16
views
0
recommends
+1 Recommend
3 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Lessons from SARS and MERS remind us of the possible therapeutic effects of implementing a siRNA strategy to target COVID‐19: Shoot the messenger!

      letter

      Read this article at

      ScienceOpenPublisherPMC
      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          1 Dear Editor, Since its first identification as a human pathogen in the Wuhan province of China in December 2019, the SARS‐CoV‐2 virus, which causes COVID‐19, has become a global pandemic with immense medical and socio‐economic costs. Like other coronaviruses, such as severe acute respiratory syndrome (SARS) and Middle East respiratory syndrome coronavirus (MERS‐CoV), SARS‐CoV‐2 is a single‐stranded positive‐sense RNA virus. The SARS‐COV‐2 as well as the SARS‐COV and MERS‐COV genomes contain several open‐reading frames (ORFs) that play an essential role in viral pathogenicity and infection. 1 , 2 , 3 Based on previous experiences with other coronaviruses, ORFs are considered to be essential for viral replication through encoding viral replicase proteins to synthesize mRNAs of subgenomic length. 1 , 3 Silencing or small/short interfering RNA (siRNA) is a gene silencing approach using a small fragment of approximately 20‐25 base pairs of double‐stranded RNA that binds to a specific site of the relevant/target messenger RNA (mRNA); siRNAs are designed to silence genes at the post‐transcriptional level (by inducing cleavage and subsequent degradation of target mRNA) and can therefore be considered as vaccines or therapeutic agents. Zheng et al 4 designed 48 siRNA sequences that potentially target the entire SARS‐CoV genome RNA, including ORFs for the translation of several key proteins. Among these, four siRNAs that could inhibit SARS‐CoV infection in foetal rhesus monkey kidney cells (FRhK‐4), both in a prophylactic and post‐infection therapeutic manner, were identified. Translating this idea to live animal experiments, Li et al 5 demonstrated a similar efficacy of siRNAs in a rhesus macaque (Macaca mulatta) SARS model. These agents, with no visible signs of toxicity, were shown to improve several symptoms of SARS‐CoV, such as fever, viral load and acute alveolar damage. Importantly, the efficacy of the siRNAs was evident at relatively small respiratory doses (10‐40 mg/kg). Similar experiments have been performed with different siRNA sequences targeting various regions of the SARS‐CoV genome. For example, He et al 6 showed that miRNAs targeting the replicase 1A region were more effective against the virus in FRhK‐4 cells. In 293 and HeLa cells, siRNAs targeting SARS‐CoV RNA‐dependent RNA polymerase (RDRP) showed therapeutic potential as well by specifically inhibiting RDRP expression. 7 In addition, this system reduced plaque formation in Vero‐E6 cells, a cell line classically used to identify and count hemorrhagic fever viruses. In these cells, siRNA to target and inhibit gene expression of SARS‐CoV spike (S) protein has been successfully utilized in vitro. 8 , 9 Similarly, siRNAs efficiently targeting S protein coding regions have been identified using FRhk‐4 cells and an in vivo rhesus macaque model of SARS‐CoV infection. 10 In line with these findings, the aforementioned efficacy of the siRNA developed by Li et al 5 was based on the S protein coding and ORF1b (NSP12) regions. Envelope (E) and membrane (M) proteins could also be (specifically) targeted, as demonstrated in SARS‐CoV‐infected FRhk‐4 cells. 11 In addition to synergistic effects that may be exhibited by different siRNAs, their therapeutic action can synergize with other currently existing antiviral agents through direct or indirect targeting common structural genes or other cellular targets. 11 , 12 In principle, several proteins encoded by the viral genome can be targeted by siRNA technology. 