2,873
Views
18
CrossRef citations to date
0
Altmetric
Commentary

miRNA activation is an endogenous gene expression pathway

Pages 826-828 | Received 17 Jan 2018, Accepted 07 Mar 2018, Published online: 03 Apr 2018

ABSTRACT

Transfection of small non-coding RNAs (sncRNAs) molecules has become a routine technique widely used for silencing gene expression by triggering post-transcriptional and transcriptional RNA interference (RNAi) pathways. Moreover, in the past decade, small activating (saRNA) sequences targeting promoter regions were also reported, thereby a RNA-based gene activation (RNAa) mechanism has been proposed. In this regard, Turner and colleagues recently discovered an endogenous microRNA (miRNA) which binds its promoter in order to upregulate its own expression. Interestingly, several miRNA-induced RNA activation (miRNAa) phenomena have since then been identified. My objective here is to introduce the reader into the emergent miRNAa research field, as well as bring together important discoveries about this unexplored transcriptional activation pathway.

In the past decade, it was found that transfection of small double stranded RNA (dsRNA) molecules targeting promoter regions of the E-cadherin, vascular endothelial growth factor (VEGF) and p21 cyclin-dependent kinase inhibitor genes induced target transcriptional activation [Citation16]. Shortly after, Janowsky and colleagues [Citation11] reported short RNA duplexes also able to induce target gene expression of the Progesterone Receptor (PR) gene. Such process was named RNA activation (RNAa), and transfected dsRNAs (19-21 nt) referred to as small activating RNAs (saRNAs) or antigen RNAs (agRNAs) ([Citation11,Citation16], Schwartz et al. 2008 [Citation24]). The transfection of synthetic saRNAs into different cell lines has allowed to induce selective activation of target developmental genes involved in important cellular phenomena such as cell differentiation and apoptosis [Citation29,Citation33], and it has been reported in organisms as diverse as nematodes and mammals [Citation8,Citation9,Citation25]. Interestingly, several enzymes required for RNA interference (RNAi) including Dicer and Argonaute (Ago) family members have been also reported to function in RNAa [Citation9,Citation16,Citation22]; however, saRNAs exhibit slower and more durable effects when compared to small interfering RNAs (siRNAs) [Citation11,Citation16]. Functionally, saRNAs function to promote the assembly of the RNA Polymerase II (RNAPII) machinery on target promoters, and subsequently recruit heterogeneous nuclear ribonucleoproteins (hnRNPs), specific coactivators and chromatin remodeling factors which concertedly act to trigger gene activation [Citation6,Citation7,Citation24].

In higher eukaryotes, three main types of regulatory small non-coding RNAs (ncRNAs) have been identified: siRNAs, microRNAs (miRNAs) and piwi-interacting RNAs (piwi-RNAs). In particular, miRNAs are known to be evolutionarily conserved ncRNAs which target both DNA and RNA to inhibit gene expression. miRNA genes are first transcribed as long pri-miRNA stem-loop structures which are subsequently cleaved by Drosha and Dicer RNAses III enzymes to generate ∼70 nt long precursor miRNAs (pre-miRNAs), and mature miRNAs (21-22 nt), respectively [Citation13]. In the cytoplasm, miRNAs inhibit translation by mRNA degradation/destabilization and translational repression, while in the nucleus these master regulators control transcript stability and recruit epigenetic remodeling factors in order to induce gene silencing [Citation2]. Nonetheless, miRNAs can act not only to suppress, but also to induce expression of target genes. In 2008, Place et al. [Citation22] observed that transfection of synthetic miR-373 mimic targeting the region upstream of the transcription start site of the E-cadherin and the cold-shock domain-containing protein C2 (CSDC2) genes induced transcriptional activation in prostate cancer cell lines. Since then further miRNA-induced activation (miRNAa) phenomena have been reported (). In murine cell lines, miR-744 and miR-1186 target promoter of the Cyclin B1 (Ccnb1) gene to induce transcriptional activation [Citation7]. Interestingly, the authors reported that knockdown of endogenous miR-744 significantly impaired mRNA expression of Ccnb1, thereby suggesting that miRNAa may represent a naturally occurring mechanism acting under physiological conditions. Likewise miRNAs involved in RNAi, activating miRNAs also require of (1) Drosha and Dicer RNase III enzymes, responsible for miRNA biogenesis, and (2) members of the Argonaute (Ago) family proteins, known to be associated with miRNAs during the formation of the effector RNA-Induced Silencing Complex (RISC) [Citation7].

