Submit or Track your Manuscript LOG-IN

Antiviral Roles of Artificial microRNAs

BJV_2_3_43-48

Commentary

Antiviral Roles of Artificial microRNAs

Huaichang Sun

College of Veterinary Medicine, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China.

Abstract | Many RNA viruses undergo rapid mutations which compromises the immune protection of conventional vaccines. RNA interference (RNAi) has becoming a feasible strategy against various virus infections. Recently, a significant advance in RNAi technology is the use of artificial microRNAs (amiRNAs) to fight virus infections. However, different strategies are needed to prevent virus variation or mutation escape. This review is intended to present the current situation of antiviral roles of amiRNAs, against variable RNA viruses in particular.

Editor | Muhammad Munir, The Pirbright Institute, Compton Laboratory, UK.

Received | April 01, 2015; Accepted | May 09, 2015; Published | June 09, 2015

*Correspondence | Huaichang Sun, Yangzhou University, Wenhui Road, Yangzhou, Jiangsu, China; E-mail | sunh@yzu.edu.cn

DOI | http://dx.doi.org/10.17582/journal.bjv/2015/2.3.43.48

Citation | Sun, H. 2015. Antiviral roles of artificial microRNAs. British Journal of Virology, 2(3): 43-48.

Introduction

RNA interference (RNAi) is a post-transcriptional gene silencing mechanism in eukaryotes (Fire et al., 1998; Ghildiyal and Zamore, 2009). Two types of small RNAs, namely small interfering RNAs (siRNAs) and microRNAs (miRNAs), are the central players in RNAi process, both of which silence gene expression by annealing to the target RNAs (Terasawa et al., 2011). Since its discovery in 1994 as an innate antiviral mechanism, RNAi has become a feasible strategy against a variety of viral infections (Arbuthnot, 2010; Zhou and Rossi, 2011).

MicroRNAs differ from siRNAs by their mechanisms of action and some features of processing. siRNAs are mostly exogenous dsRNA molecules derived from viral RNAs or artificially introduced into the cell (Chu and Rana, 2008), while miRNAs are endogenous non-coding RNAs, typically 18-22bp long, which derive from longer hairpin-shaped precursors called pre-miRNAs (Bartel, 2004). siRNAs are normally designed to be perfectly complementary to their target sequences, while miRNAs are usually fully complementary only to a small (7-8nt) “seed” sequence which determines the specific miRNA targets (Lai, 2002).

Different RNAi strategies have been proposed to inhibit virus replication in cell cultures and animal models. Among them, synthetic siRNAs can imitate endogenous triggers and have great effect for transient virus inhibition, but the toxicity and delivery difficulty prevent their clinical application (Whitehead et al., 2009). Vector-expressed siRNAs and miRNAs can enter the endogenous RNAi pathway and are processed into mature siRNAs (Boudreau et al., 2009; Paddison et al., 2002). Expression of siRNAs from RNA polymerase (pol) III promoters is the most common way to induce RNAi in cells, but the severe toxicity after administration of high doses of siRNA expression vectors has been reported (Grimm et al., 2006). These problems can be circumvented by using weaker pol II promoter to express artificial miRNSs (amiRNAs) which has several benefits, including easier expression of multiple amiRNAs in tissue-specific manners, control of the timing and level of gene silencing, and tracking of amiRNA expressing cells with correlated markers (Giering et al., 2008). Moreover, the amiRNA strategy has been studied as an antiviral approach for many viruses, including adenoviruses, rabies virus, dengue virus, and porcine reproductive and respiratory virus with minimal or no cytotoxicity (Bauer et al., 2009; Boudreau et al., 2009; Ibrisimovic et al., 2013; Israsena et al., 2009; Xie et al., 2013; Xia et al., 2013).

This brief commentary is intended to present the different amiRNA strategies against virus infections, for prevention of virus variation or mutation escaping in particular.

