key: cord-0704363-89zyu8bl authors: Zhang, Xue; Wu, Kailang; Yue, Xin; Zhu, Ying; Wu, Jianguo title: Inhibition of SARS-CoV Gene Expression by Adenovirus-Delivered Small Hairpin RNA date: 2006-11-30 journal: Intervirology DOI: 10.1159/000097391 sha: ccb32c4e7d73d0091afeafc7df3c9c6e1439be62 doc_id: 704363 cord_uid: 89zyu8bl OBJECTIVE: Severe acute respiratory syndrome (SARS) is a highly contagious and lethal disease caused by a new type of coronavirus, SARS-associated coronavirus (SARS-CoV). Currently, there is no efficient treatment and prevention for this disease. We constructed recombinant adenoviral vectors that can express shRNAs, which inhibited the expression of SARS-CoV genes effectively in mammalian cells. METHODS: In this study, we designed several plasmids that express small hairpin RNA molecules (shRNA) specifically targeting to the genes encoding for the SARS-CoV nucleocapsid (N) protein and envelope (E) protein, respectively. Effective shRNA molecules to the viral genes were screened and identified, and then constructed into adenovirus vectors. The effects of adenovirus-delivered small hairpin RNA on SARS-CoV gene expression were determined by RT-PCR, Western blot, and luciferase activity assays. RESULTS: The levels of viral mRNAs and viral proteins of the targets were significantly decreased or completely inhibited in cell lines after being infected with the recombinant adenoviruses that expressed specific shRNA molecules. CONCLUSIONS: Since many cell types can be efficiently infected by adenovirus, recombinant adenoviruses could serve as an alternative powerful tool for shRNA delivery and for gene suppression, especially when the targeted cells are resistant to transfection by DNA or RNA. With availability of high titers of adenoviruses and uniform and rapid infection, this approach would have foreseeable wide applications both in experimental biology and molecular medicine. Between the end of 2002 and June 2003, the severe epidemic disease called severe acute respiratory syndrome (SARS) broke out in China and more than 30 other countries. It was known that SARS coronavirus (SARS-CoV) is a novel human coronavirus and responsible for SARS infection [1] [2] [3] [4] . SARS-CoV is an enveloped positive-stranded RNA virus, and its genome is composed of about 29,700 nucleotides [5, 6] . The SARS genome encodes 23 putative proteins that are typical of coronavirus such as 5 -replicase (rep), spike (S), membrane (M), envelope (E), and nucleocapsid (N)-3 [7] . The N protein of SARS-CoV is 422 amino acids long, sharing only 20-30% homology with the N proteins of other coronaviruses [5, 6] . Previous studies indicated that the N proteins of other coronaviruses have a variety of functional activities [8] , including participation in transcription of the viral genome, the formation of viral core, and packaging viral RNA [9, 10] . However, no similar functional roles of SARS-CoV have been reported except that SARS-CoV N may selectively activate AP-1 pathway [11] . Furthermore, SARS-CoV N protein is capable of self-association through a C-terminal 209-amino-acid interaction domain [12, 13] . The E protein is another important structural protein of SARS-CoV. More recently, it was shown to play a major role in coronavirus assembly [14, 15] . RNA interference (RNAi) is an ancient evolutionarily conserved process [16] [17] [18] [19] . Ever since synthetic 21-23 nucleotide short interfering RNA (siRNA) was shown to induce efficient RNAi in mammalian cells, siRNA has been routinely used in gene silencing by transfection of chemically synthesized siRNA. Specific inhibition of cellular mRNA by RNAi can be triggered in mammalian cells by the introduction of siRNA. Currently, RNAi technology is reported as an ideal tool to inhibit infectious virus replication in host cells, including poliovirus [20] , HIV-1 [21, 22] , flock house virus, Rous sarcoma virus [23] , dengue virus [24] , hepatitis C virus [25] replicons, influenza virus [26] , hepatitis B virus [27, 28] and SARS-CoV [29, 30] . Adenovirus vectors have been widely used for the expression of