<iframe src="//www.googletagmanager.com/ns.html?id=GTM-5TSRKG" height="0" width="0" style="display: none; visibility: hidden">
Research Article
No access
Published Online: 19 May 2015

Modulation of Splicing by Single-Stranded Silencing RNAs

Publication: nucleic acid therapeutics
Volume 25, Issue Number 3

Abstract

Single-stranded silencing RNAs (ss-siRNAs) are chemically modified single-stranded oligonucleotides that can function through the cellular RNA interference (RNAi) machinery to modulate gene expression. Because their invention is recent, few studies have appeared describing their use and the potential of ss-siRNAs as a platform for controlling gene expression remains largely unknown. Using oligonucleotides to modulate splicing is an important area for therapeutic development and we tested the hypothesis that ss-siRNAs targeting splice sites might also be capable of directing increased production of therapeutically promising protein isoforms. Here we observe that ss-siRNAs alter splicing of dystrophin. Altered splicing requires a seed sequence complementarity to the target and expression of the RNAi factor argonaute 2. These results demonstrate that ss-siRNAs can be used to modulate splicing, providing another option for therapeutic development programs that aim to increase production of key protein isoforms. Splicing is a classical nuclear process and our data showing that it can be modulated through the action of RNA and RNAi factors offers further evidence that RNAi can take place in mammalian cell nuclei.

Get full access to this article

View all available purchase options and get full access to this article.