13 He et al 14 demonstrated the power of synergistic antiviral effects through siRNA targeting of various structural genes such as S, envelope, membrane and nucleocapsid. An additional advantage of siRNA technology is the incredibly low dose required to eliminate SARS‐CoV infection; for example, less than 60 nmol/L in Vero E6 cells 15 and 10‐40 mg/kg/daily in monkeys was sufficient for satisfactory therapeutic effects. 5 The application and potential effectiveness of siRNAs have also been evaluated in MERS‐CoV using computational models. 16 In view of angiotensin‐converting enzyme 2 (ACE2) as a recognized host cell receptor for the SARS‐CoV S protein, the development of siRNAs targeting key host proteins could hold promise. Indeed, silencing ACE2 expression in Vero E6 cells by siRNA (containing sequences homologous to a section of ACE2) significantly reduced SARS‐CoV infection. 17 Overall, the described studies on the effectiveness of specific siRNAs to battle SARS‐CoV and MERS‐CoV provide sufficient rationale to at least consider the use of siRNA strategies to target the closely related virus SARS‐CoV‐2. Despite their promising therapeutic effects, the application of higher doses of siRNAs, if so required, may be associated with some challenges, including adaptive 18 and innate immune responses, 19 , 20 unwanted target effects, and saturation of the endogenous small RNA machinery. 21 It is comforting, however, that previous data from several randomised, double‐blind, placebo‐controlled trials indicate that ALN‐RSV01 (a siRNA‐based drug) is safe to use and effective against respiratory syncytial virus infection. 22 , 23 Taken together, siRNA‐based therapeutics might be considered as an effective strategy to treat of COVID‐19. Future studies are warranted to evaluate their potential efficacy and safety. CONFLICT OF INTEREST The authors declare no competing interests. AUTHOR CONTRIBUTION Solomon Habtemariam: Formal analysis (equal); Investigation (equal). Ioana Berindan‐Neagoe: Conceptualization (equal); Investigation (equal). Cosmin Andrei Cismaru: Formal analysis (equal); Methodology (equal). Dedmer Schaafsma: Investigation (equal); Methodology (equal). Seyed Fazel Nabavi: Conceptualization (equal); Data curation (equal). Saeid Ghavami: Investigation (equal); Methodology (equal). Maciej Banach: Investigation (equal); Methodology (equal). Seyed Mohammad Nabavi: Conceptualization (equal); Formal analysis (equal). 1.1 FIGURE 1 Potential effects of siRNAs on silencing viral genes at the post‐transcriptional level in COVID‐19. Coronaviruses enter the cell via the endosomal pathway exploiting autophagy or the non‐endosomal pathway, both leading to the release of the nucleocapsid into the cytoplasm. Replication of genomic RNA takes place in double‐membrane vesicles (DMVs) shielded from host immune responses, where the translation of ORF1a/b into the replicase polyprotein 1a (pp1a) and pp1ab will take place. Papain‐like proteases (PLpro) and 3C‐like protease (3CLpro) cleave pp1a and pp1ab to produce non‐structural proteins (nsp), including replicases (RNA‐dependent RNA polymerases) and helicases. The positive‐strand genomic RNA is transcribed to form a negative‐strand template for the synthesis of new genomic RNAs and subgenomic negative‐strand templates. mRNA is synthesized and translated into producing the structural and accessory viral proteins. siRNAs can potentially silence genes at post‐transcriptional level, degrading mRNA and blocking its translation. (adapted after Zumla et al 24 ) REFERENCES 1 Menachery VD , Mitchell HD , Cockrell AS , et al. MERS‐CoV accessory ORFs play key role for infection and pathogenesis. MBio. 2017;8:e00665.28830941 2 Chen L , Zhong L . Genomics functional analysis and drug screening of SARS‐CoV‐2. Genes Dis. 2020 10.1016/j.gendis.2020.04.002 3 Yount B , Roberts RS , Sims AC , et al. Severe acute respiratory syndrome coronavirus group‐specific open reading frames encode nonessential functions for replication in cell cultures and mice. J. Virol. 