Prevention of virus variation or mutation escaping by targeting conserved sequences

Many RNA viruses undergo rapid mutations and thus escape mutants after RNAi treatment have been reported due to the sequence-specificity of RNAi mechanism (Boden et al., 2003). Unlike viral proteins, RNA dependent RNA polymerase (RDRP) is central to the replication of many RNA viruses, and thus is highly conserved between different strains. It has been shown that, however, the RDRP-targeted amiRNAs have significant inhibitory effect against the replication of different Venezuelan equine encephalitis virus strains (Bhomia et al., 2013). Foot-and-mouth disease virus (FMDV) is antigenically variable, and consists of seven serotypes and multiple subtypes. It has also been shown that the RDRP encoding 3D gene-targeted amiRNAs can efficiently inhibits FMDV replication (Du et al. 2011). Infectious bursal disease virus (IBDV), the causative agent of a highly contagious disease in chicks, is another example of variable RNA virus. The VP1 protein encoded by the small segment of IBDV RNA genome has both polymerase and capping enzyme activities. It has been shown that the anti-VP1 amiRNA has a more stable anti-IBDV effect than the amiRNA targeting main structural protein VP2 (Wang et al. 2009).

The RNA genomes of plus-strand RNA viruses have highly structured 5’- and 3’-untranslated regions (UTRs) that contain the cis-acting RNA elements involved in viral translation, replication and encapsidation (Liu et al., 2009). These conserved elements are also the good target for avoidance of virus variation or mutation escaping from RNAi. For example, Dengue viruses (DENV) have at least four known antigenically distinct DENV serotypes (DENV-1, 2, 3 and 4), and each serotype contains several phylogenetically distinct genotypes (Holmes and Burch, 2000). It has been shown that, however, the lentiviral vector-delivered tandem amiRNAs targeting the 5’- or 3’-UTR of DENV-2 genome can inhibit the viral replication in both stable and dose-dependent manners (Xie et al. 2013). Porcine reproductive and respiratory syndrome virus (PRRSV) is also notorious for strain diversity and rapid mutation (Kimman et al., 2009). Xia et al. (2013) have demonstrated that the plasmid vector-delivered amiRNAs targeting the 5’ or 3’ UTR of the viral RNA genome have potent effect against replication of three different PRRSV strains.

An internal ribosome entry site (IRES) allows for translation initiation in the middle of an mRNA sequence as part of the greater process of protein synthesis. These elements are often used by viruses when the host translation is inhibited. Chang et al. (2014) have showed that the co-expression of two IRES-targeted amiRNAs can effectively inhibit the replication of different FMDV strains. Furthermore, injection of the co-cistronic expression vector can partially or completely protect suckling mice against the challenge with prevalent PanAsia-1 and Mya98 strains of FMDV serotype O (Du et al., 2014).

Prevention of virus variation or mutation escaping by combinatorial RNAi strategy

Vector-based constructs have been developed for combinatorial RNAi approaches to silence multiple oncogenes or to enhance gene silencing efficiency. Such combinatorial RNAi approaches are particularly important for durable inhibition of escape-prone viruses. Several groups have shown that enhancement of RNAi activity can be obtained by concatenating multiple miRNA hairpins in a single construct (Sun et al., 2006; Chung et al., 2006). This approach has also been employed to inhibit pathogenic viruses that require a combinatorial RNAi attack to prevent viral escape, including HIV-1 (Liu and Berkhout, 2008) and HBV (Ely et al., 2009). In addition, the biosafety evaluation has shown that the three polycistronic amiRNAs expressed from a single construct have no apparent impact on cell proliferation, interferon response and interruption of native miRNA processing (Zhang et al. 2012). Although the siRNAs have a powerful inhibitory effect against Japanese encephalitis virus (JEV) replication, the high rate of genetic variation between JEV strains hampers their broad-spectrum application. Wu et al. (2011) have shown that tandem amiRNAs targeting the highly conserved regions of JEV genes are not only effective against the wild strains of genotypes I and genotype III, but also predicted to effective against other JEV genotypes (e.g., genotypes II and IV).