References

1.
Gagnon KT and Corey DR (2012). Argonaute and the Nuclear RNAs: new pathways for RNA-mediated control of gene expression. Nucleic Acid Ther 22:3–16.
2.
Li LC (2014). Chromatin remodeling by small RNA machinery in mammalian cells. Epigenetics 9:45–52.
3.
Castel SE and Martienssen RA (2013). RNA Interference in the nucleus: roles for small RNAs in transcription, epigenetics, and beyond. Nat Rev Genet 14:100–112.
4.
Schraivogel D and Meister G (2014). Import routes and nuclear functions of argonaute and other small RNA-silencing proteins. Trends Biochem Sci 39:420–431.
5.
White E, Schacklow M, Kamieniarz-Gdula K, Proudfoot NJ and Gullerova M (2014). Human nuclear dicer restricts the deleterious accumulation of endogenous double-stranded RNA. Nat Struc Mol Biol 21:552–559.
6.
Ross JP and Kassir Z (2014). The varied roles of nuclear argonaute-small RNA complexes and avenues for therapy. Mol Ther Nucl Acids 3:e203.
7.
Matsui M, Zhang H, Chu Y, Gagnon KT, Manoharan M, Corey DR and Janowski BA (2013). A Network of noncoding RNAs links transcription of inflammatory pathway genes COX-2 and PLAG24A. Nucleic Acids Res 41:10086–10109.
8.
Weinberg MS and Morris KV (2013). Long non-coding RNA targeting and transcriptional de-repression. Nucleic Acid Ther 23:9–4.
9.
Place RF, Li LC, Pookot D, Noonan EJ and Dahiya R (2001). MicroRNA-373 induces expression of genes with complementary promoter sequences. Proc Natl Acad Sci (USA) 105:1608–613.
10.
Kim DH, Saetrom P, Snove O and Rossi JJ (2008). MicroRNA-directed transcriptional gene silencing in mammalian cells. Proc Natl Acad Sci 105:16230–16235.
11.
Younger ST and Corey DR (2011). Transcriptional gene silencing in mammalian cells by miRNA mimics that target gene promoters. Nucl Acids Res 39:5682–5691.
12.
Robb GB, Brown KM, Khurana J and Rana TM (2005). Specific and potent RNAi in the nucleus of human cells. Nature Struct Mol Biol 12:133–37.
13.
Orht T, Muetze J, Svoboda P and P Schwille P, (2012). Intracellular localization and routing of miRNA and RNAi pathway components. Curr Top Med Chem 12:79–8.
14.
Gagnon KT, Li L, Chu Y, Janowski BA and Corey DR (2014). RNAi factors are present and active in mammalian cell nuclei. Cell Reports 6:211–221.
15.
Veltrop M and Aartsma-Rus A (2014). Antisense-mediated exon skipping: taking advantage of a trick from Mother Nature to treat rare genetic diseases. Exper Cell Res 325:50–55.
16.
Jarver P, O'Donovan L and Gait MJ (2014). A chemical view of oligonucleotides for exon skipping and related drug applications. Nucleic Acid Ther 24:37–47.
17.
Rigo F, Seth PP and Bennett CF (2014). Antisense oligonucleotide-based therapies for diseases caused by pre-mRNA processing defects. Adv Exp Med Biol 825:303–352.
18.
Battsche E and Ameyar-Zazoua M (2014). The influence of argonaute proteins on alternative RNA splicing. Wiley Interdiscip Rev. RNA 6:141–156.
19.
Liu J, Hu J, and Corey DR (2012). Expanding the action of duplex RNAs into the nucleus: redirecting alternative splicing. Nucl Acids Res 40:1240–1250.
20.
Allo M, Buggiano V, Fededa JP, Petrillo E, Schor I, de la Mata M, Agirre E, Plass M, Eyras E, et al. (2009). Control of alternative splicing through siRNA-mediated transcriptional gene silencing. Nat Struc Mol Biol 16:717–724.
21.
Ameyar-Zazoua M, Rachez C, Souidi M, Robin P, Fritsch L, Young R, Morozova N, Fenouil R, Descostes N, et al. (2014). Control of alternative splicing through siRNA-mediated transcriptional gene silencing. Nat Struc Mol Biol 19:998–1004.
22.
Allo M, Agirre E, Bessonov S, Bertucci P, Gomez Acuna L, Buggiano V, Bellora N, Singh B, Petrillo E, et al. (2104). Argonaute-1 binds transcriptional enhances and controls constitutive and alternative splicing in human cells. Proc Natl Acad Sci (USA) 111:15622–15629.
23.
Lima WF, Prakash TP, Murray HM, Kinberger GA, Li W, Chappell AE, Li CS, Murray SF, Gaus H, et al. (2012). Single-stranded siRNAs activate RNAi in animals. Cell 150: 883–894.
24.
Prakash TP, Lima WF, Murray HM, Elbashir S, Cantley W, Foster D, Jayaraman M, Chappell AE, Manoharan M, Swayze EE and Crooke ST. (2013). Lipid nanoparticles improve activity of single-stranded siRNA and gapmer antisense oligonucleotides in animals. ACS Chem Biol 8:1402–1406.
25.
Yu D, Pendergraff H, Liu J, Kordasiewicz HB, Cleveland DW, Swayze E, Lima W, Crooke ST, Prakash T and Corey DR. (2012). Single-stranded RNAs that function through RNAi are potent and allele-selective inhibitors of huntingtin expression. Cell 150:895–908.
26.
Hu J, Lui J, Yu D, Aiba Y, Lee S, Pendergraff H, Boubaker J, Arates JW, Lagier-Tourenne C, et al. (2014). Exploring the effect of sequence length and composition on allele-selective inhibition of human huntingtin expression by single-stranded silencing RNAs. Nucleic Acid Ther 24:199–209.
27.
Liu J, Yu D, Aiba Y, Pendergraff H, Swayze EE, Lima WF, Prakash TP and Corey DR. (2013). ss-siRNAs allele-selectively inhibit ataxin-3 expression: Multiple mechanisms for an alternative gene silencing strategy. Nucl Acids Res 41: 9570–9583.
28.
Hu J, Liu J, Narayannair KJ, Lackey JG, Kuchimanchi S., Rajeev KG, Manoharan M, Swayze EE, Lima WF, et al. (2014). Allele-selective inhibition of mutant atrophin-1 expression by duplex and single-stranded RNAs. Biochemistry 53,4510–4518.
29.
Matsui M, Prakash TP and Corey DR. (2013). Transcriptional silencing by single-stranded RNAs targeting a noncoding RNA that overlaps a gene promoter. ACS Chem Biol 8:122–126.
30.
Aartsma-Rus A, Bremmer-Bout M, Janson AMM, Dunnen JT, van Ommen GTB and van Deutekom JCT. (2002). Targeted exon skipping as a potential gene correction therapy for Duchenne muscular dystrophy. Neuromuscl Disord 12:S71–S77.
31.
Arechavala-Gomeza V, Graham IR, Popplewell LJ, Adams AM, Aartsma-Rus A, Kinali M, Morgan JE, Van Deutekom JC, Wilton SD, Dickson G and Muntoni F. (2007). Comparative analysis of antisense oligonucleotide sequences for targeting skipping of exon 5 during dystrophin pre-mRNA splicing in human muscle. Hum Mol Gene Ther 18:798–810.
32.
van Deutekom JC, Janson AA, Ginjaar IB, Frankhuizen WS, Aartsma-Rus A, Bremmer-Bout M, den Dunnen JT, Koop K, van der Kooi AJ, et al. (2007). Local dystrophin restoration with antisense oligonucleotide PRO051. New Eng J Med 375:2677–2686.
33.
Lu QL, Yokota T, Takeda S, Garcia L, Muntoni F and Partridge T. (2011). The status of exon skipping as a therapeutic approach to Duchennes muscular dystrophy. Mol Ther 19:9–15.
34.
Kole R and Leppert BJ (2012). Targeting mRNA splicing as a potential treatment for Duchenne muscular dystrophy. Discov Med 14:59–69.
35.
Voit T, Topaloglu H, Straub V, Muntoni F, Deconinck N, Campion G, de Kimpe SJ, Eagle M, Guglieri M, et al. (2014). Safety and efficacy of drisapersen for the treatment of Duchenne muscular dystrophy (DEMAND II): an exploratory randomised, placebo-controlled phase 2 study. Lancet Neurol 13:987–996.
36.
Hoffman EP and Mcnally EM. (2014). Exon-skipping therapy: a roadblock, detour, or a bump in the road. Sci Transl Med 6:230.
37.
Liu J, Carmell MA, Rivas FV, Marsden CG, Thomsen JM, Song JJ, Hammond SM, Joshua-Tor L and Hannon GJ. (2004). Argonaute 2 is the catalytic engine of RNAi. Science 305:1437–1441.
38.
Wang Y, Juranek S, Li H, Sheng G, Tuschl T and Patel DJ. (2008). Structure of an argonaute silencing complex with a seed-containing guide DNA and target RNA duplex. Nature 456:921–926.

