a potential therapeutic target. Mitochondrial dysfunction may contribute to accumulation of inflammatory metabolites, epigenetic alterations, post-transcriptional protein modifications, and release of mitochondrial DNA into the cytoplasm, which activates the cyclic guanosine monophosphate -adenosine monophosphate synthetase-stimulator of interferon genes signaling pathway, stimulating activation of inflammasome vesicles and transcriptional pro-inflammatory cytokine gene activation.126 Activation of immunoinflammatory pathways and reduction of negative immunoregulatory mechanisms are linked to surgery-induced delirium.127 Therefore, the inflammatory response may be a more important mediator of T cell aging associated with postoperative delirium. Notably, the timely application of drugs such as dexamethasone can inhibit the inflammatory response thereby avoid postoperative delirium and prevent T cell aging. Second, patients with a history of long-term use of benzodiazepines exhibited immunosenescence characteristics such as increased proportion of CD57+ lymphocyte subpopulations and toxicity of natural killer cells,128 demonstrating that intraoperative use of benzodiazepines may trigger postoperative delirium via the T cell aging pathway. Therefore, patients at increased risk of T cell aging, benzodiazepine use should be minimized during the perioperative period, thereby reducing the risk of delirium.

T Cell Aging and Quality of Postoperative Recovery

Numerous investigations have shown that T cell deficiency will delay the time to remission of chemotherapy-induced peripheral neuralgia, chronic arthritis, and other types of pain, whereas reconstitution with CD8+ T cells prevents the development of chronic pain.129,130 This implies that T cell aging is a risk factor for the occurrence of postoperative pain. Postoperative pain is often accompanied by negative emotions such as depression and frustration. Compared with normal mice, recombinant activation of genes 2 gene-deficient mice (mice lacking adaptive immune cells) showed significantly delayed regression of mechanical anomalous pain, spontaneous pain, and depressive-like behavior induced by complete Freund’s adjuvant. CD3 T+ transplantation can ameliorate the aforementioned pain and depression delays, whereas NSAIDs do not achieve similar effects.131 This demonstrates that T-cell regulation participates in the co-morbid mechanisms underlying the development of pain and depression and is an irreplaceable target for formulating co-morbid therapy.

Patients with acute aortic coarctation exhibited increased percentage of TEM compared with healthy volunteers, and a significant negative correlation was observed between the percentage of TEM and the time of extubation from the tracheal cannula, ICU stay, and postoperative procalcitonin levels.108 This demonstrates that the percentage of T-cell subpopulations can predict various near-term postoperative indices. The 15-item Quality of Recovery Inventory (QoR-15) is a commonly used tool for evaluating short-term postoperative recovery, including five dimensions of indicators: pain, physical comfort, physical independence, psychological support, and emotional state. As discussed, T cell aging may be linked to physical independence, pain, and emotional factors, potentially impacting the quality of postoperative recovery. However, robust RCTs are required to validate this conclusion. Additionally, T cell aging could also influence long-term prognosis. In patients with advanced non-small cell lung cancer, large numbers of circulating CD28 CD57+ KLRG1 CD8+ T cells in the peripheral blood were found to be associated with a poor clinical response to PD-1 inhibitors.132 Similarly, the ratio of peripheral blood CD8+ CD57+ lymphocytes has been recognized to predict the recurrence-free survival rate of patients with nonmuscular invasive bladder cancer treated by transurethral tumor resection combined with intravesical instillation of IL-2,133 suggesting that T cell aging has a negative effect on the long-term prognosis of patients, although fewer studies have demonstrated the association between T cell aging and long-term prognosis in non-neoplastic disease.

Perioperative Modulation of Targeted T Cell Aging

A randomized controlled trial demonstrated that 8 weeks of yoga improved the Disease Activity Score for Rheumatoid Arthritis (DAS28-ESR), reduced the proportion of Th17 cells and T cell aging subpopulations, down-regulated RORγt, IL-17, IL-6, CXCL2, and CXCR2, and up-regulated FoxP3 and TGF-β transcription, and maintained immune homeostasis.134 Another clinical study found that 6 weeks of endurance strength training enhanced the proportion and absolute value of susceptible T cell aging in elderly patients.135 The aforementioned trials indicate that engaging in moderate-intensity preoperative exercise can enhance the immune environment and decrease the proportion of senescent T cells in most older adults, ultimately leading to better postoperative outcomes (Figure 4). Furthermore, patients at higher risk for anesthesia may experience an increased likelihood of T cell aging. Clinical studies have reported that more than 12 weeks of pulmonary rehabilitation significantly improved lung function, quality of life, and T-cell immune indices such as CD3+%, CD4+%, and CD4+%/CD8+% in patients with chronic obstructive pulmonary disease compared with the normal population.136 This demonstrated that prehabilitation may be more effective in suppressing T cell aging in anesthetized high-risk patients.

Figure 4 Perioperative Regulation Targeting T Cell Aging.

Abbreviations: mTOR, Mammalian Target of Rapamycin; MAPK, Mitogen-Activated Protein Kinase; IL, Interleukin; ROS, Reactive Oxygen Species; CD, Cluster of Differentiation.

