Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
BMC Vet Res ; 17(1): 217, 2021 Jun 12.
Article in English | MEDLINE | ID: mdl-34118903

ABSTRACT

BACKGROUND: Worldwide, Porcine Reproductive and Respiratory Syndrome (PRRS) is among the diseases that cause the highest economic impact in modern pig production. PRRS was first detected in Costa Rica in 1996 and has since then severely affected the local swine industry. Studies of the molecular characterization of circulating strains, correlation with clinical records, and associations with pathogens associated with Porcine Respiratory Disease Complex (PRDC) have not been done in Costa Rica. RESULTS: Sequencing and phylogenetic analysis of ORF5 proved that PRRSV-2 was the only species detected in all locations analyzed. These sequences were grouped into three clusters. When comparing samples from San Jose, Alejuela, and Puntarenas to historical isolates of the previously described lineages (1 to 9), it has been shown that these were closely related to each other and belonged to Lineage 5, along with the samples from Heredia. Intriguingly, samples from Cartago clustered in a separate clade, phylogenetically related to Lineage 1. Epitope analysis conducted on the GP5 sequence of field isolates from Costa Rica revealed seven peptides with at least 80% amino acid sequence identity with previously described and experimentally validated immunogenic regions. Previously described epitopes A, B, and C, were detected in the Santa Barbara-Heredia isolate. CONCLUSIONS: Our data suggest that the virus has three distinct origins or introductions to the country. Future studies will elucidate how recently introduced vaccines will shape the evolutionary change of circulating field strains.


Subject(s)
Open Reading Frames/genetics , Porcine respiratory and reproductive syndrome virus/classification , Porcine respiratory and reproductive syndrome virus/genetics , Amino Acid Sequence , Animals , Costa Rica/epidemiology , Epitopes/analysis , Phylogeny , Porcine Reproductive and Respiratory Syndrome/epidemiology , Porcine Reproductive and Respiratory Syndrome/virology , Swine
2.
Trop Anim Health Prod ; 53(4): 441, 2021 Aug 18.
Article in English | MEDLINE | ID: mdl-34406521

ABSTRACT

Porcine reproductive and respiratory syndrome virus (PRRSV) causes significant economic losses to the swine industry worldwide. Little is known regarding the epidemiology of this infection in tropical countries. To address this problem in Costa Rica, a seroepidemiological study was carried out in two phases. In the first phase, a pilot study was conducted in nine farms with the clinical diagnosis of PRRSV. In total, 265 pig serum samples were collected from animals ranging in age from 1 to 15 weeks of age. This study aimed to establish the duration of maternal immunity in piglets, to identify the period of viremia, and to determine when seroconversion occurs. In the second phase, a cross-sectional serology study was performed on a representative sample of the Costa Rican national herds in the second phase. The twenty-five selected farms represent all provinces and were classified according to herd size (100 to 2000 sows). In each farm, pigs aged 8, 10, and 12 weeks were sampled, as well as gilts based on the pilot study. In total 1281 pigs were sampled across all 25 farms. The aim of the cross-sectional study was to quantify the seroprevalence of PRRSV in Costa Rican pig farms and to describe its geographical distribution in this tropical country. The prevalence of positive farms was 44% (11/25), and these farms were located in six of the seven provinces of Costa Rica. Overall, 58% (344/596) of the pigs were seropositive to PRRSV. The age of the pigs and the ecozone where farms were located were significantly related with PRRSV seroprevalence in animals and herds, respectively.


Subject(s)
Porcine Reproductive and Respiratory Syndrome , Porcine respiratory and reproductive syndrome virus , Swine Diseases , Animals , Antibodies, Viral , Costa Rica/epidemiology , Cross-Sectional Studies , Farms , Female , Pilot Projects , Porcine Reproductive and Respiratory Syndrome/epidemiology , Seroepidemiologic Studies , Swine
3.
J Virol ; 90(19): 8809-21, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27466421

