Inhibition of telomerase activity by splice-switching oligonucleotides targeting the mRNA of the telomerase catalytic subunit affects proliferation of human CD4+ T lymphocytes

https://doi.org/10.1016/j.bbrc.2018.12.186Get rights and content

Highlights

  • Splice-switching oligonucleotides inhibit telomerase activity.

  • Blocking of SRp20 and SRp40 proteins induces alternative splicing of hTERT.

  • Telomerase activity inhibition affects proliferation of human lymphocytes.

Abstract

Telomerase activity is regulated at the mRNA level by alternative splicing (AS) of its catalytic subunit hTERT. The aim of this study was to define the ability of splice-switching oligonucleotides (SSOs) that pair with hTERT pre-mRNA to induce AS and inhibit telomerase activity in human CD4+ T lymphocytes. SSOs that blocked the binding of a single splicing regulatory protein, SRp20 or SRp40, to its site within intron 8 of hTERT pre-mRNA demonstrated rather moderate capacities to induce AS and inhibit telomerase. However, a SSO that blocked the interaction of both SRp20 and SRp40 proteins with pre-mRNA was the most active. Cultivation of lymphocytes with spliced hTERT and inhibited telomerase resulted in the reduction of proliferative activity without significant induction of cell death. These results should facilitate further investigation of telomerase activity regulation, and antitelomerase SSOs could become promising agents for antiproliferative cell therapy.

Introduction

Telomerase is a ribonucleoprotein complex, which has unique reverse transcriptase activity with the main function of elongating and maintaining telomeres. Telomerase is active in most types of cancer cells, normal germ cells, stem cells, and activated lymphocytes [1]. Two main components of telomerase, the RNA component (TR) that contains the antisense template for telomere synthesis and the catalytic protein (hTERT), are enough for sufficient activity in cell-free conditions in vitro [2]. In vivo telomerase is associated with multiple regulatory proteins, which might be involved in various telomere-independent intracellular pathways (for review see Refs. [3,4]). Many different levels of regulation of the enzyme activity have been investigated [5]. Alternative splicing (AS) of hTERT mRNA is considered as one of the most precise regulators of telomerase activity in human cells. To date, 22 splice variants of hTERT have been described; however, only full-length hTERT (FL hTERT) mRNA encodes a biochemically functional enzyme [6]. In our previous work [7] we demonstrated that apoptotic endonuclease G (EndoG) could induce the deletion of exons 7 and 8 (β–variant) from hTERT mRNA, leading to a reading frame shift that results in the formation of a premature stop codon in exon 10 and the synthesis of a truncated hTERT protein [8,9]. The β– hTERT splice variant encodes inactive isoform with affected reverse transcription domain which acts as a dominant-negative protein [10,11]. EndoG could induce AS due to its RNase activity and its ability to produce the 48-mer oligonucleotide EGPO (EndoG-produced oligonucleotide) [7]. This oligonucleotide pairs with the junction site of exon 8 and intron 8 of hTERT pre-mRNA, and could induce AS in both living cells and naked cell nuclei. The action of EGPO was similar to splice-switching oligonucleotides (SSOs), which are considered a promising instrument for manipulation of protein functions [12,13].

EGPO covers two regulatory sites that are located in intron 8 of hTERT pre-mRNA, UCAUC and ACGGG, which are binding sites for SRp20 and SRp40 splicing regulator proteins, respectively [14]. We believe that base-pairing of specific oligonucleotide with pre-mRNA of hTERT can block the binding of SRp20 and SRp40 proteins to their sites, which results in the induction of the β– splice variant. The aim of this study was to investigate the ability of SSOs, which are complimentary to SRp20 or SRp40 binding sites, to induce AS of hTERT, inhibit telomerase activity and influence human lymphocytes proliferation.