trans-genes under both clinical and experimental conditions [31] . Adenovirus has several unique features, such as ability to infect both dividing and nondividing cells, lack of cell-mediated immune response against the vector, ability to integrate into a host chromosome or persist episomally. In this report, we describe the construction of a recombinant adenoviral vector that can introduce expression of shRNAs intracellularly and inhibit the expression of SARS-CoV genes effectively. The following two pairs of primers were used to amplify N and E gene from the full-length cDNA of SARS-CoV (SARS-CoV WHU, GenBank accession No. AY394850), respectively. N gene: P01: 5 -AGCTGGATCCATGTCTGATAATGGACCCCAAT-CAAAC-3 (sense) P02: 5 -AGCTGAATTCCATCATGAGTGTTTATGCCTGA-GT-3 (antisense) E gene: P03: 5 -AGCTGGATCCATGTACTCATTCGTTTCGGAAG-AAAC-3 (sense) P04: 5 -AGCTGAATTCTTAGTTCGTTTAGACCAGAAGA-TC-3 (antisense) The PCR products were cloned into BamHI and EcoRI sites of pCMV-tag2B (Stratagene) to generate pCMV-N and pCMV-E. We also used the following two pairs of primers to amplify the fragments carrying part of N or part of E gene: The PCR products were cloned into Sal I and Bgl II sites of plucF [32] to generate plasmid plucF-N ( fig. 1 a) and plucF-E ( fig. 1 b) , in which the N or E gene was fused in frame with the luciferase gene and the expression of the fusion gene was driven by the CMV promoter. All constructs were confirmed through DNA sequencing. Target sequences for shRNA were selected according to the web-based criteria and further analyzed by BLAST research to avoid significant homology with endogenous cellular genes. To construct single shRNA expression vector, two 64-nt primers, each containing a 19-nt target sequence in the sense and antisense strand from different regions of the N gene or E gene ( fig. 1 c) , were synthesized from Invitrogen. Four regions of N and four regions of E gene were selected respectively as the target sites of shRNA in this study. The sequences and locations are listed as following For each shRNA, sense and antisense oligonucleotides of selfcomplementary hairpin sequences, which contain cohesive ends for Bam HI and Hin dIII at the 5 -and 3 -ends, were synthesized and annealed by heating at 95 ° C for 10 min and slowly cooled down to room temperature. Then, eight annealed oligonucleotides were cloned into the Bam HI and Hin dIII sites of pSilencer-2.1-U6 plasmid (Amibion) according to the manufacturer's instructions (pN1shRNA, pN2shRNA, pN3shRNA, pN4shRNA, pE1shRNA, pE2shRNA, pE3shRNA, pE4shRNA). The sequences of the constructs were confirmed by DNA sequencing. To enhance the delivery efficiency of shRNAs, recombinant adenoviruses were generated as following steps: pN3shRNA, pE4shRNA and pCtrl plasmid were digested with Eco RI and Hin dIII to release small fragments containing U6 promoter and targeting sequences, which were then ligated into pAdTrack at EcoR I and Hin dIII sites to generate recombinant plasmid ADN3, ADE4 and ADCtrl, respectively. The recombinant pAdTracks were linearized with Pme I and transformed into BJ5183 cells which carry the pAdEasy-1 plasmid. Positive clones were selected and confirmed by PacI digestion. Plasmids from correct clones were amplified by transforming into DH5 ␣ cells. The resulting adenoviral DNAs were linearized with Pac I and transfected into 293T cells in a six-well plate using Sofast TM transfection reagent (Xiamen Sunma Biotechnology Co. Ltd, PR China) to produce recombinant adenoviruses ADN3, ADE4 and ADCtrl, respectively. Eight days after transfection, the recombinant virus was amplified and titered according to the manufacturer's instructions. 293T and COS7 cells were maintained in Dulbecco's modified Eagle medium (Gibco/BRL) supplemented with 100 units/ml penicillin, 100 g/ml streptomycin and 10% heat-inactivated fetal bovine serum at 37 ° C under 5% CO 2 . Cells were seeded onto plates at a density of 1.0 ! 