ABSTRACT

UNLABELLED: Foot-and-mouth disease (FMD) remains one of the most devastating livestock diseases around the world. Several serotype-specific vaccine formulations exist, but they require about 5 to 7 days to induce protective immunity. Our previous studies have shown that a constitutively active fusion protein of porcine interferon (IFN) regulatory factors (IRF) 7 and 3 [IRF7/3(5D)] strongly induced type I IFN and antiviral genes in vitro and prevented mortality in an FMD mouse model when delivered with a replication-defective adenoviral vector [Ad5-poIRF7/3(5D)]. Here, we demonstrate that pigs treated with 10(8), 10(9), or 10(10) PFU of Ad5-poIRF7/3(5D) 24 h before FMDV challenge were fully protected from FMD clinical signs and did not develop viremia, virus shedding or antibodies against FMDV nonstructural proteins. Pigs treated with Ad5-poIRF7/3(5D) had higher levels of IFN and antiviral activity in serum, and upregulated expression of several IFN-stimulated genes in peripheral blood mononuclear cells, compared to pigs treated with Ad5-Blue vector control. Importantly, treatment of porcine cultured cells with Ad5-poIRF7/3(5D) inhibited the replication of all 7 FMDV serotypes. In vitro experiments using cultured embryonic fibroblasts derived from IFN receptor knockout mice suggested that the antiviral response induced by Ad5-poIRF7/3(5D) was dependent on type I and III IFN pathways; however, experiments with mice demonstrated that a functional type I IFN pathway mediates Ad5-poIRF7/3(5D) protection conferred in vivo Our studies demonstrate that inoculation with Ad5-poIRF7/3(5D) completely protects swine against FMD by inducing a strong type I IFN response and highlights its potential application to rapidly and effectively prevent FMDV replication and dissemination. IMPORTANCE: Foot-and-mouth disease virus (FMDV) causes a fast-spreading disease that affects farm animals, with economically and socially devastating consequences. Our study shows that inoculation with a constitutively active transcription factor, namely, a fusion protein of porcine interferon (IFN) regulatory factors (IRF) 7 and 3 delivered by an adenovirus vector [Ad5-poIRF7/3(5D)], is a new effective treatment to prevent FMD in swine. Animals pretreated with Ad5-poIRF7/3(5D) 1 day before being exposed to FMDV were completely protected from viral replication and clinical disease. It is noteworthy that the doses of Ad5-poIRF7/3(5D) required for protection are lower than those previously reported for similar approaches using Ad5 vectors delivering type I, II, or III IFN, suggesting that this novel strategy would be economically appealing to counteract FMD. Our results also indicate that a dynamic interplay among different components of pigs' innate immune defenses allows potent antiviral effects after Ad5-poIF7/3(5D) administration.


Subject(s)
Foot-and-Mouth Disease Virus/immunology , Foot-and-Mouth Disease/prevention & control , Interferon Regulatory Factor-3/metabolism , Interferon Regulatory Factor-7/metabolism , Recombinant Fusion Proteins/metabolism , Swine Diseases/prevention & control , Adenoviridae/genetics , Animals , Cell Line , Drug Carriers/administration & dosage , Foot-and-Mouth Disease/pathology , Foot-and-Mouth Disease/virology , Foot-and-Mouth Disease Virus/physiology , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-7/genetics , Interferon Type I/metabolism , Interleukins/metabolism , Mice , Mice, Knockout , Recombinant Fusion Proteins/genetics , Survival Analysis , Swine , Swine Diseases/virology , Transduction, Genetic , Treatment Outcome , Virus Replication
4.
J Virol ; 88(19): 11140-53, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25031341

ABSTRACT

UNLABELLED: Several studies have demonstrated that the delivery of type I, II, or III interferons (IFNs) by inoculation of a replication-defective human adenovirus 5 (Ad5) vector expressing IFNs can effectively control foot-and-mouth disease (FMD) in cattle and swine during experimental infections. However, relatively high doses are required to achieve protection. In this study, we identified the functional properties of a porcine fusion protein, poIRF7/3(5D), as a biotherapeutic and enhancer of IFN activity against FMD virus (FMDV). We showed that poIRF7/3(5D) is a potent inducer of type I IFNs, including alpha IFN (IFN-α), IFN-ß, and IFN-ω but not type III IFN (interleukin-28B), without inducing cytotoxicity. Expression of poIRF7/3(5D) significantly and steadily reduced FMDV titers by up to 6 log10 units in swine and bovine cell lines. Treatment with an IFN receptor inhibitor (B18R) combined with an anti-IFN-α antibody neutralized the antiviral activity in the supernatants of cells transduced with an Ad5 vector expressing poIRF7/3(5D) [Ad5-poIRF7/3(5D)]. However, several transcripts with known antiviral function, including type I IFNs, were still highly upregulated (range of increase, 8-fold to over 500-fold) by poIRF7/3(5D) in the presence of B18R. Furthermore, the sera of mice treated with Ad5-poIRF7/3(5D) showed antiviral activity that was associated with the induction of high levels of IFN-α and resulted in complete protection against FMDV challenge at 6, 24, or 48 h posttreatment. This study highlights for the first time the antiviral potential of Ad5-poIRF7/3(5D) in vitro and in vivo against FMDV. IMPORTANCE: FMD remains one of the most devastating diseases that affect livestock worldwide. Effective vaccine formulations are available but are serotype specific and require approximately 7 days before they are able to elicit protective immunity. We have shown that vector-delivered IFN is an option to protect animals against many FMDV serotypes as soon as 24 h and for about 4 days postadministration. Here we demonstrate that delivery of a constitutively active transcription factor that induces the production of endogenous IFNs and potentially other antiviral genes is a viable strategy to protect against FMD.


