Elsevier

Experimental Gerontology

Volume 96, 1 October 2017, Pages 110-122
Experimental Gerontology

Senescence-associated microRNAs target cell cycle regulatory genes in normal human lung fibroblasts

https://doi.org/10.1016/j.exger.2017.06.017Get rights and content

Highlights

  • Identification of a small set of senescence-associated microRNAs (SA-miRs).

  • SA-miRs target genes regulating cell cycle progression and telomere maintenance.

  • MiR-221 is up-regulated during replicative and oxidative stress induced senescence

  • MiR-221/222 targets cell cycle effectors.

Abstract

Senescence recapitulates the ageing process at the cell level. A senescent cell stops dividing and exits the cell cycle. MicroRNAs (miRNAs) acting as master regulators of transcription, have been implicated in senescence. In the current study we investigated and compared the expression of miRNAs in young versus senescent human fibroblasts (HDFs), and analysed the role of mRNAs expressed in replicative senescent HFL-1 HDFs. Cell cycle analysis confirmed that HDFs accumulated in G1/S cell cycle phase. Nanostring analysis of isolated miRNAs from young and senescent HFL-1 showed that a distinct set of 15 miRNAs were significantly up-regulated in senescent cells including hsa-let-7d-5p, hsa-let-7e-5p, hsa-miR-23a-3p, hsa-miR-34a-5p, hsa-miR-122-5p, hsa-miR-125a-3p, hsa-miR-125a-5p, hsa-miR-125b-5p, hsa-miR-181a-5p, hsa-miR-221-3p, hsa-miR-222-3p, hsa-miR-503-5p, hsa-miR-574-3p, hsa-miR-574-5p and hsa-miR-4454. Importantly, pathway analysis of miRNA target genes down-regulated during replicative senescence in a public RNA-seq data set revealed a significant high number of genes regulating cell cycle progression, both G1/S and G2/M cell cycle phase transitions and telomere maintenance. The reduced expression of selected miRNA targets, upon replicative and oxidative-stress induced senescence, such as the cell cycle effectors E2F1, CcnE, Cdc6, CcnB1 and Cdc25C was verified at the protein and/or RNA levels. Induction of G1/S cell cycle phase arrest and down-regulation of cell cycle effectors correlated with the up-regulation of miR-221 upon both replicative and oxidative stress-induced senescence. Transient expression of miR-221/222 in HDFs promoted the accumulation of HDFs in G1/S cell cycle phase. We propose that miRNAs up-regulated during replicative senescence may act in concert to induce cell cycle phase arrest and telomere erosion, establishing a senescent phenotype.

Introduction

Cellular senescence is a state of stable and irreversible cell cycle arrest with active metabolism that normal cells undergo after a finite number of divisions, the Hayflick limit. Senescence can be triggered by intrinsic and extrinsic stimuli including telomere shortening at the end of the cell's lifespan (telomere-initiated senescence) and in response to oxidative, genotoxic or oncogenic stresses (stress-induced premature senescence; SIPS). Several effector mechanisms have been proposed to explain the manner in which diploid cells senesce, including the DNA damage response (DDR), epigenetic regulation, the secretion of a complex mixture of extracellular matrix and inflammatory cytokines generating an inflammatory microenvironment, referred to as the senescence-associated secretory phenotype (SASP), and autophagy. Senescent cells display senescence-associated β-galactosidase (SA-β-Gal) activity and undergo chromatin remodeling resulting in senescence-associated heterochromatin foci (SAHF) and DNA damage foci (SDFs). SAHFs are characterised by the presence of heterochromatin-associated histone modifications (H3K9me) and heterochromatin protein-1 (HP1), and contain E2F target genes, which SAHFs are thought to silence. Senescence is established by de-repression of the INK4/ARF locus, acting as sensor to link stress with pRb and p53 tumor suppressors (Campisi and d'Adda di Fagagna, 2007, d'Adda di Fagagna, 2008, Fumagalli and d'Adda di Fagagna, 2009, Rodier and Campisi, 2011).

