Skip to main content
Springer logoLink to Springer
. 2025 Mar 5;25(1):72. doi: 10.1007/s10238-025-01608-z

The relationship between telomere length and aging-related diseases

Xuanqi Huang 1, Leyi Huang 1, Jiaweng Lu 1, Lijuan Cheng 2,3, Du Wu 4, Linmeng Li 5, Shuting Zhang 1, Xinyue Lai 1, Lu Xu 1,3,
PMCID: PMC11882723  PMID: 40044947

Abstract

The intensifying global phenomenon of an aging population has spurred a heightened emphasis on studies on aging and disorders associated with aging. Cellular senescence and aging are known to be caused by telomere shortening. Telomere length (TL) has emerged as a biomarker under intense scrutiny, and its widespread use in investigations of diseases tied to advancing age. This review summarizes the current knowledge of the association between telomeres and aging-related diseases, explores the important contribution of dysfunctional telomeres to the development and progression of these diseases, and aims to provide valuable insights for the development of novel therapeutic strategies.

Keywords: Telomere length, Aging-related diseases, Biomarkers

Introduction

Telomeres—structure and function

Telomeres are nucleoprotein structures located at the ends of chromosomes and are composed of repetitive TTAGGG sequences [1]. They serve two essential functions: protecting chromosome termini and preventing the loss of genetic material during DNA replication. A critical component in telomere maintenance is the shelterin complex, which comprises six protein subunits—TRF1, TRF2, Tpp1, Pot1, TIN2, and Rap1 [2, 3]. This complex not only binds telomeric DNA but also facilitates its folding into a protective three-dimensional T-loop structure that safeguards the 3′–OH terminus [4]. Moreover, shelterin regulates telomerase activity at telomeres, maintaining length homeostasis and establishing the optimal telomere length [5]. Disruptions in the shelterin complex can lead to premature aging [6]. Despite shelterin’s protection, approximately 50–200 base pairs of telomeric DNA are lost with each cell division. This attrition is inherent to DNA replication because DNA polymerase synthesizes DNA only in the 5′ → 3′ direction, rendering the complete replication of chromosome ends impossible [7, 8]. In this way, telomeres act as buffers that preserve the integrity of genetic information. Because telomere length reflects cellular replicative history, telomeres are often considered markers of aging—the “mitotic clock” of human biological age [912]. When telomeres shorten beyond a critical threshold, cells enter replicative senescence, eventually triggering apoptosis [13]. Telomerase, a ribonucleoprotein enzyme complex, counteracts telomere shortening by appending repetitive sequences to chromosome ends [3]. It consists of a catalytic subunit, telomerase reverse transcriptase (TERT), and an RNA template (TERC) that directs telomeric DNA synthesis [14]. While most somatic cells exhibit low or undetectable telomerase activity, certain cell types—such as germ cells, stem cells, and cancer cells—maintain high telomerase activity to support continuous proliferation [15].

Telomeres in aging

Cellular senescence is marked by a series of complex molecular changes that lead to the gradual decline of tissue function and increased susceptibility to disease and mortality. The characteristics of cellular aging can be categorized into several key areas: changes in cellular phenotypes (such as stem cell exhaustion and senescence), disruptions in signaling pathways (including nutrient sensing distortions and intercellular communication abnormalities), organelle damage (such as mitochondrial dysfunction and loss of protein homeostasis), and genomic instability (including telomere attrition and epigenetic dysregulation) [16]. The progressive loss of physiological integrity associated with aging impairs biological functions, making it increasingly difficult for organisms to maintain homeostasis. Aging is an inevitable and irreversible process linked to the gradual decline of cellular function. In 2013, López-Otín and colleagues identified nine fundamental hallmarks of aging [16]. Building on this framework, researchers discovered three additional markers in 2023, including telomere attrition [17]. Telomere shortening is recognized as a primary indicator of aging, closely tied to the Hayflick limit—the concept that cells have a finite number of divisions. When telomeres become critically short, cells enter a senescent state, losing their ability to divide normally, which ultimately contributes to cellular arrest and dysfunction. Animal studies support this, showing that mice with longer telomeres tend to have extended lifespans, whereas those with shorter telomeres exhibit premature aging and reduced longevity [18].

DNA polymerase is an enzyme that catalyzes DNA replication, which always proceeds in the 5′ → 3′ direction. However, this enzyme can only add nucleotides to the free 3′–OH end of an existing nucleotide chain. At the very beginning of a new DNA strand, there is no available 3′–OH group for DNA polymerase to extend. To initiate DNA synthesis, RNA primers, which provide the necessary 3′–OH group, are required. After replication, these primers are removed, and the resulting gaps are filled with DNA. However, at the 5′ end of each newly synthesized daughter strand, the region previously occupied by the primer remains unfilled due to the lack of a free 3′–OH group [19]. As a result, each round of replication leads to a gradual loss of DNA. Without telomeres, this progressive DNA loss during mitosis would eventually lead to the erosion of essential genetic material [7]. When telomeres shorten to a critical length of approximately 4 kbp, cells either enter senescence or undergo apoptosis, preventing further division.

Molecular mechanisms linking telomere dysfunction to disease pathogenesis

DNA damage response

Telomere length and DNA damage response (DDR) are deeply interconnected in the regulation of cellular aging [20]. When they become critically short, they lose their ability to maintain the protective T-loop structure, exposing chromosome ends and triggering DDR pathways [21]. A hallmark of DDR activation is the phosphorylation of histone H2AX, resulting in γH2AX, which serves as a marker for DNA double-strand breaks. Proteins such as 53BP1 and MDC1 are then recruited to these sites of damage. Central to this response are the kinases ATM (ataxia telangiectasia mutated) and ATR (ATM- and Rad3-related), which act as primary sensors of DNA damage. Upon activation, they phosphorylate downstream kinases like CHK2 and CHK1, respectively. This cascade amplifies the damage signal, leading to cell cycle arrest and facilitating DNA repair processes. However, if the damage is too severe or persistent, the DDR can lead to cellular senescence (a state of irreversible cell cycle arrest) or, in some cases, apoptosis (programmed cell death) [9]. Due to the irreparable nature of critically short telomeres, the DNA damage response remains continuously activated. This persistent signaling forces the cell cycle into a permanent arrest, ultimately contributing to cellular aging and apoptosis [20, 22, 23]. Although apoptosis helps remove severely damaged cells, excessive or inappropriate apoptosis can lead to disease development [23]. Apoptosis can lead to massive cell loss and a decline in organ function [24]. For example, excessive apoptosis of cardiomyocytes or neurons can directly result in heart failure or neurodegenerative diseases. Additionally, apoptosis can cause abnormal organ repair and fibrosis. If a large number of apoptotic cells are not cleared in a timely manner, they may induce neighboring stromal cells to secrete pro-fibrotic factors (such as TGF-β), leading to compensatory fibrotic repair, which in turn causes organ hardening and functional impairment [25, 26].

However, this explanation relies solely on telomere shortening and fails to account for aging in undifferentiated or non-proliferating cells [23]. In non-proliferating cells, such as quiescent or terminally differentiated cells, telomere damage—rather than telomere shortening—is a key driver of DDR activation and age-related functional decline. Telomere-binding proteins play a crucial role in maintaining chromosome stability, but they also suppress DNA repair within the telomeric region, making it difficult to fix telomere-specific DNA damage (tDD) [27]. As a result, this unrepaired damage leads to persistent DDR signaling and the formation of telomere-associated foci (TAF). Experiments on mice have shown that as they age, the frequency of TAF increases in their intestines [20, 22, 27]. Notably, this process can occur even in non-proliferating cells with intact telomere length if they experience DNA damage. Consequently, the accumulation of telomere damage—regardless of whether telomeres have significantly shortened—can still drive cellular senescence and contribute to age-related dysfunction [28].

Oxidative stress

Telomeres are not only protective caps at the ends of chromosomes, but their damage plays a crucial role in exacerbating oxidative stress and triggering diseases [29]. When telomeres shorten or their structure is damaged, the function of the shelterin complex becomes disrupted, leading the cell to misrecognize telomeres as DNA breaks, thereby persistently activating the DDR [20, 30]. This, in turn, initiates the ATM/ATR signaling pathways and downstream effectors such as p53 and Chk1/Chk2 [20]. The prolonged activation of these damage signals drives cells into senescence or apoptosis while simultaneously inducing a pro-inflammatory response through the activation of transcription factors like NF-κB, further intensifying oxidative stress within the cell [31]. Meanwhile, telomere damage-induced cellular senescence is often accompanied by mitochondrial dysfunction, where impaired mitochondrial metabolism leads to electron leakage, resulting in the excessive production of reactive oxygen species (ROS) [32, 33]. The cellular senescence and oxidative stress environment induced by telomere damage not only reduce cellular regeneration and organ repair capacity, leading to tissue functional decline, but also compromise telomere protective functions, making chromosome ends more prone to fusion, breakage, and rearrangement [9]. Overall, telomere damage persistently activates DNA damage signaling, induces cellular senescence, and disrupts mitochondrial function, forming a positive feedback loop between ROS and telomere damage. This not only significantly exacerbates oxidative stress but also initiates and accelerates the onset and progression of various diseases through multiple molecular pathways.

Mitochondrial dysfunction

Telomere damage activates a persistent DDR, in which the activation of the p53 pathway plays a central role [20, 34]. In addition to regulating the cell cycle and inducing apoptosis, p53 downregulates key transcription factors involved in mitochondrial biogenesis and function, such as PGC-1α and PGC-1β [35]. PGC-1α/β are crucial for maintaining mitochondrial function and quantity, and when their expression is suppressed, mitochondrial renewal and repair capacity decline significantly, leading to mitochondrial dysfunction [34]. Dysfunctional mitochondria exhibit impaired energy production, resulting in ATP deficiency, which prevents cells from sustaining normal metabolic and physiological functions, ultimately driving them into apoptosis or senescence [36]. The loss of cellular function and increased cell death not only affect individual cells but also trigger chronic low-grade inflammation and cellular dysfunction at the tissue level, thereby compromising tissue repair and regeneration capacity. Over time, this persistent oxidative stress and energy-deficient environment create a foundation for the onset of various diseases. In the metabolic system, it disrupts insulin signaling, promoting the development of diabetes and other metabolic disorders [37]. And in the nervous system, mitochondrial dysfunction may accelerate neurodegenerative diseases such as Alzheimer’s disease [37, 38]. Clinical and experimental evidence further supports this connection. A population study found that individuals with primary and secondary mitochondrial dysfunction had shorter telomeres compared to healthy controls [39]. Similarly, TERC-null mice exhibited mitochondrial dysfunction and a weakened antioxidant defense system [40]. Additionally, mitochondrial DNA (mtDNA) damage, particularly deletions, may contribute to mitochondrial dysfunction and excessive ROS production, further exacerbating telomere attrition [41].

Inflammation

Telomere damage not only triggers a persistent DDR within the cell but also induces cellular senescence, leading to the formation of the senescence-associated secretory phenotype (SASP), which in turn initiates a series of complex inflammatory responses in the body [20]. First, telomere dysfunction induces cellular senescence, which leads to increased synthesis and release of inflammatory molecules such as TNF-α and IL-6 [42]. These secreted factors not only exert autocrine effects within the cell, reinforcing senescence signaling, but also influence surrounding healthy cells through paracrine effects, exposing them to inflammatory stimuli and driving them into senescence or functional dysregulation [43]. Prolonged exposure to this inflammatory environment disrupts normal intercellular communication, induces tissue remodeling, damages the extracellular matrix, and gradually leads to pathological changes such as fibrosis and sclerosis [44]. In a mouse experiment, the reduction of the telomere protective protein Trf2 in endothelial cells of young mice resulted in telomere damage and cellular senescence, accompanied by enhanced inflammatory signaling and increased oxidative stress [45]. These changes led to endothelial dysfunction and impaired glucose tolerance [46]. Consequently, inflammation induced by telomere damage contributes to endothelial dysfunction and atherosclerosis in the cardiovascular system, accelerates neurodegenerative disease progression in the nervous system, and disrupts insulin signaling in the metabolic system, promoting diabetes and other metabolic disorders [4749]. Additionally, studies indicate that YAP1 activation, through its interaction with transcription factors, regulates the expression of multiple genes involved in immune and inflammatory responses [43]. This activation may also influence the release of cytokines and inflammatory mediators such as IL-6, IL-8, and TNF-α [42], which further amplify inflammatory signaling and modulate immune functions.