Subject(s)
Adenoviridae/immunology , Foot-and-Mouth Disease Virus/immunology , Foot-and-Mouth Disease/prevention & control , Interferon Regulatory Factor-7/immunology , Recombinant Fusion Proteins/immunology , Viral Vaccines/immunology , Adenoviridae/genetics , Animals , Cattle , Cell Line , Foot-and-Mouth Disease/immunology , Foot-and-Mouth Disease/virology , Foot-and-Mouth Disease Virus/genetics , Gene Expression/immunology , Genetic Vectors , Humans , Interferon Inducers/antagonists & inhibitors , Interferon Inducers/immunology , Interferon Regulatory Factor-7/antagonists & inhibitors , Interferon Regulatory Factor-7/genetics , Interferon Type I/antagonists & inhibitors , Interferon Type I/biosynthesis , Interferon Type I/immunology , Mice , Recombinant Fusion Proteins/genetics , Swine , Vaccination , Vaccines, Synthetic , Viral Proteins/pharmacology , Viral Vaccines/administration & dosage , Virus Replication/immunology
5.
Vet Sci ; 9(6)2022 May 28.
Article in English | MEDLINE | ID: mdl-35737310

ABSTRACT

Venezuelan Equine Encephalitis virus (VEEV) is an arboviral pathogen in tropical America that causes lethal encephalitis in horses and humans. VEEV is classified into six subtypes (I to VI). Subtype I viruses are divided into epizootic (IAB and IC) and endemic strains (ID and IE) that can produce outbreaks or sporadic diseases, respectively. The objective of this study was to reconstruct the phylogeny and the molecular clock of sequences of VEEV subtype I complex and identify mutations within sequences belonging to epizootic or enzootic subtypes focusing on a sequence isolated from a mare in Costa Rica. Bayesian phylogeny of the VEEV subtype I complex tree with 110 VEEV complete genomes was analyzed. Evidence of positive selection was evaluated with Datamonkey server algorithms. The putative effects of mutations on the 3D protein structure in the Costa Rica sequence were evaluated. The phylogenetic analysis showed that Subtype IE-VEEV diverged earlier than other subtypes, Costa Rican VEEV-IE ancestors came from Nicaragua in 1963 and Guatemala in 1907. Among the observed non-synonymous mutations, only 17 amino acids changed lateral chain groups. Fourteen mutations located in the NSP3, E1, and E2 genes are unique in this sequence, highlighting the importance of E1-E2 genes in VEEV evolution.

6.
Virusdisease ; 32(2): 347-353, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33898651

ABSTRACT

We describe the first whole-genome sequence of a GA13-like isolate of avian infectious bronchitis virus CK/CR/1160/16 (MN757859), obtained in 2016 in the province of Alajuela, Costa Rica. This virus caused an outbreak with great economic impact to the local poultry industry. The genome sequence is 27 696 bp in length, with the following genome organization 5'-UTR-Pol-S-3a-3b-E-4b-4c-M-5a-5b-N-6b-3'-UTR. The complete genome sequence has the highest sequence identity (94.03%) with DMV/1639/GA9977/2019 (MK878536) from Georgia, USA, and the lowest identity (86.03%) with ck/CH/LHLJ/08-6 (KX252788), from China. Analysis of the S1 subunit indicates that the Costa Rican isolate belongs to genotype I, lineage 17 (GI-17) and displays 96.89% identity with the S1 subunit of Ga-13/14255/14 (KM087780) (USA). Possible recombination events in genes S, E, M, 4b y 4c were detected, with Massachusetts, Connecticut, Arkansas and MA5 as potential parental types. This study highlights the importance of the epidemiological and molecular surveillance of avian infectious bronchitis.