Section snippets

Cell purification and cultivation

All procedures performed in these studies that involved human participants were in accordance with the ethical standards of the institutional and/or national research committee, and with the 1964 Helsinki declaration and its later amendments or comparable ethical standards. This study was approved by the Ethical Committee at the Institute of Biomedical Chemistry; written informed consent was obtained from all participants. Blood from healthy 25–35-year-old donors (n = 4) was collected in

Cell transfection with SSO for hTERT pre-mRNA results in telomerase inhibition

In attempt to induce AS of hTERT, we transfected CD4+ T lymphocytes with one of the SSOs, which are complimentary to binding sites of the splicing regulator proteins SRp20 and SRp40 (Fig. 1A–D). Twenty-four hours post transfection, the mRNA levels of hTERT splice variants and telomerase activity were measured in transfected cells. We observed non-significant changes in total hTERT mRNA levels in cell groups transfected with each of single SSO (Fig. 1E). SRp20 24-mer SSO, which blocks only the

Discussion

Functional hTERT is detected in multiple normal somatic human cells, including lymphocytes, brain, liver, prostate, heart, and primary fibroblasts, irrespective of the degree of telomerase activity [21], which denotes its extratelomeric functions. However, the presence of a full-length hTERT variant may not be sufficient to allow hTERT activity when an abundance of spliced variants are concomitantly present. The physiological and pathological significance of hTERT splicing patterns are being

Conflicts of interest

Authors declare no conflicts of interest.

Acknowledgements

The work was performed in the framework of the Program for Basic Research of the State Academies of Sciences for 2013–2020.

References (33)

  • S.D. Perrault et al.

    Global gene expression response to telomerase in bovine adrenocortical cells

    Biochem. Biophys. Res. Commun.

    (2005)
  • J.I. Young et al.

    Telomerase expression in normal human fibroblasts stabilizes DNA 5-methylcytosine transferase I

    J. Biol. Chem.

    (2003)
  • J.W. Shay

    Role of telomeres and telomerase in aging and cancer

    Cancer Discov.

    (2016)
  • J. Zhou et al.

    Telomerase reverse transcriptase in the regulation of gene expression

    BMB Rep

    (2014)
  • C. Nicholls et al.

    Molecular regulation of telomerase activity in aging

    Protein Cell

    (2011)
  • S. Saebøe-Larssen et al.

    Characterization of novel alternative splicing sites in human telomerase reverse transcriptase (hTERT): analysis of expression and mutual correlation in mRNA isoforms from normal and tumour tissues

    BMC Mol. Biol.

    (2006)
  • Cited by (8)

    • The diffusion-driven microfluidic process to manufacture lipid-based nanotherapeutics with stealth properties for siRNA delivery

      2022, Colloids and Surfaces B: Biointerfaces
      Citation Excerpt :

      Therefore, siRNA delivery should be well-investigated to develop next-generation drugs/vaccines to treat various genetic diseases. Although new advancements in genetic engineering have significantly contributed to the design of novel genetic machinery for specific cell targeting [4,5], delivery of such materials still represents a significant barrier to the clinical applications for the treatment of genetic disorders, and the complexity of the delivery still overshadows the potential of these systems [6]. The major challenge that prevents the success of this method is the degradation of the genetic material in the physiological fluids by the action of mononuclear phagocytes as part of the immune system, glomerular filtration, and endosomes inside the cells [7].

    • Expression and regulation of telomerase in human T cell differentiation, activation, aging and diseases

      2019, Cellular Immunology
      Citation Excerpt :

      Post-transcriptional regulation by alternative splicing produces several forms of hTERT alternatively spliced products (ASPs) during development [25] and activation [14]. Among them, α-, β-, and α + β-deletion have been observed in human T cells [14,22,26–30]. Briefly, α-deletion loses 36 bp within exon 6 resulting in deletion of part of the hTERT reverse transcriptase domain; β-deletion loses 182 bp resulting in a frame shift mutation; and the α + β-deletion loses 218 bp resulting in a frame shift mutation.

    View all citing articles on Scopus
    View full text