Review
Telomerase: Key regulator of inflammation and cancer

https://doi.org/10.1016/j.phrs.2020.104726Get rights and content

Abstract

The telomerase holoenzyme, which has a highly conserved role in maintaining telomere length, has long been regarded as a high-profile target in cancer therapy due to the high dependency of the majority of cancer cells on constitutive and elevated telomerase activity for sustained proliferation and immortality. In this review, we present the salient findings in the telomerase field with special focus on the association of telomerase with inflammation and cancer. The elucidation of extra-telomeric roles of telomerase in inflammation, reactive oxygen species (ROS) generation, and cancer development further complicated the design of anti-telomerase therapy. Of note, the discovery of the unique mechanism that underlies reactivation of the dormant telomerase reverse transcriptase TERT promoter in somatic cells not only enhanced our understanding of the critical role of TERT in carcinogenesis but also opens up new intervention ideas that enable the differential targeting of cancer cells only. Despite significant effort invested in developing telomerase-targeted therapeutics, devising efficacious cancer-specific telomerase/TERT inhibitors remains an uphill task. The latest discoveries of the telomere-independent functionalities of telomerase in inflammation and cancer can help illuminate the path of developing specific anti-telomerase/TERT therapeutics against cancer cells.

Introduction

Telomeres are special regions located at the ends of chromosomes that protect chromosomal ends from DNA damage or fusion [[1], [2], [3], [4], [5]]. The telomerase holoenzyme complex maintains telomere length through the addition of G-rich repetitive sequences such as TTAGGG in vertebrates [[6], [7], [8], [9]]. In most human somatic cells, telomere length will shorten with every cell division [10,11]. When telomeres reach a critical length, replicative senescence or cell death will be triggered [[12], [13], [14]]. The physiological shortening of telomeres is a hallmark of cellular aging. Telomere shortening can be accelerated by several stimuli including oxidative stress and inflammation [[15], [16], [17], [18], [19], [20]]. Although the association between mutations in telomerase-associated genes and inflammation dysregulation is known, the mechanics behind the crosstalk between telomere/telomerase and inflammation is still unclear. In this review, we present the insights gathered over the years on the role of telomerase in inflammation and tumorigenesis.

Section snippets

Overview of telomerase

Telomerase is a ribonucleoprotein complex that consists of several components including TERT, telomerase RNA component (TERC) that serves as a template for adding telomere repeats [[21], [22], [23], [24]], the protein dyskerin, and other associated proteins including the small nucleolar RNA-associated protein (NOP1), nucleolar protein family A member 1 (GAR1), nucleolar protein family A member 2 (NHP2) and NHP3 (NOP1) [[25], [26], [27], [28]] (Fig. 1). The nucleolar protein dyskerin, encoded by

Telomerase and inflammation

Inflammation is the body’s intrinsic biological defense response to different stimuli such as pathogens and it contributes to the clearance and elimination of harmful stimuli. In general, inflammation can be divided into two groups: acute inflammation and chronic inflammation [[66], [67], [68], [69], [70], [71], [72], [73], [74], [75]]. Acute inflammation can be induced by tissue injury or microbial invasion and lasts for a relatively short duration. This response increases blood flow and

Telomerase and oxidative stress

Redox signaling plays a critical role in the regulation of physiological processes. Oxidative stress, which results from the dysregulation or disruption of redox signaling, is a hallmark of many pathologies, such as Alzheimer’s disease, Parkinson's disease and neurodegenerative diseases [[114], [115], [116], [117], [118], [119]]. The hypoxic environment of cancer cells promotes the initiation of redox signaling that can modulate their metabolism, proliferation, as well as immune response [[120]

TERT and cancer

The telomerase enzyme, which is active in proliferative cells, stem cells, germ cells but inactive in most human somatic cells, was found to be reactivated in approximately 90 % of cancer cells [169]. Known mechanisms of TERT reactivation include increased TERT copy number [170], TERT gene amplification [171], genomic rearrangements [172,173], long-range interactions [174] and several point mutations at the promoter region of the TERT gene [175] that can increase TERT mRNA expression and TERT

Telomerase-targeted cancer therapies

Telomerase has been an emerging target in cancer therapy as it is essential for the progression of different cancer types as well as cancer stem cells. The low telomerase activity in normal cells but not in cancer cells forms the basis of anti-telomerase cancer therapy. In this section, we will discuss the current telomerase-targeted cancer approaches (Fig. 6).