Prevention of virus variation or mutation escaping by targeting viral receptors

Most viruses enter the target cells via receptor-mediated endocytosis. Unlike the viral proteins which undergo rapid mutations, the cellular receptors for virus binding and/or entry are highly conserved proteins and thus can avoid the escape of gene-silencing effects by different viral strains (Fechner et al., 2007). Chicken heat-shock protein 90α (Hsp90α) has been identified as a functional component of the cellular receptor complex essential for IBDV infection (Lin et al., 2007). Yuan et al. (2012) have shown that the plasmid vector-delivered anti-Hsp90α amiRNAs have a potent inhibitory effect against IBDV infection. Furthermore, our recent studies showed that the avian adeno-associated viral vector-delivered anti-Hsp90α amiRNA was effective against three different IBDV strains (unpublished data). Various viruses, such as picornavirus, adenovirus, herpesvirus, Hantavirus, and reovirus, use integrins to initiate infection through a variety of mechanisms (Stewart and Nemerow, 2007). It has been shown that FMDV uses αvβ1, αvβ3, αvβ6 and αvβ integrins as the cellular receptors to initiate infection (Jackson et al., 2004). Recently, Du et al. (2014) have demonstrated that the plasmid vector-delivered amiRNAs targeting the αv subunit have significant inhibitory effect against FMDV replication in cells. Moreover, the amiRNA transgenic mice can be partially protected against the challenge with a large dose of FMDV. To date, at least three PRRSV receptors have been identified on porcine alveolar macrophages (PAMs), including heparan sulphate as the general attachment factor, sialoadhesin (CD169) for the viral binding and internalization, and CD163 for the viral genome release (Van Gorp et al., 2008). More recently, our group has shown that the Ad vector-delivered CD169- and CD163-targeted amiRNAs have an additive anti-PRRSV effect against different viral strains. The partial silencing effect is possibly due to alternative cellular factor(s) involved in PRRSV infection, rather than the low anti-vial activity of amiRNAs.

Cross transfer of anti-viral amiRNAs between different cells via exosomes

More recently, the exosomes derived from human and mouse cells have been shown as an efficient small RNA transfer vehicle (Valadi et al., 2007). This is very important for in vivo amiRNA delivery using viral vectors since most viruses have restricted tissue tropisms. For example, PRRSV targets the cells of monocyte-macrophage lineage, which are resistant to Ad vector transduction due to the lack of high affinity Ad receptor (Kaner et al., 1999). Our preliminary study has shown that, however, the CD163- or CD169-targeted amiRNA can be secreted from and taken up by pig cells via exosomes (Zhu et al., 2014). However, the exosome-mediated amiRNA transfer efficiencies in livestock such as pigs have not been investigated. More recently, our group showed that, after rAd transduction, the PRRSV 3’-UTR-targeted amiRNA was efficiently secreted from and taken up by different pig cell types. Moreover, the exosome-transferred amiRNA had potent anti-PRRSV effect against different viral strains (our manuscript). This indicates that pig cell-derived exosomes could also serve as an efficient small RNA transfer vehicle, including anti-viral amiRNAs.

In summary, a number of amiRNA approaches are presently available which have been proved to be the effective strategy against various virus infections, including prevention of virus variation or mutation escaping. Delivery remains the single greatest hurdle to in vivo use of the RNAi method. However, exciting advances have been made and new delivery systems under development may help to overcome these barriers.