2005;79:14909‐14922.16282490 4 Zheng BJ , Guan Y , Tang Q , et al. Prophylactic and therapeutic effects of small interfering RNA targeting SARS coronavirus. Antiviral Ther. 2004;9:365‐374. 5 Li BJ , Tang Q , Cheng D , et al. Using siRNA in prophylactic and therapeutic regimens against SARS coronavirus in Rhesus macaque. Nat Med. 2005;11:944‐951.16116432 6 He ML , Zheng B , Peng Y , et al. Inhibition of SARS‐associated coronavirus infection and replication by RNA interference. JAMA. 2003;290:2665‐2666.14645307 7 Lu A , Zhang H , Zhang X , et al. Attenuation of SARS coronavirus by a short hairpin RNA expression plasmid targeting RNA‐dependent RNA polymerase. Virology. 2004;324:84‐89.15183056 8 Zhang Y , Li T , Fu L , et al. Silencing SARS‐CoV Spike protein expression in cultured cells by RNA interference. FEBS lett. 2004;560:141‐146.14988013 9 Wu CJ , Huang HW , Liu CY , Hong CF , Chan YL . Inhibition of SARS‐CoV replication by siRNA. Antiviral Res. 2005;65:45‐48.15652970 10 Tang Q , Li B , Woodle M , Lu PY . Application of siRNA against SARS in the Rhesus macaque model Methods in Molecular Biology. Methods in Molecular Biology Totowa, New Jersey: Humana Press; 2008:139‐158. 11 He M , Chen Y , Sung JJ , et al. Development of interfering RNA agents to inhibit SARS‐associated coronavirus infection and replication. Hong Kong Med J. 2009;15:28‐31.19509435 12 Liao HI , Olson CA , Hwang S , et al. mRNA display design of fibronectin‐based intrabodies that detect and inhibit severe acute respiratory syndrome coronavirus nucleocapsid protein. J Biol Chem. 2009;284:17512‐17520.19364769 13 Åkerström S , Mirazimi A , Tan YJ . Inhibition of SARS‐CoV replication cycle by small interference RNAs silencing specific SARS proteins, 7a/7b, 3a/3b and S. Antiviral Res. 2007;73:219‐227.17112601 14 He ML , Zheng BJ , Chen Y , et al. Kinetics and synergistic effects of siRNAs targeting structural and replicase genes of SARS‐associated coronavirus. FEBS lett. 2006;580:2414‐2420.16638566 15 Shi Y , Yang DH , Xiong J , Jia J , Huang B , Jin YX . Inhibition of genes expression of SARS coronavirus by synthetic small interfering RNAs. Cell Res. 2005;15:193‐200.15780182 16 Nur SM , Hasan MA , Al Amin M , Hossain M , Sharmin T . Design of potential RNAi (miRNA and siRNA) molecules for Middle East respiratory syndrome coronavirus (MERS‐CoV) gene silencing by computational method. Interdiscipl Sci. 2015;7:257‐265. 17 Lu CY , Huang HY , Yang TH , Chang LY , Lee CY , Huang LM . siRNA silencing of angiotensin‐converting enzyme 2 reduced severe acute respiratory syndrome‐associated coronavirus replications in Vero E6 cells. Eur J Clin Microbiol Infect Dis. 2008;27:709‐715.18449585 18 Wittrup A , Lieberman J . Knocking down disease: a progress report on siRNA therapeutics. Nat Rev Genet. 2015;16:543‐552.26281785 19 Whitehead KA , Dahlman JE , Langer RS , Anderson DG . Silencing or stimulation? siRNA delivery and the immune system. Ann Rev Chem Biomol Eng. 2011;2:77‐96.22432611 20 Birmingham A , Anderson EM , Reynolds A , et al. 3′ UTR seed matches, but not overall identity, are associated with RNAi off‐targets. Nat Methods. 2006;3:199‐204.16489337 21 Grimm D , Streetz KL , Jopling CL , et al. Fatality in mice due to oversaturation of cellular microRNA/short hairpin RNA pathways. Nature. 2006;441:537‐541.16724069 22 Zamora MR , Budev M , Rolfe M , et al. RNA interference therapy in lung transplant patients infected with respiratory syncytial virus. Am J Respirat Crit Care Med. 2011;183:531‐538.20851929 23 DeVincenzo J , Lambkin‐Williams R , Wilkinson T , et al. A randomized, double‐blind, placebo‐controlled study of an RNAi‐based therapy directed against respiratory syncytial virus. Proc Natl Acad Sci. 2010;107:8800‐8805.20421463 24 Zumla A , Chan JF , Azhar EI , Hui DS , Yuen KY . Coronaviruses—drug discovery and therapeutic options. Nat Rev Drug Discov. 2016;15:327.26868298