Senescence-associated secretory phenotype

In senescent cells, the sustained activation of DDR maintains the expression of SASP. The activation of the ATM and p38 MAPK signaling pathways promotes the activation of NF-κB, which is a key transcription factor regulating SASP gene expression [31]. SASP includes the release of cytokines, chemokines, and proteases (such as IL-6, IL-8, TNF-α, and MMPs), which can reshape the surrounding tissue environment, promote chronic inflammation, and affect neighboring cells [23, 50]. Persistent local inflammation disrupts normal intercellular communication and balance, leading to extracellular matrix degradation and changes in the extracellular environment, which in turn promote pathological remodeling of tissue structure, such as loss of arterial endothelial function and liver fibrosis [51, 52]. Meanwhile, SASP leads to matrix remodeling, with proteases such as MMPs disrupting the integrity of the extracellular matrix, promoting pathological changes such as fibrosis and sclerosis [53]. Moreover, SASP factors can induce DNA damage in neighboring healthy cells, forming a positive feedback loop that further expands the damaged area [26].

In summary, telomere dysfunction accelerates disease progression in aging-related diseases primarily through mechanisms such as cellular senescence, genomic instability, and chronic inflammation.

Telomere and aging-related diseases

Metabolic diseases

Diabetes

Diabetes, encompassing type 1 and type 2 diabetes mellitus (T2DM) as well as gestational diabetes, is widespread on a global scale. The above text mentioned that telomere attrition and T2DM have a bidirectional causal relationship. Elevated oxidative stress in individuals with T2DM leads to increased DNA damage, particularly affecting telomeres. This accelerated shortening of telomeres can result in premature cellular senescence, contributing to the progression of T2DM [54]. Several cross-sectional studies have shown that patients with diabetes have shorter telomeres than those without diabetes, regardless of the type of diabetes [55]. In a population-based prospective study, researchers evaluated leucocyte relative telomere length (RTL) by quantitative polymerase chain reaction. They conducted repeat RTL measurements in 2005 (n = 558) and 2010 (n = 479), and found that low RTL was independently associated with the risk of incident T2DM [56]. Furthermore, a prospective study investigated 93 women with gestational diabetes and 186 non-diabetic women as a control group, and revealed no significant correlation between TL and gestational diabetes overall. However, in women under the age of 30, there was a certain association between TL and gestational diabetes, with a higher risk observed in those with shorter telomeres [57, 58]. Cross-sectional research findings vary, with some indicating that shorter TL is related to a twofold increased risk of gestational diabetes, while others found no connection [59]. In addition, impaired beta cell replication plays an important role in the occurrence and development of type 2 diabetes. Telomerase deficiency and consequent telomere shortening impair the replicative capacity of pancreatic β-cells [60]. This impairment leads to reduced insulin secretion and glucose intolerance [60]. Researchers built mouse models to study changes in pancreatic islet β-cell function. They discovered that in mice with type 2 diabetes, the distribution of β-cell subtypes shifted. Significantly, one subtype showed increased basal insulin secretion but a weakened response to glucose stimulation [61].

It is worth mentioning that diabetes treatment may affect TL. For instance, physical exercise, dietary control, medication, and other interventions can help alleviate oxidative stress and inflammatory conditions, consequently promoting the elongation of telomeres. This improvement in telomere length can enhance the condition and prognosis of individuals with diabetes. Silvia Canudas and colleagues conducted a systematic review of the relationship between the Mediterranean diet (MedDiet) and TL, involving 13,733 participants from five countries demonstrated a positive correlation between the MedDiet and TL [62].

Liver diseases

Non-alcoholic fatty liver disease (NAFLD) is characterized by the accumulation of fat in the liver and can progress to non-alcoholic steatohepatitis (NASH), liver fibrosis, or even hepatocellular carcinoma (HCC). Research indicates a significant association between telomere length and the progression of NAFLD [6365]. The TL likely a crucial factor in this complex connection, causing liver cells to become senescent, which in turn affects the overall health of the liver and contributes to the progression of NAFLD. The results of a controlled experimental investigation showed that the length of hepatocyte telomeres was shorter in individuals with NAFLD than in controls [66]. In a recent prospective study involving 467,848 participants from the UK Biobank, with a follow-up period of 12.83 years, 4809 patients with NAFLD were identified and TL exhibited a negative correlation with the incidence of NAFLD [65]. Moreover, a study revealed that individuals afflicted with NAFLD exhibited elongated telomeres compared to age-matched counterparts [67]. However, certain investigations have not confirmed an inverse relationship between the incidence of NAFLD and TL, which may be attributed to differences in sample size and study methodologies [68]. In comparison with non-cirrhosis controls, cirrhosis patients display shorter telomeres and an increased occurrence of telomerase mutations. These mutations often trigger uncontrolled hepatocellular proliferation, predisposing these patients to the onset of hepatocellular carcinoma [69, 70]. Furthermore, telomerase-deficient mice exposed to chronic liver injury exhibit accelerated progression of liver fibrosis [71]. This suggests that telomere dysfunction may play a crucial role in the advancement of NAFLD to liver fibrosis and liver cancer.

Hyperuricemia and gout

Hyperuricemia and gout are becoming increasingly prevalent, with hyperuricemia being the primary risk factor for gout [72]. Uric acid affects cells in multiple ways, including promoting reactive ROS production, which contributes to inflammation at deposition sites within blood vessels and joints. A recent prospective study in China conducted a longitudinal analysis of 599 participants to investigate whether shorter telomeres could predict hyperuricemia. After adjusting for risk factors, the study followed 266 participants from June 2014 to December 2021 and found that shorter leukocyte TL was significantly associated with the development of hyperuricemia during the follow-up period (odds ratio [OR] 2.347; 95% confidence interval [CI] 1.123–4.906; P = 0.023) [73]. Similarly, a comparative study examining TL in 145 gout patients and 273 healthy individuals found that gout patients had shorter telomeres and experienced greater telomere erosion at all ages. Moreover, TL was negatively correlated with the frequency of gout attacks [74].

Cardiovascular diseases

Cardiovascular disease (CVD) is one of the leading causes of death worldwide, with a complex etiology influenced by both genetic and environmental factors. CVD encompasses a range of conditions affecting the heart and blood vessels, including heart disease, hypertension, myocardial infarction, and stroke. According to the World Health Organization, CVD accounts for approximately 17.9 million deaths annually, a number that continues to rise [75]. Major risk factors for CVD include physical inactivity, an unhealthy diet, smoking, and preexisting medical conditions [76]. Numerous studies have identified a strong association between TL and heart disease [77, 78]. The pathological progression of CVD is often accompanied by oxidative stress and chronic inflammation, both of which contribute to telomere shortening and dysfunction.

Cardiomyopathy

As the heart ages, myocardial hypertrophy and fibrosis lead to increased ventricular stiffness and impaired cardiac function [79]. A meta-analysis of 18,293 patients across 12 studies found that leukocyte telomere length (LTL) can serve as a predictor of atrial fibrillation [80]. Additionally, a study involving telomere gene testing identified genetic variants associated with shorter telomeres, which were linked to an increased risk of coronary artery disease in a cohort of 22,233 patients and 64,762 controls [81]. Telomere shortening also plays a role in cardiac aging. Myocardial hypertrophy and fibrosis are characteristic features of this process, and patients with genetic hypertrophic or dilated cardiomyopathy exhibit shorter myocardial cells compared to age-matched individuals [23]. A large independent cohort study further demonstrated a correlation between LTL and the severity of hypertrophic cardiomyopathy [82]. In a prospective population-based study, Willeit et al. assessed relative LTL using PCR in adults aged 45–84 years, comparing individuals without cardiovascular disease (CVD) to those with stable coronary artery disease (CAD). Their findings revealed that LTL in CAD patients was significantly shorter than in healthy individuals of the same age [83]. These findings suggest that cardiovascular diseases may influence telomere length, with conditions such as myocardial infarction and heart failure accelerating telomere shortening. This creates a vicious cycle that exacerbates cardiac damage and functional decline. Animal studies further support this connection. Two studies in mice revealed that their naturally long telomeres offer protection against heart disease. Researchers found that mice only developed heart conditions similar to those seen in humans after their telomeres were genetically shortened to human-like lengths [34, 84]. The research also suggests that shorter telomeres may impair heart function by disrupting mitochondrial activity. When mitochondria malfunction, as observed in dilated cardiomyopathy, the resulting energy deficiency can lead to severe heart failure [34, 84].

Atherosclerosis

Atherosclerosis is indeed a chronic and progressive arterial disease characterized by inflammatory changes within the arteries. These changes are typically associated with alterations in lipid metabolism and other metabolic factors. One study revealed that patients with atherosclerosis have shorter telomeres in circulating leukocytes compared to their healthy, age-matched counterparts [85]. Furthermore, vascular smooth muscle cells (VSMCs) in atherosclerotic plaques display a significantly shorter TL than unaffected VSMCs in the same individual [23]. A reduction in TL in the of VSMCs within atherosclerotic plaques correlates with the severity of the disease. Additionally, VSMCs in atherosclerotic plaques exhibit oxidative DNA damage and express various markers of aging. Accelerated telomere attrition not only promotes atherosclerosis but also impairs vascular lesion repair, exacerbates endothelial damage, and enhances oxidative stress and inflammatory processes, thus accelerating telomere shortening-related atherosclerosis.

Hypertension

Hypertension is one of the most prevalent cardiovascular diseases, affecting individuals across all ethnicities worldwide. The relationship between TL and hypertension is thought to involve telomere shortening, which contributes to cellular aging and dysfunction, ultimately increasing the risk of hypertension [86]. Telomere dysfunction-induced cellular senescence in arterial walls may play a role in the pathogenesis of hypertension [87]. Research on spontaneously hypertensive rats (SHR) revealed that telomerase is selectively activated in the aorta, leading to alterations in telomere maintenance and the p53 checkpoint, which together promote VSMC proliferation. Inhibiting telomerase activity reduced VSMC proliferation and induced apoptosis in SHR, suggesting a potential therapeutic target for hypertension [88]. A case–control study involving 206 patients with essential hypertension (EH) and an equal number of sex-matched healthy controls found that EH patients had significantly shorter TL [89]. However, a genome-wide association study (GWAS) reported that longer TL was associated with a higher risk of hypertension—contradicting previous findings on coronary artery atherosclerosis, ischemic heart disease, myocardial infarction, and stroke [75]. These inconsistencies highlight the complex and still inconclusive relationship between TL and hypertension, underscoring the need for further research in this area.

Pulmonary diseases

Frequently encountered pulmonary diseases encompass idiopathic pulmonary fibrosis (IPF), chronic obstructive pulmonary disease (COPD), among other conditions. Numerous studies have suggested a significant correlation between pulmonary diseases and the aging process [90].

IPF

IPF is a progressive lung disease that primarily affects older individuals and ultimately leads to death. Its exact cause remains unknown [90]. The disease is characterized by excessive extracellular matrix deposition within the lung interstitium, resulting in abnormal scar formation and impaired lung function [91]. Telomere dysfunction plays a crucial role in IPF development. It accelerates fibrosis progression by inducing cellular senescence, particularly in alveolar epithelial cells, thereby reducing the lung’s regenerative capacity. This process triggers inflammatory responses and compromises stem cell function, further contributing to disease onset and progression. In experimental studies, two independent mouse models—one with severe telomere shortening due to telomerase deficiency and another with TRF1 protein deletion in alveolar type II epithelial cells—both developed pulmonary fibrosis, highlighting the direct impact of telomere dysfunction on lung pathology [92]. Genetic studies provide further evidence of this association. Approximately 40% of familial IPF cases and 25% of sporadic IPF cases are linked to shorter telomeres, often due to mutations in TERT or TERC [91]. An analysis of 73 probands from the vanderbilt familial pulmonary fibrosis registry identified heterozygous mutations in these genes in six individuals [81]. Beyond familial cases, sporadic IPF patients also exhibit telomere shortening [93]. A study comparing TL in alveolar epithelial cells from individuals with normal lung function, sporadic IPF, and IPF with telomerase mutations found that both IPF groups had significantly shorter telomeres than healthy controls [94]. Additionally, large-scale genetic research has reinforced this link. A multigene risk score analysis involving 400,000 individuals and one million controls identified pulmonary fibrosis as the disease most strongly associated with short telomeres [95]. Moreover, shorter TL has been correlated with reduced time to allograft dysfunction following lung transplantation, underscoring its clinical significance [96].