10 5 per 24-well plate or 6-well plate and grown to confluence reaching approximately 60% at the time of transfection. Cells were then transfected with 0.1 or 0.4 g of plasmid which contains N or E gene together with 0.45 or 1.2 g shRNA expression plasmids, using Sofast TM transfection reagent. The cells were harvested 48 h after transfection. For adenovirus infection, cells were incubated with recombinant virus at MOI of 10-20 at 37 ° C. After adsorption for 1-2 h, fresh growth medium was added and cells were placed in the incubator for an additional 2-3 days. 293T and COS7 cells were co-transfected with reporter plasmids and shRNA expression plasmids. Cells were washed with PBS and lysed with luciferase cell culture lysis reagent (Promega). 10 l of the cell lysates and 100 l of luciferase assay substrate (Promega) were mixed and fluorescence intensity was detected by the luminometer (Turner, TD20/20). Assays were performed in triplicate, and expressed as means 8 SD relative to vector control as 100%. 293T and COS7 cells were co-transfected with pCMV-N or pCMV-E plasmid and shRNA expression plasmids. The total RNA was then extracted from transfected cells by Trizol Reagent (Invitrogen) according to the method described in the manufacturer's manual. Reverse transcriptions were performed with total RNA as the template. Specific mRNA was amplified by RT-PCR using specific primers for the N (P01 and P02) and E (P03 and P04) gene. All product bands were visualized and quantified using Kodak 1D Imaging Systems and relative mRNA expression was estimated by normalization with ␤ -actin control. The signal intensity of the control vector was considered 100%. Cells were harvested at the indicated time points, washed once with cold PBS and lysed in lysis buffer (PBS, 0.01% EDTA, 1% Triton) containing protease inhibitor cocktail (Roche, USA). Total proteins were separated by 12% SDS-polyacrylamide gel and transferred onto a nylon membrane and incubated with anti-flag (Sigma, USA) monoclonal antibodies, followed by incubation with peroxidase-conjugated immunopure goat antimouse secondary antibody (Pierce, USA). The bands were visualized using the SuperSignal West Pico Chemiluminescent Substrate (Pierce, USA). Then, densitometric scanning of the X-ray film was performed and protein expression level was determined after normalization with ␤ -actin using Kodak 1D Imaging Systems. The signal intensity of the control vector was considered 100%. To efficiently screen shRNA molecules, selected targeting DNA sequences were fused in frame with that of luciferase gene, in which luciferase activity was supposed to represent the level of N or E mRNA expression. Cells were co-transfected with pLucF-N or pLucF-E and eight single shRNA expression vectors, respectively. Luciferase activities were then determined from both transfected 293T cells and COS-7 cells. Result showed that N1shRNA, N3shRNA, N4shRNA and E4shRNA strongly inhibited luciferase activities by 76, 45, 69 and 79%, respectively, comparing to that of random sequence-scrambled shRNA control ( fig. 2 a, b) in 293T. Similar results were obtained in COS-7 cells ( fig. 2 a, b) . These results indicated that the four shRNAs can effectively degrade the fusion mRNA of N-luciferase or E-luciferase. To determine the effects of shRNA on RNA expression level, total RNAs isolated from transfected cells were used as templates to synthesize cDNA. Specific mRNA was amplified by RT-PCR using gene-specific primers for N ( fig. 3 a, c) , or E ( fig. 3 b, d) , and ␤ -actin was used as an endogenous control. PCR products were detected on agarose gel electrophoresis, visualized under UV light and quantified using Kodak 1D Imaging Systems. Relative fig. 3 a, lane 3) , N3shRNA ( fig. 3 a, lane 4), N4shRNA ( fig. 3 a, lane 5) in 293T, respectively. The levels of E mRNA were also decreased by 89% after the treatment of E4shRNA ( fig. 3 b, lane 3) , but not by that of Silencer-2.1-U6 vector ( fig. 3 a, lane 2, fig. 3 b, lane 2) or untreated cells ( fig. 3 a, lane 1, fig. 3 b, lane 1) . Similar results were also obtained in COS-7 cells under the same conditions of shRNA treatments ( fig. 3 c, d) . The levels of N mRNA were dramatically reduced by 81, 65, and 75% in COS-7 cells after the treatment