Several studies on the mechanisms of senescence have identified a class of non-coding RNAs (ncRNAs), microRNAs (miRNAs, miRs) and long ncRNAs as important regulators of cell senescence associated gene expression programs (Bischof and Martinez-Zamudio, 2015, Gorospe and Abdelmohsen, 2011, Grillari and Grillari-Voglauer, 2010, Lanceta et al., 2010, Olivieri et al., 2013).

MiRNAs are small, non-coding, endogenous single-stranded RNA molecules of ~ 22 nucleotides (nt) length that regulate gene expression in many cellular processes at both the transcriptional and post-transcriptional levels. MiRNAs act mostly as negative regulators of mRNA translation and/or stability by binding to complementary sequences in the 3′ untranslated regions (3′ UTR) of their target mRNAs, including those genes that mediate cell cycle regulation, stress responses, differentiation, senescence, apoptosis, inflammation and have also emerged as important regulators of tumourigenesis acting as tumor promoters (oncomirs) or suppressors (Croce, 2009, d'Adda di Fagagna, 2014, Esquela-Kerscher and Slack, 2006, Esteller, 2011, Gartel and Kandel, 2008, Gorospe and Abdelmohsen, 2011, Hammond, 2007, Iorio and Croce, 2012, Leung and Sharp, 2010, Lujambio and Lowe, 2012, Ventura and Jacks, 2009, Voorhoeve, 2010, Voorhoeve and Agami, 2007). Some miRNAs regulate specific individual targets, while others can function as master regulators of key processes (Ambros, 2004, Ameres and Zamore, 2013, Bueno and Malumbres, 2011, Cech and Steitz, 2014, Gurtan and Sharp, 2013, Inui et al., 2010, Leung and Sharp, 2010).

MicroRNAs have emerged as mediators of senescence and the ageing process (Harries, 2014). Previous studies showed that the expression of several miRs is altered in senescent human lung and skin fibroblasts (HDFs) (Bischof and Martinez-Zamudio, 2015, Gorospe and Abdelmohsen, 2011, Hackl et al., 2010, Holly et al., 2015, Li et al., 2009a, Maes et al., 2009, Marthandan et al., 2016, Marthandan et al., 2015, Wang et al., 2011), in senescent endothelial cells and other cell types (Dellago et al., 2013, Di Bernardini et al., 2014, Greussing et al., 2013, Hackl et al., 2010, Napolitano et al., 2014, Zhu et al., 2013). Importantly, senescent cells of different lineages demonstrate tissue-specificity in miRNA profiles (Holly et al., 2015).

Many miRNAs act as inducers of senescence, also known as senescence-associated miRNAs (SA-miRNAs or SA-miRs) (Bischof and Martinez-Zamudio, 2015, Gorospe and Abdelmohsen, 2011). In was shown that in senescent human WI-38 and MRC-5 HDFs a group of 15 miRNAs were down-regulated and these miRNAs represented three clusters including miRs-106b/93/25, the miR-17-92 polycistron and the miR-106a-92 polycistron. Overexpression of representative miRNAs of this group promoted cell proliferation and delayed senescence (Brosh et al., 2008). Interestingly, p53 which is induced in senescence, down-regulates the expression of oncomiR-17-92 and induces the transcription of SA-miRs, miR-34a/b/c which repress a number of target genes to promote growth arrest and apoptosis (He et al., 2007, Hermeking, 2010). In senescing primary mouse and human fibroblasts the histone demethylase KDM2B is epigenetically silenced leading to the induction of the tumor suppressor miRNAs let-7b (Benhamed et al., 2012, Johnson et al., 2007, Toledano, 2013). Several other miRs, including miR-34a and miR-146 have been implicated in both replicative and oncogene-induced senescence (Bhaumik et al., 2009, Bonifacio and Jarstfer, 2010, Christoffersen et al., 2010, Maes et al., 2009, Tazawa et al., 2007). In contrast to SA-miRs, functional genetic screens have identified miRNAs that rescued HDFs from oncogene-induced senescence (Borgdorff et al., 2010) including the oncomirs, miR-372 and miR-373 (Voorhoeve et al., 2006), and miR-378 (Kooistra et al., 2014).

