Skip to main content

Canonical and non-canonical functions of the non-coding RNA component (TERC) of telomerase complex

Abstract

The telomerase complex consists of a protein component (TERT), which has reverse transcriptase activity, and an RNA component (TERC), which serves as a template for telomere synthesis. Evidence is rapidly accumulating regarding the non-canonical functions of these components in both normal or diseased cells. An oligonucleotide-based drug, the first telomerase inhibitor, secured FDA approval in June 2024. We recently summarized the non-canonical functions of TERT in viral infections and cancer. In this review, we expand on these non-canonical functions of TERC beyond telomere maintenance. Specifically, we explore TERC’s roles in cellular aging and senescence, immune regulation, genetic diseases, human cancer, as well as involvement in viral infections and host interactions. Finally, we discuss a transcription product of telomere repeats, TERRA, and explore strategies for targeting TERC as a therapeutic approach.

Introduction

Telomerase is a ribonucleoprotein polymerase (RNP) consisting of a protein component, TERT (1,132 amino acids, 127 kDa), which has reverse transcriptase activity, and an RNA component, TERC (also known as Telomerase RNA Component or TR), which serves as a template for telomere repeats (Fig. 1) [1, 2]. Initially discovered in Tetrahymena extracts, telomerase was described as a specific telomere terminal transferase involved in the de novo elongation of telomeric repeats. The human telomerase was first identified in 1989, and its function is to add the TTAGGG sequence to the ends of telomeres [3, 4]. In addition to the highly conserved TERT and TERC components, several telomerase-associated proteins are required for the functional telomerase complex. These include Ku, HSP90, telomerase-associated protein 1 (TP1), dyskerin (DKC1), telomerase Cajal body protein 1 (TCAB1), non-histone chromosome protein 2 (NHP2), nucleolar protein 10 (NOP10), and GAR1 RNP (GAR1) [5,6,7]. During early human development, telomerase is active, but its activity becomes silenced between 12 and 18 weeks of gestation, remaining low in somatic cells thereafter. In contrast, most cancer cells and stem cells exhibit relatively high telomerase activity [8,9,10,11]. While the mechanisms behind the activation or silencing of telomerase remain to be fully explored, it is widely accepted that there is a direct correlation between TERT expression and telomerase activity, as the TERC component and telomerase-associated proteins are ubiquitously expressed in most human somatic cells [12, 13]. Over the years, research has focused on TERT promoter mutations, gene copy number changes, epigenetic regulation, and transcriptional and post-transcriptional modifications to elucidate the mechanisms behind the switch in telomerase activity [14,15,16,17,18,19,20,21,22]. It has also been reported that TERC has effects on normal cell biology and disease progression [3, 23, 24].

Substantial research has focused on the role of telomerase in aging, cancer, and disease. Notably, 85% of cancer cells show telomerase activity, highlighting the therapeutic potential of targeting telomerase [9, 25, 26]. Activation of telomerase may also benefit patients with degenerative diseases [27]. However, beyond its canonical role in telomere maintenance, many non-canonical functions of telomerase have been discovered, including its involvement in the regulation of DNA replication and repair, gene expression, and cell signaling pathways. These non-canonical functions may contribute to cell cycle progression, survival, proliferation, differentiation, apoptosis, metabolism, regeneration, and tumorigenesis [22, 28, 29]. Evidence supporting the independent non-canonical roles of TERT and TERC is rapidly increasing [30,31,32]. For example, quantitative assays for TERT, TERC, and telomerase complexes in HEK293 and HeLa cells reveal approximately 240 telomerase complexes per cell, a number significantly lower than the copies of TERT and TERC. TERC levels often exceed the number of assembled telomerase RNP complexes in cancer cells (approximately 1,150 TERC molecules in HeLa cells, compared to only about 500 molecules of TERT), suggesting the existence of unassembled TERC [30]. Furthermore, the identification of 2,198 TERC-binding sites in the genome presents a substantial resource for studying the potential non-canonical functions of TERC [33]. There are reports of TERC’s alternative functions, independent of telomerase activity, in regulating gene expression and signaling pathways, with subsequent effects on cell survival, apoptosis, inflammation, and cancer promotion [31, 32]. The discovery of TERC-53 has also opened new insights into the non-canonical functions of TERC [34, 35]. Together, these findings underscore the need for further research into the non-canonical functions of TERC in human cell proliferation, differentiation, and disease progression, with significant therapeutic potential.

Fig. 1
figure 1

Canonical functions of TERT and TERC. Schematic illustration of the TERC structure and its role in the human telomerase ribonucleoprotein complex, which includes TERT, NOP10, dyskerin, GAR1, NHP2, and TCAB1. TERC provides the template (3’-CAAUCCCAAUC-5’) for hTERT to extend the telomere by adding TTAGGG DNA repeats. The H/ACA box recruits two sets of H/ACA-box-binding proteins, including NOP10, dyskerin, GAR1, and NHP2. TCAB1 is recruited through its binding with TERC and the H/ACA-box-binding proteins

Canonical functions of TERC, telomerase inhibitors

TERC is a non-coding RNA that serves as a template for telomere replication by telomerase, enabling the addition of TTAGGG repeats to maintain chromosomal stability. Its canonical role is critical for preserving telomere integrity and ensuring genomic stability, particularly in rapidly dividing cells, such as cancer cells (Fig. 1) [36]. The dependency of telomerase on TERC for its function has made it a key target in cancer research. TERC’s role extends beyond elongating telomeres, as it also stabilizes telomerase and facilitates its recruitment to telomeres [37]. Zhao et al. demonstrated that the absence of TERC (Terc -/-) in embryonic stem cells led to progressive telomere shortening and severe genomic instability across generations. Shortened telomeres disrupted heterochromatin, leading to the activation of retrotransposons like LINE1, which promoted mutations and structural variations, thus underscoring the essential function of TERC in maintaining genomic integrity through telomerase activity [38]. Telomerase inhibitors are designed to suppress the activity of telomerase, targeting either TERC, TERT, or other telomerase-associated proteins [39]. A prominent example is Imetelstat (GRN163L), an oligonucleotide that binds to TERC, preventing its interaction with TERT. By disrupting the canonical function of telomerase, Imetelstat leads to telomere shortening and ultimately triggers apoptosis or senescence in telomerase-dependent cancer cells [40, 41]. In June 2024, the US FDA approved Imetelstat (now branded as Rytelo) for adults with low- to intermediate-risk myelodysplastic syndromes (MDS) and transfusion-dependent anemia who do not respond to erythropoiesis-stimulating agents (ESAs). This approval marked the first telomerase inhibitor to secure FDA approval after 20 years and more than 20 clinical trials across various cancers, including breast cancer, lung cancer, brain cancer, and leukemia [42, 43]. Another approach involves small-molecule inhibitors such as BIBR1532, which directly block the catalytic activity of TERT, impairing telomere elongation. Similarly, stabilizing G-quadruplex structures at telomeres hinders telomerase access, indirectly affecting TERC’s template function [44]. However, these inhibitors face challenges, including potential toxicity to normal stem cells—where telomerase activity is essential—and the development of resistance mechanisms, such as activation of alternative lengthening of telomeres (ALT) pathways [39]. While initial clinical trials for solid tumors showed variable outcomes, promising results have been observed in hematological malignancies, such as myelofibrosis, highlighting the therapeutic potential of targeting TERC in specific cancer contexts [45]. In addition to its canonical role in telomere elongation, TERC has been implicated in genome stability mechanisms [46]. Using a comparative genomic approach, researchers demonstrated that TERC, as part of the telomerase complex, participates in a unique DNA double-strand break repair mechanism [46]. They identified TERC-ITSs (Interstitial Telomeric Sequences), which are sequences retrotranscribed from TERC RNA and flanked by telomeric-like repeats, inserted at break sites during vertebrate evolution. This process involves telomerase retrotranscribing its RNA template into DNA and integrating it into chromosomal loci via the non-homologous end-joining pathway. These findings highlight that, beyond elongating telomeres, TERC plays a broader role in maintaining genome integrity [46]. However, telomerase inhibitors targeting TERC could disrupt these repair mechanisms, potentially increasing genomic instability and raising critical considerations for therapeutic applications [47]. TERC has also been studied as a biomarker, with elevated levels associated with specific cancers, including astrocytoma and gastric carcinomas [48]. Gazzaniga and Blackburn further highlighted the dual role of TERC, demonstrating that its non-canonical function can protect immune cells from apoptosis independent of telomerase activity. Their findings suggest that TERC contributes to immune regulation by mitigating apoptosis through the intrinsic apoptotic pathway, separate from its role in telomere maintenance [32]. Recent research by Majumder et al. underscores the importance of TERC regulation in cancer progression. They identified the RNA-binding protein FXR1 as a critical factor stabilizing TERC in head and neck squamous cell carcinoma [45]. FXR1 binds to G-quadruplex RNA structures within TERC, maintaining its stability and promoting telomerase activity to evade senescence. Silencing FXR1 reduced TERC levels, impaired telomerase function, and induced cellular senescence, linking TERC regulation to the evasion of growth arrest in cancer cells​ [45]. Collectively, these findings emphasize the central role of TERC in telomerase activity, genome stability, and gene regulation. Targeting TERC through direct inhibitors or modulation of its regulatory pathways represents a promising avenue for cancer treatment. However, this requires careful consideration of its multifaceted functions.

Telomeric repeat-containing RNA (TERRA)

TERRA is a class of long non-coding RNAs transcribed by RNA polymerase II from telomeric and subtelomeric regions of chromosomes. Canonically, TERRA plays a critical role in maintaining genome stability and regulating telomere length [49]. The expression of TERRA varies across different cell types and telomere lengths, with its levels being regulated by chromatin state and cellular stress. In the nuclear foci at chromosome ends, TERRA interacts with telomeric chromatin and associated proteins, such as the shelterin complex. TERRA molecules are G-rich RNA, composed of 5’-UUAGGG-3’ repeats, and exhibit considerable size heterogeneity, ranging from 100 base pairs to over 9 kilobases in mammals [49]. At telomeres, TERRA performs several critical functions. It helps maintain the integrity of heterochromatin by interacting with histone-modifying enzymes, preserving the structure of telomeric chromatin. Beyond its canonical roles, TERRA influences the DNA damage response, gene expression, and certain immune regulatory processes. Moreover, TERRA has been implicated in the development of cancer and inflammatory conditions, some of which occur independently of telomerase activity. These multifaceted roles make TERRA a promising therapeutic target for diseases involving telomere dysfunction and beyond [50, 51].

TERRA also mediates the formation of R-loops, unique three-stranded nucleic acid structures that consist of a displaced single-stranded DNA molecule and an RNA-DNA hybrid. These structures naturally form during transcription and play an important role in telomere maintenance. TERRA-induced R-loops regulate telomere function by recruiting chromatin remodelers and DNA repair proteins [52]. However, when R-loops persist or form inappropriately, they can cause significant genomic instability and DNA damage. This is particularly evident in ALT (alternative lengthening of telomeres) positive cancers, where TERRA-driven R-loops accumulate at short telomeres to facilitate homologous recombination and maintain telomere length. The delicate balance between R-loop formation and resolution is vital for telomere stability [53]. Dysregulation of these structures is increasingly associated with aging and cancer, making them a key focus for therapeutic interventions targeting telomere dysfunction. Understanding the molecular players involved in R-loop formation, resolution, and their interaction with TERRA provides deeper insights into their biological significance and pathological consequences [54].

Non-canonical functions of TERC

Cell senescence

Cell aging is a process in which cells gradually lose normal physiological functions, including the ability to proliferate and differentiate [55]. Cellular senescence, a state where cells permanently cease dividing but remain viable, is a key consequence of aging [56]. Normal human cells enter senescence after a limited number of divisions, a phenomenon known as replicative senescence or the Hayflick limit [57]. In addition, cellular senescence can be induced by external or internal stressors such as oxidative stress, UV irradiation, hyperoxia, paracrine signaling from other senescent cells, and DNA damage [58,59,60,61]. This type of senescence, known as premature senescence, can occur independently of cell replication. Telomere-related proteins play direct or indirect roles in regulating both replicative and premature senescence.

Telomere maintenance, TERRA and replicative senescence

Due to the ‘end-replication problem,’ cells lose a small fragment of telomere repeats with each DNA replication, causing progressive telomere shortening. When telomeres shorten to a critical length, shelterin proteins—including TRF1, TRF2, POT1, RAP1, TIN2, and TPP1—can no longer form the intact shelterin complex, and the t-loop structure is disrupted. This exposes the loose chromosome ends with a single-strand G-rich 3’ overhang, triggering the DNA damage response (DDR) [62, 63]. Research suggests that two major DNA damage-sensing molecules, ATM and ATR kinases, are inhibited by TRF2 and TPP1-POT1, respectively [64]. The telomere DDR further recruits DDR factors, such as 53BP1 and γ-H2AX, and activates downstream p53 signaling, promoting senescence [65, 66].

Telomere length is not shortened at a fixed rate during DNA replication but is instead regulated by several telomere-related proteins. The shelterin complex and telomeric RNA (TERRA) contribute to the formation of secondary structures, including t-loops, R-loops, and G-quadruplexes (G4). These structures can impede telomere replication, requiring specific proteins to resolve them, thus influencing the rate of telomere shortening during DNA replication [67, 68]. Shelterin proteins are crucial for telomere replication. For example, TRF1 is essential for preventing telomere replication fork stalling at telomeres, while TRF2 aids by recruiting Apollo to resolve topological stress [69, 70]. Interestingly, overexpression of TRF1 and TRF2 can stall telomere replication [71]. Additionally, TRF1 and TRF2 help replication forks pass through hard-to-replicate sites in multiple ways. Down-regulation of Timeless significantly accelerates telomere shortening [72, 73]. These results suggest a probably context-dependent manner in whichTRF1 and TRF2 influencing telomere replication. Another shelterin protein, POT1, can promote telomere replication by preventing G4 structures formation [74]. The CST complex also plays important roles with shelterin in protecting telomere ends [75]. A CST complex member, Stn1, regulates DNA polymerase α loading to telomeres [76]. Other non-canonical telomere-related proteins, including helicases and single-strand DNA-binding proteins, contribute to telomere replication. For example, WRN, PIF1, and RTEL1 are involved in unwinding G4 structures at telomeres, while RTEL1 also resolves the t-loop, slowing telomere shortening [77, 78]. RNase H and ATRX can dissolve R-loops by degrading or displacing TERRA [79, 80].

In most normal human somatic cells, telomeres are not lengthened due to undetectable telomerase activity. However, the telomerase complex extends telomeres in stem cells and most cancer cells, preventing senescence. Telomere-related proteins regulate telomerase complex function [81, 82]. For instance, TIN2, a shelterin component, recruits telomerase to the telomere and enhances telomere elongation [83]. Telomerase elongation is facilitated by the inhibition of G4 formation by POT1 [84]. TPP1, another shelterin protein, also recruits telomerase to chromosome ends independently of its protective role [85, 86]. The CST complex, in contrast, competes with POT1 and TPP1 and can act as a telomerase inhibitor [87].