Chronic obstructive pulmonary disease

COPD significantly heightens morbidity and death in elderly individuals [97]. COPD accelerates the aging of lungs, marked by inflammation and oxidative stress in lung tissue and airways, along with persistent restructuring of the smaller air passages, eventually resulting in emphysema [23]. Telomere shortening can result in the aging and functional decline in lung tissue, which in turn increases the likelihood of developing COPD in patients. Small airway epithelial cells from COPD patients exhibit increased levels of telomere-associated foci (TAFs) and senescence markers compared to healthy individuals [98]. Various studies have consistently shown that the TL in the blood or lung tissue of people with COPD is generally shorter than that in the blood or lung tissue of healthy individuals [98, 99]. In COPD, telomere shortening may indicate cellular aging processes, and aged pulmonary cells may impact the progression of airflow restriction. Moreover, the presence of COPD can impact the length of telomeres through the induction of systemic inflammation [100]. Research indicates that patients with COPD have higher levels of a wide range of inflammatory mediators in their lungs and blood [101]. Shortening of telomeres leads to cellular aging and the release of inflammatory mediators, contributing to heightened incidence and severity of airway inflammation.

Bronchiectasis

Bronchiectasis is a chronic lung disease characterized by persistent inflammation and airway damage. Its development is closely linked to dysfunction and impaired repair capacity of airway epithelial cells [90]. These cells serve as the first line of defense against environmental insults and pathogens, maintaining airway integrity. Telomere attrition is thought to weaken their proliferative and reparative abilities, compromising the epithelial barrier. Histological analysis of lung tissue from affected individuals has shown shorter telomeres and increased expression of senescence markers [102]. Han et al. explored accelerated aging in bronchiectasis by examining aging-related markers in both systemic circulation and bronchial epithelium. Their findings indicate that individuals with bronchiectasis have shorter telomeres and lower levels of SIRT1 and Ku80 in peripheral blood compared to healthy controls [103].

Kidney diseases

The kidneys undergo both structural and functional alterations as they age. Various conditions such as acute kidney injury, glomerulonephritis, diabetic nephropathy, polycystic kidney disease, and chronic kidney disease (CKD) are linked to TL. TL is impacted by several variables, including oxidative stress and inflammation, which are commonly found in renal disorders.

Chronic kidney disease

Repetitive aging, stress-induced premature aging, and epigenetic abnormalities contribute to the development of CKD. Genetic damage and environmental stressors drive oxidative stress and chronic inflammation, leading to irreversible cell cycle arrest and increased secretion of the SASP [104]. Telomere shortening or damage can activate a persistent DDR, promoting cellular senescence [23]. In CKD, increased DNA damage has been observed, as evidenced by elevated DDR markers such as γH2AX in kidney tissues [105]. The disease is highly prevalent among the elderly, and numerous studies have linked shorter telomeres to CKD [106108]. One study found that for every 0.1-unit decrease in relative TL, the risk of mortality increased by 14%, independent of age, sex, kidney function, and cardiovascular risk factors [107]. Another study of over 2,000 patients with type 2 diabetes mellitus (T2DM) showed that those with shorter leukocyte TL had a 1.9-fold higher risk of albuminuria progression after adjusting for established risk factors [109]. In CKD, premature cellular aging can result from chronic inflammation and metabolic abnormalities, contributing to telomere attrition. Additionally, telomere shortening has been associated with increased inflammation and oxidative stress in CKD [108]. Both factors are common in CKD patients, and researchers speculate that telomere attrition may play a role in disease onset by exacerbating these pathological processes [110].

Kidney fibrosis

Kidney fibrosis is a hallmark clinical feature observed in nearly all cases of CKD, yet its pathophysiology remains poorly understood. The development of renal fibrosis is influenced by multiple factors, including genetic predisposition, lifestyle choices, and environmental exposures. Cellular injury often serves as the initial trigger, activating a cascade of inflammatory responses and reparative processes that culminate in collagen deposition and fibroblast proliferation, ultimately forming fibrotic scar tissue [111]. Emerging evidence suggests a strong link between telomere shortening, telomere dysfunction, and renal fibrosis [23]. Saraswati et al. demonstrated that short telomeres in mice can exacerbate the epithelial-to-mesenchymal transition (EMT), increasing renal susceptibility to folate-induced fibrosis and accelerating fibrotic progression. Additionally, the absence of Trf1, a key component of the shelterin complex, was shown to induce kidney fibrosis. Telomerase-deficient mice with shortened telomeres also developed significant renal fibrosis and impaired kidney function following minimal folic acid exposure [112].

Neurodegenerative diseases

Studies suggest a discernible correlation between TL and neurodegenerative diseases. Neurodegenerative diseases, including Alzheimer's disease (AD) and Parkinson's disease (PD), among others, are a group of conditions characterized by the gradual dysfunction or death of neurons.

Alzheimer’s disease

By 2050, AD, the most prevalent neurodegenerative disorder, is projected to affect over 100 million people worldwide, with the majority in low- and middle-income countries. Age is the primary risk factor for AD, with its impact becoming more pronounced as individuals grow older. Notably, research has demonstrated that shorter telomeres accelerate the natural aging process in humans [16]. A meta-analysis [113] of 13 studies comparing telomere length (TL) in 860 individuals with AD and 2022 controls revealed significant differences in TL between the groups. One study of 1983 participants aged 65 and older found that individuals with shorter telomeres had higher mortality rates and an increased risk of developing AD during follow-up [114]. Moreover, a significant correlation was observed between white blood cell TL and hippocampal volume. In non-demented individuals carrying the APOE3/3 genotype, a genetic variant associated with AD susceptibility, a negative correlation was identified between white blood cell TL and hippocampal volume. Additionally, AD model mice exhibited shorter blood cell telomeres compared to wild-type mice, though no such difference was observed in hippocampal telomere length [115].

Parkinson’s disease

PD, the second most prevalent neurodegenerative disorder after AD, is characterized by motor dysfunction resulting from neuronal loss in the substantia nigra and subsequent dopamine depletion in the striatum [116]. Telomere shortening in leukocytes and microglia may indirectly affect neuronal health by impairing immune cell function in the brain [117]. While no definitive population-based evidence currently establishes a direct link between PD and telomere shortening, ongoing research continues to explore this potential connection [118]. A meta-analysis of eight primary studies, including 956 individuals with PD and 1284 controls, found no statistically significant difference in TL between the two groups [119]. However, postmortem brain samples from PD patients have revealed the presence of senescent astrocytes [120]. Additionally, mice with critically short telomeres exhibited impaired neuromuscular coordination in performance tests [121]. These findings suggest a potential association between TL and PD, highlighting the need for further investigation into the biological mechanisms linking telomere dynamics and PD pathogenesis, which may inform future therapeutic strategies.

Skeletal disorders

Osteoporosis

Osteoporosis, characterized by reduced bone density and deterioration of bone microstructure [122], has been linked to the presence of senescent cells in the body [23]. Similar to many other tissues, telomeres in osteoblasts and mesenchymal stem cells (MSCs) shorten with age [7]. Telomere dysfunction and osteoporosis are also observed in premature aging syndromes, such as Werner syndrome and congenital keratinization disorders. An early cohort study by Valdes et al. examined LTL and its association with bone density in women, revealing an independent correlation between shorter LTL and decreased bone density, particularly in postmenopausal women aged 50 and above [123]. Additionally, aging is associated with increased levels of reactive oxygen species, which exacerbate oxidative stress, impair bone formation [124], and contribute to telomere shortening. However, some studies have found no significant association between leukocyte TL and osteoporosis. [125]. For instance, a health assessment study of 2750 community-dwelling elderly individuals reported no significant correlation between leukocyte TL, bone mineral density (BMD), bone loss, or fracture incidence [125]. In conclusion, further research is necessary to clarify the complex relationship between telomere length and osteoporosis.

Osteoarthritis

Osteoarthritis is defined by a decrease in bone mineral density and degeneration of bone tissue structure, leading to an increased likelihood of fractures [23]. In the context of osteoarthritis, several joint tissues, such as articular cartilage, synovium, and subchondral bone, exhibit increased numbers of senescent cells. These cells not only lose their ability to maintain tissue homeostasis but also secrete SASP factors that promote inflammation and matrix degradation, exacerbating joint degeneration [126]. Although age is recognized as a predictor of osteoarthritis, the precise underlying mechanism remains uncertain. Previous studies have revealed many aging markers in osteoarthritis [126]. Researchers found in a study on hand osteoarthritis and TLs that the shorter TL differed by approximately 11 years from those of normal people and was associated with the severity of the disease [127]. The damage and degradation of articular cartilage also correlate with TL shortening. In osteoarthritis, studies have found that many aging markers are deposited in chondrocytes [128]. Researchers showed that the shorter TL was associated with a higher degree of hand osteoarthritis and differed by approximately 11 years from that of normal individuals. There is a correlation between articular cartilage injury, deterioration, and TL shortening. A multitude of age markers have been found in chondrocytes linked to osteoarthritis. According to research, TL shortening may affect the ability of chondrocytes to divide and repair themselves. This could accelerate the deterioration of joint cartilage and increase the likelihood of osteoarthritis. After comparing the two populations, they also discovered more aging cells in the older population than in the younger group. Currently, researchers believe that extremely short telomeres provide a more accurate indicator of osteoarthritis damage than typical TL [129].

Discussion

In the past, scientists recognized telomeres as among the most accurate markers of aging, as aging diseases closely correlate with accelerated telomere shortening and telomere dysfunction [130]. Researchers discovered that during aging, DNA damage-induced changes in the rate of replication forks, commonly referred to as replication stress, can lead to asymmetric fork progression, incomplete replication, fork collapse, and genomic instability, preferentially targets telomeres with G-repeat-rich. Notably, up to half of the foci observed in stress-induced aging were located on telomeres [22], as telomeres can form stable G-quadruplexes that impede the progression of replication forks. Therefore, repairing collapsed replication forks or inhibiting the formation of G-quadruplex structures by targeting them during replication stress may represent an effective strategy to delay telomere shortening and combat the progression of age-related diseases. TL shortening appears to be associated with oxidative stress, inflammatory responses, mitochondrial dysfunction, and other aging-related factors. This association primarily arises from the fact that chronic inflammation and oxidative stress can lead to guanine oxidation, which results in telomere shortening and instability. Consequently, these processes initiate aging and contribute to the associated degeneration observed in humans [30, 131]. In addition, lifestyle is also closely related to telomeres [132]. Diet, life stress, habits, etc., will affect telomeres. One study analyzing single-molecule telomere length and telomere fusion found that the frequency of short telomeres and telomere aberrant events increases with disease progression, and samples with short telomeres exhibit large-scale genomic rearrangements [22]. Telomeres impede the progression of replication forks, which can lead to stalling. When the replication fork becomes stalled, it may collapse, resulting in the formation of DNA double-strand breaks (DSBs). Telomere aberrant can occur during both classical and alternative non-homologous end-joining repair pathways that repair DSBs. Consequently, telomere shortening associated with aging may lead to telomere aberrant, further contributing to telomere dysfunction. In this context, the inhibition of DNA repair mechanisms may prevent the aberrations associated with short telomeres [22]. Moreover, telomeres protect the ends of linear eukaryotic chromosomes from being recognized as DNA double-strand breaks. Two major protein complexes are involved in the protection of telomeres: shelterin and CST; thus, the possibility of targeting protective proteins and CSTs through small-molecule inhibitors deserves further research and exploration [22]. However, the current study cannot determine whether telomere changes are the cause or result of aging [133]. Further research is needed to analyze its causality.

The epidemiological relationship between TL and disease and mortality is not always clear and can even be contradictory. Therefore, it is necessary to specify a standardized measurement standard to specify a standard TL reference range. When analyzing the data of different studies (Fig. 1 and Table 1), we noticed a lack of a unified and standardized reference range of TL for healthy people of different ages. We found significant differences in TL reported by different studies in healthy people. Some studies have shown that TL tends to decrease with age [13], while no obvious correlation was observed in other studies. This inconsistency in data may be partly due to the use of different measurement techniques, differences in sample sources, and limitations in study design. Considering the lack of a unified and standardized reference range of TL in healthy people at different ages, we believe that it is highly important to establish such a reference range. Firstly, this will help to assess the health status of individual TLs more accurately and better identify potential health risks, aiding in prevention of chronic diseases. Second, a unified reference range can promote comparisons and data sharing between different studies, thus promoting the progress of TL research. To establish a uniform reference range for TL, we recommend a multicenter, large sample prospective cohort study. Such a study can cover healthy people of different ages, genders, ethnicities, and geographical backgrounds to ensure the representativeness and universality of the reference range. In addition, standardized measurement techniques and analysis methods should be adopted to reduce measurement errors and uncertainty in the results.