References

  • Arbuthnot, P. Harnessing RNA interference for the treatment of viral infections. Drug News Perspect. 2010 Jul-Aug; 23 (6): 341-350.
  • Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell. 2004 Jan; 116(2):281-297. http://dx.doi.org/10.1016/S0092-8674(04)00045-5
  • Bauer M, Kinkl N, Meixner A, Kremmer E, Riemenschneider M, Förstl H, Gasser T, Ueffing M. Prevention of interferon-stimulated gene expression using microRNA-designed hairpins. Gene Ther. 2009 Jan; 16(1):142-147. http://dx.doi.org/10.1038/gt.2008.123
  • Bhomia M, Sharma A, Gayen M, Gupta P, Maheshwari RK. Artificial microRNAs can effectively inhibit replication of Venezuelan equine encephalitis virus. Antiviral Res. 2013 Nov; 100(2):429-434. http://dx.doi.org/10.1016/j.antiviral.2013.08.010
  • Boden D, Pusch O, Lee F, Tucker L, Ramratnam B. Human immunodeficiency virus type 1 escape from RNA interference. J Virol. 2003; 77:11531-11535. http://dx.doi.org/10.1128/JVI.77.21.11531-11535.2003
  • Boudreau RL, Martins I, Davidson BL. Artificial microRNAs as siRNA shuttles: improved safety as compared to shRNAs in vitro and in vivo. Mol Ther. 2009; 17:169-175. http://dx.doi.org/10.1038/mt.2008.231
  • Chang Y, Dou Y, Bao H, Luo X, Liu X, Mu K, Liu Z, Liu X, Cai X1. Multiple microRNAs targeted to internal ribosome entry site against foot-and-mouth disease virus infection in vitro and in vivo. Virol J. 2014 Jan; 11:1-12. http://dx.doi.org/10.1186/1743-422X-11-1
  • Chu CY, Rana TM. Potent RNAi by short RNA triggers. RNA. 2008 Sep; 14(9):1714-1719. http://dx.doi.org/10.1261/rna.1161908
  • Chung KH, Hart CC, Al-Bassam S, Avery A, Taylor J, Patel PD, Vojtek AB, Turner DL. Polycistronic RNA polymerase II expression vectors for RNA interference based on BIC/miR-155. Nucleic Acids Res. 2006 Apr; 34(7):e53. http://dx.doi.org/10.1093/nar/gkl143
  • Du J, Gao S, Luo J, Zhang G, Cong G, Shao J, Lin T, Cai X, Chang H. Effective inhibition of foot-and-mouth disease virus (FMDV) replication in vitro by vector-delivered microRNAs targeting the 3D gene. Virol J. 2011 Jun; 8: 292. http://dx.doi.org/10.1186/1743-422X-8-292
  • Du J, Guo X, Gao S, Luo J, Gong X, Hao C, Yang B, Lin T, Shao J, Cong G, Chang H. Induction of protection against foot-and-mouth disease virus in cell culture and transgenic suckling mice by miRNA targeting integrin αv receptor. J Biotechnol. 2014 Oct; 187:154-161. http://dx.doi.org/10.1016/j.jbiotec.2014.07.001
  • Ely A, Naidoo T, Arbuthnot P. Efficient silencing of gene expression with modular trimeric Pol II expression cassettes comprising microRNA shuttles. Nucleic Acids Res. 2009 Jul; 37(13):e91. http://dx.doi.org/10.1093/nar/gkp446
  • Fechner H, Pinkert S, Wang X, Sipo I, Suckau L, Kurreck J, Dorner A, Sollerbrant K, Zeichhardt H. Coxsackievirus B3 and adenovirus infections of cardiac cells are efficiently inhibited by vector-mediated RNA interference targeting their common receptor. Gene Ther. 2007 Jun; 14: 960–971. http://dx.doi.org/10.1038/sj.gt.3302948
  • Fire A, Xu S, Montgomery MK, Kostas SA, Driver SE, Mello CC. Potent and specific genetic interference by double-stranded RNA in Caenorhabditis elegans. Nature. 1998: 391:806-811. http://dx.doi.org/10.1038/35888
  • Ghildiyal M, Zamore PD. Small silencing RNAs: an expanding universe. Nat. Rev Genet. 2009 Feb; 10:94-108. http://dx.doi.org/10.1038/nrg2504