Here, we investigated the expression of miRNAs in human lung diploid fibroblasts (HDFs), HFL-1, undergoing replicative senescence (RS) and in their young counterparts, and analysed the targets of the SA-miRs exclusively expressed in senescent HFL-1 and MRC-5 HDFs. Comparisons and bioinformatics analysis revealed a significant high number of down-regulated genes regulating cell cycle progression, both G1/S and G2/M cell cycle transitions, including CcnE, E2F1, Cdc6, CcnB1 and Cdc25C and telomere maintenance, both of which govern senescence and cancer. Moreover, transient overexpression of one of the SA-miRs, miR-221/222 retarded cell proliferation by targeting cell cycle effector gene expression.

Section snippets

Cell culture

Early passage normal human lung fibroblast strains (HDFs) HFL-1 (PDL24), and MRC-5 (PDL26) were cultured in Dulbecco's modified Eagle medium (DMEM) with low glucose (Hyclone), supplemented with 10% foetal bovine serum (FBS) (Gibco Life Technologies), 2 mM l-glutamine, 1% non-essential amino acids (NEAA), 100 units/ml penicillin and 100 μg/ml streptomycin. Cells were cultured at 37 °C, 5% CO2.

To determine the lifespan of both HFL-1 and MRC-5 HDFs, we estimated their cumulative number of population

Induction of replicative senescence in normal human embryonic lung fibroblasts

Replicative senescence (RS) of normal human lung fibroblasts (HDFs) was induced by continuously subculturing early passage HFL-1 at PDL26 and MRC-5 at PDL28 cells until they reached the end of their lifespan, ranging from 48 to 52 PDLs, and from 53 to 57 PDLs, respectively. The onset of senescence of HFL-1 HDFs was at PDL48 (Fig. 1A), and that of MRC-5 at PDL53 (Supplementary Fig. 1A).

Young HFL-1 at PDL28 and senescent HFL-1 at PDL48 were subjected to cell cycle analysis and to

Discussion

MicroRNAs negatively regulate gene expression and have emerged as important regulators of cellular senescence. MiRNAs have been shown to affect cell cycle progression by acting either as oncogenes or tumor suppressors, and several miRNAs that target the p53/p21 and the p16/pRb senescence pathways, the SASP-related pathway and transcription factors have been identified (Bischof and Martinez-Zamudio, 2015, Gorospe and Abdelmohsen, 2011), in several cell types including fibroblasts (Grillari and

Conflict of interest

The authors have no conflicts of interests.

Acknowledgements

We thank Professor Reuven Agami, and Dr. Pieter Breugel, Division of Biological Stress Response, The Netherlands Cancer Institute, Amsterdam, The Netherlands, for providing the MSCV-blast and MSCV-miR-221/222 and retroviral constructs.

This research has been, in part, co-financed by the European Union (European Social Fund - ESF) and Greek National Funds through the Operational Program “Education and Lifelong Learning” of the National Strategic Reference Framework (NSRF) - Research Funding

References (98)

  • J. Grillari et al.

    Novel modulators of senescence, aging, and longevity: small non-coding RNAs enter the stage

    Exp. Gerontol.

    (2010)
  • A.M. Gurtan et al.

    The role of miRNAs in regulating gene expression networks

    J. Mol. Biol.

    (2013)
  • G.S. Kim et al.

    Cdc6 localizes to S- and G2-phase centrosomes in a cell cycle-dependent manner

    Biochem. Biophys. Res. Commun.

    (2015)
  • K. Lafferty-Whyte et al.

    Pathway analysis of senescence-associated miRNA targets reveals common processes to different senescence induction mechanisms

    Biochim. Biophys. Acta (BBA) - Mol. Basis Dis.

    (2009)
  • J. Lanceta et al.

    MicroRNA group disorganization in aging

    Exp. Gerontol.