Alternative Lengthening of Telomeres (ALT) is another mechanism to extend telomeres [88]. This pathway is dependent on homology-directed repair (HDR) mediated by RAD51 or RAD52 and regulated by several telomere-related proteins in a context-dependent manner [89, 90]. Shelterin proteins TRF2 and RAP1 inhibit ALT by preventing telomere “ultrabright” formations, which allows HDR factors to localize and form D-loops [91, 92]. Paradoxically, TRF2, RAP1, TIN2, and TRF1 can also support ALT by facilitating the formation of ALT-associated promyelocytic leukemia bodies (APBs) [93, 94]. Furthermore, R-loops promote senescence in some cells by hindering telomere replication and triggering HDR in ALT-positive cells by inducing DSBs at telomeres [79, 80]. This recruits R-loop-regulating proteins such as RNase H, ATRX, RAD51AP1, and Npl3, facilitating ALT-dependent telomere lengthening [88, 95,96,97]. In ALT-negative cells, RAD51-mediated HDR results in uncapped telomere fusion, leading to stable senescence and genomic instability [98].

These mechanisms play crucial roles in telomere length maintenance during cell proliferation and the onset of replicative senescence.

Telomere and premature senescence

Premature senescence can arise from persistent DDR induced by stress [99]. Telomeres are particularly vulnerable to DNA damage and may form persistent DNA damage foci unrelated to replication-induced telomere shortening [100, 101]. Telomere-related proteins regulate stress-induced telomere DNA damage (tDD) and DDR.

Telomeric 8-oxo-guanine (8oxoG), a common damage caused by oxidative stress, activates ATM and ATR signaling to trigger senescence. Acute 8oxoG can even cause telomere loss and crisis [102, 103]. This process detaches approximately 50% of TRF1 and TRF2 from telomeres, leaving telomeric DNA unprotected [104]. MTH1 removes telomeric 8oxoG, thereby protecting telomeres and enhancing telomerase activity [105, 106].

Telomere-related proteins and mitochondrial Functions also play a role in regulating cell aging. In skeletal muscle fibers, TRF2 loss leads to postmitotic cell senescence by downregulating mitochondrial SIRT3 expression, causing mitochondrial dysfunction [107]. Interestingly, FOXO3a can replace TRF2 at telomeres, preventing telomere deprotection [108]. In cancer cells, TIN2 localizes in mitochondria and regulates various metabolic pathways [109]. Additionally, under chronic oxidative stress (such as H2O2 exposure, X-ray irradiation, or excitotoxic glutamate exposure), TERT relocates to the cytosol and mitochondria, protecting mitochondrial DNA while downregulating telomerase activity for telomere elongation [110, 111].

Studies show that telomere damage accumulates persistently during cell aging, suggesting that such damage is difficult to repair [101, 112]. This phenomenon is largely regulated by shelterin proteins. For example, TRF2 inhibits the repair of telomeric single-strand breaks (SSBs). In human fibroblasts, accumulation of stress-induced SSBs drives cellular senescence [113, 114]. DNA double-strand breaks (DSBs) are repaired through classical non-homologous end-joining (c-NHEJ), backup non-homologous end-joining (b-NHEJ), or HDR [115]. However, NHEJ of telomeres increases the risk of chromosomal fusion and requires strict regulation. TRF2 binds to a c-NHEJ factor, Ku, to inhibit telomere NHEJ [116]. TRF2 and RAP1 also inhibit another c-NHEJ factor, DNA-dependent protein kinase, while allowing counteraction with b-NHEJ [117]. Activation of protein phosphatase magnesium-dependent 1 delta (PPM1D) phosphorylates TRF2, enhancing its interaction with TIN2, and promotes the binding of NHEJ factor 53BP1 to telomeres [118]. TRF2-dependent R-loop formation with TERRA recruits RAD52 [119], while telomerase complex protein TCAB1 recruits RNF168, BRCA1, and RAD51, promoting HDR [120].

Telomere-related proteins and senescence-related pathways

Telomere is linked to cell senescence not only through the DDR but also via interactions with several key senescence- related pathways. Telomere position effects (TPE) link telomere length to gene expression regulation [121]. It has been shown that telomeres can reach chromosome sites 10 Mb away, enriching TRF2 at the promoter regions of genes and regulating gene expression [122]. In CD4+ T cells and promyelocytic cancer cells, TPE was found to affect the expression of TERT [123].

Telomerase complex proteins regulate senescence-related pathways. TERT reduces the ROS level by upregulating GSH: GSSG ratio [124], enhance PINK1 mitochondrial localization, and promote mitophagy [125]. Telomerase complex, including hTERT and TERC, can bind to NF-κB and enhance NF-κB-dependent gene expression [126, 127]. Dyskerin 1 participates in telomerase complex by combining to TERC and interacting with Reptin [128]. Dyskerin 1 loss inhibit ribosome production and lead to p53-mediated cell-cycle arrest [129], while reptin bind to p53 and inhibit its expression [130].

Immune cell regulation

Mutations in TERC and elevated TERC copy numbers are associated with congenital dyskeratosis, aplastic anemia [131, 132], and other genetic diseases [133,134,135]. Notably, patients with these conditions often exhibit immunological dysfunction [136, 137]. TERC has been shown to function in immune regulation independent of its telomerase activity (Table 1) [32, 138,139,140].

Table 1 Canonical and non-canonical functions of TERC in Immune Cell Regulation

Gazzaniga and Blackburn [32] investigated TERC’s non-canonical role in immune cell survival, revealing mechanisms that do not depend on its enzymatic activity or telomere elongation. They demonstrated that TERC protects CD4 + T cells from apoptosis induced by dexamethasone, implicating TERC in the intrinsic apoptotic pathway. Knockdown of TERC resulted in activation of pro-apoptotic markers such as Bim and increased caspase-9 and caspase-3/7 activity, confirming TERC’s anti-apoptotic role. Importantly, this function was independent of telomere length or DNA damage, suggesting TERC’s involvement in immune cell homeostasis under stress [32].

Liu et al. [141] further explored TERC’s non-canonical immune-regulatory functions, focusing on its ability to modulate inflammatory responses independently of telomerase activity. They found that TERC upregulates inflammatory cytokines such as IL-6, IL-8, and TNF-α by activating the NF-κB signaling pathway. TERC binds to specific gene promoters (e.g., TYROBP, USP16, TPRG1L, and LIN37) through RNA-DNA triplex formation, enhancing the transcription of these inflammation-related genes. Elevated TERC expression was also correlated with chronic inflammatory diseases such as type II diabetes and multiple sclerosis, where both TERC and its target genes are overexpressed. These findings position TERC as a potential regulator of immune responses and a therapeutic target in inflammation-driven conditions [141].

Germline mutations in TERC: canonical and non-canonical implications

Germline mutations in TERC gene have been implicated in a spectrum of disorders collectively known as telomere biology disorders (TBDs). These mutations primarily affect telomere maintenance, leading to canonical disease manifestations. However, emerging evidence suggests non-canonical roles for TERC, expanding our understanding of its function in human biology and disease.

Canonical implications of TERC mutations

The canonical effects of TERC mutations are primarily related to impaired telomere maintenance. One of the most well-characterized TBDs is dyskeratosis congenita (DC), an inherited bone marrow failure syndrome. Vulliamy et al. first identified heterozygous TERC mutations in autosomal dominant DC, demonstrating that TERC haploinsufficiency leads to telomere shortening and disease [142]. More recently, it has been shown that zebrafish TERC and human TERC (hTERC) can serve as transcription factors that recruit RNA polymerase II, thereby regulating the expression of myeloid genes. This suggests that TERC’s non-canonical functions are closely tied to DC [147]. TERC mutations have also been found in patients with isolated aplastic anemia, highlighting the variable expressivity of TBDs [148]. These findings have expanded the phenotypic spectrum of TERC mutations to include idiopathic pulmonary fibrosis (IPF) and liver disease [149, 150].

The canonical effects of TERC mutations occur through reduced telomerase activity and accelerated telomere shortening. This leads to premature senescence in highly proliferative tissues, which explains the classic triad of mucocutaneous features, bone marrow failure, and cancer predisposition seen in DC [151]. Interestingly, the same TERC mutation can lead to different phenotypes within a family, suggesting that genetic and environmental modifiers influence disease expression [152].

Non-canonical implications of TERC mutations

Recent studies have uncovered non-canonical implications of TERC mutations. Beyond its role in telomere elongation, TERC has been shown to influence gene expression through interactions with chromatin-modifying complexes. For instance, Chu et al. demonstrated that TERC can bind to chromatin at non-telomeric sites and regulate the transcription of target genes [153]. This suggests that TERC mutations may affect cellular functions independently of telomere length.

Additionally, TERC has been implicated in mitochondrial function. Sahin et al. showed that telomere dysfunction can impair mitochondrial biogenesis and function through p53-mediated repression of PGC-1α and PGC-1β [154]. Although this study focused on TERC knockout mice, it raises the possibility that TERC mutations in humans might similarly affect mitochondrial homeostasis, contributing to the complex phenotypes observed in TBDs.

TERC’s role in cellular senescence extends beyond telomere shortening. Recent work has demonstrated that TERC can regulate cellular senescence through both telomerase-dependent and -independent mechanisms. For example, manipulating TERC levels through overexpression or depletion affects cellular senescence and proliferation even in telomerase-deficient cells (TERC-/-), suggesting non-canonical functions in senescence regulation [35]. These findings suggest that TERC mutations could impact tissue homeostasis through mechanisms beyond simple telomere attrition, potentially contributing to the diverse phenotypes observed in TBDs.

Understanding both the canonical and non-canonical implications of TERC mutations is crucial for improving the diagnosis and treatment of TBDs. Genetic testing for TERC mutations has become an important diagnostic tool for patients with suspected TBDs [155]. Furthermore, emerging therapies targeting telomerase or telomere biology, such as small molecule telomerase activators or gene therapy approaches, hold promise for treating these disorders [156].

TERT and TERC in cancer

TERC demonstrates non-canonical functions in cancer, promoting tumorigenesis and progression through mechanisms that are unrelated to telomere elongation. These roles are multifaceted and involve the regulation of gene expression, cellular signaling, and interactions within the tumor microenvironment [157,158,159]. The role of TERC in cancer development has been particularly elucidated in HPV-related cancers. The E6 and E7 oncogenes from high-risk human papillomavirus (HPV) DNA are present in nearly all cervical cancers, and HPV infection is essential for cancer initiation and development [160]. While E6 and E7 proteins are required for host cell immortalization, they are not sufficient for full cell transformation and tumorigenesis. This suggests that other host factors contribute to malignant transformation and progression. In general, no commonly occurring mutations have been identified in cervical cancer initiation or progression [161]. However, telomerase activity increases in cervical dysplasia due to E6-induced TERT transcription and direct interactions with TERT [162,163,164,165,166,167,168,169,170,171]. We and others have shown increased telomerase activity in cells immortalized by the high-risk HPV E6 and E7 oncogenes via three different mechanisms: activation of hTERT, stabilization of hTERT mRNA, and direct interaction with hTERT protein [162,163,164,165,166,167,168,169,170,171]. TERT is also increasingly expressed in the cascade of cervical dysplasia [172]. Our previous studies revealed a non-canonical function of TERT in HPV-induced immortalization, where TERT regulates cellular gene expression and HPV promoter activity independent of its telomerase activity [22, 162, 173]. Interestingly, TERC, the RNA component of the telomerase complex, is amplified and overexpressed in nearly all human cervical cancers (> 90%) [174, 175]. Studies have shown that TERC expression is elevated in 20–21% of mild dysplasia (CIN I), 50–68% of moderate dysplasia (CIN II), 81–82% of severe dysplasia (CIN III), and 95–100% in invasive cancers [176, 177]. Recently, we demonstrated that high expression of both TERT and TERC led to increased cell growth, anchorage-independent growth in soft agar, and tumor formation in immunodeficient mice [178]. This represents a key progressive step in the conversion of normal to malignant cells. In cells overexpressing both TERT and TERC, the molecular interplay between HPV oncogenes and telomerase is important for tumorigenesis [178]. These data suggest that TERT and TERC play critical roles in the multistep development of human cervical cancer through both telomerase-dependent and -independent pathways.

Sun et al. [157] uncovered a non-canonical role for TERC in the progression of non-small cell lung cancer (NSCLC). Beyond its role in telomere maintenance, TERC facilitates the nuclear localization of TERT. TERC mediates this process by promoting TERT’s interaction with other telomerase subunits, such as SMG6, in the cytoplasm. This process, supported by the nuclear RNA export factor NXF1, is vital for assembling a functional telomerase complex that maintains telomeres and regulates transcription. Elevated TERC levels, driven by promoter hypomethylation, are associated with more advanced cancer stages and poor patient outcomes [157]. Knockdown of TERC disrupts the assembly and nuclear transport of the telomerase complex, resulting in telomere shortening, decreased NSCLC cell proliferation, and reduced expression of oncogenic factors such as c-Myc, Cyclin D1, and VEGF [157]. These findings highlight TERC as a crucial contributor to NSCLC progression and a promising therapeutic target [157]. ​ Wu and associates [158] reported a non-canonical role for TERC in regulating cell division. The study demonstrates that TERC and the PI3K-AKT signaling pathway are involved in a positive feedback loop. TERC enhances AKT phosphorylation and downstream signaling by transcriptionally activating genes such as PDPK1, a crucial regulator of AKT activation. In turn, activated AKT suppresses the transcription factor FOXO1, which negatively regulates TERC expression, thereby maintaining the feedback loop. This mechanism is implicated in activated CD4 + T cells and normal somatic cells, where TERC-induced AKT activation is essential for cell proliferation. Their research also suggests that TERC overexpression may contribute to tumor progression by upregulating the expression of oncogenic targets such as IL4R and EGFR [158]. Cheng and colleagues [159] identified a novel non-canonical role for TERC in the mitochondria, where RNASET2 converts it into TERC-53 and exports it to the cytosol. Cytosolic TERC-53 levels respond to mitochondrial dysfunction, serving as an indicator of mitochondrial states [159]. This highlights a new mitochondrial retrograde signaling mechanism that connects TERC to cellular regulation and potentially to cancer progression. Jin et al. [179] identified a non-canonical role of TERC in suppressing PD-L1 expression independently of telomerase activity. TERC reduces the stability of PD-L1 mRNA by downregulating the RNA-binding protein HuR, which stabilizes PD-L1 transcripts. Elevated TERC levels accelerate PD-L1 mRNA degradation, inhibiting immune escape mechanisms in cancer cells. The study also demonstrated that the FoxO1 inhibitor AS1842856 could upregulate TERC, counteracting chemotherapy-induced PD-L1 expression, offering potential for combination cancer therapy strategies [179]. Kheimar and colleagues [180] showed that TERC overexpression promotes tumor formation in a virus-induced cancer model. Using recombinant Marek’s disease virus, the researchers replaced the viral telomerase RNA (vTR) with cellular TERC and observed that TERC overexpression restored tumor formation and metastasis in the absence of vTR. This effect was independent of viral replication efficiency, suggesting a telomere-independent mechanism. These findings provide direct evidence that cellular TERC contributes to oncogenesis when overexpressed, highlighting its potential role in virus-induced cancer progression [180].