Fig. 1.

Fig. 1

Telomere length and aging-related diseases. A schematic overview of the aging-related diseases discussed in this Review, organized by organs or systems. AD, Alzheimer’s disease; CKD, chronic kidney disease; COPD, chronic obstructive pulmonary disease; IPF, idiopathic pulmonary fibrosis; NAFLD, non-alcoholic fatty liver disease; PD, Parkinson’s disease; T2D, type 2 diabetes

Table 1.

Associations between telomere length and aging-related diseases

Disease Study type Method Expression of TL Cases Controls References
Type No Type No
T2DM Meta-Analysis q-PCR T/S, short without T2DM 606 N/A N/A [56]
T2DM Observational q-FISH T/S, short T2DM AND CVD 204 without T2DM and CVD/without T2DM but CVD 258/798 [139]
T2DM MR q-PCR T/S, short T2DM 569 without T2DM 448 [140]
T2DM Randomized Controlled Trial q-PCR T/S, short CHD and Mediterranean diet/CHD and low-fat diet 502/500 without CHD 462 [141]
T2DM Observational q-PCR T/S, short T2DM 930 without T2DM 867 [142]
GDM Observational q-PCR T/S, short GDM 93 without GDM 186 [57]
DN Observational Southern Blot kb, short T1DM 132 without DN 44 [143]
NAFLD Observational q-PCR T/S, short NAFLD 60 without NAFLD 23 [64]
NAFLD Observational q-FISH T/S, short NAFLD 70 without NAFLD 60 [66]
CLD Observational q-PCR T/S, short CLD 521 without CLD 600 [69]
Gout Observational q-PCR T/S, short GOUT 145 without Gout 273 [74]
Hyperuricemia MR q-PCR T/S, short Hyperuricemia 488/400 N/A N/A [144]
CAD Observational q-PCR T/S, < Q1 CAD 566 N/A N/A [145]
CAD MR N/A N/A CAD 22,233 without CAD 64,762 [81]
CAD Observational Southern Blot kb, short CAD 484 without CAD 1058 [146]
HCM Observational q-PCR T/S, short HCM 56 without HCM 59 [82]
CVD Observational q-PCR T/S, short PAD and T2DM 241 without PAD but T2DM 249 [147]
CA Observational Southern Blot kb, short CA and hypertension 90 without Carotid plaque but hypertension 73 [148]
Hypertension Observational Southern Blot kb, short male with hypertension 120 female with hypertension 73 [149]
IPF Observational q-FISH Kb, short IIP 62 without IIP 400 [94]
IPF Observational q-PCR Kb, short Dallas IPF cohort/Chicago IPF cohort/San Francisco IPF cohort 149/139/54 Dallas Cohorts: without IPF 221 [150]
COPD Meta-Analysis N/A N/A COPD 934 without COPD 15,846 [99]
COPD Observational q-PCR Kb, short COPD and smoking 70 smoker without COPD 73 [151]
Bronchiectasis Observational q-PCR T/S, short Bronchiectasis 8 without Bronchiectasis 7 [102]
CKD Randomized Controlled Trial q-PCR T/S, short CKD and n-3 FA/CKD and CoQ/CKD and n-3 FA + CoQ 19/21/18 CKD and placebo 15 [152]
CKD Observational q-PCR T/S, Q1-Q4 CKD 4955 N/A N/A [153]
AD Observational q-PCR T/S, short AD 32 Stable aMCI/Healthy people 13/20 [154]
AD Observational q-PCR T/S, Q1-Q5 aMCI 137 without aMCI 137 [155]
PD Observational q-PCR T/S, short PD 168 without PD 30 [156]
OP Observational Southern Blot kb, long OP 2150 without OP 2150 [123]
OA Observational Southern Blot kb, short OA 160 without OA 926 [127]

AD: Alzheimer’s disease; aMCI: Amnestic mild cognitive impairment; CA: Carotid atherosclerosis; CAD: Coronary artery disease; CKD: Chronic kidney disease; CLD: Chronic liver disease; COPD: Chronic obstructive pulmonary disease; CVD: Cardiovascular disease; DN: Diabetic nephropathy; GDM: Gestational diabetes mellitus; HCM: Hypertrophic cardiomyopathy; IIP: Idiopathic interstitial pneumonias IPF: Idiopathic pulmonary fibrosis; NAFLD: Non-alcohol-related fatty liver disease; OA: Osteoarthritis; OP: Osteoporosis; PD: Parkinson's disease; T2DM: Type 2 diabetes mellitus

Although the previous text did not mention cancer, cancer is closely related to telomeres and their associated factors [134]. When short telomeres induce the DDR, they can lead to chromosomal end-to-end fusions, breakages, and rearrangements, resulting in chromosomal instability [9, 20]. Chromosomal instability is one of the hallmarks of cancer [135]. Additionally, DDR promotes cellular senescence. Senescent cells secrete a variety of pro-inflammatory cytokines, growth factors, and proteases, collectively known as the SASP. For example, SASP factors can alter the tissue microenvironment, making it more conducive to cancer development [136]. These factors can stimulate nearby cells to proliferate more actively, enhance angiogenesis (the growth of new blood vessels), and create a permissive environment for cancer cells to thrive. Furthermore, if senescent cells are not cleared by the immune system, they can accumulate over time, exacerbating inflammation and further promoting cancer progression. Cancer cells typically exhibit uncontrolled cell proliferation and unlimited division capabilities, attributed in part to their aberrant regulation of telomerase. Previous research has highlighted the crucial and intricate roles of TERT and its various subtypes in telomerase activity [137]. A study investigated 14,830 samples, and among 6,835 cases of cancer, 73% expressed TERT [138]. While there is substantial evidence suggesting an association between TL and age-related diseases, establishing a causal relationship requires longitudinal studies and the evaluation of other markers of telomere biology to elucidate the role of telomeres and telomerase in cancer.

Although telomeres are widely recognized as one of the precise biomarkers of aging, the complexity and cost of TL measurement methods pose challenges that limit their application in clinical practice. However, with ongoing technological advancements, we anticipate that TL measurements could be more extensively utilized in clinical diagnostics and therapeutics.

Potential interventions

Telomeres, the protective caps at the ends of chromosomes, play a crucial role in maintaining genomic stability and cellular lifespan [1]. As cells divide, telomeres progressively shorten, eventually leading to cellular senescence or apoptosis. Interventions targeting telomere biology hold potential for addressing age-related diseases and extending healthy lifespan.

Strategies for telomerase activation

Small molecule activator

In recent years, certain small-molecule compounds have been found to have the potential to activate telomerase activity. For example, components extracted from traditional Chinese herbs (such as TA-65) have shown telomere-lengthening effects in both in vitro studies and animal models [157]. The primary mechanism of action involves upregulating the expression of TERT, thereby slowing telomere shortening and the process of cellular aging. It is important to note that while telomerase activation may help delay aging, its high activity in most cancer cells raises concerns about the potential risk of inducing abnormal cell proliferation with long-term use. Therefore, rigorous safety assessments should be conducted before clinical application [157].

Gene therapy strategy

Gene therapy introduces the TERT gene into the body using viral vectors or other delivery systems, thereby directly enhancing intracellular telomerase activity [158]. A study by Bernardes de Jesus et al. in a mouse model demonstrated that this strategy not only extends telomeres but also improves tissue function, delays aging symptoms, and, to some extent, prolongs lifespan [159]. Moreover, the study did not identify significant cancer risks during the evaluation process. However, due to the inherent challenges of gene therapy, such as immune responses and long-term regulatory issues, further research is needed before its application in humans [159].

Strategies for lifestyle change

Balanced diet

A balanced diet (such as the Mediterranean diet) is rich in antioxidants, fiber, polyphenols, and omega-3 fatty acids, which help reduce oxidative stress and chronic inflammation in the body, thereby protecting telomeres from damage [160, 161]. Research has shown that a high antioxidant diet not only helps maintain telomere length, but is also associated with a reduced risk of cardiovascular disease, metabolic syndrome, and other conditions [162]. Therefore, a reasonable dietary structure is considered an important non-pharmacological intervention to delay cellular aging.

Moderate exercise

Regular and moderate exercise can improve cardiovascular health, regulate endocrine function, and reduce inflammation levels, thereby indirectly protecting telomeres [163]. During physical activity, the body releases various beneficial factors that enhance antioxidant defense and cellular repair capacity. Numerous epidemiological studies have shown that individuals with higher levels of physical activity generally have longer leukocyte telomeres, providing important evidence for the role of exercise interventions in anti-aging [164].

Stress management

Chronic psychological stress is considered one of the key external factors accelerating telomere shortening, primarily through the prolonged activation of the hypothalamic–pituitary–adrenal (HPA) axis [165, 166]. Under persistent stress, the body continuously secretes cortisol, a stress hormone that not only increases oxidative stress levels but also promotes the release of inflammatory factors, leading to negative effects on cells and accelerating telomere shortening [167]. To counter these adverse effects, various stress management strategies have been proposed and have shown potential in protecting telomeres. For example, mindfulness meditation helps individuals focus on the present moment, thereby reducing anxiety and emotional fluctuations [168]. Studies have shown that after a period of mindfulness training, participants experience significantly lower cortisol levels and improved inflammatory biomarkers [169]. Additionally, practicing yoga, which integrates physical postures, breath control, and meditation, can help alleviate both physical and psychological tension. Deep breathing exercises activate the parasympathetic nervous system, reducing heart rate and blood pressure, thereby mitigating stress responses. Lastly, maintaining good sleep quality is also crucial, as individuals with sufficient and high-quality sleep tend to exhibit lower stress levels and longer leukocyte telomeres [170].

Conclusions

We have outlined the connection between TL and aging-related disorders here. TL reduction is directly linked to the development and occurrence of many chronic diseases, including diabetes, cancer, and cardiovascular disease. This emphasizes how crucial TL is for preserving cellular health and function. Environmental factors such as stress, pollution, and poor lifestyles are linked to aberrant telomere shortening, providing additional evidence of the influence of lifestyle on cellular health. Therefore, an improved lifestyle may facilitate the maintenance of healthy TLs and lower the risk of disease. Despite numerous studies suggesting that TL is a prognostic indicator for aging-related disorders and mortality, clinical practice currently underutilizes its detection. Strengthening communication between the laboratory and clinical settings is essential for laying the groundwork for its clinical application.

Acknowledgements

We thank the Key Laboratory of Aging and Cancer Biology of Zhejiang Province, Hangzhou Wuyunshan Hospital, Zhuji People’s Hospital of Zhejiang Province, for their support.

Author contributions

X.H. and L.X. were involved in the conceptualization; X.H., L.H., and S.Z. contributed to writing—preparation of the original draft; X.H., J.L., and L.X. wrote the original draft; X.L., L.L., and L.C. contributed to the review and editing; D.W. and L.X. supervised and had leadership responsibility for the planning and execution of the research activity, including mentorship to the core team. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by grants from the Department of Science and Technology of Zhejiang Province (2023C03166) and Hangzhou Normal University (4285C50223204005).

Data availability

No datasets were generated or analyzed during the current study.

Declarations

Conflict of interest

The authors declare no competing interests.

Informed consent

Not applicable.