Information & Authors

Information

Published In

cover image Nucleic Acid Therapeutics
nucleic acid therapeutics
Volume 25Issue Number 3June 2015
Pages: 113 - 120
PubMed: 25757055

History

Published in print: June 2015
Published online: 19 May 2015
Published ahead of print: 10 March 2015
Accepted: 2 February 2015
Received: 11 December 2014

Permissions

Request permissions for this article.

Topics

Authors

Affiliations

Jing Liu
Department of Pharmacology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas.
Department of Biochemistry, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas.
Jiaxin Hu
Department of Pharmacology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas.
Department of Biochemistry, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas.
Jessica A. Hicks
Department of Pharmacology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas.
Department of Biochemistry, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas.
Thazha P. Prakash
ISIS Pharmaceuticals, Carlsbad, California.
David R. Corey
Department of Pharmacology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas.
Department of Biochemistry, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas.

Notes

Address correspondence to:David Corey, PhDDepartment of PharmacologyUniversity of Texas SouthwesternMedical Center at Dallas6001 Forest ParkDallas, TX 75390E-mail: [email protected]

Author Disclosure Statement

T.P. Prakash is an employee of ISIS Pharmaceutical.

Metrics & Citations

Metrics

Citations

Export citation

Select the format you want to export the citations of this publication.

View Options

Get Access

Access content

To read the fulltext, please use one of the options below to sign in or purchase access.

Society Access

If you are a member of a society that has access to this content please log in via your society website and then return to this publication.

Restore your content access

Enter your email address to restore your content access:

Note: This functionality works only for purchases done as a guest. If you already have an account, log in to access the content to which you are entitled.

View options

PDF/EPUB

View PDF/ePub

Full Text

View Full Text

Media

Figures

Other

Tables

Share

Share

Copy the content Link

Share on social media

Back to Top