Small molecule telomerase inhibitors such as the non-nucleoside reverse transcriptase inhibitor BIBR1532 was found to inhibit telomerase

Discussion

Although it has only been approximately 35 years after the discovery of telomerase function in telomere elongation, our understanding of telomerase function has increased tremendously within this short span of time. As presented in this review, TERT is implicated in non-canonical roles including modulation of inflammation; its dual role in oxidative stress and subsequent downstream effects such as mitochondrial DNA damage and ROS generation; the contribution of exogenous and endogenous factors

Innovation

Emerging non-canonical roles of telomerase, which include fine-tuning inflammation, reactive oxygen species generation, and cancer development, can potentially broaden the range of telomerase-targeted strategies and circumvent the limitations of conventional therapeutic approaches. Examples of novel therapeutic strategies that have been developed include combination therapy comprising of telomerase inhibitors and antioxidants, as well as selective inhibition of TERT transcription in cancer

Declaration of Competing Interest

The authors declare no conflict of interest.

Acknowledgments

This work was supported by a National Research Foundation of Korea (NRF) grant funded by the Korean government (MSIP) (NRF-2018R1D1A1B07042969). We thank the Agency for Science Technology and Research (A*STAR) and Singapore National Research Foundation (NRF-CRP17-2017-02) for funding and support to the V.T. laboratory.

References (220)

  • S.C. Akincilar et al.

    Quantitative assessment of telomerase components in cancer cell lines

    FEBS Lett.

    (2015)
  • M.Z. Levy et al.

    Telomere end-replication problem and cell aging

    J. Mol. Biol.

    (1992)
  • J.Z. Ye et al.

    TIN2 binds TRF1 and TRF2 simultaneously and stabilizes the TRF2 complex on telomeres

    J. Biol. Chem.

    (2004)
  • Y. Doksani et al.

    Super-resolution fluorescence imaging of telomeres reveals TRF2-dependent T-loop formation

    Cell

    (2013)
  • J.D. Griffith et al.

    Mammalian telomeres end in a large duplex loop

    Cell

    (1999)
  • L. Hayflick et al.

    The serial cultivation of human diploid cell strains

    Exp. Cell Res.

    (1961)
  • L. Hayflick

    The limited in vitro lifetime of human diploid cell strains

    Exp. Cell Res.

    (1965)
  • N. Serakinci et al.

    Telomere stability and telomerase in mesenchymal stem cells

    Biochimie

    (2008)
  • S.R. Sousa et al.

    Heterozygous TERT gene mutation associated with familial idiopathic pulmonary fibrosis

    Respir. Med. Case Rep.

    (2019)
  • H.K. Keerthy et al.

    Novel synthetic biscoumarins target tumor necrosis factor-α in hepatocellular carcinoma in vitro and in vivo

    J. Biol. Chem.

    (2014)
  • C.Q. Wang et al.

    Disruption of Runx1 and Runx3 leads to bone marrow failure and leukemia predisposition due to transcriptional and DNA repair defects

    Cell Rep.

    (2014)
  • K.S. Ahn et al.

    Reversal of chemoresistance and enhancement of apoptosis by statins through down-regulation of the NF-κB pathway

    Biochem. Pharmacol.

    (2008)
  • A.W. Chua et al.

    Butein downregulates chemokine receptor CXCR4 expression and function through suppression of NF-κB activation in breast and pancreatic tumor cells

    Biochem. Pharmacol.

    (2010)
  • K.S. Ahn et al.

    Genetic deletion of NAD(P)H:quinone oxidoreductase 1 abrogates activation of nuclear factor-κB, IκBα kinase, c-Jun N-terminal kinase, Akt, p38, and p44/42 mitogen-activated protein kinases and potentiates apoptosis

    J. Biol. Chem.

    (2006)
  • E.H. Blackburn

    Structure and function of telomeres

    Nature

    (1991)
  • R.K. Moyzis et al.

    A highly conserved repetitive DNA sequence, (TTAGGG)n, present at the telomeres of human chromosomes

    Proc. Natl. Acad. Sci. U.S.A

    (1988)
  • R.J. O’Sullivan et al.

    Telomeres: protecting chromosomes against genome instability

    Nat. Rev. Mol. Cell Biol.

    (2010)
  • J. Lingner et al.

    Reverse transcriptase motifs in the catalytic subunit of telomerase

    Science

    (1997)
  • T.M. Nakamura et al.

    Telomerase catalytic subunit homologs from fission yeast and human

    Science

    (1997)
  • C.B. Harley et al.

    Telomeres shorten during ageing of human fibroblasts

    Nature

    (1990)
  • N.D. Hastie et al.