Footnotes

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

References

  • 1.Turner KJ, Vasu V, Griffin DK. Telomere biology and human phenotype. Cells. 2019. 10.3390/cells8010073. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.de Lange T. Shelterin-mediated telomere protection. Annu Rev Genet. 2018;52:223–47. 10.1146/annurev-genet-032918-021921. [DOI] [PubMed] [Google Scholar]
  • 3.Lim CJ, Cech TR. Shaping human telomeres: from shelterin and CST complexes to telomeric chromatin organization. Nat Rev Mol Cell Biol. 2021;22:283–98. 10.1038/s41580-021-00328-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Blasco MA. Telomere length, stem cells and aging. Nat Chem Biol. 2007;3:640–9. 10.1038/nchembio.2007.38. [DOI] [PubMed] [Google Scholar]
  • 5.Lin J, Epel E. Stress and telomere shortening: insights from cellular mechanisms. Ageing Res Rev. 2022;73: 101507. 10.1016/j.arr.2021.101507. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.van Steensel B, Smogorzewska A, de Lange T. TRF2 protects human telomeres from end-to-end fusions. Cell. 1998;92:401–13. 10.1016/s0092-8674(00)80932-0. [DOI] [PubMed] [Google Scholar]
  • 7.Herrmann M, Pusceddu I, Marz W, Herrmann W. Telomere biology and age-related diseases. Clin Chem Lab Med. 2018;56:1210–22. 10.1515/cclm-2017-0870. [DOI] [PubMed] [Google Scholar]
  • 8.Chan SR, Blackburn EH. Telomeres and telomerase. Philos Trans R Soc Lond B Biol Sci. 2004;359:109–21. 10.1098/rstb.2003.1370. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Hemann MT, Strong MA, Hao LY, Greider CW. The shortest telomere, not average telomere length, is critical for cell viability and chromosome stability. Cell. 2001;107:67–77. 10.1016/s0092-8674(01)00504-9. [DOI] [PubMed] [Google Scholar]
  • 10.Shay JW, Wright WE. Telomeres and telomerase: three decades of progress. Nat Rev Genet. 2019;20:299–309. 10.1038/s41576-019-0099-1. [DOI] [PubMed] [Google Scholar]
  • 11.De Meyer T, Nawrot T, Bekaert S, De Buyzere ML, Rietzschel ER, Andres V. Telomere length as cardiovascular aging biomarker: JACC review topic of the week. J Am Coll Cardiol. 2018;72:805–13. 10.1016/j.jacc.2018.06.014. [DOI] [PubMed] [Google Scholar]
  • 12.Schneider CV, Schneider KM, Teumer A, Rudolph KL, Hartmann D, Rader DJ, Strnad P. Association of telomere length with risk of disease and mortality. JAMA Intern Med. 2022;182:291–300. 10.1001/jamainternmed.2021.7804. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Blackburn EH, Greider CW, Szostak JW. Telomeres and telomerase: the path from maize, tetrahymena and yeast to human cancer and aging. Nat Med. 2006;12:1133–8. 10.1038/nm1006-1133. [DOI] [PubMed] [Google Scholar]
  • 14.Wang Y, Susac L, Feigon J. Structural biology of telomerase. Cold Spring Harb Perspect Biol. 2019. 10.1101/cshperspect.a032383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Bernardes de Jesus B, Blasco MA. Telomerase at the intersection of cancer and aging. Trends Genet. 2013;29:513–20. 10.1016/j.tig.2013.06.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Lopez-Otin C, Blasco MA, Partridge L, Serrano M, Kroemer G. The hallmarks of aging. Cell. 2013;153:1194–217. 10.1016/j.cell.2013.05.039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Lopez-Otin C, Blasco MA, Partridge L, Serrano M, Kroemer G. Hallmarks of aging: an expanding universe. Cell. 2023;186:243–78. 10.1016/j.cell.2022.11.001. [DOI] [PubMed] [Google Scholar]
  • 18.Tomas-Loba A, Flores I, Fernandez-Marcos PJ, Cayuela ML, Maraver A, Tejera A, Borras C, Matheu A, Klatt P, Flores JM, et al. Telomerase reverse transcriptase delays aging in cancer-resistant mice. Cell. 2008;135:609–22. 10.1016/j.cell.2008.09.034. [DOI] [PubMed] [Google Scholar]
  • 19.Watson JD. Origin of concatemeric T7 DNA. Nat New Biol. 1972;239:197–201. 10.1038/newbio239197a0. [DOI] [PubMed] [Google Scholar]
  • 20.Di Micco R, Krizhanovsky V, Baker D, d’Adda di Fagagna F. Cellular senescence in ageing: from mechanisms to therapeutic opportunities. Nat Rev Mol Cell Biol. 2021;22:75–95. 10.1038/s41580-020-00314-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Cesare AJ, Karlseder J. A three-state model of telomere control over human proliferative boundaries. Curr Opin Cell Biol. 2012;24:731–8. 10.1016/j.ceb.2012.08.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Hewitt G, Jurk D, Marques FD, Correia-Melo C, Hardy T, Gackowska A, Anderson R, Taschuk M, Mann J, Passos JF. Telomeres are favoured targets of a persistent DNA damage response in ageing and stress-induced senescence. Nat Commun. 2012;3:708. 10.1038/ncomms1708. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Rossiello F, Jurk D, Passos JF, d’Adda di Fagagna F. Telomere dysfunction in ageing and age-related diseases. Nat Cell Biol. 2022;24:135–47. 10.1038/s41556-022-00842-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Papathanassoglou ED, Moynihan JA, Ackerman MH. Does programmed cell death (apoptosis) play a role in the development of multiple organ dysfunction in critically ill patients? a review and a theoretical framework. Crit Care Med. 2000;28:537–49. 10.1097/00003246-200002000-00042. [DOI] [PubMed] [Google Scholar]
  • 25.Hinz B, Lagares D. Evasion of apoptosis by myofibroblasts: a hallmark of fibrotic diseases. Nat Rev Rheumatol. 2020;16:11–31. 10.1038/s41584-019-0324-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Su L, Dong Y, Wang Y, Wang Y, Guan B, Lu Y, Wu J, Wang X, Li D, Meng A, et al. Potential role of senescent macrophages in radiation-induced pulmonary fibrosis. Cell Death Dis. 2021;12:527. 10.1038/s41419-021-03811-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Fumagalli M, Rossiello F, Clerici M, Barozzi S, Cittaro D, Kaplunov JM, Bucci G, Dobreva M, Matti V, Beausejour CM, et al. Telomeric DNA damage is irreparable and causes persistent DNA-damage-response activation. Nat Cell Biol. 2012;14:355–65. 10.1038/ncb2466. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Victorelli S, Passos JF. Telomeres and cell senescence - size matters not. EBioMedicine. 2017;21:14–20. 10.1016/j.ebiom.2017.03.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Levstek T, Trebusak Podkrajsek K. Telomere attrition in chronic kidney diseases. Antioxidants (Basel). 2023. 10.3390/antiox12030579. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.von Zglinicki T, Saretzki G, Docke W, Lotze C. Mild hyperoxia shortens telomeres and inhibits proliferation of fibroblasts: a model for senescence? Exp Cell Res. 1995;220:186–93. 10.1006/excr.1995.1305. [DOI] [PubMed] [Google Scholar]
  • 31.Zhao Y, Simon M, Seluanov A, Gorbunova V. DNA damage and repair in age-related inflammation. Nat Rev Immunol. 2023;23:75–89. 10.1038/s41577-022-00751-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Rose J, Brian C, Woods J, Pappa A, Panayiotidis MI, Powers R, Franco R. Mitochondrial dysfunction in glial cells: Implications for neuronal homeostasis and survival. Toxicology. 2017;391:109–15. 10.1016/j.tox.2017.06.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Zorov DB, Juhaszova M, Sollott SJ. Mitochondrial reactive oxygen species (ROS) and ROS-induced ROS release. Physiol Rev. 2014;94:909–50. 10.1152/physrev.00026.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Sahin E, Colla S, Liesa M, Moslehi J, Muller FL, Guo M, Cooper M, Kotton D, Fabian AJ, Walkey C, et al. Telomere dysfunction induces metabolic and mitochondrial compromise. Nature. 2011;470:359–65. 10.1038/nature09787. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Moslehi J, DePinho RA, Sahin E. Telomeres and mitochondria in the aging heart. Circ Res. 2012;110:1226–37. 10.1161/CIRCRESAHA.111.246868. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Harrington JS, Ryter SW, Plataki M, Price DR, Choi AMK. Mitochondria in health, disease, and aging. Physiol Rev. 2023;103:2349–422. 10.1152/physrev.00058.2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Schapira AH. Mitochondrial diseases. Lancet. 2012;379:1825–34. 10.1016/S0140-6736(11)61305-6. [DOI] [PubMed] [Google Scholar]
  • 38.Lin MT, Beal MF. Mitochondrial dysfunction and oxidative stress in neurodegenerative diseases. Nature. 2006;443:787–95. 10.1038/nature05292. [DOI] [PubMed] [Google Scholar]
  • 39.Gonzales-Ebsen AC, Gregersen N, Olsen RK. Linking telomere loss and mitochondrial dysfunction in chronic disease. Front Biosci (Landmark Ed). 2017;22:117–27. 10.2741/4475. [DOI] [PubMed] [Google Scholar]
  • 40.Trifunovic A, Wredenberg A, Falkenberg M, Spelbrink JN, Rovio AT, Bruder CE, Bohlooly YM, Gidlof S, Oldfors A, Wibom R, et al. Premature ageing in mice expressing defective mitochondrial DNA polymerase. Nature. 2004;429:417–23. 10.1038/nature02517. [DOI] [PubMed] [Google Scholar]
  • 41.Passos JF, Saretzki G, von Zglinicki T. DNA damage in telomeres and mitochondria during cellular senescence: is there a connection? Nucleic Acids Res. 2007;35:7505–13. 10.1093/nar/gkm893. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Coppe JP, Desprez PY, Krtolica A, Campisi J. The senescence-associated secretory phenotype: the dark side of tumor suppression. Annu Rev Pathol. 2010;5:99–118. 10.1146/annurev-pathol-121808-102144. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Chakravarti D, Hu B, Mao X, Rashid A, Li J, Li J, Liao WT, Whitley EM, Dey P, Hou P, et al. Telomere dysfunction activates YAP1 to drive tissue inflammation. Nat Commun. 2020;11:4766. 10.1038/s41467-020-18420-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Unamuno X, Gomez-Ambrosi J, Ramirez B, Rodriguez A, Becerril S, Valenti V, Moncada R, Silva C, Salvador J, Fruhbeck G, et al. NLRP3 inflammasome blockade reduces adipose tissue inflammation and extracellular matrix remodeling. Cell Mol Immunol. 2021;18:1045–57. 10.1038/s41423-019-0296-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Bloom SI, Liu Y, Tucker JR, Islam MT, Machin DR, Abdeahad H, Thomas TG, Bramwell RC, Lesniewski LA, Donato AJ. Endothelial cell telomere dysfunction induces senescence and results in vascular and metabolic impairments. Aging Cell. 2023;22: e13875. 10.1111/acel.13875. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Hung YJ, Lee CH, Chu NF, Shieh YS. Plasma protein growth arrest-specific 6 levels are associated with altered glucose tolerance, inflammation, and endothelial dysfunction. Diabetes Care. 2010;33:1840–4. 10.2337/dc09-1073. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Tansey MG, Wallings RL, Houser MC, Herrick MK, Keating CE, Joers V. Inflammation and immune dysfunction in Parkinson disease. Nat Rev Immunol. 2022;22:657–73. 10.1038/s41577-022-00684-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Leng F, Edison P. Neuroinflammation and microglial activation in Alzheimer disease: where do we go from here? Nat Rev Neurol. 2021;17:157–72. 10.1038/s41582-020-00435-y. [DOI] [PubMed] [Google Scholar]
  • 49.Back M, Yurdagul A Jr, Tabas I, Oorni K, Kovanen PT. Inflammation and its resolution in atherosclerosis: mediators and therapeutic opportunities. Nat Rev Cardiol. 2019;16:389–406. 10.1038/s41569-019-0169-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Tchkonia T, Zhu Y, van Deursen J, Campisi J, Kirkland JL. Cellular senescence and the senescent secretory phenotype: therapeutic opportunities. J Clin Invest. 2013;123:966–72. 10.1172/JCI64098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Hammerich L, Tacke F. Hepatic inflammatory responses in liver fibrosis. Nat Rev Gastroenterol Hepatol. 2023;20:633–46. 10.1038/s41575-023-00807-x. [DOI] [PubMed] [Google Scholar]