    (2010)
  • A.K. Leung et al.

    MicroRNA functions in stress responses

    Mol. Cell

    (2010)
  • G. Li et al.

    Alterations in microRNA expression in stress-induced cellular senescence

    Mech. Ageing Dev.

    (2009)
  • K.J. Livak et al.

    Analysis of relative gene expression data using real-time quantitative PCR and the 2 − ΔΔCT method

    Methods

    (2001)
  • F. Olivieri et al.

    MicroRNAs linking inflamm-aging, cellular senescence and cancer

    Ageing Res. Rev.

    (2013)
  • T.G. Petrakis et al.

    Exploring and exploiting the systemic effects of deregulated replication licensing

    Semin. Cancer Biol.

    (2016)
  • L. Poliseno et al.

    MicroRNAs modulate the angiogenic properties of HUVECs

    Blood

    (2006)
  • S. Roush et al.

    The let-7 family of microRNAs

    Trends Cell Biol.

    (2008)
  • A. Sfikas et al.

    The canonical NF-kappaB pathway differentially protects normal and human tumor cells from ROS-induced DNA damage

    Cell. Signal.

    (2012)
  • H. Toledano

    The role of the heterochronic microRNA let-7 in the progression of aging

    Exp. Gerontol.

    (2013)
  • A. Tzatsos et al.

    Lysine-specific demethylase 2B (KDM2B)-let-7-enhancer of zester homolog 2 (EZH2) pathway regulates cell cycle progression and senescence in primary cells

    J. Biol. Chem.

    (2011)
  • A. Ventura et al.

    MicroRNAs and cancer: short RNAs go a long way

    Cell

    (2009)
  • P.M. Voorhoeve

    MicroRNAs: oncogenes, tumor suppressors or master regulators of cancer heterogeneity?

    Biochim. Biophys. Acta

    (2010)
  • P.M. Voorhoeve et al.

    Classifying microRNAs in cancer: the good, the bad and the ugly

    Biochim. Biophys. Acta

    (2007)
  • P.M. Voorhoeve et al.

    A genetic screen implicates miRNA-372 and miRNA-373 as oncogenes in testicular germ cell tumors

    Cell

    (2006)
  • H. Yin et al.

    Progress on the relationship between miR-125 family and tumorigenesis

    Exp. Cell Res.

    (2015)
  • F. d'Adda di Fagagna

    Living on a break: cellular senescence as a DNA-damage response

    Nat. Rev. Cancer

    (2008)
  • F. d'Adda di Fagagna et al.

    A DNA damage checkpoint response in telomere-initiated senescence

    Nature

    (2003)
  • F. d'Adda di Fagagna et al.

    Functional links between telomeres and proteins of the DNA-damage response

    Genes Dev.

    (2004)
  • E.A. Afanasyeva et al.

    MicroRNA miR-885-5p targets CDK2 and MCM5, activates p53 and inhibits proliferation and survival

    Cell Death Differ.

    (2011)
  • V. Ambros

    The functions of animal microRNAs

    Nature

    (2004)
  • S.L. Ameres et al.

    Diversifying microRNA sequence and function

    Nat. Rev. Mol. Cell Biol.

    (2013)
  • J. Bartkova et al.

    Oncogene-induced senescence is part of the tumorigenesis barrier imposed by DNA damage checkpoints

    Nature

    (2006)
  • M. Benhamed et al.

    Senescence is an endogenous trigger for microRNA-directed transcriptional gene silencing in human cells

    Nat. Cell Biol.

    (2012)
  • D. Bhaumik et al.

    MicroRNAs miR-146a/b negatively modulate the senescence-associated inflammatory mediators IL-6 and IL-8

    Aging (Albany NY)

    (2009)
  • O. Bischof et al.

    MicroRNAs and lncRNAs in senescence: a re-view

    IUBMB Life

    (2015)
  • L.N. Bonifacio et al.

    MiRNA profile associated with replicative senescence, extended cell culture, and ectopic telomerase expression in human foreskin fibroblasts

    PLoS One

    (2010)
  • V. Borgdorff et al.