Viruses and host interactions

Early reports indicating a reduced papilloma frequency in TERC-deficient cells suggested a potential correlation between TERC and viruses [181]. Subsequent discoveries of virus-encoded telomerase RNA have revealed mechanistic crosstalk between viruses and TERC. For instance, Marek’s disease herpesvirus can encode a TERC-like viral telomerase RNA, which has been shown to promote tumorigenesis [182]. Notably, this function persists even when the viral telomerase RNA is replaced by cellular TERC [180], and both human TERC and viral telomerase RNA exhibit anti-apoptotic effects [183], indicating functional similarities between telomeric and viral RNA.

The correlation between TERC gene amplification and viral signals has been studied in HPV-infected cells. Simultaneous FISH detection of TERC gene copies and integrated HPV in uterine cervix lesions revealed that HPV integration is associated with an increase in TERC gene copy number [184, 185]. Later studies that co-detected TERC lncRNA and HPV E6/E7 mRNA supported this correlation [186]. These studies primarily focused on the co-detection of HPV and TERC as biomarkers for cervical cancer, rather than exploring the underlying mechanisms of this correlation. Furthermore, their findings were mostly limited to lesion samples. However, the development of HPV E6/E7 immortalized keratinocytes has enabled more in-depth in vivo studies on the HPV-TERC interaction. These studies indicated that the simultaneous overexpression of human TERT and TERC could induce tumorigenesis in E6/E7 immortalized cells [178].

Recent research on telomere-associated proteins has also indirectly connected TERC to viruses mechanistically. Dyskerin 1 (DKC1), a telomerase component that binds TERC to modulate telomere maintenance [187], has been shown to pseudouridylate EBER2, an Epstein-Barr virus (EBV)-encoded noncoding RNA, which is crucial for efficient viral lytic replication [188]. Another telomerase component, NHP2, which stabilizes telomeres [189], was found to be upregulated in cells overexpressing the hepatitis B virus X protein (HBx). Knocking down NHP2 inhibited the proliferation of HBx over-expressed cells [190]. TCAB1, a protein that binds and recruits TERC to the Cajal body [191], was also upregulated by EBV to promote the DNA damage response [192]. TPP1, a shelterin component that indirectly binds TERC and recruits telomerase to telomeres [193], is degraded by herpes simplex virus 1 (HSV-1)-encoded E3 ubiquitin ligase [194]. NOP2, a TERC-binding protein associated with catalytically active telomerase [195], has shown essential functions in association with two types of viruses. The HIV-1 Tat protein can be replaced by NOP2 at its binding site on HIV-1 TAR RNA, promoting viral latency [196]. Similarly, HBx has been reported to be post-transcriptionally regulated by NOP2, which adds five-methylcytosine to HBV mRNA, essential for HBV mRNA transportation and HBx translation [197].

Over the past few decades, TRF2, a shelterin component that modulates telomeric maintenance along with TERC [198], has emerged as a key target for multiple types of viruses. The EBV life cycle has been shown to be regulated by TRF2 and other telomeric proteins through modulation of replication at EBV OriP [199]. Later studies revealed that TRF2 is displaced by EBV nuclear antigen (EBNA)-1 [200] and downregulated by EBV latent membrane protein 1 (LMP1) [201]. TRF2 is also post-transcriptionally inhibited during hepatitis C virus (HCV) infection through p53-dependent Siah-1a ubiquitination [202]. In cells infected with human herpesviruses 6 A and 6B, TRF2 is recruited to viral replication compartments (VRCs) to facilitate viral integration [203]. More recently, a study on SARS-CoV-2 infection reported its effect on downregulating TRF2 [204]. Collectively, these findings suggest that TERC shares structural similarities with viral RNA, is correlated with viral latent integration, and overlaps in its reliance on telomere-associated proteins that are crucial for viral life cycles.

TERC as a target for cancer therapy

In normal somatic cells, telomerase activity is minimal or undetectable, whereas telomerase is frequently upregulated in cancer cells [9]. This makes telomerase a promising therapeutic target for selectively eliminating tumor cells while minimizing adverse effects on healthy tissues.

Small molecule inhibitors and vaccines targeting telomerase are the most common drug candidates in development. The connection between cancer progression and the noncanonical functions of telomerase further strengthens the rationale for targeting telomerase as an anticancer strategy. This approach aims to disrupt the feed-forward regulatory mechanisms that sustain chronic inflammation and oncogenic processes, as well as to mitigate the oncogenic signaling pathways upregulated by telomerase.

Several key signaling pathways involved in tumorigenesis have been linked to telomerase activity, including Wnt/β-catenin [205, 206], NF-κB [205, 207], and DNA damage response (DDR) [208]. For example, the irreversible telomerase inhibitor NU-1 has been shown to impair DNA double-strand break repair by downregulating DDR-related gene expression in the colorectal cancer cell line CT26 [208]. Additionally, novel strategies are emerging to inhibit the interactions between TERT and β-catenin [209] or NF-κB [210], which could complement telomerase-targeting therapies. Further, the non-canonical functions of telomerase components like Reptin and Pontin, which interact with key oncogenic factors such as c-Myc and β-catenin [211,212,213], also present new targets for cancer therapy. Inhibiting these non-canonical functions of TERC could provide an innovative therapeutic approach to suppress tumor growth. Targeting tumor cell survival and proliferation processes that operate independently of telomere elongation could make cancer therapies more effective. Inhibiting telomerase’s catalytic activity alongside its noncanonical functions may induce senescence in proliferating tumor cells by critically shortening telomeres while simultaneously curbing the activation of oncogenic signaling pathways.

These strategies hold promise for inhibiting telomerase-positive tumors with minimal cytotoxic effects on normal tissues, potentially offering an advantage over current anticancer treatment modalities. Further research into the noncanonical roles of telomerase components, their mechanisms, interacting partners, and potential coactivators is essential for the development of effective therapies targeting telomerase-positive cancers.

One promising strategy involves the use of ligands that induce the formation of quadruplex structures in telomeric DNA, such as telomestatin, RHPS4, TMPyP4, and BRACO-19 [214]. In glioblastoma stem-like cells, RHPS4 has been shown to suppress proliferation independently of telomeric dysfunction, suggesting a differential effect in cancer therapy [215]. However, given that G-rich DNA is abundant throughout the genome, particularly in the promoter regions of oncogenes, the use of G4 ligands may pose risks of off-target effects [216, 217]. TMPyP4, for example, inhibits TERT expression and telomerase activity in humans by downregulating TERT transcription [218, 219].

THIO, 6-thio-dG, also named 6-thio-2’-deoxyguanosine is a nucleotide analog that has the potential to induce telomere dysfunction in telomerase-positive cells by being incorporated into telomeric DNA [220]. In non-small cell lung cancer (NSCLC) xenografts, 6-thio-dG has been shown to inhibit tumor growth by promoting telomere damage [221]. This therapeutic effect has been validated in melanoma [221]. This therapeutic effect has been validated in melanoma [222, 223] and Gliomas [224], both in vitro and in vivo.

Additionally, 6-thio-dG has been found to reduce chemotherapy resistance in EGFR inhibitors. Research demonstrated that NSCLC cells resistant to erlotinib, paclitaxel/carboplatin, and gemcitabine/cisplatin became sensitive to 6-thio-dG treatment [25]. More recent studies have shown that resistance to the EGFR mutant inhibitor Osimertinib in NSCLC involves TERT elevation, and combining Osimertinib with 6-thio-dG induces apoptosis in these resistant cells [225].

6-thio-dG has also shown potential in treating therapy-resistant pediatric brain tumors. It effectively inhibits tumor growth in pediatric high-risk group-3 xenografts from neuroblastoma cell lines and orthotopic patient-derived models of diffuse intrinsic pontine glioma [226, 227]. Additionally, combining 6-thio-dG with anti-VEGF and anti-PD-L1 treatments has significantly improved efficacy in hepatocellular carcinoma [228, 229], particularly through the action of CD8 + T cells.

Using compounds that inhibit TERT DNA-binding sounds less efficient than targeting active TERT sites directly [230, 231]. GRN163L (Imetelstat) binds directly to the TERC component in the catalytic site of the telomerase enzyme inhibiting telomerase [232]. It improves the efficacy of chemotherapy and induce cell death in patient-derived xenografts of Acute myeloid leukemia (AML) [233, 234]. Imetelstat showed a reasonable effect in phase II study of lower-risk myelodysplastic patients [235]. In a phase III study, 40% of patients who are refractory to erythropoiesis-stimulating agents or have relapsed achieved transfusion independence at least 8 weeks [43]. BIBR1532 compound binds to the active sites of TERT non-competitively, inhibiting its telomerase activity [236, 237]. A potential anticancer therapeutic strategy was suggested by Amin et al. who found that cellular proliferation in a zebrafish model was short-term inhibited by BIBR1532 [238]. Supporting this finding, a new BIBR1532-based analogue, demonstrated a stronger anticancer activity in the Ehrlich carcinoma model [232]. The findings suggest that telomerase-targeting therapy exhibits significant potential; however, additional research is necessary to mitigate adverse drug reactions.

Lastly, TERT-derived vaccines have emerged as a promising approach to elicit an immune response against various cancers [239]. Several vaccine candidates are currently in clinical trials, either as single agents [240], or in combination with UV1 [241] or HR2822 [242], and in conjunction with checkpoint inhibitors [243,244,245]. TERT vaccines have demonstrated substantial therapeutic efficacy in several cancer types through the recognition of CD4+/CD8 + T cells. However, further investigation is required to assess their safety profile and broader applicability across different cancer types [246,247,248].

Future aspects

Investigating the non-canonical roles of TERC presents exciting opportunities for advancing our understanding of its functions in cell biology, cancer progression, immune modulation, and therapeutic development. While TERC is widely recognized for its role in telomere maintenance, it is also implicated in various other cellular processes. TERC interacts with numerous molecular pathways, contributing to transcription regulation and chromatin remodeling (Fig. 2) [249]. These interactions are vital for maintaining chromatin stability and ensuring the integrity of the genome. Despite this, the non-telomeric functions of TERC remain relatively underexplored. This gap in knowledge highlights the need for further research to fully map how TERC interacts with proteins and chromatin across different cell types. Recent advances in sequencing techniques are providing high-throughput tools that can significantly enhance our understanding of TERC’s non-canonical roles. For instance, Chromatin Isolation by RNA Purification (ChIRP) has proven effective in enriching RNA-binding proteins and DNA fragments associated with specific long noncoding RNAs, including TERC. Using ChIRP, it was discovered that hTERC binds to several Wnt pathway genes, suggesting a potential regulatory role in cellular signaling [33]. However, additional studies are needed to confirm these findings and elucidate the full scope of TERC’s involvement in these processes. Moreover, a recent study using hTERT ChIP-sequencing has shown that TERT can promote the expression of DNMT3b, a DNA methyltransferase, influencing overall DNA methylation patterns [250]. This finding suggests a possible link between telomerase and epigenetic regulation, but whether these non-canonical functions are dependent on TERC remains an open question.

As research progresses, understanding these alternative functions of TERC could provide valuable insights into its broader role in cellular regulation, cancer progression, and immune response, potentially unveiling new therapeutic strategies.

Fig. 2
figure 2

Non-canonical functions of TERC. (a) Biological processes that can be regulated by TERC according to current research. (b) Schematic illustration of potential mechanisms by which TERC performs known and unknown non-canonical functions

In cancer, TERC’s role extends well beyond telomere elongation. It may contribute to tumorigenesis through non-canonical pathways, including inflammation, immune system evasion, and genomic instability [251, 252]. These mechanisms, often less explored, could significantly impact tumor progression, especially in cancers that are less reliant on traditional telomerase activity. Understanding these processes could offer a new perspective on TERC’s contribution to cancer biology, potentially reshaping our approach to tumor progression. Beyond its role in cancer development, TERC holds great promise as a diagnostic biomarker. This is particularly true in malignancies where TERC’s non-canonical functions play a more prominent role [253]. Advancements in detection technologies will be pivotal in unlocking TERC’s potential for early cancer diagnosis, providing more accurate and accessible tools for clinicians.

The therapeutic implications of TERC’s non-canonical functions are expansive. In addition to traditional telomerase inhibitors, therapies designed to modulate TERC’s regulatory roles could offer innovative strategies for cancer and other diseases [251]. Potential approaches include small molecules targeting RNA, antisense oligonucleotides, or CRISPR-based tools aimed at modulating TERC expression—either by disrupting or enhancing its function. Furthermore, TERC’s unique role as a regulatory RNA opens the door for engineering TERC for therapeutic purposes. Synthetic versions could be designed to counteract pathological processes or even promote tissue regeneration [254,255,256]. Integrating TERC-targeted therapies with conventional cancer treatments may also yield synergistic effects, particularly for aggressive or treatment-resistant cancers. This multi-pronged approach could improve treatment outcomes, providing new hope for patients facing challenging diagnoses.

Data availability

The authors have nothing to report.

References

  1. Nakamura TM, Morin GB, Chapman KB, Weinrich SL, Andrews WH, Lingner J, Harley CB, Cech TR. Telomerase catalytic subunit homologs from fission yeast and human. Science. 1997;277:955–9.

    Article  CAS  PubMed  Google Scholar 

  2. Autexier C, Lue NF. The structure and function of telomerase reverse transcriptase. Annu Rev Biochem. 2006;75:493–517.

    Article  CAS  PubMed  Google Scholar 

  3. Greider CW, Blackburn EH. Identification of a specific telomere terminal transferase activity in Tetrahymena extracts. Cell. 1985;43:405–13.

    Article  CAS  PubMed  Google Scholar 

  4. Morin GB. The human telomere terminal transferase enzyme is a ribonucleoprotein that synthesizes TTAGGG repeats. Cell. 1989;59:521–9.

    Article  CAS  PubMed  Google Scholar 

  5. Harrington L, McPhail T, Mar V, Zhou W, Oulton R, Bass MB, Arruda I, Robinson MO. A mammalian telomerase-associated protein. Science. 1997;275:973–7.