  • 52.Grunewald M, Kumar S, Sharife H, Volinsky E, Gileles-Hillel A, Licht T, Permyakova A, Hinden L, Azar S, Friedmann Y, et al. Counteracting age-related VEGF signaling insufficiency promotes healthy aging and extends life span. Science. 2021. 10.1126/science.abc8479. [DOI] [PubMed] [Google Scholar]
  • 53.Guccini I, Revandkar A, D’Ambrosio M, Colucci M, Pasquini E, Mosole S, Troiani M, Brina D, Sheibani-Tezerji R, Elia AR, et al. Senescence reprogramming by TIMP1 deficiency promotes prostate cancer metastasis. Cancer Cell. 2021;39(68–82):e69. 10.1016/j.ccell.2020.10.012. [DOI] [PubMed] [Google Scholar]
  • 54.Zhou Y, Ning Z, Lee Y, Hambly BD, McLachlan CS. Shortened leukocyte telomere length in type 2 diabetes mellitus: genetic polymorphisms in mitochondrial uncoupling proteins and telomeric pathways. Clin Transl Med. 2016;5:8. 10.1186/s40169-016-0089-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Jeanclos E, Krolewski A, Skurnick J, Kimura M, Aviv H, Warram JH, Aviv A. Shortened telomere length in white blood cells of patients with IDDM. Diabetes. 1998;47:482–6. 10.2337/diabetes.47.3.482. [DOI] [PubMed] [Google Scholar]
  • 56.Willeit P, Raschenberger J, Heydon EE, Tsimikas S, Haun M, Mayr A, Weger S, Witztum JL, Butterworth AS, Willeit J, et al. Leucocyte telomere length and risk of type 2 diabetes mellitus: new prospective cohort study and literature-based meta-analysis. PLoS ONE. 2014;9: e112483. 10.1371/journal.pone.0112483. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Lin Y, Zhu Y, Wu J, Hinkle SN, Rawal S, Han J, Weir NL, Tsai MY, Zhang C. A Prospective study of leukocyte telomere length and risk of gestational diabetes in a multiracial cohort. Epidemiology. 2019;30(2):S10–6. 10.1097/EDE.0000000000001081. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Cheng F, Carroll L, Joglekar MV, Januszewski AS, Wong KK, Hardikar AA, Jenkins AJ, Ma RCW. Diabetes, metabolic disease, and telomere length. Lancet Diabetes Endocrinol. 2021;9:117–26. 10.1016/S2213-8587(20)30365-X. [DOI] [PubMed] [Google Scholar]
  • 59.Weng Q, Deng K, Wu F, Gan M, Li J, Dai Y, Jiang Y, Chen J, Dai J, Ma H, et al. Leukocyte telomere length, lipid parameters and gestational diabetes risk: a case-control study in a Chinese population. Sci Rep. 2019;9:8483. 10.1038/s41598-019-44968-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Kuhlow D, Florian S, von Figura G, Weimer S, Schulz N, Petzke KJ, Zarse K, Pfeiffer AF, Rudolph KL, Ristow M. Telomerase deficiency impairs glucose metabolism and insulin secretion. Aging (Albany NY). 2010;2:650–8. 10.18632/aging.100200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Dorrell C, Schug J, Canaday PS, Russ HA, Tarlow BD, Grompe MT, Horton T, Hebrok M, Streeter PR, Kaestner KH, et al. Human islets contain four distinct subtypes of beta cells. Nat Commun. 2016;7:11756. 10.1038/ncomms11756. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Canudas S, Becerra-Tomas N, Hernandez-Alonso P, Galie S, Leung C, Crous-Bou M, De Vivo I, Gao Y, Gu Y, Meinila J, et al. Mediterranean diet and telomere length: a systematic review and meta-analysis. Adv Nutr. 2020;11:1544–54. 10.1093/advances/nmaa079. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Wiemann SU, Satyanarayana A, Tsahuridu M, Tillmann HL, Zender L, Klempnauer J, Flemming P, Franco S, Blasco MA, Manns MP, et al. Hepatocyte telomere shortening and senescence are general markers of human liver cirrhosis. FASEB J. 2002;16:935–42. 10.1096/fj.01-0977com. [DOI] [PubMed] [Google Scholar]
  • 64.Shin HK; Park JH; Yu JH; Jin YJ; Suh YJ; Lee JW; Kim W; Korean Nonalcoholic Fatty Liver Study, G. Association between telomere length and hepatic fibrosis in non-alcoholic fatty liver disease. Sci Rep. 2021;11:18004. 10.1038/s41598-021-97385-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Tang L, Li D, Ma Y, Cui F, Wang J, Tian Y. The association between telomere length and non-alcoholic fatty liver disease: a prospective study. BMC Med. 2023;21:427. 10.1186/s12916-023-03136-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Aravinthan A, Scarpini C, Tachtatzis P, Verma S, Penrhyn-Lowe S, Harvey R, Davies SE, Allison M, Coleman N, Alexander G. Hepatocyte senescence predicts progression in non-alcohol-related fatty liver disease. J Hepatol. 2013;58:549–56. 10.1016/j.jhep.2012.10.031. [DOI] [PubMed] [Google Scholar]
  • 67.Zhang M, Hu ML, Huang JJ, Xia SS, Yang Y, Dong K. Association of leukocyte telomere length with non-alcoholic fatty liver disease in patients with type 2 diabetes. Chin Med J (Engl). 2019;132:2927–33. 10.1097/CM9.0000000000000559. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Wojcicki JM, Rehkopf D, Epel E, Rosenthal P. Shorter leukocyte telomere length in relation to presumed nonalcoholic fatty liver disease in Mexican-American Men in NHANES 1999–2002. Int J Hepatol. 2017;2017:8435178. 10.1155/2017/8435178. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Hartmann D, Srivastava U, Thaler M, Kleinhans KN, N’Kontchou G, Scheffold A, Bauer K, Kratzer RF, Kloos N, Katz SF, et al. Telomerase gene mutations are associated with cirrhosis formation. Hepatology. 2011;53:1608–17. 10.1002/hep.24217. [DOI] [PubMed] [Google Scholar]
  • 70.Nault JC, Ningarhari M, Rebouissou S, Zucman-Rossi J. The role of telomeres and telomerase in cirrhosis and liver cancer. Nat Rev Gastroenterol Hepatol. 2019;16:544–58. 10.1038/s41575-019-0165-3. [DOI] [PubMed] [Google Scholar]
  • 71.Alves-Paiva RM, Kajigaya S, Feng X, Chen J, Desierto M, Wong S, Townsley DM, Donaires FS, Bertola A, Gao B, et al. Telomerase enzyme deficiency promotes metabolic dysfunction in murine hepatocytes upon dietary stress. Liver Int. 2018;38:144–54. 10.1111/liv.13529. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Dalbeth N, Gosling AL, Gaffo A, Abhishek A. Gout Lancet. 2021;397:1843–55. 10.1016/S0140-6736(21)00569-9. [DOI] [PubMed] [Google Scholar]
  • 73.Qi M, Yu J, Ping F, Xu L, Li W, Zhang H, Li Y. Leukocyte telomere length independently predicts hyperuricemia risk in a longitudinal study of the Chinese population. Nutr Metab Cardiovasc Dis. 2023. 10.1016/j.numecd.2023.10.004. [DOI] [PubMed] [Google Scholar]
  • 74.Vazirpanah N, Kienhorst LBE, Van Lochem E, Wichers C, Rossato M, Shiels PG, Dalbeth N, Stamp LK, Merriman TR, Janssen M, et al. Patients with gout have short telomeres compared with healthy participants: association of telomere length with flare frequency and cardiovascular disease in gout. Ann Rheum Dis. 2017;76:1313–9. 10.1136/annrheumdis-2016-210538. [DOI] [PubMed] [Google Scholar]
  • 75.Deng Y, Li Q, Zhou F, Li G, Liu J, Lv J, Li L, Chang D. Telomere length and the risk of cardiovascular diseases: a Mendelian randomization study. Front Cardiovasc Med. 2022;9:1012615. 10.3389/fcvm.2022.1012615. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Bhattacharyya J, Mihara K, Bhattacharjee D, Mukherjee M. Telomere length as a potential biomarker of coronary artery disease. Indian J Med Res. 2017;145:730–7. 10.4103/0971-5916.216974. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Zimnitskaya OV, Petrova MM, Lareva NV, Cherniaeva MS, Al-Zamil M, Ivanova AE, Shnayder NA. Leukocyte telomere length as a molecular biomarker of coronary heart disease. Genes (Basel). 2022. 10.3390/genes13071234. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Niu Z, Wen X, Buka SL, Wang M, Tian L, Loucks EB, Kubzansky LD, Mu L. Associations of telomere length at birth with predicted atherosclerotic lesions and cardiovascular disease risk factors in midlife: a 40-year longitudinal study. Atherosclerosis. 2021;333:67–74. 10.1016/j.atherosclerosis.2021.08.013. [DOI] [PubMed] [Google Scholar]
  • 79.Chang ACY, Chang ACH, Kirillova A, Sasagawa K, Su W, Weber G, Lin J, Termglinchan V, Karakikes I, Seeger T, et al. Telomere shortening is a hallmark of genetic cardiomyopathies. Proc Natl Acad Sci U S A. 2018;115:9276–81. 10.1073/pnas.1714538115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Zheng Y, Zhang N, Wang Y, Wang F, Li G, Tse G, Liu T. Association between leucocyte telomere length and the risk of atrial fibrillation: an updated systematic review and meta-analysis. Ageing Res Rev. 2022;81: 101707. 10.1016/j.arr.2022.101707. [DOI] [PubMed] [Google Scholar]
  • 81.Codd V, Nelson CP, Albrecht E, Mangino M, Deelen J, Buxton JL, Hottenga JJ, Fischer K, Esko T, Surakka I, et al. Identification of seven loci affecting mean telomere length and their association with disease. Nat Genet. 2013;45:422–7. 10.1038/ng.2528. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Chatterjee S, de Gonzalo-Calvo D, Derda AA, Schimmel K, Sonnenschein K, Bavendiek U, Bauersachs J, Bar C, Thum T. Leukocyte telomere length correlates with hypertrophic cardiomyopathy severity. Sci Rep. 2018;8:11227. 10.1038/s41598-018-29072-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Willeit P, Willeit J, Brandstatter A, Ehrlenbach S, Mayr A, Gasperi A, Weger S, Oberhollenzer F, Reindl M, Kronenberg F, et al. Cellular aging reflected by leukocyte telomere length predicts advanced atherosclerosis and cardiovascular disease risk. Arterioscler Thromb Vasc Biol. 2010;30:1649–56. 10.1161/ATVBAHA.110.205492. [DOI] [PubMed] [Google Scholar]
  • 84.Theodoris CV, Mourkioti F, Huang Y, Ranade SS, Liu L, Blau HM, Srivastava D. Long telomeres protect against age-dependent cardiac disease caused by NOTCH1 haploinsufficiency. J Clin Invest. 2017;127:1683–8. 10.1172/JCI90338. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Samani NJ, Boultby R, Butler R, Thompson JR, Goodall AH. Telomere shortening in atherosclerosis. Lancet. 2001;358:472–3. 10.1016/S0140-6736(01)05633-1. [DOI] [PubMed] [Google Scholar]
  • 86.Liu P, Zhang Y, Ma L. Telomere length and associated factors in older adults with hypertension. J Int Med Res. 2019;47:5465–74. 10.1177/0300060519882570. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Morgan RG, Ives SJ, Walker AE, Cawthon RM, Andtbacka RH, Noyes D, Lesniewski LA, Richardson RS, Donato AJ. Role of arterial telomere dysfunction in hypertension: relative contributions of telomere shortening and telomere uncapping. J Hypertens. 2014;32:1293–9. 10.1097/HJH.0000000000000157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Cao Y.; Li H.; Mu F.T.; Ebisui O.; Funder J.W.; Liu, J.P. Telomerase activation causes vascular smooth muscle cell proliferation in genetic hypertension. FASEB J. 2002;16:96–8. 10.1096/cj.01-0447fje. [DOI] [PubMed] [Google Scholar]
  • 89.Cheng G, Wang L, Dai M, Wei F, Xu D. Shorter Leukocyte Telomere Length coupled with lower expression of Telomerase Genes in patients with Essential Hypertension. Int J Med Sci. 2020;17:2180–6. 10.7150/ijms.48456. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Schneider JL, Rowe JH, Garcia-de-Alba C, Kim CF, Sharpe AH, Haigis MC. The aging lung: physiology, disease, and immunity. Cell. 2021;184:1990–2019. 10.1016/j.cell.2021.03.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Zhang K, Xu L, Cong YS. Telomere dysfunction in idiopathic pulmonary fibrosis. Front Med (Lausanne). 2021;8: 739810. 10.3389/fmed.2021.739810. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Povedano JM, Martinez P, Flores JM, Mulero F, Blasco MA. Mice with pulmonary fibrosis driven by telomere dysfunction. Cell Rep. 2015;12:286–99. 10.1016/j.celrep.2015.06.028. [DOI] [PubMed] [Google Scholar]