7.
Microbiol Resour Announc ; 9(1)2020 Jan 02.
Article in English | MEDLINE | ID: mdl-31896625

ABSTRACT

We report the draft genome sequence of the multidrug-resistant Escherichia coli strain PTA A1517-5, isolated from a wastewater treatment plant in Costa Rica. The genome consists of 4,927,375 bp with a GC content of 50.57% and a total of 4,853 genes. This strain harbors bla CTX-M-115, bla CMY-2, aminoglycoside, tetracycline, and sulfonamide resistance genes.

8.
Microbiol Resour Announc ; 8(36)2019 Sep 05.
Article in English | MEDLINE | ID: mdl-31488528

ABSTRACT

The first complete coding sequence of the Venezuelan equine encephalitis virus IE, isolated from a Costa Rican mare with severe encephalitis, was confirmed by histological and viral whole-genome analyses. The isolated virus grouped in the Pacific cluster.

9.
PLoS One ; 13(2): e0190977, 2018.
Article in English | MEDLINE | ID: mdl-29390015

ABSTRACT

Foot-and-mouth disease (FMD) is a severe infection caused by a picornavirus that affects livestock and wildlife. Persistence in ruminants is a well-documented feature of Foot-and-mouth disease virus (FMDV) pathogenesis and a major concern for disease control. Persistently infected animals harbor virus for extended periods, providing a unique opportunity to study within-host virus evolution. This study investigated the genetic dynamics of FMDV during persistent infections of naturally infected Asian buffalo. Using next-generation sequencing (NGS) we obtained 21 near complete FMDV genome sequences from 12 sub-clinically infected buffalo over a period of one year. Four animals yielded only one virus isolate and one yielded two isolates of different serotype suggesting a serial infection. Seven persistently infected animals yielded more than one virus of the same serotype showing a long-term intra-host viral genetic divergence at the consensus level of less than 2.5%. Quasi-species analysis showed few nucleotide variants and non-synonymous substitutions of progeny virus despite intra-host persistence of up to 152 days. Phylogenetic analyses of serotype Asia-1 VP1 sequences clustered all viruses from persistent animals with Group VII viruses circulating in Pakistan in 2011, but distinct from those circulating on 2008-2009. Furthermore, signature amino acid (aa) substitutions were found in the antigenically relevant VP1 of persistent viruses compared with viruses from 2008-2009. Intra-host purifying selective pressure was observed, with few codons in structural proteins undergoing positive selection. However, FMD persistent viruses did not show a clear pattern of antigenic selection. Our findings provide insight into the evolutionary dynamics of FMDV populations within naturally occurring subclinical and persistent infections that may have implications to vaccination strategies in the region.


Subject(s)
Buffaloes , Foot-and-Mouth Disease Virus/genetics , Foot-and-Mouth Disease/virology , Genome, Viral , Amino Acid Sequence , Animals , Cattle , Foot-and-Mouth Disease/epidemiology , Foot-and-Mouth Disease/transmission , Foot-and-Mouth Disease Virus/isolation & purification , Foot-and-Mouth Disease Virus/pathogenicity , Phylogeny , RNA, Viral/genetics , Sequence Alignment
10.
Antiviral Res ; 125: 8-13, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26592975

ABSTRACT

Type I interferons (IFNs) are key mediators of the innate antiviral response in mammalian cells. Elongation initiation factor 4E binding proteins (4E-BPs) are translational controllers of interferon regulatory factor 7 (IRF-7), the "master regulator" of IFN transcription. Previous studies have suggested that mouse cells depleted of 4E-BPs are more sensitive to IFNß treatment and had lower viral loads as compared to wild type (WT) cells. However, such approach has not been tested as an antiviral strategy in livestock species. In this study, we tested the antiviral activity of porcine cells depleted of 4E-BP1 by a Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/CRISPR-associated protein-9 nuclease (Cas9) genome engineering system. We found that 4E-BP1 knockout (KO) porcine cells had increased expression of IFNα and ß, IFN stimulated genes, and significant reduction in vesicular stomatitis virus titer as compare to WT cells. No phenotypical changes associated with CRISPR/Cas9 manipulation were observed in 4E-BP1 KO cells. This work highlights the use of the CRISPR/Cas9 system to enhance the antiviral response in porcine cells.