    Telomere reduction in human colorectal carcinoma and with ageing

    Nature

    (1990)
  • M.A. Blasco

    Telomeres and human disease: ageing, cancer and beyond

    Nat. Rev. Genet.

    (2005)
  • K. Collins et al.

    Telomerase in the human organism

    Oncogene

    (2002)
  • W.E. Wright et al.

    Telomere biology in aging and cancer

    J. Am. Geriatr. Soc.

    (2005)
  • S. Kawanishi et al.

    Mechanism of telomere shortening by oxidative stress

    Ann. N. Y. Acad. Sci.

    (2004)
  • S. Raeisi et al.

    Oxidative stress-induced renal telomere shortening as a mechanism of cyclosporine-induced nephrotoxicity

    J. Biochem. Mol. Toxicol.

    (2018)
  • D. Jurk et al.

    Chronic inflammation induces telomere dysfunction and accelerates ageing in mice

    Nat. Commun.

    (2014)
  • S.L. Weinrich et al.

    Reconstitution of human telomerase with the template RNA component hTR and the catalytic protein subunit hTRT

    Nat. Genet.

    (1997)
  • Q. Zhang et al.

    Architecture of human telomerase RNA

    Proc. Natl. Acad. Sci. U.S.A.

    (2011)
  • C.W. Greider et al.

    A telomeric sequence in the RNA of Tetrahymena telomerase required for telomere repeat synthesis

    Nature

    (1989)
  • E.D. Egan et al.

    Biogenesis of telomerase ribonucleoproteins

    Rna

    (2012)
  • J.C. Schmidt et al.

    Human telomerase: biogenesis, trafficking, recruitment, and activation

    Genes Dev.

    (2015)
  • A.S. Venteicher et al.

    A human telomerase holoenzyme protein required for Cajal body localization and telomere synthesis

    Science

    (2009)
  • M.B. Ozturk et al.

    Current insights to regulation and role of telomerase in human diseases

    Antioxidants Basel (Basel)

    (2017)
  • C.M. Koh et al.

    Telomerase regulates MYC-driven oncogenesis independent of its reverse transcriptase activity

    J. Clin. Invest.

    (2015)
  • E. Khattar et al.

    Telomerase reverse transcriptase promotes cancer cell proliferation by augmenting tRNA expression

    J. Clin. Invest.

    (2016)
  • Y. Maida et al.

    An RNA-dependent RNA polymerase formed by TERT and the RMRP RNA

    Nature

    (2009)
  • A. Ghosh et al.

    Telomerase directly regulates NF-κB-dependent transcription

    Nat. Cell Biol.

    (2012)
  • K. Hoffmeyer et al.

    Wnt/β-Catenin signaling regulates telomerase in stem cells and cancer cells

    Science

    (2012)
  • K.Y. Sarin et al.

    Conditional telomerase induction causes proliferation of hair follicle stem cells

    Nature

    (2005)
  • Cited by (33)

    • Implications of cancer stem cells in diabetes and pancreatic cancer

      2023, Life Sciences
      Citation Excerpt :

      In the telomerase activity in humans, there are two subunits in it – one is the human telomerase reverse transcriptase (hTERT), and the other one is the human telomerase RNA (hTR) [219]. The telomerase inhibitor Imetelstat, an oligonucleotide telomerase antisense inhibitor, basically prevents the holoenzyme assembly in telomerase by binding with the RNA component by competing with the catalytic component of telomerase enzyme and binds to the region with high affinity [220]. Imetelstat has been successful past the second clinical trial phase and is effective in treating and preventing further growth of cancer cells in breast cancer, myelomas, etc. [220].

    • A zeta potential-based homogeneous assay for amplified detection of telomerase in cancer cells

      2022, Sensors and Actuators B: Chemical
      Citation Excerpt :

      Telomerase is a ribonucleoprotein complex composed of telomerase reverse transcriptase (TERT) and template RNA component (TERC), which is responsible for maintaining telomere stability and regulating cell growth and proliferation. Generally, normal human cells will reach the Hayflick limit after 50–70 times of division, resulting in cell aging and death [1,2]. However, in more than 85% of cases in cancers, telomerase is overexpressed, and high levels of telomerase will maintain telomere length by adding 5′-TTAGGG-3′ sequence repeats to the ends of the telomere to maintain the proliferation of cancer cells [3,4].

    View all citing articles on Scopus
    1

    Both Lele Wu and Kerem Fidan contributed equally to this study.

    View full text