          Related collections

          Most cited references14

          • Record: found
          • Abstract: found
          • Article: not found

          3' UTR seed matches, but not overall identity, are associated with RNAi off-targets.

          Off-target gene silencing can present a notable challenge in the interpretation of data from large-scale RNA interference (RNAi) screens. We performed a detailed analysis of off-targeted genes identified by expression profiling of human cells transfected with small interfering RNA (siRNA). Contrary to common assumption, analysis of the subsequent off-target gene database showed that overall identity makes little or no contribution to determining whether the expression of a particular gene will be affected by a given siRNA, except for near-perfect matches. Instead, off-targeting is associated with the presence of one or more perfect 3' untranslated region (UTR) matches with the hexamer or heptamer seed region (positions 2-7 or 2-8) of the antisense strand of the siRNA. These findings have strong implications for future siRNA design and the application of RNAi in high-throughput screening and therapeutic development.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Severe acute respiratory syndrome coronavirus group-specific open reading frames encode nonessential functions for replication in cell cultures and mice.

            SARS coronavirus (SARS-CoV) encodes several unique group-specific open reading frames (ORFs) relative to other known coronaviruses. To determine the significance of the SARS-CoV group-specific ORFs in virus replication in vitro and in mice, we systematically deleted five of the eight group-specific ORFs, ORF3a, OF3b, ORF6, ORF7a, and ORF7b, and characterized recombinant virus replication and gene expression in vitro. Deletion of the group-specific ORFs of SARS-CoV, either alone or in various combinations, did not dramatically influence replication efficiency in cell culture or in the levels of viral RNA synthesis. The greatest reduction in virus growth was noted following ORF3a deletion. SARS-CoV spike (S) glycoprotein does not encode a rough endoplasmic reticulum (rER)/Golgi retention signal, and it has been suggested that ORF3a interacts with and targets S glycoprotein retention in the rER/Golgi apparatus. Deletion of ORF3a did not alter subcellular localization of the S glycoprotein from distinct punctuate localization in the rER/Golgi apparatus. These data suggest that ORF3a plays little role in the targeting of S localization in the rER/Golgi apparatus. In addition, insertion of the 29-bp deletion fusing ORF8a/b into the single ORF8, noted in early-stage SARS-CoV human and civet cat isolates, had little if any impact on in vitro growth or RNA synthesis. All recombinant viruses replicated to wild-type levels in the murine model, suggesting that either the group-specific ORFs play little role in in vivo replication efficiency or that the mouse model is not of sufficient quality for discerning the role of the group-specific ORFs in disease origin and development.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              A randomized, double-blind, placebo-controlled study of an RNAi-based therapy directed against respiratory syncytial virus.