Subject(s)
Antiviral Agents/pharmacology , CRISPR-Associated Proteins/metabolism , CRISPR-Cas Systems , Eukaryotic Initiation Factor-4E/deficiency , Animals , Cell Line , Clustered Regularly Interspaced Short Palindromic Repeats , Eukaryotic Initiation Factor-4E/genetics , Eukaryotic Initiation Factor-4E/metabolism , Gene Expression Regulation , Gene Knockout Techniques , Interferon-alpha/biosynthesis , Interferon-alpha/genetics , Interferon-beta/biosynthesis , Interferon-beta/genetics , Swine , Vesicular stomatitis Indiana virus
11.
J Interferon Cytokine Res ; 36(1): 37-47, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26418342

ABSTRACT

The genetic regulatory network controlling the innate immune system is well understood in many species. However, the role of the epigenetic mechanisms underlying the expression of immunoregulatory genes is less clear, especially in livestock species. Histone H3 lysine 9 dimethylation (H3K9me2) is an epigenetic modification associated with transcriptional silencing within the euchromatin regions. Euchromatic histone-lysine N-methyltransferase 2 (EHMT2; also known as G9a) is a crucial enzyme responsible for regulating the dynamics of this epigenetic modification. It has been shown that histone modifications play a role in regulating type I interferon (IFN) response. In the present study, we investigated the role of EHMT2 in the epigenetic regulation of bovine antiviral innate immunity and explored its therapeutic potential against viral infections. We evaluated the effects of pharmacological and RNAi-mediated inhibition of EHMT2 on the transcription of IFN-ß and other IFN-inducible antiviral genes, as well as its effect on foot-and-mouth disease virus (FMDV) and vesicular stomatitis virus (VSV) replication in bovine cells. We show that treatment of primary bovine cells with the synthetic EHMT2 inhibitor (UNC0638) either before or shortly after virus infection resulted in a significant increase in transcript levels of bovine IFN-ß (boIFN-ß; 300-fold) and other IFN-inducible genes, including IFN-stimulated gene 15 (ISG-15), myxovirus resistance 1 (Mx-1), Mx-2, RIG-I, 2',5'-oligoadenylate synthetase 1 (OAS-1), and protein kinase R (PKR). Expression of these factors correlated with a significant decrease in VSV and FMDV viral titers. Our data confirm the involvement of EHMT2 in the epigenetic regulation of boIFN-ß and demonstrate the activation of a general antiviral state after EHMT2 inhibition.


Subject(s)
Epigenesis, Genetic , Foot-and-Mouth Disease Virus/drug effects , Histocompatibility Antigens/immunology , Histone-Lysine N-Methyltransferase/immunology , Interferon-beta/immunology , Vesicular stomatitis Indiana virus/drug effects , 2',5'-Oligoadenylate Synthetase/genetics , 2',5'-Oligoadenylate Synthetase/immunology , Animals , Cattle , Cell Line , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/immunology , Epithelial Cells/drug effects , Epithelial Cells/immunology , Epithelial Cells/virology , Euchromatin/chemistry , Euchromatin/drug effects , Euchromatin/metabolism , Fetus , Fibroblasts/drug effects , Fibroblasts/immunology , Fibroblasts/virology , Foot-and-Mouth Disease Virus/growth & development , Foot-and-Mouth Disease Virus/immunology , Histocompatibility Antigens/genetics , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Histone-Lysine N-Methyltransferase/genetics , Immunity, Innate , Interferon-beta/pharmacology , Myxovirus Resistance Proteins/genetics , Myxovirus Resistance Proteins/immunology , Poly I-C/pharmacology , Primary Cell Culture , Quinazolines/pharmacology , RNA, Messenger/genetics , RNA, Messenger/immunology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Transcription, Genetic , Ubiquitins/genetics , Ubiquitins/immunology , Vesicular stomatitis Indiana virus/growth & development , Vesicular stomatitis Indiana virus/immunology , eIF-2 Kinase/genetics , eIF-2 Kinase/immunology
12.
mSystems ; 1(3)2016.
Article in English | MEDLINE | ID: mdl-27822536