Based on the mechanisms of T cell aging, future research should aim to develop agents for inhibiting inflammation, improving mitochondrial function, and autophagy. Meanwhile, strategies that alleviate postoperative neuropathy by improving T cell aging should be explored. It has also been reported that IL-15 treatment improved mitochondrial adaptation of Treg cells in HIV-infected patients, thereby restoring peroxisome proliferation-activated receptor-γ-coactivator 1α and mitochondrial transcription factor A. It also stimulated mitochondrial biosynthesis and enhanced the antiviral efficacy of HIV-specific CD8+ T cells.137 Metformin upregulates the expression of mitochondrial transcription factor A and mitochondrial function in CD8+ T cells in mice, thereby inhibiting Mycobacterium tuberculosis infection, and also improves TH17 inflammatory features by activating autophagy and improving mitochondrial bioenergetics in T cells.138 It is currently being investigated in clinical studies for potential anti-aging treatment.139 Treatment with ROS scavengers restored the immunosuppressive functions of senescence and DNA damage-binding protein 1- and Cullin4- associated factor 1-deficient Treg cells.48 Targeting the NF-κB, mTORC, and p38 MAPK pathways in the aging-associated secretory phenotype may inhibit inflammation, and further research is advocated to explore these possibilities. Animal experiments have shown that rapamycin improves the quality and intensity of CD8+ T cell memory responses to viral infection in mice.140 Findings from clinical trials have revealed that a low-dose mTOR inhibitor RTB101 is well-tolerated and up-regulates IFN-induced antiviral responses in elderly patients, making it an ideal agent for preventing aging.141 Inhibition of p38 MAPK partially restored the proliferative capacity of TRMRA by improving telomerase activity, and thus p38 MAPK is also a potential research direction.142 Chronic consumption of resveratrol has been shown to reverse pro-inflammatory cytokine profiles and oxidative DNA damage in senescent hybrid mice.131 Regarding the enhancement of autophagy, spermine intake emerges as a promising research avenue, given that autophagy deficiency is linked to reduced levels of nicotinamide dinucleotide and a decline in spermine, which is necessary for the hypo-uridylation-dependent translation of pro-autophagic proteins. Animal experiments have revealed that spermidine promotes homeostatic differentiation of Treg cells in the intestine,143 but subsequent clinical trials are needed to validate its feasibility as a treatment for T cell aging and prevent postoperative neuropathy. Anti-CD25 therapy maintains the infantile pattern of T cells by depleting Treg or by shifting chromatin accessibility to a younger individual state, thereby regulating neurological functions such as cognition.123,144 Tribbles homolog 2 is an important regulator of T cell naivety, and agents that stabilize its activity have shown potential to treat neurodegenerative diseases in clinical studies.47,145

Personalized adoptive neuro-immunotherapy is a novel personalized cellular immunotherapy for patients with aging, tumors, neurodegenerative diseases, and chronic infections.146 The therapy initially evaluates the responsiveness of various neurotransmitters and neuropeptides to T cells in the patient’s peripheral blood, assessing both beneficial increases or decreases in T cell protein expression levels and enhancements in T cell functional responses. Subsequently, a substantial number of T cells are isolated from the peripheral blood and infused back into the body after being exposed to the appropriate neurotransmitters and neuropeptides.147 The T cells interact with neurotransmitters or neuropeptides and does not involve unnatural activation, expansion, genetic manipulation, or other processes, making a safe intervention.

Conclusions

In the aging process, immune organ degeneration, long-term infection, inflammation aging, mitochondrial dysfunction, protein homeostasis imbalance, and epigenetic alterations are the underlying causes of T cell aging, which will lead to phenotypes such as T cell senescence, reduced T cell plasticity, imbalance of the ratio of naïve T cells to memory T cells and defective TCR pools. Future studies are anticipated to clarify the relationship between the triggers of T cell aging and to distinguish between primary and secondary changes. This understanding will aid in identifying optimal molecular targets that may slow or interrupt the aging of T cells. In the future, further research can be carried out in the following areas to deepen the understanding of the clinical significance of T cell aging: First, preoperative and intraoperative factors, such as hypertension, diabetes, acute respiratory distress syndrome, hypoxemia, depression and pain, nervous system diseases, obesity, tumor, autoimmune diseases, anesthetic methods and medications, as well as types of surgery, have been identified as predictive factors for high-risk groups of T cell aging. However, there is no robust clinical prediction model available to evaluate the relative importance of various perioperative indicators for T cell aging. Future studies should focus on developing rigorous prediction models to quantify the risks associated with these preoperative and intraoperative factors. Second, preliminary basic experiments and limited clinical research suggests that T cell aging may be linked to multiple adverse outcomes, such as postoperative neurological disorders. However, additional large-scale clinical investigations are required to elucidate the relationship between T cell aging and overall postoperative recovery quality, as well as long-term prognosis. Our research team is currently conducting a cohort study examining the association between T cell aging and postoperative recovery quality in elderly patients with colorectal cancer. This study will help directly investigate the association between T cell aging and postoperative recovery quality, as well as screen for clinically significant indicators of T cell aging. Third, surgery and anesthesia may potentially harm the human body and accelerate T cell aging, which warrants further exploration. Finally, prehabilitation, pharmacological interventions, and personalized adoptive neuroimmunotherapies can improve clinical prognosis by regulating T cell aging. In the future, a comprehensive and tailored strategy for regulating T cell aging could be developed for individual patients. For anesthesiologists, appropriate nerve blocks (eg, stellate ganglion block) and specific anesthesia techniques (eg, intravenous anesthesia instead of volatile anesthesia) may influence the state of T cell aging and contribute to improved clinical prognosis. In summary, given the increasing global aging population, in-depth research into T cell aging is of great significance for facilitating perioperative transitions and achieving healthy aging.

Abbreviations

IL, Interleukin; OS, Oxidative Stress; TCRs T, Cell Receptors; CMV, Cytomegalovirus; mTORC1, Mammalian target of rapamycin 1; TNF, Tumor Necrosis Factor; TEMRA Cells, Terminal Effector Memory T Cells; ROS, Reactive Oxygen Species; IFN-I, Interferon Type I; NF-κB, Nuclear factor-kappaB; CD, Cluster of Differentiation; KLRG1, Killer Cell Lectin-like Receptor Subfamily G1; PD-1, Programmed Death 1; Tregs, Regulatory T cells; CCR 7, Chemokine Receptor 7; TEM, Effector memory T cells; MAPK, Mitogen-Activated Protein Kinase; T2D, Type 2 Diabetes; CX3CR1, C-X3-C-Motif Ligand 1; AD, Alzheimer’s Disease; PD, Parkinson’s Disease; RA, Rheumatoid arthritis; SLE, Systemic lupus erythematosus; IBD, Inflammatory bowel disease.

Author Contributions

All authors made a significant contribution to the work reported, whether that is in the conception, study design, execution, acquisition of data, analysis and interpretation, or in all these areas; took part in drafting, revising or critically reviewing the article; gave final approval of the version to be published; have agreed on the journal to which the article has been submitted; and agree to be accountable for all aspects of the work.