    Article  CAS  PubMed  Google Scholar 

  6. Holt SE, Aisner DL, Baur J, Tesmer VM, Dy M, Ouellette M, Trager JB, Morin GB, Toft DO, Shay JW, Wright WE, White MA. Functional requirement of p23 and Hsp90 in telomerase complexes. Genes Dev. 1999;13:817–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Shay JW. Role of telomeres and telomerase in aging and Cancer. Cancer Discov. 2016;6:584–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Hornsby PJ. Telomerase and the aging process. Exp Gerontol. 2007;42:575–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Kim NW, Piatyszek MA, Prowse KR, Harley CB, West MD, Ho PL, Coviello GM, Wright WE, Weinrich SL, Shay JW. Specific association of human telomerase activity with immortal cells and cancer. Science. 1994;266:2011–5.

    Article  CAS  PubMed  Google Scholar 

  10. Wright WE, Piatyszek MA, Rainey WE, Byrd W, Shay JW. Telomerase activity in human germline and embryonic tissues and cells. Dev Genet. 1996;18:173–9.

    Article  CAS  PubMed  Google Scholar 

  11. Ulaner GA, Hu JF, Vu TH, Giudice LC, Hoffman AR. Tissue-specific alternate splicing of human telomerase reverse transcriptase (hTERT) influences telomere lengths during human development. Int J Cancer. 2001;91:644–9.

    Article  CAS  PubMed  Google Scholar 

  12. Ito H, Kyo S, Kanaya T, Takakura M, Koshida K, Namiki M, Inoue M. Detection of human telomerase reverse transcriptase messenger RNA in voided urine samples as a useful diagnostic tool for bladder cancer. Clin Cancer Res. 1998;4:2807–10.

    CAS  PubMed  Google Scholar 

  13. Avilion AA, Piatyszek MA, Gupta J, Shay JW, Bacchetti S, Greider CW. Human telomerase RNA and telomerase activity in immortal cell lines and tumor tissues. Cancer Res. 1996;56:645–50.

    CAS  PubMed  Google Scholar 

  14. Huang FW, Hodis E, Xu MJ, Kryukov GV, Chin L, Garraway LA. Highly recurrent TERT promoter mutations in human melanoma. Science. 2013;339:957–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Barthel FP, Wei W, Tang M, Martinez-Ledesma E, Hu X, Amin SB, Akdemir KC, Seth S, Song X, Wang Q, Lichtenberg T, Hu J, Zhang J, Zheng S, Verhaak RG. Systematic analysis of telomere length and somatic alterations in 31 cancer types. Nat Genet. 2017;49:349–57.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Kawashima M, Kojima M, Ueda Y, Kurihara S, Hiyama E. Telomere biology including TERT rearrangements in neuroblastoma: a useful indicator for surgical treatments. J Pediatr Surg. 2016;51:2080–5.

    Article  PubMed  Google Scholar 

  17. Guilleret I, Yan P, Grange F, Braunschweig R, Bosman FT, Benhattar J. Hypermethylation of the human telomerase catalytic subunit (hTERT) gene correlates with telomerase activity. Int J Cancer. 2002;101:335–41.

    Article  CAS  PubMed  Google Scholar 

  18. Lewis KA, Tollefsbol TO. Regulation of the Telomerase Reverse Transcriptase Subunit through Epigenetic mechanisms. Front Genet. 2016;7:83.

    Article  PubMed  PubMed Central  Google Scholar 

  19. Cong YS, Wright WE, Shay JW. 2002. Human telomerase and its regulation. Microbiol Mol Biol Rev 66:407– 25, table of contents.

  20. Kim W, Ludlow AT, Min J, Robin JD, Stadler G, Mender I, Lai TP, Zhang N, Wright WE, Shay JW. Regulation of the human telomerase gene TERT by Telomere position effect-over long distances (TPE-OLD): implications for aging and Cancer. PLoS Biol. 2016;14:e2000016.

    Article  PubMed  PubMed Central  Google Scholar 

  21. Leao R, Apolonio JD, Lee D, Figueiredo A, Tabori U, Castelo-Branco P. Mechanisms of human telomerase reverse transcriptase (hTERT) regulation: clinical impacts in cancer. J Biomed Sci. 2018;25:22.

    Article  PubMed  PubMed Central  Google Scholar 

  22. Rasouli S, Dakic A, Wang QE, Mitchell D, Blakaj DM, Putluri N, Li J, Liu X. Noncanonical functions of telomerase and telomeres in viruses-associated cancer. J Med Virol. 2024;96:e29665.

    Article  CAS  PubMed  Google Scholar 

  23. Kuglik P, Kasikova K, Smetana J, Vallova V, Lastuvkova A, Moukova L, Cvanova M, Brozova L. Molecular cytogenetic analyses of hTERC (3q26) and MYC (8q24) genes amplifications in correlation with oncogenic human papillomavirus infection in Czech patients with cervical intraepithelial neoplasia and cervical carcinomas. Neoplasma. 2015;62:130–9.

    Article  CAS  PubMed  Google Scholar 

  24. Cao Y, Bryan TM, Reddel RR. Increased copy number of the TERT and TERC telomerase subunit genes in cancer cells. Cancer Sci. 2008;99:1092–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Mender I, LaRanger R, Luitel K, Peyton M, Girard L, Lai TP, Batten K, Cornelius C, Dalvi MP, Ramirez M, Du W, Wu LF, Altschuler SJ, Brekken R, Martinez ED, Minna JD, Wright WE, Shay JW. Telomerase-mediated strategy for overcoming Non-small Cell Lung Cancer targeted therapy and Chemotherapy Resistance. Neoplasia. 2018;20:826–37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Shay JW, Wright WE. Telomerase therapeutics for cancer: challenges and new directions. Nat Rev Drug Discov. 2006;5:577–84.

    Article  CAS  PubMed  Google Scholar 

  27. Harley CB. Telomerase therapeutics for degenerative diseases. Curr Mol Med. 2005;5:205–11.

    Article  CAS  PubMed  Google Scholar 

  28. Segal-Bendirdjian E, Geli V. Non-canonical roles of telomerase: unraveling the Imbroglio. Front Cell Dev Biol. 2019;7:332.

    Article  PubMed  PubMed Central  Google Scholar 

  29. Nassir N, Hyde GJ, Baskar R. A telomerase with novel non-canonical roles: TERT controls cellular aggregation and tissue size in Dictyostelium. PLoS Genet. 2019;15:e1008188.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Xi L, Cech TR. Inventory of telomerase components in human cells reveals multiple subpopulations of hTR and hTERT. Nucleic Acids Res. 2014;42:8565–77.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Kedde M, le Sage C, Duursma A, Zlotorynski E, van Leeuwen B, Nijkamp W, Beijersbergen R, Agami R. Telomerase-independent regulation of ATR by human telomerase RNA. J Biol Chem. 2006;281:40503–14.

    Article  CAS  PubMed  Google Scholar 

  32. Gazzaniga FS, Blackburn EH. An antiapoptotic role for telomerase RNA in human immune cells independent of telomere integrity or telomerase enzymatic activity. Blood. 2014;124:3675–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Chu C, Qu K, Zhong FL, Artandi SE, Chang HY. Genomic maps of long noncoding RNA occupancy reveal principles of RNA-chromatin interactions. Mol Cell. 2011;44:667–78.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Cheng Y, Liu P, Zheng Q, Gao G, Yuan J, Wang P, Huang J, Xie L, Lu X, Tong T, Chen J, Lu Z, Guan J, Wang G. Mitochondrial trafficking and Processing of Telomerase RNA TERC. Cell Rep. 2018;24:2589–95.

    Article  CAS  PubMed  Google Scholar 

  35. Zheng Q, Liu P, Gao G, Yuan J, Wang P, Huang J, Xie L, Lu X, Di F, Tong T, Chen J, Lu Z, Guan J, Wang G. Mitochondrion-processed TERC regulates senescence without affecting telomerase activities. Protein Cell. 2019;10:631–48.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Jafri MA, Ansari SA, Alqahtani MH, Shay JW. Roles of telomeres and telomerase in cancer, and advances in telomerase-targeted therapies. Genome Med. 2016;8:1–18.

    Article  Google Scholar 

  37. Shay JW, Wright WE. Role of telomeres and telomerase in cancer. Sem Cancer Biol. 2011;21:349–53.

    Article  CAS  Google Scholar 

  38. Zhao N, Yin G, Liu C, Zhang W, Shen Y, Wang D, Lin Z, Yang J, Mao J, Guo R. Critically short telomeres derepress retrotransposons to promote genome instability in embryonic stem cells. Cell Discovery. 2023;9:45.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Guterres AN, Villanueva J. Targeting telomerase for cancer therapy. Oncogene. 2020;39:5811–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Baerlocher GM, Oppliger Leibundgut E, Ottmann OG, Spitzer G, Odenike O, McDevitt MA, Röth A, Daskalakis M, Burington B, Stuart M. Telomerase inhibitor imetelstat in patients with essential thrombocythemia. N Engl J Med. 2015;373:920–8.

    Article  CAS  PubMed  Google Scholar 

  41. Asai A, Oshima Y, Yamamoto Y, Uochi T, Kusaka H, Akinaga S, Yamashita Y, Pongracz K, Pruzan R, Wunder E, Piatyszek M, Li SH, Chin AC, Harley CB, Gryaznov S. A novel telomerase template antagonist (GRN163) as a potential anticancer agent. Cancer Res. 2003;63:3931–9.

    CAS  PubMed  Google Scholar 

  42. Patnaik MM. Telomerase inhibition in haematological neoplasms-are we ready for primetime? Lancet. 2024;403:220–2.

    Article  CAS  PubMed  Google Scholar 

  43. Platzbecker U, Santini V, Fenaux P, Sekeres MA, Savona MR, Madanat YF, Diez-Campelo M, Valcarcel D, Illmer T, Jonasova A, Belohlavkova P, Sherman LJ, Berry T, Dougherty S, Shah S, Xia Q, Sun L, Wan Y, Huang F, Ikin A, Navada S, Feller F, Komrokji RS, Zeidan AM. Imetelstat in patients with lower-risk myelodysplastic syndromes who have relapsed or are refractory to erythropoiesis-stimulating agents (IMerge): a multinational, randomised, double-blind, placebo-controlled, phase 3 trial. Lancet. 2024;403:249–60.

    Article  CAS  PubMed  Google Scholar 

  44. Damm K, Hemmann U, Garin-Chesa P, Hauel N, Kauffmann I, Priepke H, Niestroj C, Daiber C, Enenkel B, Guilliard B. A highly selective telomerase inhibitor limiting human cancer cell proliferation. The EMBO journal; 2001.

  45. Majumder M, House R, Palanisamy N, Qie S, Day TA, Neskey D, Diehl JA, Palanisamy V. RNA-binding protein FXR1 regulates p21 and TERC RNA to bypass p53-mediated cellular senescence in OSCC. PLoS Genet. 2016;12:e1006306.

    Article  PubMed  PubMed Central  Google Scholar 

  46. Sola L, Nergadze SG, Cappelletti E, Piras FM, Giulotto E, Santagostino M. Telomeric-like repeats flanked by sequences retrotranscribed from the telomerase RNA inserted at DNA double-strand Break sites during Vertebrate Genome Evolution. Int J Mol Sci. 2021;22:11048.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Xu Y, Goldkorn A. Telomere and telomerase therapeutics in cancer. Genes. 2016;7:22.

    Article  PubMed  PubMed Central  Google Scholar 

  48. Sallinen P, Miettinen H, Sallinen SL, Haapasalo H, Helin H, Kononen J. Increased expression of telomerase RNA component is associated with increased cell proliferation in human astrocytomas. Am J Pathol. 1997;150:1159–64.

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Azzalin CM, Reichenbach P, Khoriauli L, Giulotto E, Lingner J. Telomeric repeat-containing RNA and RNA surveillance factors at mammalian chromosome ends. Science. 2007;318:798–801.

    Article  CAS  PubMed  Google Scholar 

  50. Schoeftner S, Blasco MA. Developmentally regulated transcription of mammalian telomeres by DNA-dependent RNA polymerase II. Nat Cell Biol. 2008;10:228–36.

    Article  CAS  PubMed  Google Scholar 

  51. Canale P, Campolo J, Borghini A, Andreassi MG. Long telomeric repeat-containing RNA (TERRA): Biological functions and challenges in Vascular Aging and Disease. Biomedicines. 2023;11:3211.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Gong Y, Liu Y. R-loops at chromosome ends: from formation, regulation, and cellular consequence. Cancers. 2023;15:2178.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Heaphy CM, Subhawong AP, Hong S-M, Goggins MG, Montgomery EA, Gabrielson E, Netto GJ, Epstein JI, Lotan TL, Westra WH. Prevalence of the alternative lengthening of telomeres telomere maintenance mechanism in human cancer subtypes. Am J Pathol. 2011;179:1608–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Brickner JR, Garzon JL, Cimprich KA. Walking a tightrope: the complex balancing act of R-loops in genome stability. Mol Cell. 2022;82:2267–97.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Guo J, Huang XQ, Dou L, Yan MJ, Shen T, Tang WQ, Li J. Aging and aging-related diseases: from molecular mechanisms to interventions and treatments. Signal Transduct Target Therapy. 2022;7:40.

    Google Scholar 

  56. van Deursen JM. The role of senescent cells in ageing. Nature. 2014;509:439–46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Hayflick L, Moorhead PS. SERIAL CULTIVATION OF HUMAN DIPLOID CELL STRAINS. Exp Cell Res. 1961;25:585–.

    Article  CAS  PubMed  Google Scholar 

  58. Toussaint O, Medrano EE, von Zglinicki T. Cellular and molecular mechanisms of stress-induced premature senescence (SIPS) of human diploid fibroblasts and melanocytes. Exp Gerontol. 2000;35:927–45.

    Article  CAS  PubMed  Google Scholar 

  59. Kornienko JS, Smirnova IS, Pugovkina NA, Ivanova JS, Shilina MA, Grinchuk TM, Shatrova AN, Aksenov ND, Zenin VV, Nikolsky NN, Lyublinskaya OG. High doses of synthetic antioxidants induce premature senescence in cultivated mesenchymal stem cells. Sci Rep. 2019;9:13.

    Article  Google Scholar 

  60. Huang WJ, Hickson LJ, Eirin A, Kirkland JL, Lerman LO. Cellular senescence: the good, the bad and the unknown. Nat Rev Nephrol. 2022;18:611–27.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Olivieri F, Albertini MC, Orciani M, Ceka A, Cricca M, Procopio AD, Bonafè M. DNA damage response (DDR) and senescence: shuttled inflamma-miRNAs on the stage of inflamm-aging. Oncotarget. 2015;6:35509–21.

    Article  PubMed  PubMed Central  Google Scholar 

  62. Wang Y, Sharpless N, Chang S. p16 < SUP > INK4a protects against dysfunctional telomere-induced ATR-dependent DNA damage responses. J Clin Invest. 2013;123:4489–501.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Palm W, de Lange T. How Shelterin protects mammalian telomeres. Annu Rev Genet. 2008;42:301–34.