  • Giering JC, Grimm D, Storm TA, Kay MA. Expression of shRNA from a tissue-specific pol II promoter is an effective and safe RNAi therapeutic. Mol Ther. 2008 Sep; 16(9):1630-1636. http://dx.doi.org/10.1038/mt.2008.144
  • Grimm D, Streetz KL, Jopling CL, Storm TA, Pandey K, Davis CR, Marion P, Salazar F, Kay MA. Fatality in mice due to oversaturation of cellular microRNA/short hairpin RNA pathways. Nature. 2006 May; 441(7092):537-541. http://dx.doi.org/10.1038/nature04791
  • Holmes EC, Burch SS. The causes and consequences of genetic variation in dengue virus. Trends Microbiol. 2000 Feb; 8:74–77. http://dx.doi.org/10.1016/S0966-842X(99)01669-8
  • Ibrisimovic M, Kneidinger D, Lion T, Klein R. An adenoviral vector-based expression and delivery system for the inhibition of wild-type adenovirus replication by artificial microRNAs. Antiviral Res. 2013 Jan; 97:10-23. http://dx.doi.org/10.1016/j.antiviral.2012.10.008
  • Israsena N, Supavonwong P, Ratanasetyuth N, Khawplod P, Hemachudha, T. Inhibition of rabies virus replication by multiple artificial microRNAs. Antiviral Res. 2009 Otc; 84:76-83.
  • Jackson T, Clark S, Berryman S, Burman A, Cambier S, Mu D, Nishimura S, King AMQ. Integrin αvβ functions as a receptor for foot-and-mouth disease virus: role of the β-chain cytodomain in integrin-mediated infection. J Virol. 2004 May; 78: 4533–4540. http://dx.doi.org/10.1128/JVI.78.9.4533-4540.2004
  • Kaner RJ, Worgall S, Leopold PL, Stolze E, Milano E, Hidaka C, Ramalingam R, Hackett NR, Singh R, Bergelson J, Finberg R, Falck-Pedersen E, Crystal RG. Modification of the genetic program of human alveolar macrophages by adenovirus vectors in vitro is feasible but inefficient, limited in part by the low level of expression of the coxsackie/adenovirus receptor. Am J Respir Cell Mol Biol. 1999 Mar; 20(3):361-370. http://dx.doi.org/10.1165/ajrcmb.20.3.3398
  • Kimman TG, Cornelissen LA, Moormann RJ, Rebel JM, Stockhofe-Zurwieden N. Challenges for porcine reproductive and respiratory syndrome virus (PRRSV) vaccinology. Vaccine. 2009 Jun; 27(28): 3704-3718. http://dx.doi.org/10.1016/j.vaccine.2009.04.022
  • Lai EC. Micro RNAs are complementary to 3’ UTR sequence motifs that mediate negative post-transcriptional regulation. Nat Genet. 2002 Apr; 30(4):363-364. http://dx.doi.org/10.1038/ng865
  • Lin TW, Lo CW, Lai SY, Fan RJ, Lo CJ, Chou YM, Thiruvengadam R, Wang AH, Wang MY. Chicken heat shock protein 90 is a component of the putative cellular receptor complex of infectious bursal disease virus. J Virol. 2007; 81(16):8730-8741.
  • Liu YP, Berkhout B. Combinatorial RNAi strategies against HIV-1 and other escape-prone viruses. Int J BioSci Technol. 2008; 1:1-10.
  • Liu Y, Wimmer E, Paul AV. Cis-acting RNA elements in human and animal plus-strand RNA viruses. Biochim Biophys Acta. 2009 Sep-Oct; 1789(9-10):495-517. http://dx.doi.org/10.1016/j.bbagrm.2009.09.007
  • Paddison PJ, Caudy AA, Bernstein E, Hannon GJ, Conklin DS. Short hairpin RNAs (shRNAs) induce sequence-specific silencing in mammalian cells. Genes Dev. 2002 Apr; 16:948-958. http://dx.doi.org/10.1101/gad.981002
  • Stewart PL, Nemerow GR. Cell integrins: commonly used receptors for diverse viral pathogens. Trends Microbiol. 2007 Nov; 15 (11): 500–507. http://dx.doi.org/10.1016/j.tim.2007.10.001