Table 1. miRNAs reported to target sequences and induce (or maintain) gene expression. The species from which cell lines/lineages have been derived are indicated in parentheses.

It was not until 2014 that was reported the first experimental evidence of a miRNA acting endogenously to induce target gene expression. In nematodes, Turner and colleagues showed that miRNA lin-4 targets a putative lin-4-complementary element (LCE) situated at its promoter to trigger autoactivation. This evolutionary conserved miRNA (lin-4) was the first miRNA discovered, thereby its effects on gene expression are nowadays well known, especially its ability to regulate the activity of major developmental genes involved in post-embryonic growth [Citation14]. Importantly, Turner et al. [Citation25] observed that lin-4 mediates the recruitment of RNAPII to its own promoter and it is sufficient to trigger lin-4 miRNA autoactivation. Relevant research reports on miRNA activation phenomena were made using nematodes as experimental organism models. In C. elegans, it also was found that primary transcripts of let-7 miRNA interact with ALG-1 (Argonaute Like Gene 1), and this complex is responsible for processing of mature miRNAs from their corresponding precursors [Citation34]. Moreover, activating miRNAs have also been reported to induce target host gene expression. In sarcoma cells, the Insulin-like growth factor 2 (IGF2) gene is upregulated by a miRNA localized within one of its introns, miR-483-5p, which targets the 5’ UTR of IGF2 mRNA transcripts [Citation19]. IGF2 is a tissue-specific maternally imprinted gene whose overexpression has been associated with tumorigenesis [Citation21]. Thus, miR-483-5p-mediated IGF2 gene expression would indicate that miRNAa plays an important role in the orchestration of complex epigenetic pathways such as involved in the control of gene imprinting. In support to this view, Liu et al. [Citation19] showed that miR-483-5p induces transcriptional activation through interaction of IGF2 with RHA (also known as DHX9), a RNA helicase which have been recently identified as a key component of the RNA-Induced Transcriptional Activation (RITA) complex [Citation23].

According to the experimental data, miRNAa does not fit a single model. Matsui et al. [Citation20] proposed that endogenous miR-589 induces target gene expression of the cyclooxygenase-2 (COX2) and phospholipase A2 (PLA2G4A) genes via a binding loop mechanism which includes gene promoters, nascent promoter COX2 transcript, Argonaute-2 (Ago2) and GW182, the last two being core components of the miRNA-induced silencing complex (miRISC) [Citation1]. It is important to note that the COX2 and PLA2G4A genes are located at a distance of only 149 kb, and they encode enzymes involved in the same inflammatory pathway. Likewise synthetic saRNAs, activating miRNAs may also produce site-specific epigenetic modifications [Citation7,Citation9,Citation11,Citation16,Citation20]. In HEK293T cell lines, miR-24-1 overexpression is sufficient to trigger histone 3 lysine 27 acetylation (H3K27ac) at target enhancers [Citation28]. In contrast, however, miRNAa targeted to TATA-box motifs seem not to involve epigenetic marks at promoters sequences, as reported during activation of the interleukin-2 (IL-2) gene [Citation31]. In this regard, authors propose that miRNAs targeting TATA-box motifs might enhance the processivity of the RNAPII through the stabilization of the transcriptional machinery.