    Article  CAS  PubMed  Google Scholar 

  64. Kibe T, Zimmermann M, de Lange T. TPP1 blocks an ATR-Mediated resection mechanism at telomeres. Mol Cell. 2016;61:236–46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. di Fagagna FD, Reaper PM, Clay-Farrace L, Fiegler H, Carr P, von Zglinicki T, Saretzki G, Carter NP, Jackson SP. A DNA damage checkpoint response in telomere-initiated senescence. Nature. 2003;426:194–8.

    Article  Google Scholar 

  66. Takai H, Smogorzewska A, de Lange T. DNA damage foci at dysfunctional telomeres. Curr Biol. 2003;13:1549–56.

    Article  CAS  PubMed  Google Scholar 

  67. Mir SM, Tehrani SS, Goodarzi G, Jamalpoor Z, Asadi J, Khelghati N, Qujeq D, Maniati M. Shelterin Complex at telomeres: implications in Ageing. Clin Interv Aging. 2020;15:827–39.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Herr LM, Schaffer ED, Fuchs KF, Datta A, Brosh RM Jr. Replication stress as a driver of cellular senescence and aging. Commun Biology. 2024;7:19.

    Article  Google Scholar 

  69. Sfeir A, Kosiyatrakul ST, Hockemeyer D, MacRae SL, Karlseder J, Schildkraut CL, de Lange T. Mammalian telomeres resemble Fragile sites and require TRF1 for efficient replication. Cell. 2009;138:90–103.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Ye J, Lenain C, Bauwens S, Rizzo A, Saint-Léger A, Poulet A, Benarroch D, Magdinier F, Morere J, Amiard S, Verhoeyen E, Britton S, Calsou P, Salles B, Bizard A, Nadal M, Salvati E, Sabatier L, Wu YL, Biroccio A, Londoño-Vallejo A, Giraud-Panis MJ, Gilson E. TRF2 and Apollo Cooperate with topoisomerase 2α to protect human telomeres from replicative damage. Cell. 2010;142:230–42.

    Article  CAS  PubMed  Google Scholar 

  71. Ohki R, Ishikawa F. Telomere-bound TRF1 and TRF2 stall the replication fork at telomeric repeats. Nucleic Acids Res. 2004;32:1627–37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Leman AR, Noguchi E. Local and global functions of Timeless and Tipin in replication fork protection. Cell Cycle. 2012;11:3945–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Leman AR, Dheekollu J, Deng Z, Lee SW, Das MM, Lieberman PM, Noguchi E. Timeless preserves telomere length by promoting efficient DNA replication through human telomeres. Cell Cycle. 2012;11:2337–47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Wang H, Nora GJ, Ghodke H, Opresko PL. Single molecule studies of physiologically relevant Telomeric tails reveal POT1 mechanism for promoting G-quadruplex unfolding. J Biol Chem. 2011;286:7479–89.

    Article  CAS  PubMed  Google Scholar 

  75. 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.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Huang CH, Dai XY, Chai WH. Human Stn1 protects telomere integrity by promoting efficient lagging-strand synthesis at telomeres and mediating C-strand fill-in. Cell Res. 2012;22:1681–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Mohaghegh P, Karow JK, Brosh RM, Bohr VA, Hickson ID. The Bloom’s and Werner’s syndrome proteins are DNA structure-specific helicases. Nucleic Acids Res. 2001;29:2843–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Vannier JB, Pavicic-Kaltenbrunner V, Petalcorin MIR, Ding H, Boulton SJ. RTEL1 dismantles T loops and counteracts telomeric G4-DNA to maintain Telomere Integrity. Cell. 2012;149:795–806.

    Article  CAS  PubMed  Google Scholar 

  79. Flynn RL, Cox KE, Jeitany M, Wakimoto H, Bryll AR, Ganem NJ, Bersani F, Pineda JR, Suvà ML, Benes CH, Haber DA, Boussin FD, Zou L. Alternative lengthening of telomeres renders cancer cells hypersensitive to ATR inhibitors. Science. 2015;347:273–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Arora R, Lee Y, Wischnewski H, Brun CM, Schwarz T, Azzalin CM. RNaseH1 regulates TERRA-telomeric DNA hybrids and telomere maintenance in ALT tumour cells. Nat Commun. 2014;5:11.

    Article  Google Scholar 

  81. Günes C, Rudolph KL. The role of telomeres in stem cells and Cancer. Cell. 2013;152:390–3.

    Article  PubMed  Google Scholar 

  82. Huda A, Arakawa H, Mazzucco G, Galli M, Petrocelli V, Casola S, Chen L, Doksani Y. The telomerase reverse transcriptase elongates reversed replication forks at telomeric repeats. Sci Adv. 2023;9:12.

    Article  Google Scholar 

  83. Frank AK, Tran DC, Qu RW, Stohr BA, Segal DJ, Xu LF. The shelterin TIN2 subunit mediates recruitment of telomerase to telomeres. PLoS Genet. 2015;11:19.

    Article  Google Scholar 

  84. Lee JY, Okumus B, Kim DS, Ha TJ. Extreme conformational diversity in human telomeric DNA. Proc Natl Acad Sci USA. 2005;102:18938–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Nandakumar J, Bell CF, Weidenfeld I, Zaug AJ, Leinwand LA, Cech TR. The TEL patch of telomere protein TPP1 mediates telomerase recruitment and processivity. Nature. 2012;492:285–.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Zhong FL, Batista LFZ, Freund A, Pech MF, Venteicher AS, Artandi SE. TPP1 OB-Fold domain controls Telomere maintenance by recruiting telomerase to chromosome ends. Cell. 2012;150:481–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Chen LY, Redon S, Lingner J. The human CST complex is a terminator of telomerase activity. Nature. 2012;488:540–.

    Article  CAS  PubMed  Google Scholar 

  88. Kaminski N, Wondisford AR, Kwon Y, Lynskey ML, Bhargava R, Barroso-González J, García-Expósito L, He BX, Xu M, Mellacheruvu D, Watkins SC, Modesti M, Miller KM, Nesvizhskii AI, Zhang HY, Sung P, O’Sullivan RJ. RAD51AP1 regulates ALT-HDR through chromatin-directed homeostasis of TERRA. Mol Cell. 2022;82:4001–.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Pizzul P, Rinaldi C, Bonetti D. The multistep path to replicative senescence onset: zooming on triggering and inhibitory events at telomeric DNA. Front Cell Dev Biology. 2023;11:7.

    Article  Google Scholar 

  90. Zhang JM, Zou L. Alternative lengthening of telomeres: from molecular mechanisms to therapeutic outlooks. Cell Bioscience. 2020;10:9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Liang FS, Rai R, Sodeinde T, Chang SY. TRF2-RAP1 represses RAD51-dependent homology-directed telomere repair by promoting BLM-mediated D-loop unwinding and inhibiting BLM-DNA2-dependent 5′-end resection. Nucleic Acids Res. 2024;52:9695–709.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Rai R, Biju K, Sun W, Sodeinde T, Al-Hiysat A, Morgan J, Ye X, Li X, Chen Y, Chang S. Homology directed telomere clustering, ultrabright telomere formation and nuclear envelope rupture in cells lacking TRF2 < SUP > B and RAP1. Nat Commun. 2023;14:20.

    Google Scholar 

  93. Jiang WQ, Zhong ZH, Henson JD, Reddel RR. Identification of candidate alternative lengthening of telomeres genes by methionine restriction and RNA interference. Oncogene. 2007;26:4635–47.

    Article  CAS  PubMed  Google Scholar 

  94. Yu J, Lan JP, Wang C, Wu Q, Zhu YY, Lai XY, Sun J, Jin CJ, Huang H. PML3 interacts with TRF1 and is essential for ALT-associated PML bodies assembly in U2OS cells. Cancer Lett. 2010;291:177–86.

    Article  CAS  PubMed  Google Scholar 

  95. Pérez-Martínez L, Wagner T, Luke B. Telomere Interacting proteins and TERRA Regulation. Front Genet. 2022;13:8.

    Article  Google Scholar 

  96. Pérez-Martínez L, Öztürk M, Butter F, Luke B. Npl3 stabilizes R-loops at telomeres to prevent accelerated replicative senescence. EMBO Rep. 2020;21:12.

    Article  Google Scholar 

  97. Graf M, Bonetti D, Lockhart A, Serhal K, Kellner V, Maicher A, Jolivet P, Teixeira MT, Luke B. Telomere length determines TERRA and R-Loop Regulation through the cell cycle. Cell. 2017;170:72–.

    Article  CAS  PubMed  Google Scholar 

  98. Ghadaouia S, Olivier MA, Martinez A, Kientega T, Qin J, Lambert-Lanteigne P, Cardin GB, Autexier C, Malaquin N, Rodier F. Homologous recombination-mediated irreversible genome damage underlies telomere-induced senescence. Nucleic Acids Res. 2021;49:11690–707.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Chen JH, Hales CN, Ozanne SE. DNA damage, cellular senescence and organismal ageing: causal or correlative? Nucleic Acids Res. 2007;35:7417–28.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Rossiello F, Jurk D, Passos JF, di Fagagna FD. Telomere dysfunction in ageing and age-related diseases. Nat Cell Biol. 2022;24:135–47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Hewitt G, Jurk D, Marques FDM, 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;3:9.

    Article  Google Scholar 

  102. Fouquerel E, Barnes RP, Uttam S, Watkins SC, Bruchez MP, Opresko PL. Targeted and persistent 8-Oxoguanine base damage at Telomeres promotes Telomere loss and Crisis. Mol Cell. 2019;75:117–.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Barnes RP, de Rosa M, Thosar SA, Detwiler AC, Roginskaya V, Van Houten B, Bruchez MP, Stewart-Ornstein J, Opresko PL. Telomeric 8-oxo-guanine drives rapid premature senescence in the absence of telomere shortening. Nat Struct Mol Biol. 2022;29:639–.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Opresko PL, Fan JH, Danzy S, Wilson DM, Bohr VA. Oxidative damage in telomeric DNA disrupts recognition by TRF1 and TRF2. Nucleic Acids Res. 2005;33:1230–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Fouquerel E, Lormand J, Bose A, Lee HT, Kim GS, Li JF, Sobol RW, Freudenthal BD, Myong S, Opresko PL. Oxidative guanine base damage regulates human telomerase activity. Nat Struct Mol Biol. 2016;23:1092–100.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Sanford SL, Welfer GA, Freudenthal BD, Opresko PL. Mechanisms of telomerase inhibition by oxidized and therapeutic dNTPs. Nat Commun. 2020;11:12.

    Article  Google Scholar 

  107. Robin JD, Burbano MSJ, Peng H, Croce O, Thomas JL, Laberthonniere C, Renault V, Lototska L, Pousse M, Tessier F, Bauwens S, Leong W, Sacconi S, Schaeffer L, Magdinier F, Ye J, Gilson E. Mitochondrial function in skeletal myofibers is controlled by a TRF2-SIRT3 axis over lifetime. Aging Cell. 2020;19:16.

    Article  Google Scholar 

  108. Burbano MSJ, Robin JD, Bauwens S, Martin M, Donati E, Martínez L, Lin PP, Sacconi S, Magdinier F, Gilson E. Non-canonical telomere protection role of FOXO3a of human skeletal muscle cells regulated by the TRF2-redox axis. Commun Biology. 2023;6:11.

    Google Scholar 

  109. Chen LY, Zhang Y, Zhang QF, Li HZ, Luo ZH, Fang HZ, Kim SH, Qin L, Yotnda P, Xu JM, Tu BP, Bai YD, Zhou SY. Mitochondrial localization of Telomeric protein TIN2 links Telomere Regulation to Metabolic Control. Mol Cell. 2012;47:839–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Ahmed S, Passos JF, Birket MJ, Beckmann T, Brings S, Peters H, Birch-Machin MA, von Zglinicki T, Saretzki G. Telomerase does not counteract telomere shortening but protects mitochondrial function under oxidative stress. J Cell Sci. 2008;121:1046–53.

    Article  CAS  PubMed  Google Scholar 

  111. Eitan E, Braverman C, Tichon A, Gitler D, Hutchison ER, Mattson MP, Priel E. Excitotoxic and Radiation stress increase TERT levels in the Mitochondria and Cytosol of cerebellar purkinje neurons. Cerebellum. 2016;15:509–17.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Fumagalli M, Rosiello F, Clerici M, Barozzi S, Cittaro D, Kaplunov JM, Bucci G, Dobreva M, Matti V, Beausejour CM, Herbig U, Longhese MP, di Fagagna FD. Telomeric DNA damage is irreparable and causes persistent DNA-damage-response activation. Nat Cell Biol. 2012;14:355–.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Von Zglinicki T, Pilger R, Sitte N. Accumulation of single-strand breaks is the major cause of telomere shortening in human fibroblasts. Free Radic Biol Med. 2000;28:64–74.

    Article  Google Scholar 

  114. Richter T, Saretzki G, Nelson G, Melcher M, Olijslagers S, von Zglinicki T. TRF2 overexpression diminishes repair of telomeric single-strand breaks and accelerates telomere shortening in human fibroblasts. Mech Ageing Dev. 2007;128:340–5.

    Article  CAS  PubMed  Google Scholar 

  115. Ceccaldi R, Rondinelli B, D’Andrea AD. Repair pathway choices and consequences at the double-strand break. Trends Cell Biol. 2016;26:52–64.

    Article  CAS  PubMed  Google Scholar 

  116. Ribes-Zamora A, Indiviglio SM, Mihalek I, Williams CL, Bertuch AA. TRF2 Interaction with Ku Heterotetramerization Interface gives insight into c-NHEJ Prevention at Human telomeres. Cell Rep. 2013;5:194–206.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Bombarde O, Boby C, Gomez D, Frit P, Giraud-Panis MJ, Gilson E, Salles B, Calsou P. TRF2/RAP1 and DNA-PK mediate a double protection against joining at telomeric ends. EMBO J. 2010;29:1573–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Storchova R, Palek M, Palkova N, Veverka P, Brom T, Hofr C, Macurek L. Phosphorylation of TRF2 promotes its interaction with TIN2 and regulates DNA damage response at telomeres. Nucleic Acids Res. 2023;51:1154–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Tan J, Duan M, Yadav T, Phoon L, Wang XY, Zhang JM, Zou L, Lan L. An R-loop-initiated CSB-RAD52-POLD3 pathway suppresses ROS-induced telomeric DNA breaks. Nucleic Acids Res. 2020;48:1285–300.

    Article  CAS  PubMed  Google Scholar 

  120. Hedström E, Pederiva C, Farnebo J, Nodin B, Jirström K, Brennan DJ, Farnebo M. Downregulation of the cancer susceptibility protein WRAP53β in epithelial ovarian cancer leads to defective DNA repair and poor clinical outcome. Cell Death Dis. 2015;6:9.