of N1shRNA ( fig. 3 c, lane 2), N3shRNA ( fig. 3 c, lane 3) and N4shRNA ( fig. 3 c, lane 4) , respectively. The levels of E mRNA in COS-7 cells were also reduced by 91% after the treatment of E4shRNA ( fig. 3 d, lane 3) , but To determine effects of shRNAs on viral RNA expression, total RNAs isolated from transfected cells were used as templates to synthesize cDNA. Specific mRNA was amplified by RT-PCR using specific primers for the N gene ( fig. 3a, c, e) , the E gene-specific primers ( fig. 3b, d, f) , and the ␤ -actin gene (as an endogenous control). PCR products were detected on agarose gel electrophoresis and visualized under UV light. not by that of pSilencer-2.1-U6 vector ( fig. 3 c, lane 1, fig. 3 d, lane 2) or untreated cells ( fig. 3 c, lane 5, fig. 3 d, lane 1) . To determine whether shRNAs specifically reduce N or E protein expression, Western blot was applied to analyze the levels of flag-tagged N and E protein in 293T ( fig. 4 a, b) and COS-7 ( fig. 4 c, d) cells using anti-flag antibody. The results showed that the decrease level of pro-teins was correlated to the RT-PCR assays in both cell lines. To enhance the effectiveness of shRNA delivery and investigate the therapeutic feasibility of shRNA, we generated shRNA-expressing recombinant adenovirus ADN3, ADE4, ADN1, and ADN4 (data not shown), which target different locations of both N and E gene. We also generated a negative control which expresses random se- . 2 c, d) . Transduction of the shRNA-expressing adenoviruses dose-dependently reduced the expression of their target genes. The target mRNAs and proteins were analyzed by RT-PCR ( fig. 3 e, f) and Western blot analysis ( fig. 4 e, f) in parallel, results indicated that further reduction was observed after transduction of shRNA-expressing adenoviruses comparing to that transfection of shRNA-expressing plasmids. SARS is a newly emerging infectious disease. It is usually characterized by fever, dry cough, myalgia, and mild sore throat, which progresses to a typical pneumonia. Over 8,000 SARS cases and 770 SARS-related deaths were reported to WHO from over 26 countries around the world (http://www.who.int/csr/sars/country/en). SARS is dangerous for its high morbidity and mortality rates. SARS is caused by a newly identified virus within the family Coronaviridae . This virus has been designated as the SARS coronavirus (SARS-CoV). At present, several potential SARS therapies are under development, such as vaccines [33, 34] and interferons [35, 36] . The use of RNAi technology in the therapy of SARS would be advantageous in its specificity for the target gene with minimized side effects. It has the potential to prove useful as novel treatments for virus-induced diseases such as HIV-1, although the problem of how to efficiently deliver small interfering RNA expression vectors, or the small interfering RNAs themselves to target host cells, remains to be resolved. The interference of SARS-CoV genes expression induced by the use of siRNA duplexes is transient. And because of the high sequence specificity of siRNA-mediated RNA degradation, antiviral efficacy of siRNA directed to viral genome will be largely limited by emergence of escape variants resistant to siRNA due to high mutation rates of virus, especially RNA viruses such as poliovirus, hepatitis C virus and SARS-CoV. The problem can be cir-cumvented by targeting multiple essential genes of the viruses and increasing the delivery efficiency of siRNAs. Among all the approaches the most perhaps desirable one is choosing the adenoviral vector expression system. Our results have demonstrated that inhibition of SARS-CoV genes can be achieved by adenovirus-delivered siRNAs. In our studies, we have examined gene inhibition by the adenoviral system in COS-7 cells. It is possible that this approach may be applicable to other types of cells as well, especially primary cells that have only a limited lifespan and be resistant to transfection. The high efficiency of adenoviral infection of a variety of host cell types may render this approach a convenient method to achieve gene inhibition