Funding

This work was supported by the National Natural Science Foundation of China (82171217, 81771181, 81571065), the Beijing Natural Science Foundation (7202053), and the Beijing Friendship Hospital Zhongzi Project (YYZZ202332).

Disclosure

The authors have no conflicts of interest to disclose in this work.

References

1. Xu Y, Wang Z, Li S, et al. An in-depth understanding of the role and mechanisms of T cells in immune organ aging and age-related diseases. Sci China Life Sci. 2025;68(2):328–353. doi:10.1007/s11427-024-2695-x

2. Bosch DJ, Nieuwenhuijs-Moeke GJ, van Meurs M, Abdulahad WH, Struys M. Immune modulatory effects of nonsteroidal anti-inflammatory drugs in the perioperative period and their consequence on postoperative outcome. Anesthesiology. 2022;136(5):843–860. doi:10.1097/ALN.0000000000004141

3. Ackerman RS, Luddy KA, Icard BE, Pineiro Fernandez J, Gatenby RA, Muncey AR. The effects of anesthetics and perioperative medications on immune function: a narrative review. Anesth Analg. 2021;133(3):676–689.

4. Mittelbrunn M, Kroemer G. Hallmarks of T cell aging. Nat Immunol. 2021;22(6):687–698.

5. Gustafson CE, Kim C, Weyand CM, Goronzy JJ. Influence of immune aging on vaccine responses. J Allergy Clin Immunol. 2020;145(5):1309–1321.

6. Zhang H, Weyand CM, Goronzy JJ. Hallmarks of the aging T-cell system. FEBS J. 2021;288(24):7123–7142. doi:10.1111/febs.15770

7. Seddon B, Yates AJ. The natural history of naive T cells from birth to maturity. Immunol Rev. 2018;285(1):218–232. doi:10.1111/imr.12694

8. den Braber I, Mugwagwa T, Vrisekoop N, et al. Maintenance of peripheral naive T cells is sustained by thymus output in mice but not humans. Immunity. 2012;36(2):288–297. doi:10.1016/j.immuni.2012.02.006

9. Bains I, Antia R, Callard R, Yates AJ. Quantifying the development of the peripheral naive CD4+ T-cell pool in humans. Blood. 2009;113(22):5480–5487. doi:10.1182/blood-2008-10-184184

10. Sandstedt M, Chung RWS, Skoglund C, et al. Complete fatty degeneration of thymus associates with male sex, obesity and loss of circulating naive CD8(+) T cells in a Swedish middle-aged population. Immun Ageing. 2023;20(1):45. doi:10.1186/s12979-023-00371-7

11. Nasi M, Troiano L, Lugli E, et al. Thymic output and functionality of the IL-7/IL-7 receptor system in centenarians: implications for the neolymphogenesis at the limit of human life. Aging Cell. 2006;5(2):167–175. doi:10.1111/j.1474-9726.2006.00204.x

12. Coder BD, Wang H, Ruan L, Su DM. Thymic involution perturbs negative selection leading to autoreactive T cells that induce chronic inflammation. J Immunol. 2015;194(12):5825–5837. doi:10.4049/jimmunol.1500082

13. Ucar D, Marquez EJ, Chung CH, et al. The chromatin accessibility signature of human immune aging stems from CD8(+) T cells. J Exp Med. 2017;214(10):3123–3144. doi:10.1084/jem.20170416

14. Pawelec G, Larbi A, Derhovanessian E. Senescence of the human immune system. J Comp Pathol. 2010;142(Suppl 1):S39–44. doi:10.1016/j.jcpa.2009.09.005

15. Hadrup SR, Strindhall J, Kollgaard T, et al. Longitudinal studies of clonally expanded CD8 T cells reveal a repertoire shrinkage predicting mortality and an increased number of dysfunctional cytomegalovirus-specific T cells in the very elderly. J Immunol. 2006;176(4):2645–2653. doi:10.4049/jimmunol.176.4.2645

16. Goronzy JJ, Weyand CM. Successful and Maladaptive T Cell Aging. Immunity. 2017;46(3):364–378. doi:10.1016/j.immuni.2017.03.010

17. Nguyen LN, Nguyen LNT, Zhao J, et al. Immune activation induces telomeric DNA damage and promotes short-lived effector T cell differentiation in chronic HCV infection. Hepatology. 2021;74(5):2380–2394. doi:10.1002/hep.32008

18. Dang X, Cao D, Zhao J, et al. Mitochondrial topoisomerase 1 inhibition induces topological DNA damage and T cell dysfunction in patients with chronic viral infection. Front Cell Infect Microbiol. 2022;12:1026293. doi:10.3389/fcimb.2022.1026293

19. Chambers ES, Akbar AN. Can blocking inflammation enhance immunity during aging? J Allergy Clin Immunol. 2020;145(5):1323–1331. doi:10.1016/j.jaci.2020.03.016

20. White JT, Cross EW, Burchill MA, et al. Virtual memory T cells develop and mediate bystander protective immunity in an IL-15-dependent manner. Nat Commun. 2016;7:11291. doi:10.1038/ncomms11291

21. Mogilenko DA, Shpynov O, Andhey PS, et al. Comprehensive profiling of an aging immune system reveals clonal GZMK(+) CD8(+) T cells as conserved hallmark of inflammaging. Immunity. 2021;54(1):99–115e12. doi:10.1016/j.immuni.2020.11.005

22. Shen-Orr SS, Furman D, Kidd BA, et al. Defective signaling in the JAK-STAT pathway tracks with chronic inflammation and cardiovascular risk in aging humans. Cell Syst. 2016;3(4):374–84e4. doi:10.1016/j.cels.2016.09.009

23. Callender LA, Carroll EC, Bober EA, Akbar AN, Solito E, Henson SM. Mitochondrial mass governs the extent of human T cell senescence. Aging Cell. 2020;19(2):e13067. doi:10.1111/acel.13067

24. Miwa S, Kashyap S, Chini E, von Zglinicki T. Mitochondrial dysfunction in cell senescence and aging. J Clin Invest. 2022;132(13). doi:10.1172/JCI158447

25. Sanderson SL, Simon AK. In aged primary T cells, mitochondrial stress contributes to telomere attrition measured by a novel imaging flow cytometry assay. Aging Cell. 2017;16(6):1234–1243. doi:10.1111/acel.12640

26. Escrig-Larena JI, Delgado-Pulido S, Mittelbrunn M. Mitochondria during T cell aging. Semin Immunol. 2023;69:101808.

27. Ramstead AG, Wallace JA, Lee SH, et al. Mitochondrial pyruvate carrier 1 promotes peripheral T cell homeostasis through metabolic regulation of thymic development. Cell Rep. 2020;30(9):2889–99e6. doi:10.1016/j.celrep.2020.02.042

28. Arata Y, Watanabe A, Motosugi R, et al. Defective induction of the proteasome associated with T-cell receptor signaling underlies T-cell senescence. Genes Cells. 2019;24(12):801–813.