miRNAs are critical and versatile regulators that function in the nucleus to regulate gene expression, although how they do it is still an open question. In regard to the miRNAa mechanism, it has shown to require: 1- core components of the miRNA silencing pathway (i.e., Ago1, Ago2, Dicer and Drosha enzymes), 2- enrichment of RNAPII at target sites, and 3- epigenetic transcriptional reprogramming [Citation4,Citation7,Citation9,Citation10,Citation20,Citation22]. There are, however, important question about miRNAa, namely, how activating miRNAs act under physiological conditions, in which cellular processes/circumstances they are transcribed, etc. Importantly, advanced insight into RNA-based regulatory mechanisms would allow design effective and switchable strategies for controlling transcriptional networks. For instance, malfunctioning of the RNAi pathway by miRNA deregulation has been found to be associated with progression of tissue-specific tumors and faulty silencing of specific oncogenes, thereby activating miRNAs might be used to produce miRNAa-mediated transcriptional activation of target tumor suppressors. In conclusion, miRNAa is one of the hottest topics in current molecular biology and it is expected that a more deeply understanding about this emergent field render important insights into genome function, as well as promising therapeutic applications.

Disclosure of potential conflicts of interest

No potential conflicts of interest were disclosed.

References

  • Carthew RW, Sontheimer EJ. Origins and mechanisms of miRNAs and siRNAs. Cell. 2009;136(4):642–655.
  • Catalanotto C, Cogoni C, Zardo G. MicroRNA in control of gene expression: an overview of nuclear functions. Int J Mol Sci. 2016;17(10):1712.
  • Chaluvally-Raghavan P, Jeong KJ, Pradeep S, et al. Direct upregulation of STAT3 by microRNA-551b-3p deregulates growth and metastasis of ovarian cancer. Cell Rep. 2016;15(7):1493–1504.
  • Dharap A, Pokrzywa C, Murali S, et al. MicroRNA miR-324-3p induces promoter-mediated expression of RelA gene. PLoS one. 2013;8(11):e79467.
  • Gernapudi R, Wolfson B, Zhang Y, et al. MicroRNA 140 promotes expression of long noncoding RNA NEAT1 in adipogenesis. Mol Cell Biol. 2016;36(1):30–38.
  • Hu J, Chen Z, Xia D, et al. Promoter-associated small double-stranded RNA interacts with heterogeneous nuclear ribonucleoprotein A2/B1 to induce transcriptional activation. Biochem J. 2012;447:407–416.
  • Huang V, Place RF, Portnoy V, et al. Upregulation of Cyclin B1 by miRNA and its implications in cancer. Nucleic Acids Res. 2012;40(4):1695–1707.
  • Huang V, Qin Y, Wang J, et al. RNAa is conserved in mammalian cells. PloS one. 2010;5(1):e8848.
  • Huang V, Zheng J, Qi Z, et al. Ago1 Interacts with RNA polymerase II and binds to the promoters of actively transcribed genes in human cancer cells. PLoS genet. 2013;9(9):e1003821.
  • Huang V.. Endogenous miRNAa: miRNA-Mediated Gene Upregulation. In RNA Activation (pp. .p. 65–79.). 2017. Springer; Singapore:
  • Janowski BA, Younger ST, Hardy DB, et al. Activating gene expression in mammalian cells with promoter-targeted duplex RNAs. Nat Chem Biol. 2007;3(3):166–173.
  • Kang MR, Park KH, Yang JO, et al. miR-6734 up-regulates p21 gene expression and induces cell cycle arrest and apoptosis in colon cancer cells. PLoS one. 2016;11(8):e0160961.
  • Lai EC. Micro RNAs are complementary to 3’ UTR sequence motifs that mediate negative post-transcriptional regulation. Nat genet. 2002;30(4):363.