through cellular expression of shRNAs. Also it provides the possibility that we apply this technique in animal model in our further investigations. In conclusion, we developed a simple shRNA delivery strategy by combination of well-defined U6 promoter and conventional pAdEasy-1 adenovirus system. Our results demonstrate significant inhibition of viral genes can be achieved in mammalian cells. Since many cell types can be efficiently infected by adenovirus, recombinant adenoviruses could serve as an alternative powerful tool for delivery of shRNA and for gene suppression, especially when the targeted cells are resistant to transfection by DNA or RNA. In addition, our findings also provide solid evidence for development of an anti-SARS-CoV therapy using adenovirus-delivered small hairpin RNA, which may enhance anti-SARS-CoV efficacy and overcome drug resistance. With availability of high titers of adenoviruses and uniform and rapid infection, this technology will have a foreseeable wide application both in experimental biology and molecular medicine. Identification of a novel coronavirus in patients with severe acute respiratory syndrome Coronavirus as a possible cause of severe acute respiratory syndrome Clinical progression and viral load in a community outbreak of coronavirus-associated SARS pneumonia: a prospective study A novel coronavirus associated with severe acute respiratory syndrome Characterization of a novel coronavirus associated with severe acute respiratory syndrome The genome sequence of the SARS-associated coronavirus Philadelphia, Lippincott Williams & Wilkins Ribonucleoprotein-like structures from coronavirus particles The molecular biology of coronaviruses The nucleocapsid protein of coronavirus mouse hepatitis virus interacts with the cellular heterogeneous nuclear ribonucleoprotein A1 in vitro and in vivo Activation of AP-1 signal transduction pathway by SARS coronavirus nucleocapsid protein A highly conserved region of the Sendai virus nucleocapsid protein contributes to the NP-NP binding domain Hantavirus nucleocapsid protein coiled-coil domains The production of recombinant infectious DI particles of a murine coronavirus in the absence of helper virus Rottier PJ: Nucleocapsid-independent assembly of coronavirus-like particles by co-expression of viral envelope protein genes Fire A: RNA-triggered gene silencing Interfering with gene expression RNAi: Nature abhors a double-strand RNA interference Potent and specific inhibition of human immunodeficiency virus type 1 replication by RNA interference Modulation of HIV-1 replication by RNA interference Expression of small interfering RNAs targeted against HIV-1 rev transcripts in human cells Bushman FD: Inhibition of retroviral pathogenesis by RNA interference RNA silencing of dengue virus type 2 replication in transformed C6/36 mosquito cells transcribing an inverted-repeat RNA derived from the virus genome Interference of hepatitis C virus RNA replication by short interfering RNAs RNA interference of influenza virus production by directly targeting mRNA for degradation and indirectly inhibiting all viral RNA transcription Short interfering RNA-directed inhibition of hepatitis B virus replication Effective expression of small interfering RNA in human cells Attenuation of SARS coronavirus by a short hairpin RNA expression plasmid targeting RNA-dependent RNA polymerase Small interfering RNA inhibits SARS-CoV nucleocapsid gene expression in cultured cells and mouse muscles Use of adeno-associated viral vector for delivery of small interfering RNA Inhibition of hepatitis B virus gene expression by single and dual small interfering RNA treatment Effects of a SARS-associated coronavirus vaccine in monkeys Potent neutralization of severe acute respiratory syndrome coronavirus by a human mAb to S1 protein that blocks receptor association Treatment of SARS with human interferons Pegylated interferon-␣ protects type 1 pneumocytes against SARS coronavirus infection in macaques This work was supported by research grants from the National Natural Science Foundation of China to J. Wu