  • 93.Alder JK, Armanios M. Telomere-mediated lung disease. Physiol Rev. 2022;102:1703–20. 10.1152/physrev.00046.2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Alder JK, Chen JJ, Lancaster L, Danoff S, Su SC, Cogan JD, Vulto I, Xie M, Qi X, Tuder RM, et al. Short telomeres are a risk factor for idiopathic pulmonary fibrosis. Proc Natl Acad Sci U S A. 2008;105:13051–6. 10.1073/pnas.0804280105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Haycock PC, Burgess S, Nounu A, Zheng J, Okoli GN, Bowden J, Wade KH, Timpson NJ, Evans DM, et al. Association between telomere length and risk of cancer and non-neoplastic diseases: a mendelian randomization study. JAMA Oncol. 2017;3:636–51. 10.1001/jamaoncol.2016.5945. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Newton CA, Kozlitina J, Lines JR, Kaza V, Torres F, Garcia CK. Telomere length in patients with pulmonary fibrosis associated with chronic lung allograft dysfunction and post-lung transplantation survival. J Heart Lung Transplant. 2017;36:845–53. 10.1016/j.healun.2017.02.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Disease GBD, Injury I, Prevalence C. Global, regional, and national incidence, prevalence, and years lived with disability for 354 diseases and injuries for 195 countries and territories, 1990–2017: a systematic analysis for the global burden of disease study 2017. Lancet. 2018;392:1789–858. 10.1016/S0140-6736(18)32279-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Ahmad T, Sundar IK, Tormos AM, Lerner CA, Gerloff J, Yao H, Rahman I. Shelterin telomere protection protein 1 reduction causes telomere attrition and cellular senescence via sirtuin 1 deacetylase in chronic obstructive pulmonary disease. Am J Respir Cell Mol Biol. 2017;56:38–49. 10.1165/rcmb.2016-0198OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Albrecht E, Sillanpaa E, Karrasch S, Alves AC, Codd V, Hovatta I, Buxton JL, Nelson CP, Broer L, Hagg S, et al. Telomere length in circulating leukocytes is associated with lung function and disease. Eur Respir J. 2014;43:983–92. 10.1183/09031936.00046213. [DOI] [PubMed] [Google Scholar]
  • 100.Savale L, Chaouat A, Bastuji-Garin S, Marcos E, Boyer L, Maitre B, Sarni M, Housset B, Weitzenblum E, Matrat M, et al. Shortened telomeres in circulating leukocytes of patients with chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2009;179:566–71. 10.1164/rccm.200809-1398OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Gan WQ, Man SF, Senthilselvan A, Sin DD. Association between chronic obstructive pulmonary disease and systemic inflammation: a systematic review and a meta-analysis. Thorax. 2004;59:574–80. 10.1136/thx.2003.019588. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Birch J, Victorelli S, Rahmatika D, Anderson RK, Jiwa K, Moisey E, Ward C, Fisher AJ, De Soyza A, Passos JF. Telomere dysfunction and senescence-associated pathways in bronchiectasis. Am J Respir Crit Care Med. 2016;193:929–32. 10.1164/rccm.201510-2035LE. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Han XR, Cen LJ, Pan CX, Lin ZH, Li HM, Zhang RL, Huang Y, Gao YH, Guan WJ. Decreased systemic and airway sirtuin 1 expression in adults with bronchiectasis. Front Med (Lausanne). 2021;8: 768770. 10.3389/fmed.2021.768770. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Wang WJ, Cai GY, Chen XM. Cellular senescence, senescence-associated secretory phenotype, and chronic kidney disease. Oncotarget. 2017;8:64520–33. 10.18632/oncotarget.17327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Hayashi K, Hishikawa A, Hashiguchi A, Azegami T, Yoshimoto N, Nakamichi R, Tokuyama H, Itoh H. Association of glomerular DNA damage and DNA methylation with one-year eGFR decline in IgA nephropathy. Sci Rep. 2020;10:237. 10.1038/s41598-019-57140-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Park S, Lee S, Kim Y, Cho S, Kim K, Kim YC, Han SS, Lee H, Lee JP, Joo KW, et al. A Mendelian randomization study found causal linkage between telomere attrition and chronic kidney disease. Kidney Int. 2021;100:1063–70. 10.1016/j.kint.2021.06.041. [DOI] [PubMed] [Google Scholar]
  • 107.Fazzini F, Lamina C, Raschenberger J, Schultheiss UT, Kotsis F, Schönherr S, Weissensteiner H, Forer L, Steinbrenner I, Meiselbach H, et al. Results from the German chronic kidney disease (GCKD) study support association of relative telomere length with mortality in a large cohort of patients with moderate chronic kidney disease. Kidney Int. 2020;98:488–97. 10.1016/j.kint.2020.02.034. [DOI] [PubMed] [Google Scholar]
  • 108.Kronenberg F. Telomere length and chronic kidney disease: cause or consequence? Kidney Int. 2021;100:980–3. 10.1016/j.kint.2021.08.013. [DOI] [PubMed] [Google Scholar]
  • 109.Gurung RL, Yar M, Liu S, Liu JJ, Lim SC. Short leukocyte telomere length predicts albuminuria progression in individuals with type 2 diabetes. Kidney Int Rep. 2018;3:592–601. 10.1016/j.ekir.2017.12.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Akinnibosun OA, Maier MC, Eales J, Tomaszewski M, Charchar FJ. Telomere therapy for chronic kidney disease. Epigenomics. 2022;14:1039–54. 10.2217/epi-2022-0073. [DOI] [PubMed] [Google Scholar]
  • 111.Kang HM, Ahn SH, Choi P, Ko YA, Han SH, Chinga F, Park AS, Tao J, Sharma K, Pullman J, et al. Defective fatty acid oxidation in renal tubular epithelial cells has a key role in kidney fibrosis development. Nat Med. 2015;21:37–46. 10.1038/nm.3762. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Saraswati S, Martinez P, Grana-Castro O, Blasco MA. Short and dysfunctional telomeres sensitize the kidneys to develop fibrosis. Nat Aging. 2021;1:269–83. 10.1038/s43587-021-00040-8. [DOI] [PubMed] [Google Scholar]
  • 113.Forero DA, Gonzalez-Giraldo Y, Lopez-Quintero C, Castro-Vega LJ, Barreto GE, Perry G. Meta-analysis of telomere length in Alzheimer’s disease. J Gerontol A Biol Sci Med Sci. 2016;71:1069–73. 10.1093/gerona/glw053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Honig LS, Kang MS, Schupf N, Lee JH, Mayeux R. Association of shorter leukocyte telomere repeat length with dementia and mortality. Arch of Neurol. 2012. 10.1001/archneurol.2012.1541. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Martinez-Gonzalez K, Islas-Hernandez A, Martinez-Ezquerro JD, Bermudez-Rattoni F, Garcia-delaTorre P. Telomere length and oxidative stress variations in a murine model of Alzheimer’s disease progression. Eur J Neurosci. 2020;52:4863–74. 10.1111/ejn.14877. [DOI] [PubMed] [Google Scholar]
  • 116.Hou Y, Dan X, Babbar M, Wei Y, Hasselbalch SG, Croteau DL, Bohr VA. Ageing as a risk factor for neurodegenerative disease. Nat Rev Neurol. 2019;15:565–81. 10.1038/s41582-019-0244-7. [DOI] [PubMed] [Google Scholar]
  • 117.Weng NP. Telomeres and immune competency. Curr Opin Immunol. 2012;24:470–5. 10.1016/j.coi.2012.05.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Schurks M, Buring J, Dushkes R, Gaziano JM, Zee RY, Kurth T. Telomere length and Parkinson’s disease in men: a nested case-control study. Eur J Neurol. 2014;21:93–9. 10.1111/ene.12252. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Forero DA, Gonzalez-Giraldo Y, Lopez-Quintero C, Castro-Vega LJ, Barreto GE, Perry G. Telomere length in Parkinson’s disease: a meta-analysis. Exp Gerontol. 2016;75:53–5. 10.1016/j.exger.2016.01.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Chinta SJ, Woods G, Demaria M, Rane A, Zou Y, McQuade A, Rajagopalan S, Limbad C, Madden DT, Campisi J, et al. Cellular senescence is induced by the environmental neurotoxin paraquat and contributes to neuropathology linked to Parkinson’s disease. Cell Rep. 2018;22:930–40. 10.1016/j.celrep.2017.12.092. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Whittemore K, Derevyanko A, Martinez P, Serrano R, Pumarola M, Bosch F, Blasco MA. Telomerase gene therapy ameliorates the effects of neurodegeneration associated to short telomeres in mice. Aging (Albany NY). 2019;11:2916–48. 10.18632/aging.101982. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Fragkiadaki P, Nikitovic D, Kalliantasi K, Sarandi E, Thanasoula M, Stivaktakis PD, Nepka C, Spandidos DA, Tosounidis T, Tsatsakis A. Telomere length and telomerase activity in osteoporosis and osteoarthritis. Exp Ther Med. 2020;19:1626–32. 10.3892/etm.2019.8370. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Valdes AM, Richards JB, Gardner JP, Swaminathan R, Kimura M, Xiaobin L, Aviv A, Spector TD. Telomere length in leukocytes correlates with bone mineral density and is shorter in women with osteoporosis. Osteoporos Int. 2007;18:1203–10. 10.1007/s00198-007-0357-5. [DOI] [PubMed] [Google Scholar]
  • 124.Ponzetti M, Rucci N. Updates on osteoimmunology: what’s new on the cross-talk between bone and immune system. Front Endocrinol (Lausanne). 2019;10:236. 10.3389/fendo.2019.00236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Sanders JL, Cauley JA, Boudreau RM, Zmuda JM, Strotmeyer ES, Opresko PL, Hsueh WC, Cawthon RM, Li R, Harris TB, et al. Leukocyte telomere length is not associated with BMD, osteoporosis, or fracture in older adults: results from the health, aging and body composition study. J Bone Miner Res. 2009;24:1531–6. 10.1359/jbmr.090318. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.McCulloch K, Litherland GJ, Rai TS. Cellular senescence in osteoarthritis pathology. Aging Cell. 2017;16:210–8. 10.1111/acel.12562. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Zhai G, Aviv A, Hunter DJ, Hart DJ, Gardner JP, Kimura M, Lu X, Valdes AM, Spector TD. Reduction of leucocyte telomere length in radiographic hand osteoarthritis: a population-based study. Ann Rheum Dis. 2006;65:1444–8. 10.1136/ard.2006.056903. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Price JS, Waters JG, Darrah C, Pennington C, Edwards DR, Donell ST, Clark IM. The role of chondrocyte senescence in osteoarthritis. Aging Cell. 2002;1:57–65. 10.1046/j.1474-9728.2002.00008.x. [DOI] [PubMed] [Google Scholar]
  • 129.Harbo M, Delaisse JM, Kjaersgaard-Andersen P, Soerensen FB, Koelvraa S, Bendix L. The relationship between ultra-short telomeres, aging of articular cartilage and the development of human hip osteoarthritis. Mech Ageing Dev. 2013;134:367–72. 10.1016/j.mad.2013.07.002. [DOI] [PubMed] [Google Scholar]
  • 130.Vaiserman A, Krasnienkov D. telomere length as a marker of biological age: State-of-the-Art, open issues, and future perspectives. Front Genet. 2020;11: 630186. 10.3389/fgene.2020.630186. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Hall DB, Holmlin RE, Barton JK. Oxidative DNA damage through long-range electron transfer. Nature. 1996;382:731–5. 10.1038/382731a0. [DOI] [PubMed] [Google Scholar]