    Article  Google Scholar 

  121. Lee KH, Kim DY, Kim W. Regulation of Gene expression by Telomere position effect. Int J Mol Sci. 2021;22:13.

    Article  Google Scholar 

  122. Robin JD, Ludlow AT, Batten K, Magdinier F, Stadler G, Wagner KR, Shay JW, Wright WE. Telomere position effect: regulation of gene expression with progressive telomere shortening over long distances. Genes Dev. 2014;28:2464–76.

    Article  PubMed  PubMed Central  Google Scholar 

  123. Kim W, Shay JW. Long-range telomere regulation of gene expression: Telomere looping and telomere position effect over long distances (TPE-OLD). Differentiation. 2018;99:1–9.

    Article  PubMed  Google Scholar 

  124. Indran IR, Hande MP, Pervaiz S. hTERT overexpression alleviates intracellular ROS production, improves mitochondrial function, and inhibits ROS-Mediated apoptosis in Cancer cells. Cancer Res. 2011;71:266–76.

    Article  CAS  PubMed  Google Scholar 

  125. Shin WH, Chung KC. Human telomerase reverse transcriptase positively regulates mitophagy by inhibiting the processing and cytoplasmic release of mitochondrial PINK1. Cell Death Dis. 2020;11:14.

    Article  Google Scholar 

  126. Ghosh A, Saginc G, Leow SC, Khattar E, Shin EM, Yan TD, Wong M, Zhang ZZ, Li GL, Sung WK, Zhou JB, Chng WJ, Li S, Liu E, Tergaonkar V. Telomerase directly regulates NF-κB-dependent transcription. Nat Cell Biol. 2012;14:1270–.

    Article  CAS  PubMed  Google Scholar 

  127. Wu LL, Fidan K, Um JY, Ahn KS. Telomerase: key regulator of inflammation and cancer. Pharmacol Res. 2020;155:12.

    Article  Google Scholar 

  128. Venteicher AS, Meng ZJ, Mason PJ, Veenstra TD, Artandi SE. Identification of ATPases pontin and reptin as telomerase components essential for holoenzyme assembly. Cell. 2008;132:945–57.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. O’Brien R, Tran SL, Maritz MF, Liu B, Kong CF, Purgato S, Yang C, Murray J, Russell AJ, Flemming CL, von Jonquieres G, Pickett HA, London WB, Haber M, Gunaratne PH, Norris MD, Perini G, Fletcher JI, MacKenzie KL. <i > MYC-Driven neuroblastomas are addicted to a telomerase-independent function of Dyskerin</i >. Cancer Res. 2016;76:3604–17.

    Article  PubMed  Google Scholar 

  130. Healy AR, Houston DR, Remnant L, Huart AS, Brychtova V, Maslon MM, Meers O, Muller P, Krejci A, Blackburn EA, Vojtesek B, Hernychova L, Walkinshaw MD, Westwood NJ, Hupp TR. Discovery of a novel ligand that modulates the protein-protein interactions of the AAA plus superfamily oncoprotein reptin. Chem Sci. 2015;6:3109–16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Marrone A, Sokhal P, Walne A, Beswick R, Kirwan M, Killick S, Williams M, Marsh J, Vulliamy T, Dokal I. Functional characterization of novel telomerase RNA (TERC) mutations in patients with diverse clinical and pathological presentations. Haematologica. 2007;92:1013.

    Article  CAS  PubMed  Google Scholar 

  132. Vulliamy TJ, Marrone A, Knight SW, Walne A, Mason PJ, Dokal I. Mutations in dyskeratosis congenita: their impact on telomere length and the diversity of clinical presentation. Blood. 2006;107:2680–5.

    Article  CAS  PubMed  Google Scholar 

  133. Biron-Shental T, Kitay-Cohen Y, Tene T, Sharony R, Amiel A. Increased TERC gene copy number in amniocytes from fetuses with trisomy 18 or a sex chromosome aneuploidy. Gene. 2012;506:46–9.

    Article  CAS  PubMed  Google Scholar 

  134. Laish I, Katz H, Sulayev Y, Liberman M, Naftali T, Benjaminov F, Stein A, Kitay-Cohen Y, Biron-Shental T, Konikoff F. Increased TERC gene copy number and cells in senescence in primary sclerosing cholangitis compared to colitis and control patients. Gene. 2013;529:245–9.

    Article  CAS  PubMed  Google Scholar 

  135. Mohamad Ashari ZS, Sulong S, Hassan R, Husin A, Sim GA, Wahid S. Low level of TERC gene amplification between chronic myeloid leukaemia patients resistant and respond to imatinib mesylate treatment. Asian Pac J Cancer Prev. 2014;15:1863–9.

    Article  PubMed  Google Scholar 

  136. Baran I, Nalcaci R, Kocak M. Dyskeratosis congenita: clinical report and review of the literature. Int J Dental Hygiene. 2010;8:68–74.

    Article  CAS  Google Scholar 

  137. Li J, Ge M, Lu S, Shi J, Li X, Wang M, Huang J, Shao Y, Huang Z, Zhang J. Pro-inflammatory effects of the Th1 chemokine CXCL10 in acquired aplastic anaemia. Cytokine. 2017;94:45–51.

    Article  CAS  PubMed  Google Scholar 

  138. Vulliamy TJ, Kirwan MJ, Beswick R, Hossain U, Baqai C, Ratcliffe A, Marsh J, Walne A, Dokal I. Differences in disease severity but similar telomere lengths in genetic subgroups of patients with telomerase and shelterin mutations. PLoS ONE. 2011;6:e24383.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Collopy LC, Walne AJ, Cardoso S, de la Fuente J, Mohamed M, Toriello H, Tamary H, Ling AJ, Lloyd T, Kassam R. Triallelic and epigenetic-like inheritance in human disorders of telomerase. Blood J Am Soc Hematol. 2015;126:176–84.

    CAS  Google Scholar 

  140. Yamaguchi H, Sakaguchi H, Yoshida K, Yabe M, Yabe H, Okuno Y, Muramatsu H, Takahashi Y, Yui S, Shiraishi Y. Clinical and genetic features of dyskeratosis congenita, cryptic dyskeratosis congenita, and Hoyeraal-Hreidarsson syndrome in Japan. Int J Hematol. 2015;102:544–52.

    Article  PubMed  Google Scholar 

  141. Liu H, Yang Y, Ge Y, Liu J, Zhao Y. TERC promotes cellular inflammatory response independent of telomerase. Nucleic Acids Res. 2019;47:8084–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Vulliamy T, Marrone A, Goldman F, Dearlove A, Bessler M, Mason PJ, Dokal I. The RNA component of telomerase is mutated in autosomal dominant dyskeratosis congenita. Nature. 2001;413:432–5.

    Article  CAS  PubMed  Google Scholar 

  143. Vulliamy T, Marrone A, Dokal I, Mason PJ. Association between aplastic anaemia and mutations in telomerase RNA. Lancet. 2002;359:2168–70.

    Article  CAS  PubMed  Google Scholar 

  144. Armanios MY, Chen JJ-L, Cogan JD, Alder JK, Ingersoll RG, Markin C, Lawson WE, Xie M, Vulto I, Phillips IIIJA. Telomerase mutations in families with idiopathic pulmonary fibrosis. N Engl J Med. 2007;356:1317–26.

    Article  CAS  PubMed  Google Scholar 

  145. Wong LS, Oeseburg H, de Boer RA, van Gilst WH, van Veldhuisen DJ, van der Harst P. Telomere biology in cardiovascular disease: the TERC–/– mouse as a model for heart failure and ageing. Cardiovascular Res. 2009;81:244–52.

    Article  CAS  Google Scholar 

  146. Sarek G, Marzec P, Margalef P, Boulton SJ. Molecular basis of telomere dysfunction in human genetic diseases. Nat Struct Mol Biol. 2015;22:867–74.

    Article  CAS  PubMed  Google Scholar 

  147. García-Castillo J, Alcaraz-Pérez F, Martínez-Balsalobre E, García-Moreno D, Rossmann MP, Fernández-Lajarín M, Bernabé-García M, Pérez-Oliva AB, Rodríguez-Cortez VC, Bueno C, Adatto I, Agarwal S, Menéndez P, Zon LI, Mulero V, Cayuela ML. Telomerase RNA recruits RNA polymerase II to target gene promoters to enhance myelopoiesis. Proc Natl Acad Sci USA. 2021;118:11.

    Article  Google Scholar 

  148. Yamaguchi H, Baerlocher GM, Lansdorp PM, Chanock SJ, Nunez O, Sloand E, Young NS. Mutations of the human telomerase RNA gene (TERC) in aplastic anemia and myelodysplastic syndrome. Blood. 2003;102:916–8.

    Article  CAS  PubMed  Google Scholar 

  149. Calado RT, Brudno J, Mehta P, Kovacs JJ, Wu C, Zago MA, Chanock SJ, Boyer TD, Young NS. Constitutional telomerase mutations are genetic risk factors for cirrhosis. Hepatology. 2011;53:1600–7.

    Article  CAS  PubMed  Google Scholar 

  150. Armanios MY, Chen JJ, Cogan JD, Alder JK, Ingersoll RG, Markin C, Lawson WE, Xie M, Vulto I, Phillips JA 3rd, Lansdorp PM, Greider CW, Loyd JE. Telomerase mutations in families with idiopathic pulmonary fibrosis. N Engl J Med. 2007;356:1317–26.

    Article  CAS  PubMed  Google Scholar 

  151. Savage SA. Dyskeratosis congenita and telomere biology disorders. Hematol Am Soc Hematol Educ Program. 2022;2022:637–48.

    Article  Google Scholar 

  152. Alder JK, Hanumanthu VS, Strong MA, DeZern AE, Stanley SE, Takemoto CM, Danilova L, Applegate CD, Bolton SG, Mohr DW, Brodsky RA, Casella JF, Greider CW, Jackson JB, Armanios M. Diagnostic utility of telomere length testing in a hospital-based setting. Proc Natl Acad Sci U S A. 2018;115:E2358–65.

    Article  PubMed  PubMed Central  Google Scholar 

  153. Chu HP, Cifuentes-Rojas C, Kesner B, Aeby E, Lee HG, Wei C, Oh HJ, Boukhali M, Haas W, Lee JT. TERRA RNA antagonizes ATRX and protects telomeres. Cell. 2017;170:86–e10116.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Sahin E, Colla S, Liesa M, Moslehi J, Muller FL, Guo M, Cooper M, Kotton D, Fabian AJ, Walkey C, Maser RS, Tonon G, Foerster F, Xiong R, Wang YA, Shukla SA, Jaskelioff M, Martin ES, Heffernan TP, Protopopov A, Ivanova E, Mahoney JE, Kost-Alimova M, Perry SR, Bronson R, Liao R, Mulligan R, Shirihai OS, Chin L, DePinho RA. Telomere dysfunction induces metabolic and mitochondrial compromise. Nature. 2011;470:359–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Nagpal N, Agarwal S. Telomerase RNA processing: implications for human health and disease. Stem Cells. 2020. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/stem.3270.

    Article  PubMed  Google Scholar 

  156. Townsley DM, Dumitriu B, Liu D, Biancotto A, Weinstein B, Chen C, Hardy N, Mihalek AD, Lingala S, Kim YJ, Yao J, Jones E, Gochuico BR, Heller T, Wu CO, Calado RT, Scheinberg P, Young NS. Danazol Treatment for Telomere diseases. N Engl J Med. 2016;374:1922–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  157. Sun H, Li X, Long Q, Wang X, Zhu W, Chen E, Zhou W, Yang H, Huang C, Deng W. 2024. TERC promotes non-small cell lung cancer progression by facilitating the nuclear localization of TERT. Iscience 27.

  158. Wu S, Ge Y, Lin K, Liu Q, Zhou H, Hu Q, Zhao Y, He W, Ju Z. Telomerase RNA TERC and the PI3K-AKT pathway form a positive feedback loop to regulate cell proliferation independent of telomerase activity. Nucleic Acids Res. 2022;50:3764–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  159. Cheng Y, Liu P, Zheng Q, Gao G, Yuan J, Wang P, Huang J, Xie L, Lu X, Tong T. Mitochondrial trafficking and processing of telomerase RNA TERC. Cell Rep. 2018;24:2589–95.

    Article  CAS  PubMed  Google Scholar 

  160. zur Hausen H. Papillomaviruses and cancer: from basic studies to clinical application. Nat Rev Cancer. 2002;2:342–50.

    Article  CAS  PubMed  Google Scholar 

  161. Network CGAR, Medicine AECo, Services AB, Hospital BC, Medicine BCo H, BRIoCo A, BIfRo, Centre CMSGS, School HM, Services HFGCCRICCH, Biotechnology HIf, ILSbio LLC, Medicine IUSo, Virology IH, Biology IS, Consortium IG, Biomedical L, Hospital MG, University MGIW, MCo W, Center CMUS, Center MSKC, University MM et al. OHS, Samuel Oschin Comprehensive Cancer Institute Cd-SMC, International S, System SJsCH, University EELBIoMIoTH, Hospital RIaNCs, University SKCCCaJH, Bergen Uo, Center UoTMAC, Hospital UoAT, Birmingham UoAa, University of California Iv, Cruz UoCS, Center UoKM, Lausanne Uo, Center UoNMHS, Hill UoNCaC, Center UoOHS,. 2017. Integrated genomic and molecular characterization of cervical cancer. Nature 543:378–384.

  162. Miller J, Dakic A, Chen R, Palechor-Ceron N, Dai Y, Kallakury B, Schlegel R, Liu X. HPV16 E7 protein and hTERT proteins defective for telomere maintenance cooperate to immortalize human keratinocytes. PLoS Pathog. 2013;9:e1003284.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  163. Liu X, Dakic A, Zhang Y, Dai Y, Chen R, Schlegel R. HPV E6 protein interacts physically and functionally with the cellular telomerase complex. Proc Natl Acad Sci U S A. 2009;106:18780–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  164. Katzenellenbogen R. 2017. Telomerase Induction in HPV Infection and Oncogenesis. Viruses 9.

  165. Katzenellenbogen RA. Activation of telomerase by HPVs. Virus Res. 2017;231:50–5.

    Article  CAS  PubMed  Google Scholar 

  166. Katzenellenbogen RA, Vliet-Gregg P, Xu M, Galloway DA. Cytoplasmic poly(A) binding proteins regulate telomerase activity and cell growth in human papillomavirus type 16 E6-expressing keratinocytes. J Virol. 2010;84:12934–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  167. Vliet-Gregg PA, Hamilton JR, Katzenellenbogen RA. NFX1-123 and human papillomavirus 16E6 increase notch expression in keratinocytes. J Virol. 2013;87:13741–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  168. Vliet-Gregg PA, Hamilton JR, Katzenellenbogen RA. Human papillomavirus 16E6 and NFX1-123 potentiate notch signaling and differentiation without activating cellular arrest. Virology. 2015;478:50–60.