29. Stepensky P, Rensing-Ehl A, Gather R, et al. Early-onset Evans syndrome, immunodeficiency, and premature immunosenescence associated with tripeptidyl-peptidase II deficiency. Blood. 2015;125(5):753–761.

30. Swadling L, Pallett LJ, Diniz MO, et al. Human liver memory CD8(+) T cells use autophagy for tissue residence. Cell Rep. 2020;30(3):687–98e6.

31. Bektas A, Schurman SH, Gonzalez-Freire M, et al. Age-associated changes in human CD4(+) T cells point to mitochondrial dysfunction consequent to impaired autophagy. Aging. 2019;11(21):9234–9263. doi:10.18632/aging.102438

32. Li Y, Shen Y, Hohensinner P, et al. Deficient activity of the nuclease MRE11A induces T cell aging and promotes arthritogenic effector functions in patients with rheumatoid arthritis. Immunity. 2016;45(4):903–916. doi:10.1016/j.immuni.2016.09.013

33. Cheung P, Vallania F, Warsinske HC, et al. Single-cell chromatin modification profiling reveals increased epigenetic variations with aging. Cell. 2018;173(6):1385–97e14. doi:10.1016/j.cell.2018.03.079

34. Salumets A, Tserel L, Rumm AP, et al. Epigenetic quantification of immunosenescent CD8(+) TEMRA cells in human blood. Aging Cell. 2022;21(5):e13607. doi:10.1111/acel.13607

35. Kim C, Jadhav RR, Gustafson CE, et al. Defects in antiviral T cell responses inflicted by aging-associated miR-181a deficiency. Cell Rep. 2019;29(8):2202–16e5. doi:10.1016/j.celrep.2019.10.044

36. Qian F, Wang X, Zhang L, et al. Age-associated elevation in TLR5 leads to increased inflammatory responses in the elderly. Aging Cell. 2012;11(1):104–110. doi:10.1111/j.1474-9726.2011.00759.x

37. Zhao Y, Simon M, Seluanov A, Gorbunova V. DNA damage and repair in age-related inflammation. Nat Rev Immunol. 2023;23(2):75–89.

38. Huang M, Wang Y, Fang L, et al. T cell senescence: a new perspective on immunotherapy in lung cancer. Front Immunol. 2024;15:1338680. doi:10.3389/fimmu.2024.1338680

39. Henson SM, Riddell NE, Akbar AN. Properties of end-stage human T cells defined by CD45RA re-expression. Curr Opin Immunol. 2012;24(4):476–481. doi:10.1016/j.coi.2012.04.001

40. Larbi A, Fulop T. From “truly naive” to “exhausted senescent” T cells: when markers predict functionality. Cytometry A. 2014;85(1):25–35. doi:10.1002/cyto.a.22351

41. Pereira BI, De Maeyer RPH, Covre LP, et al. Sestrins induce natural killer function in senescent-like CD8(+) T cells. Nat Immunol. 2020;21(6):684–694. doi:10.1038/s41590-020-0643-3

42. Akbar AN, Borthwick N, Salmon M, et al. The significance of low bcl-2 expression by CD45RO T cells in normal individuals and patients with acute viral infections. The role of apoptosis in T cell memory. J Exp Med. 1993;178(2):427–438. doi:10.1084/jem.178.2.427

43. Callender LA, Carroll EC, Beal RWJ, et al. Human CD8(+) EMRA T cells display a senescence-associated secretory phenotype regulated by p38 MAPK. Aging Cell. 2018;17(1). doi:10.1111/acel.12675

44. Weng NP, Akbar AN, Goronzy J. CD28(-) T cells: their role in the age-associated decline of immune function. Trends Immunol. 2009;30(7):306–312.

45. Henson SM, Lanna A, Riddell NE, et al. p38 signaling inhibits mTORC1-independent autophagy in senescent human CD8(+) T cells. J Clin Invest. 2014;124(9):4004–4016. doi:10.1172/JCI75051

46. Mold JE, Reu P, Olin A, et al. Cell generation dynamics underlying naive T-cell homeostasis in adult humans. PLoS Biol. 2019;17(10):e3000383. doi:10.1371/journal.pbio.3000383

47. Cao W, Sturmlechner I, Zhang H, et al. TRIB2 safeguards naive T cell homeostasis during aging. Cell Rep. 2023;42(3):112195. doi:10.1016/j.celrep.2023.112195

48. Guo Z, Wang G, Wu B, et al. DCAF1 regulates Treg senescence via the ROS axis during immunological aging. J Clin Invest. 2020;130(11):5893–5908.

49. Fang L, Liu K, Liu C, et al. Tumor accomplice: t cell exhaustion induced by chronic inflammation. Front Immunol. 2022;13:979116.

50. Hu B, Li G, Ye Z, et al. Transcription factor networks in aged naive CD4 T cells bias lineage differentiation. Aging Cell. 2019;18(4):e12957.