  • Lee RC, Feinbaum RL, Ambros V. The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell. 1993;75(5):843–854.
  • Li C, Ge Q, Liu J, et al. Effects of miR-1236-3p and miR-370-5p on activation of p21 in various tumors and its inhibition on the growth of lung cancer cells. Tumour Biol. 2017a;39(6):1010428317710824.
  • Li LC, Okino ST, Zhao H, et al. Small dsRNAs induce transcriptional activation in human cells. Proc Natl Acad Sci USA. 2006;103:17337–17342.
  • Li S, Zhu Y, Liang Z, et al. Up-regulation of p16 by miR-877-3p inhibits proliferation of bladder cancer. Oncotarget. 2016;7(32):51773.
  • Li S, Wang C, Yu X, et al. miR-3619-5p inhibits prostate cancer cell growth by activating CDKN1A expression. Oncol Rep. 2017b;37(1):241–248.
  • Liu M, Roth A, Yu M, et al. The IGF2 intronic miR-483 selectively enhances transcription from IGF2 fetal promoters and enhances tumorigenesis. Genes Dev. 2013;27(23):2543–2548.
  • Matsui M, Chu Y, Zhang H, et al. 2013; Promoter RNA links transcriptional regulation of inflammatory pathway genes. Nucleic Acids Res.41(22):10086–10109.
  • Ogawa O, Eccles MR, Szeto J, et al. Relaxation of insulin-like growth factor II gene imprinting implicated in Wilms' tumour. Nature. 1993;362(6422):749.
  • Place RF, Li LC, Pookot D, et al. MicroRNA-373 induces expression of genes with complementary promoter sequences. Proc Natl Acad Sci USA. 2008;105(5):1608–1613.
  • Portnoy V, Lin SHS, Li KH, et al. saRNA-guided Ago2 targets the RITA complex to promoters to stimulate transcription. Cell Res. 2016;26:320–335.
  • Schwartz JC. Younger ST, Nguyen NB, Hardy DB, Monia BP, Corey DR, Janowski BA Antisense transcripts are targets for activating small RNAs. Nat. Struct Mol Biol. 2008;15(8): 842–848.
  • Turner M, Jiao A, Slack FJ. Autoregulation of lin-4 microRNA transcription by RNA activation (RNAa) in C. elegans Cell Cycle. 2014;13(5):772–781.
  • Wang C, Chen Z, Ge Q, et al. Up-regulation of p21(WAF1/ CIP1) by miRNAs and its implications in bladder cancer cells. FEBS Lett. 2014;588(24):4654–4664.
  • Wang C, Tang K, Li Z, et al. Targeted p21(WAF1/CIP1) activation by miR-1236 inhibits cell proliferation and correlates with favorable survival in renal cell carcinoma. Urol Oncol. 2016;34(2):59 e23–34.
  • Xiao M, Li J, Li W, et al. MicroRNAs Activate Gene Transcription Epigenetically as an Enhancer Trigger. RNA Biol. 2017;14(10): 1326–1334.
  • Yang K, Zheng XY, Qin J, et al. Up-regulation of p21 WAF1/Cip1 by saRNA induces G1-phase arrest and apoptosis in T24 human bladder cancer cells. Cancer Lett. 2008;265(2):206–214.
  • Zhang Y, Fan M, Geng G, et al. A novel HIV-1-encoded microRNA enhances its viral replication by targeting the TATA box region. Retrovirology. 2014a;11:23.
  • Zhang Y, Fan M, Zhang X, et al. Cellular microRNAs up-regulate transcription via interaction with promoter TATA-box motifs. RNA .2014b;20(12):1878–1889.
  • Zhang Y, Liu W, Chen Y, et al. A Cellular MicroRNA Facilitates Regulatory T Lymphocyte Development by Targeting the FOXP3 Promoter TATA-Box Motif. J Immunol. 2018;200(3):1053–1063.
  • Zheng L, Wang L, Gan J, et al. RNA activation: promise as a new weapon against cancer. Cancer Lett. 2014;355(1):18–24.
  • Zisoulis DG, Kai ZS, Chang RK, et al. Auto-regulation of miRNA biogenesis by let-7 and Argonaute. Nature. 2012;486(7404):541.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.