    Article  CAS  PubMed  Google Scholar 

  169. Zhang Y, Dakic A, Chen R, Dai Y, Schlegel R, Liu X. Direct HPV E6/Myc interactions induce histone modifications, Pol II phosphorylation, and hTERT promoter activation. Oncotarget. 2017;8:96323.

    Article  PubMed  PubMed Central  Google Scholar 

  170. Liu X, Roberts J, Dakic A, Zhang Y, Schlegel R. HPV E7 contributes to the telomerase activity of immortalized and tumorigenic cells and augments E6-induced hTERT promoter function. Virology. 2008;375:611–23.

    Article  CAS  PubMed  Google Scholar 

  171. Veldman T, Liu X, Yuan H, Schlegel R. Human papillomavirus E6 and Myc proteins associate in vivo and bind to and cooperatively activate the telomerase reverse transcriptase promoter. Proc Natl Acad Sci U S A. 2003;100:8211–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  172. Pańczyszyn A, Boniewska-Bernacka E, Głąb G. Telomeres and telomerase during Human Papillomavirus-Induced Carcinogenesis. Mol Diagn Ther. 2018;22:421–30.

    Article  PubMed  PubMed Central  Google Scholar 

  173. Miller J, Dakic A, Spurgeon M, Saenz F, Kallakury B, Zhao B, Zhang J, Zhu J, Ma Q, Xu Y, Lambert P, Schlegel R, Riegel AT, Liu X. AIB1 is a novel target of the high-risk HPV E6 protein and a biomarker of cervical cancer progression. J Med Virol. 2022;94:3962–77.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  174. Heselmeyer K, Schröck E, du Manoir S, Blegen H, Shah K, Steinbeck R, Auer G, Ried T. Gain of chromosome 3q defines the transition from severe dysplasia to invasive carcinoma of the uterine cervix. Proc Natl Acad Sci U S A. 1996;93:479–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  175. Heselmeyer-Haddad K, Janz V, Castle PE, Chaudhri N, White N, Wilber K, Morrison LE, Auer G, Burroughs FH, Sherman ME, Ried T. Detection of genomic amplification of the human telomerase gene (TERC) in cytologic specimens as a genetic test for the diagnosis of cervical dysplasia. Am J Pathol. 2003;163:1405–16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  176. Liu H, Liu S, Wang H, Xie X, Chen X, Zhang X, Zhang Y. Genomic amplification of the human telomerase gene (hTERC) associated with human papillomavirus is related to the progression of uterine cervical dysplasia to invasive cancer. Diagn Pathol. 2012;7:147.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  177. Jiang J, Wei LH, Li YL, Wu RF, Xie X, Feng YJ, Zhang G, Zhao C, Zhao Y, Chen Z. Detection of TERC amplification in cervical epithelial cells for the diagnosis of high-grade cervical lesions and invasive cancer: a multicenter study in China. J Mol Diagn. 2010;12:808–17.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  178. Wang A, Zhou D, Krawczyk E, Li T, Simic V, Lu J, Liu X, Schlegel R, Yuan H. Overexpression of the telomerase holoenzyme induces EMT and tumorigenesis of HPV-immortalized keratinocytes. J Med Virol. 2023;95:e28681.

    Article  CAS  PubMed  Google Scholar 

  179. Jin H, Chen Y, Ren J, Huang J, Zhao Y, Liu H. TERC suppresses PD-L1 expression by downregulating RNA binding protein HuR. Sci China Life Sci. 2022;65:2505–16.

    Article  CAS  PubMed  Google Scholar 

  180. Kheimar A, Trimpert J, Groenke N, Kaufer BB. Overexpression of cellular telomerase RNA enhances virus-induced cancer formation. Oncogene. 2019;38:1778–86.

    Article  CAS  PubMed  Google Scholar 

  181. González-Suárez E, Samper E, Flores JM, Blasco MA. Telomerase-deficient mice with short telomeres are resistant to skin tumorigenesis. Nat Genet. 2000;26:114–7.

    Article  PubMed  Google Scholar 

  182. Trapp S, Parcells MS, Kamil JP, Schumacher D, Tischer BK, Kumar PM, Nair VK, Osterrieder N. A virus-encoded telomerase RNA promotes malignant T cell lymphomagenesis. J Exp Med. 2006;203:1307–17.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  183. Kheimar A, Trapp-Fragnet L, Conradie AM, Bertzbach LD, You Y, Sabsabi MA, Kaufer BB. Viral and cellular telomerase RNAs possess host-specific anti-apoptotic functions. Microbiol Spectr. 2023;11:e0188723.

    Article  PubMed  Google Scholar 

  184. Hopman AH, Theelen W, Hommelberg PP, Kamps MA, Herrington CS, Morrison LE, Speel EJ, Smedts F, Ramaekers FC. Genomic integration of oncogenic HPV and gain of the human telomerase gene TERC at 3q26 are strongly associated events in the progression of uterine cervical dysplasia to invasive cancer. J Pathol. 2006;210:412–9.

    Article  CAS  PubMed  Google Scholar 

  185. Kuglik P, Smetana J, Vallova V, Moukova L, Kasikova K, Cvanova M, Brozova L. Genome-wide screening of DNA copy number alterations in cervical carcinoma patients with CGH plus SNP microarrays and HPV-FISH. Int J Clin Exp Pathol. 2014;7:5071–82.

    CAS  PubMed  PubMed Central  Google Scholar 

  186. Zhao H, He Y, Fan B, Wang Y, Wu YM. Human papillomavirus E6E7 mRNA and TERC lncRNA in situ detection in cervical scraped cells and cervical disease progression assessment. Virol J. 2022;19:18.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  187. Garus A, Autexier C. Dyskerin: an essential pseudouridine synthase with multifaceted roles in ribosome biogenesis, splicing, and telomere maintenance. RNA. 2021;27:1441–58.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  188. Henry BA, Marchand V, Schlegel BT, Helm M, Motorin Y, Lee N. Pseudouridylation of Epstein-Barr virus noncoding RNA EBER2 facilitates lytic replication. RNA. 2022;28:1542–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  189. Shay JW, Wright WE. Telomeres and telomerase: three decades of progress. Nat Rev Genet. 2019;20:299–309.

    Article  CAS  PubMed  Google Scholar 

  190. Tang S, Wu W, Wan H, Wu X, Chen H. Knockdown of NHP2 inhibits hepatitis B virus X protein-induced hepatocarcinogenesis via repressing TERT expression and disrupting the stability of telomerase complex. Aging. 2020;12:19365–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  191. Tycowski KT, Shu MD, Kukoyi A, Steitz JA. A conserved WD40 protein binds the cajal body localization signal of scaRNP particles. Mol Cell. 2009;34:47–57.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  192. Wang K, Ge YC, Ni C, Cui BM, Du JT, Zhang B, Hu XY, Chen J, Xiao LY, Sun CK, Li Y. Epstein-Barr virus-induced up-regulation of TCAB1 is involved in the DNA damage response in nasopharyngeal carcinoma. Sci Rep. 2017;7:10.

    Google Scholar 

  193. Nandakumar J, Cech TR. Finding the end: recruitment of telomerase to telomeres. Nat Rev Mol Cell Biol. 2013;14:69–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  194. Deng Z, Kim ET, Vladimirova O, Dheekollu J, Wang Z, Newhart A, Liu D, Myers JL, Hensley SE, Moffat J, Janicki SM, Fraser NW, Knipe DM, Weitzman MD, Lieberman PM. HSV-1 remodels host telomeres to facilitate viral replication. Cell Rep. 2014;9:2263–78.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  195. Hong J, Lee JH, Chung IK. Telomerase activates transcription of cyclin D1 gene through an interaction with NOL1. J Cell Sci. 2016;129:1566–79.

    Article  CAS  PubMed  Google Scholar 

  196. Kong W, Biswas A, Zhou D, Fiches G, Fujinaga K, Santoso N, Zhu J. Nucleolar protein NOP2/NSUN1 suppresses HIV-1 transcription and promotes viral latency by competing with Tat for TAR binding and methylation. PLoS Pathog. 2020;16:e1008430.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  197. Ding S, Liu H, Liu L, Ma L, Chen Z, Zhu M, Zhang X, Hao H, Zuo L, Yang J, Wu X, Zhou P, Huang F, Zhu F, Guan W. Epigenetic addition of m. Cell Death Dis. 2024;15:39.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  198. de Lange T. Shelterin-mediated Telomere Protection. Annu Rev Genet. 2018;52:223–47.

    Article  PubMed  Google Scholar 

  199. Deng Z, Atanasiu C, Burg JS, Broccoli D, Lieberman PM. Telomere repeat binding factors TRF1, TRF2, and hRAP1 modulate replication of Epstein-Barr virus OriP. J Virol. 2003;77:11992–2001.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  200. Kamranvar SA, Masucci MG. The Epstein-Barr virus nuclear antigen-1 promotes telomere dysfunction via induction of oxidative stress. Leukemia. 2011;25:1017–25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  201. Lajoie V, Lemieux B, Sawan B, Lichtensztejn D, Lichtensztejn Z, Wellinger R, Mai S, Knecht H. LMP1 mediates multinuclearity through downregulation of shelterin proteins and formation of telomeric aggregates. Blood. 2015;125:2101–10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  202. Nguyen LN, Zhao J, Cao D, Dang X, Wang L, Lian J, Zhang Y, Jia Z, Wu XY, Morrison Z, Xie Q, Ji Y, Zhang Z, El Gazzar M, Ning S, Moorman JP, Yao ZQ. Inhibition of TRF2 accelerates telomere attrition and DNA damage in naïve CD4 T cells during HCV infection. Cell Death Dis. 2018;9:900.

    Article  PubMed  PubMed Central  Google Scholar 

  203. Gilbert-Girard S, Gravel A, Collin V, Wight DJ, Kaufer BB, Lazzerini-Denchi E, Flamand L. Role for the shelterin protein TRF2 in human herpesvirus 6A/B chromosomal integration. PLoS Pathog. 2020;16:e1008496.

    Article  PubMed  PubMed Central  Google Scholar 

  204. Victor J, Deutsch J, Whitaker A, Lamkin EN, March A, Zhou P, Botten JW, Chatterjee N. SARS-CoV-2 triggers DNA damage response in Vero E6 cells. Biochem Biophys Res Commun. 2021;579:141–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  205. Yin L, Hubbard AK, Giardina C. NF-kappa B regulates transcription of the mouse telomerase catalytic subunit. J Biol Chem. 2000;275:36671–5.

    Article  CAS  PubMed  Google Scholar 

  206. Zhang Y, Toh L, Lau P, Wang X. Human telomerase reverse transcriptase (hTERT) is a novel target of the Wnt/beta-catenin pathway in human cancer. J Biol Chem. 2012;287:32494–511.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  207. Ghosh A, Saginc G, Leow SC, Khattar E, Shin EM, Yan TD, Wong M, Zhang Z, Li G, Sung WK, Zhou J, Chng WJ, Li S, Liu E, Tergaonkar V. Telomerase directly regulates NF-kappaB-dependent transcription. Nat Cell Biol. 2012;14:1270–81.

    Article  CAS  PubMed  Google Scholar 

  208. Liu Y, Betori RC, Pagacz J, Frost GB, Efimova EV, Wu D, Wolfgeher DJ, Bryan TM, Cohen SB, Scheidt KA, Kron SJ. Targeting telomerase reverse transcriptase with the covalent inhibitor NU-1 confers immunogenic radiation sensitization. Cell Chem Biology. 2022;29:1517–.

    Article  CAS  Google Scholar 

  209. Clevers H, Nusse R. Wnt/beta-catenin signaling and disease. Cell. 2012;149:1192–205.

    Article  CAS  PubMed  Google Scholar 

  210. Ding D, Xi P, Zhou J, Wang M, Cong YS. Human telomerase reverse transcriptase regulates MMP expression independently of telomerase activity via NF-kappaB-dependent transcription. FASEB J. 2013;27:4375–83.

    Article  CAS  PubMed  Google Scholar 

  211. Low KC, Tergaonkar V. Telomerase: central regulator of all of the hallmarks of cancer. Trends Biochem Sci. 2013;38:426–34.

    Article  CAS  PubMed  Google Scholar 

  212. Wu KJ, Grandori C, Amacker M, Simon-Vermot N, Polack A, Lingner J, Dalla-Favera R. Direct activation of TERT transcription by c-MYC. Nat Genet. 1999;21:220–4.

    Article  CAS  PubMed  Google Scholar 

  213. Choi J, Southworth LK, Sarin KY, Venteicher AS, Ma W, Chang W, Cheung P, Jun S, Artandi MK, Shah N, Kim SK, Artandi SE. TERT promotes epithelial proliferation through transcriptional control of a myc- and wnt-related developmental program. PLoS Genet. 2008;4:e10.

    Article  PubMed  PubMed Central  Google Scholar 

  214. Balasubramanian S, Hurley LH, Neidle S. Targeting G-quadruplexes in gene promoters: a novel anticancer strategy? Nat Rev Drug Discov. 2011;10:261–75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  215. Berardinelli F, Tanori M, Muoio D, Buccarelli M, di Masi A, Leone S, Ricci-Vitiani L, Pallini R, Mancuso M, Antoccia A. G-quadruplex ligand RHPS4 radiosensitizes glioblastoma xenograft in vivo through a differential targeting of bulky differentiated- and stem-cancer cells. J Exp Clin Cancer Res. 2019;38:311.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  216. Tsai RX, Fang KC, Yang PC, Hsieh YH, Chiang IT, Chen Y, Lee HG, Lee JT, Chu HC. TERRA regulates DNA G-quadruplex formation and ATRX recruitment to chromatin. Nucleic Acids Res. 2022;50:12217–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  217. Shankar U, Mishra SK, Jain N, Tawani A, Yadav P, Kumar A. Ni(+ 2) permease system of Helicobacter pylori contains highly conserved G-quadruplex motifs. Infect Genet Evol. 2022;101:105298.

    Article  CAS  PubMed  Google Scholar 

  218. Konieczna N, Romaniuk-Drapala A, Lisiak N, Toton E, Paszel-Jaworska A, Kaczmarek M, Rubis B. 2019. Telomerase inhibitor TMPyP4 alters Adhesion and Migration of breast-Cancer cells MCF7 and MDA-MB-231. Int J Mol Sci 20.

  219. Cheng MJ, Cao YG. TMPYP4 exerted antitumor effects in human cervical cancer cells through activation of p38 mitogen-activated protein kinase. Biol Res. 2017;50:24.

    Article  PubMed  PubMed Central  Google Scholar 

  220. Mender I, Gryaznov S, Dikmen ZG, Wright WE, Shay JW. Induction of telomere dysfunction mediated by the telomerase substrate precursor 6-thio-2’-deoxyguanosine. Cancer Discov. 2015;5:82–95.