51. Cao W, Fang F, Gould T, et al. Ecto-NTPDase CD39 is a negative checkpoint that inhibits follicular helper cell generation. J Clin Invest. 2020;130(7):3422–3436. doi:10.1172/JCI132417

52. Kim C, Hu B, Jadhav RR, et al. Activation of miR-21-regulated pathways in immune aging selects against signatures characteristic of memory T cells. Cell Rep. 2018;25(8):2148–62e5. doi:10.1016/j.celrep.2018.10.074

53. Goronzy JJ, Li G, Yu M, Weyand CM. Signaling pathways in aged T cells - a reflection of T cell differentiation, cell senescence and host environment. Semin Immunol. 2012;24(5):365–372. doi:10.1016/j.smim.2012.04.003

54. Lanfermeijer J, Borghans JAM, van Baarle D. How age and infection history shape the antigen-specific CD8(+) T-cell repertoire: implications for vaccination strategies in older adults. Aging Cell. 2020;19(11):e13262. doi:10.1111/acel.13262

55. Egorov ES, Kasatskaya SA, Zubov VN, et al. The changing landscape of naive T cell receptor repertoire with human aging. Front Immunol. 2018;9:1618. doi:10.3389/fimmu.2018.01618

56. Kared H, Tan SW, Lau MC, et al. Immunological history governs human stem cell memory CD4 heterogeneity via the Wnt signaling pathway. Nat Commun. 2020;11(1):821. doi:10.1038/s41467-020-14442-6

57. Williams-Gray CH, Wijeyekoon RS, Scott KM, Hayat S, Barker RA, Jones JL. Abnormalities of age-related T cell senescence in Parkinson’s disease. J Neuroinflammation. 2018;15(1):166. doi:10.1186/s12974-018-1206-5

58. Goronzy JJ, Weyand CM. Mechanisms underlying T cell ageing. Nat Rev Immunol. 2019;19(9):573–583. doi:10.1038/s41577-019-0180-1

59. Kouli A, Jensen M, Papastavrou V, et al. T lymphocyte senescence is attenuated in Parkinson’s disease. J Neuroinflammation. 2021;18(1):228. doi:10.1186/s12974-021-02287-9

60. Zhang JD, Patel MB, Song YS, et al. A novel role for type 1 angiotensin receptors on T lymphocytes to limit target organ damage in hypertension. Circ Res. 2012;110(12):1604–1617. doi:10.1161/CIRCRESAHA.111.261768

61. Youn JC, Yu HT, Lim BJ, et al. Immunosenescent CD8+ T cells and C-X-C chemokine receptor type 3 chemokines are increased in human hypertension. Hypertension. 2013;62(1):126–133. doi:10.1161/HYPERTENSIONAHA.113.00689

62. Pan XX, Wu F, Chen XH, et al. T-cell senescence accelerates angiotensin II-induced target organ damage. Cardiovasc Res. 2021;117(1):271–283. doi:10.1093/cvr/cvaa032

63. Guzik TJ, Hoch NE, Brown KA, et al. Role of the T cell in the genesis of angiotensin II induced hypertension and vascular dysfunction. J Exp Med. 2007;204(10):2449–2460. doi:10.1084/jem.20070657

64. Sim BC, Kang YE, You SK, et al. Hepatic T-cell senescence and exhaustion are implicated in the progression of fatty liver disease in patients with type 2 diabetes and mouse model with nonalcoholic steatohepatitis. Cell Death Dis. 2023;14(9):618. doi:10.1038/s41419-023-06146-8

65. Lau EYM, Carroll EC, Callender LA, et al. Type 2 diabetes is associated with the accumulation of senescent T cells. Clin Exp Immunol. 2019;197(2):205–213. doi:10.1111/cei.13344

66. Callender LA, Carroll EC, Garrod-Ketchley C, et al. Altered nutrient uptake causes mitochondrial dysfunction in senescent CD8(+) EMRA T cells during type 2 diabetes. Front Aging. 2021;2:681428. doi:10.3389/fragi.2021.681428

67. Philippe M, Gatterer H, Burtscher M, et al. Concentric and Eccentric Endurance Exercise Reverse Hallmarks of T-Cell Senescence in Pre-diabetic Subjects. Front Physiol. 2019;10:684. doi:10.3389/fphys.2019.00684

68. Ju SH, Lim JY, Song M, et al. Distinct effects of rosuvastatin and rosuvastatin/ezetimibe on senescence markers of CD8+ T cells in patients with type 2 diabetes mellitus: a randomized controlled trial. Front Endocrinol. 2024;15:1336357. doi:10.3389/fendo.2024.1336357

69. Bouadma L, Wiedemann A, Patrier J, et al. Immune alterations in a patient with SARS-CoV-2-related acute respiratory distress syndrome. J Clin Immunol. 2020;40(8):1082–1092. doi:10.1007/s10875-020-00839-x

70. Alvarez-Heredia P, Reina-Alfonso I, Dominguez-Del-Castillo JJ, et al. Accelerated T-cell immunosenescence in cytomegalovirus-seropositive individuals after severe acute respiratory syndrome coronavirus 2 infection. J Infect Dis. 2023;228(5):576–585. doi:10.1093/infdis/jiad119

71. Wang JS, Lin CT. Systemic hypoxia promotes lymphocyte apoptosis induced by oxidative stress during moderate exercise. Eur J Appl Physiol. 2010;108(2):371–382. doi:10.1007/s00421-009-1231-2

72. Brown FF, Bigley AB, Sherry C, et al. Training status and sex influence on senescent T-lymphocyte redistribution in response to acute maximal exercise. Brain Behav Immun. 2014;39:152–159. doi:10.1016/j.bbi.2013.10.031

73. Simon MS, Ioannou M, Arteaga-Henriquez G, et al. Premature T cell aging in major depression: a double hit by the state of disease and cytomegalovirus infection. Brain Behav Immun Health. 2023;29:100608. doi:10.1016/j.bbih.2023.100608

74. Swiatkowski M, Rybakowski JK. Depression and T lymphocytes in patients with irritable bowel syndrome. J Affect Disord. 1993;28(3):199–202. doi:10.1016/0165-0327(93)90105-S

75. Shirakawa K, Yan X, Shinmura K, et al. Obesity accelerates T cell senescence in murine visceral adipose tissue. J Clin Invest. 2016;126(12):4626–4639. doi:10.1172/JCI88606