    Multiple microRNAs rescue from Ras-induced senescence by inhibiting p21(Waf1/Cip1)

    Oncogene

    (2010)
  • L.R. Borlado et al.

    CDC6: from DNA replication to cell cycle checkpoints and oncogenesis

    Carcinogenesis

    (2008)
  • S. Boronat et al.

    Linking mitosis with S-phase: Cdc6 at play

    Cell Cycle

    (2008)
  • R. Brosh et al.

    p53-repressed miRNAs are involved with E2F in a feed-forward loop promoting proliferation

    Mol. Syst. Biol.

    (2008)
  • D.M. Burns et al.

    CPEB and two poly(A) polymerases control miR-122 stability and p53 mRNA translation

    Nature

    (2011)
  • J. Campisi et al.

    Cellular senescence: when bad things happen to good cells

    Nat. Rev. Mol. Cell Biol.

    (2007)
  • N.R. Christoffersen et al.

    p53-independent upregulation of miR-34a during oncogene-induced senescence represses MYC

    Cell Death Differ.

    (2010)
  • C.M. Croce

    Causes and consequences of microRNA dysregulation in cancer

    Nat. Rev. Genet.

    (2009)
  • Cited by (49)

    • Role of hypoxia in cellular senescence

      2023, Pharmacological Research
    • Lin28b delays vasculature aging by reducing platelet-derived growth factor-beta resistance in senescent vascular smooth muscle cells

      2023, Atherosclerosis
      Citation Excerpt :

      There is also debate about the role of let-7 in aging. Let-7d and let-7e are upregulated as biomarkers in human dermal senescent fibroblasts [26]. Other researchers have reported that let-7a represses the translation of p66Shc, p52Shc, and p46Shc, delaying replicative senescence in human diploid fibroblasts [42].

    • microRNA-34 family: From mechanism to potential applications

      2022, International Journal of Biochemistry and Cell Biology
      Citation Excerpt :

      Yang et al. reported that miR-34 inhibited autophagy by directly inhibiting the expression of autophagy related protein Atg9 (Yang et al., 2013). The expression of miR-34a-5p was significantly up-regulated in skin fibroblasts affected telomere maintenance and cell life cycle at all stages (Markopoulos et al., 2017). Blackstone et al. found that miR-34 promoted the generation of wrinkles by degenerating collagen in the extracellular matrix when matrix metalloproteinases were activated and stimulated by ultraviolet B radiation (Blackstone et al., 2020).

    • Genes and pathways involved in senescence bypass identified by functional genetic screens

      2021, Mechanisms of Ageing and Development
      Citation Excerpt :

      ARF contributes to activation of p53 by promoting MDM2 degradation and induces cell cycle arrest (Campisi and d’Adda di Fagagna, 2007; d’Adda di Fagagna, 2008; d’Adda di Fagagna et al., 2003; Fumagalli and d’Adda di Fagagna, 2009; Gorgoulis and Halazonetis, 2010; Halazonetis et al., 2008; Martinez-Zamudio et al., 2017b; McHugh and Gil, 2017; Ogrunc et al., 2014; Rodier and Campisi, 2011; Saab, 2011; Salama et al., 2014; Sharpless and Sherr, 2015). Noncoding RNAs also control the senescence responses via a variety of mechanisms (Abdelmohsen et al., 2013; Benhamed et al., 2012; Borgdorff et al., 2010; Dimri et al., 2013; Gorgoulis et al., 2019; Kooistra et al., 2014; Lee et al., 2017; Li et al., 2018; Markopoulos et al., 2017a, b; Neault et al., 2016; Xu et al., 2019). Telomeres are repetitive nucleotide sequences at the chromosome ends that, along with specialised proteins, maintain genome integrity and chromosome stability (Bandaria et al., 2016).

    View all citing articles on Scopus
    1

    Georgios Markopoulos, Eugenia Roupakia and Maria Tokamani have contributed equally to this article.

    View full text