              RNA interference (RNAi) is a natural mechanism regulating protein expression that is mediated by small interfering RNAs (siRNA). Harnessing RNAi has potential to treat human disease; however, clinical evidence for the effectiveness of this therapeutic approach is lacking. ALN-RSV01 is an siRNA directed against the mRNA of the respiratory syncytial virus (RSV) nucleocapsid (N) protein and has substantial antiviral activity in a murine model of RSV infection. We tested the antiviral activity of ALN-RSV01 in adults experimentally infected with wild-type RSV. Eighty-eight healthy subjects were enrolled into a randomized, double-blind, placebo-controlled trial. A nasal spray of ALN-RSV01 or saline placebo was administered daily for 2 days before and for 3 days after RSV inoculation. RSV was measured serially in nasal washes using several different viral assays. Intranasal ALN-RSV01 was well tolerated, exhibiting a safety profile similar to saline placebo. The proportion of culture-defined RSV infections was 71.4 and 44.2% in placebo and ALN-RSV01 recipients, respectively (P = 0.009), representing a 38% decrease in the number of infected and a 95% increase in the number of uninfected subjects. The acquisition of infection over time was significantly lower in ALN-RSV01 recipients (P = 0.007 and P = 0.03, viral culture and PCR, respectively). Multiple logistic regression analysis showed that the ALN-RSV01 antiviral effect was independent of other factors, including preexisting RSV antibody and intranasal proinflammatory cytokine concentrations. ALN-RSV01 has significant antiviral activity against human RSV infection, thus establishing a unique proof-of-concept for an RNAi therapeutic in humans and providing the basis for further evaluation in naturally infected children and adults.
                Bookmark

                Author and article information

                Contributors
                Nabavi208@gmail.com
                Journal
                J Cell Mol Med
                J. Cell. Mol. Med
                10.1111/(ISSN)1582-4934
                JCMM
                Journal of Cellular and Molecular Medicine
                John Wiley and Sons Inc. (Hoboken )
                1582-1838
                1582-4934
                17 July 2020
                : 10.1111/jcmm.15652
                Affiliations
                [ 1 ] Pharmacognosy Research Laboratories and Herbal Analysis Services University of Greenwich Kent UK
                [ 2 ] Research Center for functional Genomics, Biomedicine and Translational Medicine “Iuliu Hatieganu” University of Medicine and Pharmacy Cluj‐Napoca Romania
                [ 3 ] Department of Functional Sciences, Immunology and Allergology “Iuliu Hatieganu” University of Medicine and Pharmacy Cluj‐Napoca Romania
                [ 4 ] Science Impact Winnipeg MB Canada
                [ 5 ] Applied Biotechnology Research Center Baqiyatallah University of Medical Sciences Tehran Iran
                [ 6 ] Division of Translational Medicine, Baqiyatallah Hospital Baqiyatallah University of Medical Sciences Tehran Iran
                [ 7 ] Department of Human Anatomy and Cell Sciences Rady Faculty of Health Sciences Max Rady College of Medicine University of Manitoba Winnipeg Canada
                [ 8 ] Department of Hypertension Medical University of Lodz Lodz Poland
                [ 9 ] Polish Mother's Memorial Hospital Research Institute (PMMHRI) Lodz Poland
                Author notes
                [*] [* ] Correspondence

                Seyed Mohammad Nabavi, Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran

                Email: Nabavi208@ 123456gmail.com

                Author information
                https://orcid.org/0000-0001-6743-2244
                https://orcid.org/0000-0001-5828-1325
                https://orcid.org/0000-0001-5716-8101
                https://orcid.org/0000-0003-0898-7973
                https://orcid.org/0000-0002-4945-9651
                https://orcid.org/0000-0001-5948-508X
                https://orcid.org/0000-0001-6690-6874
                https://orcid.org/0000-0001-8859-5675
                Article
                JCMM15652
                10.1111/jcmm.15652
                7405483
                32677763
                f5e1ed59-f56a-4883-aa96-f1b2c9b669b9
                © 2020 The Authors. Journal of Cellular and Molecular Medicine published by Foundation for Cellular and Molecular Medicine and John Wiley & Sons Ltd

                This is an open access article under the terms of the http://creativecommons.org/licenses/by/4.0/ License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited.

                History
                : 02 June 2020
                : 30 June 2020
                Page count
                Figures: 1, Tables: 0, Pages: 3, Words: 5295
                Categories
                Letter to the Editor
                Letter to the Editors
                Custom metadata
                2.0
                corrected-proof
                Converter:WILEY_ML3GV2_TO_JATSPMC version:5.8.6 mode:remove_FC converted:05.08.2020

                Molecular medicine
                Molecular medicine

                Comments

                Comment on this article