76. Shirakawa K, Sano M. Drastic transformation of visceral adipose tissue and peripheral CD4 T cells in obesity. Front Immunol. 2022;13:1044737. doi:10.3389/fimmu.2022.1044737

77. Vick LV, Collins CP, Khuat LT, et al. Aging augments obesity-induced thymic involution and peripheral T cell exhaustion altering the “obesity paradox”. Front Immunol. 2022;13:1012016. doi:10.3389/fimmu.2022.1012016

78. Mundstock E, Sarria EE, Zatti H, et al. Effect of obesity on telomere length: systematic review and meta-analysis. Obesity. 2015;23(11):2165–2174. doi:10.1002/oby.21183

79. Kaushik P, Anderson JT. Obesity: epigenetic aspects. Biomol Concepts. 2016;7(3):145–155. doi:10.1515/bmc-2016-0010

80. Wang X, Zhu H, Snieder H, et al. Obesity related methylation changes in DNA of peripheral blood leukocytes. BMC Med. 2010;8:87. doi:10.1186/1741-7015-8-87

81. Jongbloed F, Meijers RW, IJzermans JNM, et al. Effects of bariatric surgery on telomere length and T-cell aging. Int J Obes. 2019;43(11):2189–2199. doi:10.1038/s41366-019-0351-y

82. Pena E, Leon-Mengibar J, Powell TR, Caixas A, Cardoner N, Rosa A. Telomere length in patients with obesity submitted to bariatric surgery: a systematic review. Eur Eat Disord Rev. 2021;29(6):842–853. doi:10.1002/erv.2865

83. Fessler J, Angiari S. The role of T cell senescence in neurological diseases and its regulation by cellular metabolism. Front Immunol. 2021;12:706434. doi:10.3389/fimmu.2021.706434

84. Haegert DG. Multiple sclerosis: a disorder of altered T-cell homeostasis. Mult Scler Int. 2011;2011:461304. doi:10.1155/2011/461304

85. Yildiz O, Schroth J, Tree T, et al. Senescent-like blood lymphocytes and disease progression in amyotrophic lateral sclerosis. Neurol Neuroimmunol Neuroinflamm. 2023;10(1). doi:10.1212/NXI.0000000000200042

86. Pellicano M, Larbi A, Goldeck D, et al. Immune profiling of Alzheimer patients. J Neuroimmunol. 2012;242(1–2):52–59. doi:10.1016/j.jneuroim.2011.11.005

87. Gate D, Saligrama N, Leventhal O, et al. Clonally expanded CD8 T cells patrol the cerebrospinal fluid in Alzheimer’s disease. Nature. 2020;577(7790):399–404. doi:10.1038/s41586-019-1895-7

88. Panossian LA, Porter VR, Valenzuela HF, et al. Telomere shortening in T cells correlates with Alzheimer’s disease status. Neurobiol Aging. 2003;24(1):77–84. doi:10.1016/S0197-4580(02)00043-X

89. Monsonego A, Zota V, Karni A, et al. Increased T cell reactivity to amyloid beta protein in older humans and patients with Alzheimer disease. J Clin Invest. 2003;112(3):415–422. doi:10.1172/JCI200318104

90. Browne TC, McQuillan K, McManus RM, O’Reilly JA, Mills KH, Lynch MA. IFN-gamma production by amyloid beta-specific Th1 cells promotes microglial activation and increases plaque burden in a mouse model of Alzheimer’s disease. J Immunol. 2013;190(5):2241–2251. doi:10.4049/jimmunol.1200947

91. Sulzer D, Alcalay RN, Garretti F, et al. T cells from patients with parkinson’s disease recognize alpha-synuclein peptides. Nature. 2017;546(7660):656–661. doi:10.1038/nature22815

92. Tedeschi V, Paldino G, Kunkl M, et al. CD8(+) T cell senescence: lights and shadows in viral infections, autoimmune disorders and cancer. Int J mol Sci. 2022;23(6):3374. doi:10.3390/ijms23063374

93. Onyema OO, Decoster L, Njemini R, et al. Shifts in subsets of CD8+ T-cells as evidence of immunosenescence in patients with cancers affecting the lungs: an observational case-control study. BMC Cancer. 2015;15:1016. doi:10.1186/s12885-015-2013-3

94. Ager CR, Zhang M, Chaimowitz M, et al. KLRG1 marks tumor-infiltrating CD4 T cell subsets associated with tumor progression and immunotherapy response. J Immunother Cancer. 2023;11(9):e006782. doi:10.1136/jitc-2023-006782

95. Onyema OO, Decoster L, Njemini R, et al. Chemotherapy-induced changes and immunosenescence of CD8+ T-cells in patients with breast cancer. Anticancer Res. 2015;35(3):1481–1489.

96. Lu Y, Ruan Y, Hong P, et al. T-cell senescence: a crucial player in autoimmune diseases. Clin Immunol. 2023;248:109202. doi:10.1016/j.clim.2022.109202

97. Melis L, Van Praet L, Pircher H, Venken K, Elewaut D. Senescence marker killer cell lectin-like receptor G1 (KLRG1) contributes to TNF-alpha production by interaction with its soluble E-cadherin ligand in chronically inflamed joints. Ann Rheum Dis. 2014;73(6):1223–1231. doi:10.1136/annrheumdis-2013-203881

98. Kalim H, Wahono CS, Permana BPO, Pratama MZ, Handono K. Association between senescence of T cells and disease activity in patients with systemic lupus erythematosus. Reumatologia. 2021;59(5):292–301. doi:10.5114/reum.2021.110318

99. Dai SX, Gu HX, Lin QY, et al. Decreased CD8+CD28+/CD8+CD28- T cell ratio can sensitively predict poor outcome for patients with complicated crohn disease. Medicine. 2017;96(26):e7247. doi:10.1097/MD.0000000000007247

100. Konstantis G, Tsaousi G, Kitsikidou E, Zacharoulis D, Pourzitaki C. The immunomodulatory effect of various anaesthetic practices in patients undergoing gastric or colon cancer surgery: a systematic review and meta-analysis of randomized clinical trials. J Clin Med. 2023;12(18):6027. doi:10.3390/jcm12186027

101. Siekmann W, Eintrei C, Magnuson A, et al. Surgical and not analgesic technique affects postoperative inflammation following colorectal cancer surgery: a prospective, randomized study. Colorectal Dis. 2017;19(6):O186–O95. doi:10.1111/codi.13643

102. Zhou M, Liu W, Peng J, Wang Y. Impact of propofol epidural anesthesia on immune function and inflammatory factors in patients undergoing gastric cancer surgery. Am J Transl Res. 2021;13(4):3064–3073.