  • 132.Kivimaki M, Bartolomucci A, Kawachi I. The multiple roles of life stress in metabolic disorders. Nat Rev Endocrinol. 2023;19:10–27. 10.1038/s41574-022-00746-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Tedone E, Huang E, O’Hara R, Batten K, Ludlow AT, Lai TP, Arosio B, Mari D, Wright WE, Shay JW. Telomere length and telomerase activity in T cells are biomarkers of high-performing centenarians. Aging Cell. 2019;18: e12859. 10.1111/acel.12859. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Suram A, Kaplunov J, Patel PL, Ruan H, Cerutti A, Boccardi V, Fumagalli M, Di Micco R, Mirani N, Gurung RL, et al. Oncogene-induced telomere dysfunction enforces cellular senescence in human cancer precursor lesions. EMBO J. 2012;31:2839–51. 10.1038/emboj.2012.132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Bailey SM, Murnane JP. Telomeres, chromosome instability and cancer. Nucleic Acids Res. 2006;34:2408–17. 10.1093/nar/gkl303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Faget DV, Ren Q, Stewart SA. Unmasking senescence: context-dependent effects of SASP in cancer. Nat Rev Cancer. 2019;19:439–53. 10.1038/s41568-019-0156-2. [DOI] [PubMed] [Google Scholar]
  • 137.Borah S, Xi L, Zaug AJ, Powell NM, Dancik GM, Cohen SB, Costello JC, Theodorescu D, Cech TR. Cancer. TERT promoter mutations and telomerase reactivation in urothelial cancer. Science. 2015;347:1006–10. 10.1126/science.1260200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Barthel FP, Wei W, Tang M, Martinez-Ledesma E, Hu X, Amin SB, Akdemir KC, Seth S, Song X, Wang Q, et al. Systematic analysis of telomere length and somatic alterations in 31 cancer types. Nat Genet. 2017;49:349–57. 10.1038/ng.3781. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Boehm BO, Moller P, Hogel J, Winkelmann BR, Renner W, Rosinger S, Seelhorst U, Wellnitz B, Marz W, Melzner J, et al. Lymphocytes of type 2 diabetic women carry a high load of stable chromosomal aberrations: a novel risk factor for disease-related early death. Diabetes. 2008;57:2950–7. 10.2337/db08-0274. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Salpea KD, Talmud PJ, Cooper JA, Maubaret CG, Stephens JW, Abelak K, Humphries SE. Association of telomere length with type 2 diabetes, oxidative stress and UCP2 gene variation. Atherosclerosis. 2010;209:42–50. 10.1016/j.atherosclerosis.2009.09.070. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Ojeda-Rodriguez A, Rangel-Zuniga OA, Arenas-de Larriva AP, Gutierrez-Mariscal FM, Torres-Pena JD, Romero-Cabrera JL, Podadera-Herreros A, Garcia-Fernandez H, Porras-Perez E, Luque RM, et al. Telomere length as biomarker of nutritional therapy for prevention of type 2 diabetes mellitus development in patients with coronary heart disease: CORDIOPREV randomised controlled trial. Cardiovasc Diabetol. 2024;23:98. 10.1186/s12933-024-02175-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Xiao F, Zheng X, Cui M, Shi G, Chen X, Li R, Song Z, Rudolph KL, Chen B, Ju Z. Telomere dysfunction-related serological markers are associated with type 2 diabetes. Diabetes Care. 2011;34:2273–8. 10.2337/dc10-2431. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Fyhrquist F.; Tiitu A.; Saijonmaa O.; Forsblom C.; Groop, P.H.; FinnDiane Study, G. Telomere length and progression of diabetic nephropathy in patients with type 1 diabetes. J Intern Med. 2010;267:278–86. 10.1111/j.1365-2796.2009.02139.x. [DOI] [PubMed] [Google Scholar]
  • 144.Lv Z, Cui J, Zhang J. Associations between serum urate and telomere length and inflammation markers: evidence from UK Biobank cohort. Front Immunol. 2022;13:1065739. 10.3389/fimmu.2022.1065739. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Hammadah M, Al Mheid I, Wilmot K, Ramadan R, Abdelhadi N, Alkhoder A, Obideen M, Pimple PM, Levantsevych O, Kelli HM, et al. Telomere shortening, regenerative capacity, and cardiovascular outcomes. Circ Res. 2017;120:1130–8. 10.1161/CIRCRESAHA.116.309421. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Brouilette SW, Moore JS, McMahon AD, Thompson JR, Ford I, Shepherd J, Packard CJ, Samani NJ. West of scotland coronary prevention study, G. Telomere length, risk of coronary heart disease, and statin treatment in the West of Scotland primary prevention study: a nested case-control study. Lancet. 2007;369:107–14. 10.1016/S0140-6736(07)60071-3. [DOI] [PubMed] [Google Scholar]
  • 147.Raschenberger J, Kollerits B, Hammerer-Lercher A, Rantner B, Stadler M, Haun M, Klein-Weigel P, Fraedrich G, Kronenberg F. The association of relative telomere length with symptomatic peripheral arterial disease: results from the CAVASIC study. Atherosclerosis. 2013;229:469–74. 10.1016/j.atherosclerosis.2013.05.027. [DOI] [PubMed] [Google Scholar]
  • 148.Benetos A, Gardner JP, Zureik M, Labat C, Xiaobin L, Adamopoulos C, Temmar M, Bean KE, Thomas F, Aviv A. Short telomeres are associated with increased carotid atherosclerosis in hypertensive subjects. Hypertension. 2004;43:182–5. 10.1161/01.HYP.0000113081.42868.f4. [DOI] [PubMed] [Google Scholar]
  • 149.Benetos A, Okuda K, Lajemi M, Kimura M, Thomas F, Skurnick J, Labat C, Bean K, Aviv A. Telomere length as an indicator of biological aging: the gender effect and relation with pulse pressure and pulse wave velocity. Hypertension. 2001;37:381–5. 10.1161/01.hyp.37.2.381. [DOI] [PubMed] [Google Scholar]
  • 150.Stuart BD, Lee JS, Kozlitina J, Noth I, Devine MS, Glazer CS, Torres F, Kaza V, Girod CE, Jones KD, et al. Effect of telomere length on survival in patients with idiopathic pulmonary fibrosis: an observational cohort study with independent validation. Lancet Respir Med. 2014;2:557–65. 10.1016/S2213-2600(14)70124-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Cordoba-Lanus E, Cazorla-Rivero S, Espinoza-Jimenez A, De-Torres JP, Pajares MJ, Aguirre-Jaime A, Celli B, Casanova C. Telomere shortening and accelerated aging in COPD: findings from the BODE cohort. Respir Res. 2017;18:59. 10.1186/s12931-017-0547-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Barden A, O’Callaghan N, Burke V, Mas E, Beilin LJ, Fenech M, Irish AB, Watts GF, Puddey IB, Huang RC, et al. n-3 Fatty acid supplementation and leukocyte telomere length in patients with chronic kidney disease. Nutrients. 2016;8:175. 10.3390/nu8030175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Fazzini F, Lamina C, Raschenberger J, Schultheiss UT, Kotsis F, Schonherr S, Weissensteiner H, Forer L, Steinbrenner I, Meiselbach H, et al. Results from the German chronic kidney disease (GCKD) study support association of relative telomere length with mortality in a large cohort of patients with moderate chronic kidney disease. Kidney Int. 2020;98:488–97. 10.1016/j.kint.2020.02.034. [DOI] [PubMed] [Google Scholar]
  • 154.Moverare-Skrtic S, Johansson P, Mattsson N, Hansson O, Wallin A, Johansson JO, Zetterberg H, Blennow K, Svensson J. Leukocyte telomere length (LTL) is reduced in stable mild cognitive impairment but low LTL is not associated with conversion to Alzheimer’s disease: a pilot study. Exp Gerontol. 2012;47:179–82. 10.1016/j.exger.2011.12.005. [DOI] [PubMed] [Google Scholar]
  • 155.Roberts RO, Boardman LA, Cha RH, Pankratz VS, Johnson RA, Druliner BR, Christianson TJ, Roberts LR, Petersen RC. Short and long telomeres increase risk of amnestic mild cognitive impairment. Mech Ageing Dev. 2014;141–142:64–9. 10.1016/j.mad.2014.10.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Degerman S, Domellof M, Landfors M, Linder J, Lundin M, Haraldsson S, Elgh E, Roos G, Forsgren L. Long leukocyte telomere length at diagnosis is a risk factor for dementia progression in idiopathic parkinsonism. PLoS ONE. 2014;9: e113387. 10.1371/journal.pone.0113387. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Bernardes De Jesus B, Schneeberger K, Vera E, Tejera A, Harley CB, Blasco MA. The telomerase activator TA-65 elongates short telomeres and increases health span of adult/old mice without increasing cancer incidence. Aging Cell. 2011;10:604–21. 10.1111/j.1474-9726.2011.00700.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Dolcetti R, De Rossi A. Telomere/telomerase interplay in virus-driven and virus-independent lymphomagenesis: pathogenic and clinical implications. Med Res Rev. 2012;32:233–53. 10.1002/med.20211. [DOI] [PubMed] [Google Scholar]
  • 159.Bernardes de Jesus B, Vera E, Schneeberger K, Tejera AM, Ayuso E, Bosch F, Blasco MA. Telomerase gene therapy in adult and old mice delays aging and increases longevity without increasing cancer. EMBO Mol Med. 2012;4:691–704. 10.1002/emmm.201200245. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Ruiz-Leon AM, Lapuente M, Estruch R, Casas R. Clinical advances in immunonutrition and atherosclerosis: a review. Front Immunol. 2019;10:837. 10.3389/fimmu.2019.00837. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Nikiphorou E, Philippou E. Nutrition and its role in prevention and management of rheumatoid arthritis. Autoimmun Rev. 2023;22: 103333. 10.1016/j.autrev.2023.103333. [DOI] [PubMed] [Google Scholar]
  • 162.Moores CJ, Fenech M, O’Callaghan NJ. Telomere dynamics: the influence of folate and DNA methylation. Ann N Y Acad Sci. 2011;1229:76–88. 10.1111/j.1749-6632.2011.06101.x. [DOI] [PubMed] [Google Scholar]
  • 163.Puterman E, Epel ES, Lin J, Blackburn EH, Gross JJ, Whooley MA, Cohen BE. Multisystem resiliency moderates the major depression-telomere length association: findings from the Heart and Soul Study. Brain Behav Immun. 2013;33:65–73. 10.1016/j.bbi.2013.05.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Cherkas LF, Hunkin JL, Kato BS, Richards JB, Gardner JP, Surdulescu GL, Kimura M, Lu X, Spector TD, Aviv A. The association between physical activity in leisure time and leukocyte telomere length. Arch Intern Med. 2008;168:154–8. 10.1001/archinternmed.2007.39. [DOI] [PubMed] [Google Scholar]
  • 165.Tomiyama AJ, O’Donovan A, Lin J, Puterman E, Lazaro A, Chan J, Dhabhar FS, Wolkowitz O, Kirschbaum C, Blackburn E, et al. Does cellular aging relate to patterns of allostasis? An examination of basal and stress reactive HPA axis activity and telomere length. Physiol Behav. 2012;106:40–5. 10.1016/j.physbeh.2011.11.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Aulinas A, Ramirez MJ, Barahona MJ, Mato E, Bell O, Surralles J, Webb SM. Telomeres and endocrine dysfunction of the adrenal and GH/IGF-1 axes. Clin Endocrinol (Oxf). 2013;79:751–9. 10.1111/cen.12310. [DOI] [PubMed] [Google Scholar]
  • 167.Osler M, Bendix L, Rask L, Rod NH. Stressful life events and leucocyte telomere length: Do lifestyle factors, somatic and mental health, or low grade inflammation mediate this relationship? Results from a cohort of Danish men born in 1953. Brain Behav Immun. 2016;58:248–53. 10.1016/j.bbi.2016.07.154. [DOI] [PubMed] [Google Scholar]
  • 168.Hoge EA, Bui E, Mete M, Dutton MA, Baker AW, Simon NM. Mindfulness-based stress reduction vs escitalopram for the treatment of adults with anxiety disorders: a randomized clinical trial. JAMA Psychiat. 2023;80:13–21. 10.1001/jamapsychiatry.2022.3679. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Ewais T, Begun J, Kenny M, Rickett K, Hay K, Ajilchi B, Kisely S. A systematic review and meta-analysis of mindfulness based interventions and yoga in inflammatory bowel disease. J Psychosom Res. 2019;116:44–53. 10.1016/j.jpsychores.2018.11.010. [DOI] [PubMed] [Google Scholar]
  • 170.Kim KS, Kwak JW, Lim SJ, Park YK, Yang HS, Kim HJ. Oxidative stress-induced telomere length shortening of circulating leukocyte in patients with obstructive sleep apnea. Aging Dis. 2016;7:604–13. 10.14336/AD.2016.0215. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

No datasets were generated or analyzed during the current study.


Articles from Clinical and Experimental Medicine are provided here courtesy of Springer

RESOURCES