ABSTRACT

Several biosafety level 3 and/or 4 (BSL-3/4) pathogens are high-consequence, single-stranded RNA viruses, and their genomes, when introduced into permissive cells, are infectious. Moreover, many of these viruses are select agents (SAs), and their genomes are also considered SAs. For this reason, cDNAs and/or their derivatives must be tested to ensure the absence of infectious virus and/or viral RNA before transfer out of the BSL-3/4 and/or SA laboratory. This tremendously limits the capacity to conduct viral genomic research, particularly the application of next-generation sequencing (NGS). Here, we present a sequence-independent method to rapidly amplify viral genomic RNA while simultaneously abolishing both viral and genomic RNA infectivity across multiple single-stranded positive-sense RNA (ssRNA+) virus families. The process generates barcoded DNA amplicons that range in length from 300 to 1,000 bp, which cannot be used to rescue a virus and are stable to transport at room temperature. Our barcoding approach allows for up to 288 barcoded samples to be pooled into a single library and run across various NGS platforms without potential reconstitution of the viral genome. Our data demonstrate that this approach provides full-length genomic sequence information not only from high-titer virion preparations but it can also recover specific viral sequence from samples with limited starting material in the background of cellular RNA, and it can be used to identify pathogens from unknown samples. In summary, we describe a rapid, universal standard operating procedure that generates high-quality NGS libraries free of infectious virus and infectious viral RNA. IMPORTANCE This report establishes and validates a standard operating procedure (SOP) for select agents (SAs) and other biosafety level 3 and/or 4 (BSL-3/4) RNA viruses to rapidly generate noninfectious, barcoded cDNA amenable for next-generation sequencing (NGS). This eliminates the burden of testing all processed samples derived from high-consequence pathogens prior to transfer from high-containment laboratories to lower-containment facilities for sequencing. Our established protocol can be scaled up for high-throughput sequencing of hundreds of samples simultaneously, which can dramatically reduce the cost and effort required for NGS library construction. NGS data from this SOP can provide complete genome coverage from viral stocks and can also detect virus-specific reads from limited starting material. Our data suggest that the procedure can be implemented and easily validated by institutional biosafety committees across research laboratories.

13.
Elife ; 4: e09790, 2015 Jul 29.
Article in English | MEDLINE | ID: mdl-26222499

ABSTRACT

By genetically engineering pigs to degrade a crucial viral protein, livestock can be made less susceptible to foot and mouth disease virus.


Subject(s)
Animals, Genetically Modified , Antiviral Agents/metabolism , Disease Resistance , Foot-and-Mouth Disease Virus/immunology , Foot-and-Mouth Disease/prevention & control , RNA, Small Interfering/metabolism , Animals
14.
Antiviral Res ; 95(2): 150-8, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22640779

ABSTRACT

Vesicular stomatitis virus (VSV) causes great economic impact to livestock industry and is a prototype for studying non-segmented negative-stranded RNA (NSNR) viruses. In this study, we evaluated the antiviral potential of unique short-hairpin RNA (shRNA) targeting genes that form the ribonuclear protein (RNP) complex of VSV serotype Indiana (VSIV). We used lentiviral vectors to construct cell lines that stably expressed one of seven shRNAs targeting the RNP genes of VSIV, namely nucleocapsid (N), phosphoprotein (P), or polymerase (L). We reported two N-shRNA sequences targeting the 5' or 3' end of N that significantly reduced N, P, and L viral transcripts (p<0.001), reduced viral protein expression, and reduced the viral particles shed in Vero cells (p<0.01). When we analyzed the sequence diversity in the target region of this N-shRNA from two field isolates, we detected a single base substitution outside the seed region. We also reported five other shRNA sequences targeting components of the viral RNA that significantly reduce N, P, and L viral transcripts (p<0.001) but failed to efficiently impair viral replication. The differences in the efficiency of the shRNAs tested were not due to mismatches within the target region in the genome of VSIV. Although partial silencing of viral transcripts by single shRNAs impaired but did not block VSIV replication, the combination of the shRNAs identified here into a multiple shRNA vector may result in inhibition of viral replication. These data contribute to ongoing development of RNAi-based technologies to combat viral diseases.


Subject(s)
Antiviral Agents/pharmacology , Biological Products/pharmacology , RNA, Small Interfering/pharmacology , Vesiculovirus/drug effects , Virus Replication/drug effects , Animals , Cell Line , Chlorocebus aethiops , Drug Carriers , Gene Expression , Genetic Vectors , Lentivirus/genetics , RNA, Small Interfering/genetics , Transduction, Genetic , Vero Cells
SELECTION OF CITATIONS
SEARCH DETAIL