103. Hagen M, Derudder E. Inflammation and the alteration of B-cell physiology in aging. Gerontology. 2020;66(2):105–113. doi:10.1159/000501963

104. Li X, Li C, Zhang W, Wang Y, Qian P, Huang H. Inflammation and aging: signaling pathways and intervention therapies. Signal Transduct Target Ther. 2023;8(1):239. doi:10.1038/s41392-023-01502-8

105. Hogarth K, Tarazi D, Maynes JT. The effects of general anesthetics on mitochondrial structure and function in the developing brain. Front Neurol. 2023;14:1179823. doi:10.3389/fneur.2023.1179823

106. Sauce D, Larsen M, Fastenackels S, et al. Evidence of premature immune aging in patients thymectomized during early childhood. J Clin Invest. 2009;119(10):3070–3078. doi:10.1172/JCI39269

107. Taves MD, Ashwell JD. Effects of sex steroids on thymic epithelium and thymocyte development. Front Immunol. 2022;13:975858. doi:10.3389/fimmu.2022.975858

108. Li H, Wang PF, Luo W, et al. CD36-mediated ferroptosis destabilizes CD4(+) T cell homeostasis in acute Stanford type-A aortic dissection. Cell Death Dis. 2024;15(9):669. doi:10.1038/s41419-024-07022-9

109. Zhou Y, Ju H, Hu Y, et al. Tregs dysfunction aggravates postoperative cognitive impairment in aged mice. J Neuroinflammation. 2023;20(1):75. doi:10.1186/s12974-023-02760-7

110. Pereira BI, Devine OP, Vukmanovic-Stejic M, et al. Senescent cells evade immune clearance via HLA-E-mediated NK and CD8(+) T cell inhibition. Nat Commun. 2019;10(1):2387. doi:10.1038/s41467-019-10335-5

111. Hazeldine J, Hampson P, Lord JM. Reduced release and binding of perforin at the immunological synapse underlies the age-related decline in natural killer cell cytotoxicity. Aging Cell. 2012;11(5):751–759. doi:10.1111/j.1474-9726.2012.00839.x

112. Wang TW, Johmura Y, Suzuki N, et al. Blocking PD-L1-PD-1 improves senescence surveillance and ageing phenotypes. Nature. 2022;611(7935):358–364. doi:10.1038/s41586-022-05388-4

113. Desdin-Mico G, Soto-Heredero G, Aranda JF, et al. T cells with dysfunctional mitochondria induce multimorbidity and premature senescence. Science. 2020;368(6497):1371–1376. doi:10.1126/science.aax0860

114. Kaya T, Mattugini N, Liu L, et al. CD8(+) T cells induce interferon-responsive oligodendrocytes and microglia in white matter aging. Nat Neurosci. 2022;25(11):1446–1457. doi:10.1038/s41593-022-01183-6

115. Groh J, Knopper K, Arampatzi P, et al. Accumulation of cytotoxic T cells in the aged CNS leads to axon degeneration and contributes to cognitive and motor decline. Nat Aging. 2021;1(4):357–367. doi:10.1038/s43587-021-00049-z

116. Broux B, Pannemans K, Zhang X, et al. CX(3)CR1 drives cytotoxic CD4(+)CD28(-) T cells into the brain of multiple sclerosis patients. J Autoimmun. 2012;38(1):10–19. doi:10.1016/j.jaut.2011.11.006

117. van Nierop GP, van Luijn MM, Michels SS, et al. Phenotypic and functional characterization of T cells in white matter lesions of multiple sclerosis patients. Acta Neuropathol. 2017;134(3):383–401. doi:10.1007/s00401-017-1744-4

118. Saresella M, Calabrese E, Marventano I, et al. Increased activity of Th-17 and Th-9 lymphocytes and a skewing of the post-thymic differentiation pathway are seen in Alzheimer’s disease. Brain Behav Immun. 2011;25(3):539–547. doi:10.1016/j.bbi.2010.12.004

119. Brigas HC, Ribeiro M, Coelho JE, et al. IL-17 triggers the onset of cognitive and synaptic deficits in early stages of Alzheimer’s disease. Cell Rep. 2021;36(9):109574. doi:10.1016/j.celrep.2021.109574

120. Markovic-Plese S, Cortese I, Wandinger KP, McFarland HF, Martin R. CD4+CD28- costimulation-independent T cells in multiple sclerosis. J Clin Invest. 2001;108(8):1185–1194. doi:10.1172/JCI200112516

121. Lima TS. Beyond an inflammatory mediator: interleukin-1 in neurophysiology. Exp Physiol. 2023;108(7):917–924. doi:10.1113/EP090780

122. Veldhoen M, Uyttenhove C, van Snick J, et al. Transforming growth factor-beta ‘reprograms’ the differentiation of T helper 2 cells and promotes an interleukin 9-producing subset. Nat Immunol. 2008;9(12):1341–1346. doi:10.1038/ni.1659

123. Da Mesquita S, Herz J, Wall M, et al. Aging-associated deficit in CCR7 is linked to worsened glymphatic function, cognition, neuroinflammation, and beta-amyloid pathology. Sci Adv. 2021;7(21). doi:10.1126/sciadv.abe4601

124. Yang Y, Zhang W, Liu Y, et al. Mitochondrial dysfunction of peripheral platelets as a predictive biomarker for postoperative delirium in elderly patients. Ann Neurol. 2024;96(1):74–86. doi:10.1002/ana.26918

125. Lu J, Liang F, Bai P, et al. Blood tau-PT217 contributes to the anesthesia/surgery-induced delirium-like behavior in aged mice. Alzheimers Dement. 2023;19(9):4110–4126. doi:10.1002/alz.13118