    Article  CAS  PubMed  Google Scholar 

  221. Pineiro-Hermida S, Bosso G, Sanchez-Vazquez R, Martinez P, Blasco MA. Telomerase deficiency and dysfunctional telomeres in the lung tumor microenvironment impair tumor progression in NSCLC mouse models and patient-derived xenografts. Cell Death Differ. 2023;30:1585–600.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  222. Zhang G, Wu LW, Mender I, Barzily-Rokni M, Hammond MR, Ope O, Cheng C, Vasilopoulos T, Randell S, Sadek N, Beroard A, Xiao M, Tian T, Tan J, Saeed U, Sugarman E, Krepler C, Brafford P, Sproesser K, Murugan S, Somasundaram R, Garman B, Wubbenhorst B, Woo J, Yin X, Liu Q, Frederick DT, Miao B, Xu W, Karakousis GC, Xu X, Schuchter LM, Mitchell TC, Kwong LN, Amaravadi RK, Lu Y, Boland GM, Wei Z, Nathanson K, Herbig U, Mills GB, Flaherty KT, Herlyn M, Shay JW. Induction of Telomere Dysfunction prolongs Disease Control of Therapy-Resistant Melanoma. Clin Cancer Res. 2018;24:4771–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  223. Reyes-Uribe P, Adrianzen-Ruesta MP, Deng Z, Echevarria-Vargas I, Mender I, Saheb S, Liu Q, Altieri DC, Murphy ME, Shay JW, Lieberman PM, Villanueva J. Exploiting TERT dependency as a therapeutic strategy for NRAS-mutant melanoma. Oncogene. 2018;37:4058–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  224. Yu S, Wei S, Savani M, Lin X, Du K, Mender I, Siteni S, Vasilopoulos T, Reitman ZJ, Ku Y, Wu D, Liu H, Tian M, Chen Y, Labrie M, Charbonneau CM, Sugarman E, Bowie M, Hariharan S, Waitkus M, Jiang W, McLendon RE, Pan E, Khasraw M, Walsh KM, Lu Y, Herlyn M, Mills G, Herbig U, Wei Z, Keir ST, Flaherty K, Liu L, Wu K, Shay JW, Abdullah K, Zhang G, Ashley DM. A modified nucleoside 6-Thio-2’-Deoxyguanosine exhibits Antitumor Activity in Gliomas. Clin Cancer Res. 2021;27:6800–14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  225. Chen Z, Vallega KA, Wang DS, Quan ZH, Fan SQ, Wang QM, Leal T, Ramalingam SS, Sun SY. Inhibition of hTERT/telomerase/telomere mediates therapeutic efficacy of osimertinib in EGFR mutant lung cancer. J Exp Med. 2024;221:23.

    Article  Google Scholar 

  226. Sengupta S, Sobo M, Lee K, Senthil Kumar S, White AR, Mender I, Fuller C, Chow LML, Fouladi M, Shay JW, Drissi R. Induced Telomere damage to treat telomerase expressing therapy-resistant Pediatric Brain tumors. Mol Cancer Ther. 2018;17:1504–14.

    Article  CAS  PubMed  Google Scholar 

  227. Fischer-Mertens J, Otte F, Roderwieser A, Rosswog C, Kahlert Y, Werr L, Hellmann AM, Berding M, Chiu B, Bartenhagen C, Fischer M. Telomerase-targeting compounds Imetelstat and 6-thio-dG act synergistically with chemotherapy in high-risk neuroblastoma models. Cell Oncol (Dordr). 2022;45:991–1003.

    Article  CAS  PubMed  Google Scholar 

  228. Mender I, Siteni S, Barron S, Flusche AM, Kubota N, Yu C, Cornelius C, Tedone E, Maziveyi M, Grichuk A, Venkateswaran N, Conacci-Sorrell M, Hoshida Y, Kang R, Tang D, Gryaznov S, Shay JW. Activating an adaptive Immune response with a telomerase-mediated Telomere Targeting Therapeutic in Hepatocellular Carcinoma. Mol Cancer Ther. 2023;22:737–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  229. Mender I, Zhang A, Ren Z, Han C, Deng Y, Siteni S, Li H, Zhu J, Vemula A, Shay JW, Fu YX. Telomere stress potentiates STING-Dependent anti-tumor immunity. Cancer Cell. 2020;38:400–11. e6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  230. Sullivan HJ, Readmond C, Radicella C, Persad V, Fasano TJ, Wu C. Binding of Telomestatin, TMPyP4, BSU6037, and BRACO19 to a telomeric G-Quadruplex-duplex hybrid probed by all-Atom Molecular Dynamics Simulations with Explicit Solvent. ACS Omega. 2018;3:14788–806.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  231. Mulholland K, Wu C. Binding of Telomestatin to a telomeric G-Quadruplex DNA probed by all-Atom Molecular Dynamics Simulations with Explicit Solvent. J Chem Inf Model. 2016;56:2093–102.

    Article  CAS  PubMed  Google Scholar 

  232. Al-Karmalawy AA, Nafie MS, Shaldam MA, Elmaaty AA, Antar SA, El-Hamaky AA, Saleh MA, Elkamhawy A, Tawfik HO. Ligand-based design on the dog-bone-shaped BIBR1532 pharmacophoric features and synthesis of Novel analogues as Promising Telomerase inhibitors with in Vitro and in vivo evaluations. J Med Chem. 2023;66:777–92.

    Article  CAS  PubMed  Google Scholar 

  233. Barwe SP, Huang F, Kolb EA, Gopalakrishnapillai A. 2022. Imetelstat induces leukemia stem cell death in Pediatric Acute myeloid leukemia patient-derived xenografts. J Clin Med 11.

  234. Bruedigam C, Porter AH, Song A, Vroeg In de Wei G, Stoll T, Straube J, Cooper L, Cheng G, Kahl VFS, Sobinoff AP, Ling VY, Jebaraj BMC, Janardhanan Y, Haldar R, Bray LJ, Bullinger L, Heidel FH, Kennedy GA, Hill MM, Pickett HA, Abdel-Wahab O, Hartel G, Lane SW. Imetelstat-mediated alterations in fatty acid metabolism to induce ferroptosis as a therapeutic strategy for acute myeloid leukemia. Nat Cancer. 2024;5:47–65.

    Article  CAS  PubMed  Google Scholar 

  235. Steensma DP, Fenaux P, Van Eygen K, Raza A, Santini V, Germing U, Font P, Diez-Campelo M, Thepot S, Vellenga E, Patnaik MM, Jang JH, Varsos H, Bussolari J, Rose E, Sherman L, Sun L, Wan Y, Dougherty S, Huang F, Feller F, Rizo A, Platzbecker U. Imetelstat achieves meaningful and durable transfusion independence in high transfusion-burden patients with Lower-Risk Myelodysplastic syndromes in a phase II study. J Clin Oncol. 2021;39:48–56.

    Article  CAS  PubMed  Google Scholar 

  236. Dratwa M, Wysoczanska B, Lacina P, Kubik T, Bogunia-Kubik K. TERT-Regulation and roles in Cancer formation. Front Immunol. 2020;11:589929.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  237. Liu B, He Y, Wang Y, Song H, Zhou ZH, Feigon J. Structure of active human telomerase with telomere shelterin protein TPP1. Nature. 2022;604:578–83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  238. Amin A, Morello M, Petrara MR, Rizzo B, Argenton F, De Rossi A, Giunco S. 2023. Short-term TERT inhibition impairs Cellular Proliferation via a Telomere length-independent mechanism and can be exploited as a potential Anticancer Approach. Cancers (Basel) 15.

  239. Kailashiya C, Sharma HB, Kailashiya J. Telomerase based anticancer immunotherapy and vaccines approaches. Vaccine. 2017;35:5768–75.

    Article  CAS  PubMed  Google Scholar 

  240. Jo JH, Kim YT, Choi HS, Kim HG, Lee HS, Choi YW, Kim DU, Lee KH, Kim EJ, Han JH, Lee SO, Park CH, Choi EK, Kim JW, Cho JY, Lee WJ, Moon HR, Park MS, Kim S, Song SY. Efficacy of GV1001 with gemcitabine/capecitabine in previously untreated patients with advanced pancreatic ductal adenocarcinoma having high serum eotaxin levels (KG4/2015): an open-label, randomised, phase 3 trial. Br J Cancer. 2024;130:43–52.

    Article  CAS  PubMed  Google Scholar 

  241. Ellingsen EB, O’Day S, Mezheyeuski A, Gromadka A, Clancy T, Kristedja TS, Milhem M, Zakharia Y. Clinical activity of combined telomerase vaccination and Pembrolizumab in Advanced Melanoma: results from a phase I Trial. Clin Cancer Res. 2023;29:3026–36.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  242. Brunsvig PF, Aamdal S, Gjertsen MK, Kvalheim G, Markowski-Grimsrud CJ, Sve I, Dyrhaug M, Trachsel S, Moller M, Eriksen JA, Gaudernack G. Telomerase peptide vaccination: a phase I/II study in patients with non-small cell lung cancer. Cancer Immunol Immunother. 2006;55:1553–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  243. Ellingsen EB, Bounova G, Kerzeli I, Anzar I, Simnica D, Aamdal E, Guren T, Clancy T, Mezheyeuski A, Inderberg EM, Mangsbo SM, Binder M, Hovig E, Gaudernack G. Characterization of the T cell receptor repertoire and melanoma tumor microenvironment upon combined treatment with ipilimumab and hTERT vaccination. J Transl Med. 2022;20:419.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  244. Gridelli C, Ciuleanu T, Domine M, Szczesna A, Bover I, Cobo M, Kentepozidis N, Zarogoulidis K, Kalofonos C, Kazarnowisz A, Korozan M, de Las Penas R, Majem M, Chella A, Griesinger F, Bournakis E, Sadjadian P, Kotsakis A, Chinet T, Syrigos KN, Correale P, Gallou C, Jamet JM, Vetsika EK, Kosmatopoulos K, Georgoulias V. Vx-001-201 trial t. 2020. Clinical activity of a htert (vx-001) cancer vaccine as post-chemotherapy maintenance immunotherapy in patients with stage IV non-small cell lung cancer: final results of a randomised phase 2 clinical trial. Br J Cancer 122:1461–6.

  245. Filaci G, Fenoglio D, Nole F, Zanardi E, Tomasello L, Aglietta M, Del Conte G, Carles J, Morales-Barrera R, Guglielmini P, Scagliotti G, Signori A, Parodi A, Kalli F, Astone G, Ferrera F, Altosole T, Lamperti G, Criscuolo D, Gianese F, Boccardo F. Telomerase-based GX301 cancer vaccine in patients with metastatic castration-resistant prostate cancer: a randomized phase II trial. Cancer Immunol Immunother. 2021;70:3679–92.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  246. Dosset M, Godet Y, Vauchy C, Beziaud L, Lone YC, Sedlik C, Liard C, Levionnois E, Clerc B, Sandoval F, Daguindau E, Wain-Hobson S, Tartour E, Langlade-Demoyen P, Borg C, Adotevi O. Universal cancer peptide-based therapeutic vaccine breaks tolerance against telomerase and eradicates established tumor. Clin Cancer Res. 2012;18:6284–95.

    Article  CAS  PubMed  Google Scholar 

  247. Jansons J, Skrastina D, Kurlanda A, Petkov S, Avdoshina D, Kuzmenko Y, Krotova O, Trofimova O, Gordeychuk I, Sominskaya I, Isaguliants M. 2021. Reciprocal inhibition of immunogenic performance in mice of two potent DNA immunogens targeting HCV-Related Liver Cancer. Microorganisms 9.

  248. Negrini S, De Palma R, Filaci G. Anti-cancer immunotherapies targeting telomerase. Cancers. 2020;12:22.

    Article  Google Scholar 

  249. Akincilar SC, Chan CHT, Ng QF, Fidan K, Tergaonkar V. Non-canonical roles of canonical telomere binding proteins in cancers. Cell Mol Life Sci. 2021;78:4235–57.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  250. Shim HS, Iaconelli J, Shang XY, Li JX, Lan ZD, Jiang S, Nutsch K, Beyer BA, Lairson LL, Boutin AT, Bollong MJ, Schultz PG, Depinho RA. TERT activation targets DNA methylation and multiple aging hallmarks. Cell. 2024;187:27.

    Article  Google Scholar 

  251. Li Y, Tergaonkar V. Noncanonical functions of telomerase: implications in telomerase-targeted cancer therapies. Cancer Res. 2014;74:1639–44.

    Article  CAS  PubMed  Google Scholar 

  252. Romaniuk A, Paszel-Jaworska A, Totoń E, Lisiak N, Hołysz H, Królak A, Grodecka-Gazdecka S, Rubiś B. The non-canonical functions of telomerase: to turn off or not to turn off. Mol Biol Rep. 2019;46:1401–11.

    Article  CAS  PubMed  Google Scholar 

  253. Jafri MA, Ansari SA, Alqahtani MH, Shay JW. Roles of telomeres and telomerase in cancer, and advances in telomerase-targeted therapies. Genome Med. 2016;8:18.

    Article  Google Scholar 

  254. Chen X, Zhong S, Zhan Y, Zhang X. CRISPR–Cas9 applications in T cells and adoptive T cell therapies. Cell Mol Biol Lett. 2024;29:52.

    Article  PubMed  PubMed Central  Google Scholar 

  255. Khuu A, Verreault M, Colin P, Tran H, Idbaih A. Clinical applications of antisense oligonucleotides in Cancer: a focus on Glioblastoma. Cells. 2024;13:1869.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  256. Steinle H, Weber J, Stoppelkamp S, Grosse-Berkenbusch K, Golombek S, Weber M, Canak-Ipek T, Trenz S-M, Schlensak C, Avci-Adali M. Delivery of synthetic mRNAs for tissue regeneration. Adv Drug Deliv Rev. 2021;179:114007.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We acknowledge The Ohio State University Comprehensive Cancer Center (OSUCCC) for the start-up funds.

Funding

This work was supported by The Ohio State University Comprehensive Cancer Center and the National Institutes of Health under grant numbers P30CA016058, R01CA222148, R33CA258016, R01CA276474, U01CA278927.

Author information

Authors and Affiliations

Authors

Contributions

Chongwen Cao, Weiyi Gong, Yuanlong Shuai, Sara Rosauli, Qianyun Ge, Anam Khan, Danyal Daneshdoust, Rani Mahyoob: literature search, writing. Chongwen Cao: Figures. Rani Mahyoob: organizing and coordinate; Danyal Daneshdoust: finalizing, interpretation and editing. Yun-ling Zheng, Nagireddy Putluri, Gennady Shvets, Jenny Li: interpretation and editing, finalizing. Xuefeng Liu: conceptual design, summary, writing and editing, and final review.

Corresponding author

Correspondence to Xuefeng Liu.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors have nothing to report.

Additional information

Publisher’s note

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

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Cao, C., Gong, W., Shuai, Y. et al. Canonical and non-canonical functions of the non-coding RNA component (TERC) of telomerase complex. Cell Biosci 15, 30 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13578-025-01367-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13578-025-01367-0

Keywords