  • Sun D, Melegari M, Sridhar S, Rogler CE, Zhu L. Multi-miRNA hairpin method that improves gene knockdown efficiency and provides linked multi-gene knockdown. Biotechniques. 2006 Jul; 41(1):59-63. http://dx.doi.org/10.2144/000112203
  • Terasawa K, Shimizu K, Tsujimoto G. Synthetic Pre-miRNA-Based shRNA as Potent RNAi Triggers. J Nucleic Acids. 2011 Jun; 2011:131579.
  • Valadi H, Ekström K, Bossios A, Sjöstrand M, Lee JJ, Lötval, JO. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat Cell Biol. 2007 Jun; 9(6): 654–659. http://dx.doi.org/10.1038/ncb1596
  • Van Gorp H, Van Breedam W, Delputte PL, Neuwynck HI. Sialoadhesin and CD163 join forces during entry of the porcine reproductive and respiratory syndrome virus. J Gen Virol. 2008 Dec; 89(Pt 12):2943-2953. http://dx.doi.org/10.1099/vir.0.2008/005009-0
  • Wang Y, Sun H, Shen P, Zhang X, Xia X. Effective inhibition of infectious bursal disease virus replication by recombinant avian adeno-associated virus-delivered microRNAs. J Gen Virol. 2009 Jun; 90(Pt 6):1417-1722. http://dx.doi.org/10.1099/vir.0.010520-0
  • Whitehead KA, Langer R, Anderson DG. Knocking down barriers: advances in siRNA delivery. Nat Rev Drug Discov. 2009; 8:129-138. http://dx.doi.org/10.1038/nrd2919-c1 http://dx.doi.org/10.1038/nrd2742
  • Wu Z, Xue Y, Wang B, Du J, Jin Q. Broad-spectrum antiviral activity of RNA interference against four genotypes of Japanese encephalitis virus based on single microRNA polycistrons. PLoS One. 2011; 6(10):e26304. http://dx.doi.org/10.1371/journal.pone.0026304
  • Xia B, Song H, Chen Y, Zhang X, Xia X, Sun H. Efficient inhibition of porcine reproductive and respiratory syndrome virus replication by artificial microRNAs targeting the untranslated regions. Arch Virol. 2013 Apr; 158:55-61. http://dx.doi.org/10.1007/s00705-012-1455-z
  • Xie PW, Xie Y, Zhang XJ, Huang H, He LN, Wang XJ, Wang SQ. Inhibition of Dengue virus 2 replication by artificial micrornas targeting the conserved regions. Nucleic Acid Ther. 2013 Aug; 23(4):244-252.
  • Yuan W, Zhang X, Xia X, Sun H. Inhibition of infectious bursal disease virus infection by artificial microRNAs targeting chicken heat-shock protein 90. J Gen Virol. 2012 Apr; 93(Pt 4):876-879. http://dx.doi.org/10.1099/vir.0.039172-0
  • Zhang T, Cheng T, Wei L, Cai Y, Yeo AE, Han J, Yuan YA, Zhang J, Xia N. Efficient inhibition of HIV-1 replication by an artificial polycistronic miRNA construct. Virol J. 2012 Jun; 9:118. http://dx.doi.org/10.1186/1743-422X-9-118
  • Zhou J, Rossi JJ. Progress in RNAi-based antiviral therapeutics. Methods Mol Biol. 2011; 721:67-75. http://dx.doi.org/10.1007/978-1-61779-037-9_4
  • Zhu L, Song H, Zhang X, Xia X, Sun H. Inhibition of porcine reproductive and respiratory syndrome virus infection by recombinant adenovirus- and/or exosome-delivered the artificial microRNAs targeting sialoadhesin andCD163 receptors. Virol J. 2014 Dec; 11(1): 225. http://dx.doi.org/10.1186/s12985-014-0225-9
  • Zhu L, Bao L, Zhang X, Xia X, Sun H. The exosome-mediated transfer and anti-viral effect of 3’ untranslated region-targeted artificial microRNA against porcine reproductive and respiratory syndrome virus replication. Virus Res. 2015 (manuscript).

To share on other social networks, click on any share button. What are these?

Hosts and Viruses

December

Vol.10, Pages 1-71

Featuring

Click here for more

Subscribe Today

Receive free updates on new articles, opportunities and benefits


Subscribe Unsubscribe