126. Soto-Heredero G, Gomez de Las Heras MM, Gabande-Rodriguez E, Oller J, Mittelbrunn M. Glycolysis - a key player in the inflammatory response. FEBS J. 2020;287(16):3350–3369. doi:10.1111/febs.15327

127. Thisayakorn P, Thipakorn Y, Tantavisut S, Sirivichayakul S, Maes M. Delirium due to hip fracture is associated with activated immune-inflammatory pathways and a reduction in negative immunoregulatory mechanisms. BMC Psychiatry. 2022;22(1):369. doi:10.1186/s12888-022-04021-y

128. Lechin F, van der Dijs B, Benaim M. Benzodiazepines: tolerability in elderly patients. Psychother Psychosom. 1996;65(4):171–182. doi:10.1159/000289072

129. Baddack-Werncke U, Busch-Dienstfertig M, Gonzalez-Rodriguez S, et al. Cytotoxic T cells modulate inflammation and endogenous opioid analgesia in chronic arthritis. J Neuroinflammation. 2017;14(1):30. doi:10.1186/s12974-017-0804-y

130. Laumet G, Edralin JD, Dantzer R, Heijnen CJ, Kavelaars A. Cisplatin educates CD8+ T cells to prevent and resolve chemotherapy-induced peripheral neuropathy in mice. Pain. 2019;160(6):1459–1468. doi:10.1097/j.pain.0000000000001512

131. Laumet G, Edralin JD, Dantzer R, Heijnen CJ, Kavelaars A. CD3(+) T cells are critical for the resolution of comorbid inflammatory pain and depression-like behavior. Neurobiol Pain. 2020;7:100043. doi:10.1016/j.ynpai.2020.100043

132. Ferrara R, Naigeon M, Auclin E, et al. Circulating T-cell immunosenescence in patients with advanced non-small cell lung cancer treated with single-agent PD-1/PD-L1 inhibitors or platinum-based chemotherapy. Clin Cancer Res. 2021;27(2):492–503. doi:10.1158/1078-0432.CCR-20-1420

133. Characiejus D, Pasukoniene V, Jacobs JJ, et al. Prognostic significance of peripheral blood CD8highCD57+ lymphocytes in bladder carcinoma patients after intravesical IL-2. Anticancer Res. 2011;31(2):699–703.

134. Gautam S, Kumar R, Kumar U, Kumar S, Luthra K, Dada R. Yoga maintains Th17/Treg cell homeostasis and reduces the rate of T cell aging in rheumatoid arthritis: a randomized controlled trial. Sci Rep. 2023;13(1):14924. doi:10.1038/s41598-023-42231-w

135. Cao Dinh H, Njemini R, Onyema OO, et al. Strength endurance training but not intensive strength training reduces senescence-prone T cells in peripheral blood in community-dwelling elderly women. J Gerontol a Biol Sci Med Sci. 2019;74(12):1870–1878. doi:10.1093/gerona/gly229

136. Ma Y, Chen Y, Zhang N, et al. Efficacy and safety of pulmonary rehabilitation training on lung function, quality of life, and T cell immune function in patients with stable chronic obstructive pulmonary disease: a randomized controlled trial. Ann Palliat Med. 2022;11(5):1774–1785. doi:10.21037/apm-22-451

137. Younes SA, Talla A, Pereira Ribeiro S, et al. Cycling CD4+ T cells in HIV-infected immune nonresponders have mitochondrial dysfunction. J Clin Invest. 2018;128(11):5083–5094. doi:10.1172/JCI120245

138. Bohme J, Martinez N, Li S, et al. Metformin enhances anti-mycobacterial responses by educating CD8+ T-cell immunometabolic circuits. Nat Commun. 2020;11(1):5225. doi:10.1038/s41467-020-19095-z

139. Kulkarni AS, Gubbi S, Barzilai N. Benefits of metformin in attenuating the hallmarks of aging. Cell Metab. 2020;32(1):15–30. doi:10.1016/j.cmet.2020.04.001

140. Chen Y, Xu Z, Sun H, et al. Regulation of CD8(+) T memory and exhaustion by the mTOR signals. Cell mol Immunol. 2023;20(9):1023–1039. doi:10.1038/s41423-023-01064-3

141. Mannick JB, Teo G, Bernardo P, et al. Targeting the biology of ageing with mTOR inhibitors to improve immune function in older adults: phase 2b and Phase 3 randomised trials. Lancet Healthy Longev. 2021;2(5):e250–e62. doi:10.1016/S2666-7568(21)00062-3

142. Henson SM, Macaulay R, Riddell NE, Nunn CJ, Akbar AN. Blockade of PD-1 or p38 MAP kinase signaling enhances senescent human CD8(+) T-cell proliferation by distinct pathways. Eur J Immunol. 2015;45(5):1441–1451. doi:10.1002/eji.201445312

143. Carriche GM, Almeida L, Stuve P, et al. Regulating T-cell differentiation through the polyamine spermidine. J Allergy Clin Immunol. 2021;147(1):335–48e11. doi:10.1016/j.jaci.2020.04.037

144. Zhang H, Jadhav RR, Cao W, et al. Aging-associated HELIOS deficiency in naive CD4(+) T cells alters chromatin remodeling and promotes effector cell responses. Nat Immunol. 2023;24(1):96–109. doi:10.1038/s41590-022-01369-x

145. Guo L, Li X, Gould T, Wang ZY, Cao W. T cell aging and Alzheimer’s disease. Front Immunol. 2023;14:1154699. doi:10.3389/fimmu.2023.1154699

146. Levite M. T cells plead for rejuvenation and amplification; with the brain’s neurotransmitters and neuropeptides we can make it happen. Front Immunol. 2021;12:617658. doi:10.3389/fimmu.2021.617658

147. Levite M. Neuro faces of beneficial T cells: essential in brain, impaired in aging and neurological diseases, and activated functionally by neurotransmitters and neuropeptides. Neural Regen Res. 2023;18(6):1165–1178. doi:10.4103/1673-5374.357903

Creative Commons License © 2025 The Author(s). This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution - Non Commercial (unported, 4.0) License. By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.