Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Cell Mol Life Sci ; 81(1): 148, 2024 Mar 20.
Article in English | MEDLINE | ID: mdl-38509419

ABSTRACT

Propagation of viruses requires interaction with host factors in infected cells and repression of innate immune responses triggered by the host viral sensors. Cytosolic DNA sensing pathway of cyclic GMP-AMP synthase (cGAS) and stimulator of interferon genes (STING) is a major component of the antiviral response to DNA viruses, also known to play a relevant role in response to infection by RNA viruses, including foot-and-mouth disease virus (FMDV). Here, we provide supporting evidence of cGAS degradation in swine cells during FMDV infection and show that the two virally encoded proteases, Leader (Lpro) and 3Cpro, target cGAS for cleavage to dampen the cGAS/STING-dependent antiviral response. The specific target sequence sites on swine cGAS were identified as Q140/T141 for the FMDV 3Cpro and the KVKNNLKRQ motif at residues 322-330 for Lpro. Treatment of swine cells with inhibitors of the cGAS/STING pathway or depletion of cGAS promoted viral infection, while overexpression of a mutant cGAS defective for cGAMP synthesis, unlike wild type cGAS, failed to reduce FMDV replication. Our findings reveal a new mechanism of RNA viral antagonism of the cGAS-STING innate immune sensing pathway, based on the redundant degradation of cGAS through the concomitant proteolytic activities of two proteases encoded by an RNA virus, further proving the key role of cGAS in restricting FMDV infection.


Subject(s)
Foot-and-Mouth Disease Virus , Animals , Swine , Foot-and-Mouth Disease Virus/metabolism , Peptide Hydrolases/metabolism , Signal Transduction , Immunity, Innate , Endopeptidases/genetics , Endopeptidases/metabolism , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , Antiviral Agents/metabolism
2.
Front Immunol ; 14: 1166725, 2023.
Article in English | MEDLINE | ID: mdl-37063925

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of a potentially severe respiratory disease, the coronavirus disease 2019 (COVID-19), an ongoing pandemic with limited therapeutic options. Here, we assessed the anti-coronavirus activity of synthetic RNAs mimicking specific domains in the non-coding regions of the foot-and-mouth disease virus (FMDV) genome (ncRNAs). These molecules are known to exert broad-spectrum antiviral activity in cell culture, mice and pigs effectively triggering the host innate immune response. The ncRNAs showed potent antiviral activity against SARS-CoV-2 after transfection in human intestinal Caco-2 and lung epithelium Calu-3 2B4 cells. When the in vivo efficacy of the FMDV ncRNAs was assessed in K18-hACE2 mice, administration of naked ncRNA before intranasal SARS-CoV-2 infection significantly decreased the viral load and the levels of pro-inflammatory cytokines in the lungs compared with untreated infected mice. The ncRNAs were also highly efficacious when assayed against common human HCoV-229E and porcine transmissible gastroenteritis virus (TGEV) in hepatocyte-derived Huh-7 and swine testis ST cells, respectively. These results are a proof of concept of the pan-coronavirus antiviral activity of the FMDV ncRNAs including human and animal divergent coronaviruses and potentially enhance our ability to fight future emerging variants.


Subject(s)
COVID-19 , Foot-and-Mouth Disease Virus , Male , Animals , Humans , Swine , Mice , Antiviral Agents/pharmacology , Foot-and-Mouth Disease Virus/genetics , Caco-2 Cells , SARS-CoV-2/genetics , RNA, Untranslated
3.
Antimicrob Agents Chemother ; 67(4): e0170322, 2023 04 18.
Article in English | MEDLINE | ID: mdl-36975844

ABSTRACT

Antiviral compounds targeting cellular metabolism are part of the therapeutic arsenal to control the spread of virus infection, either as sole treatment or in combination with direct-acting antivirals (DAA) or vaccines. Here, we describe the effect of two of them, lauryl gallate (LG) and valproic acid (VPA) both exhibiting a wide antiviral spectrum, against infection by coronaviruses such as HCoV-229E, HCoV-OC43, and SARS-CoV-2. A consistent 2 to 4-log-decrease in virus yields was observed in the presence of each antiviral, with an average IC50 value of 1.6 µM for LG and 7.2 mM for VPA. Similar levels of inhibition were observed when adding the drug 1 h before adsorption, at the time of infection or 2 h after infection, supporting a postvirus entry mechanism of action. The specificity of the antiviral effect of LG against SARS-CoV-2, relative to other related compounds such as gallic acid (G) and epicatechin gallate (ECG), predicted to be better inhibitors according to in silico studies, was also demonstrated. The combined addition of LG, VPA, and remdesivir (RDV), a DAA with a proven effect against human coronaviruses, resulted in a robust synergistic effect between LG and VPA, and to a lesser extent between the other drug combinations. These findings reinforce the interest of these wide antiviral spectrum host-targeted compounds as a first line of defense against viral diseases or as a vaccine complement to minimize the gap in antibody-mediated protection evoked by vaccines, either in the case of SARS-CoV-2 or for other possible emerging viruses.


Subject(s)
COVID-19 , Coronavirus 229E, Human , Coronavirus OC43, Human , Hepatitis C, Chronic , Humans , Antiviral Agents/pharmacology , SARS-CoV-2
4.
Methods Mol Biol ; 2465: 125-135, 2022.
Article in English | MEDLINE | ID: mdl-35118619

ABSTRACT

The ncRNAs are short RNA transcripts with sequence and structure resembling that of specific domains in the non-coding regions of the foot-and-mouth disease (FMD) virus (FMDV ) genome. These synthetic molecules induce a robust antiviral response and have been shown to enhance the immune response and protection induced by an FMD inactivated vaccine in pigs. Here, we describe the method for ncRNAs synthesis, formulation, and delivery into mice and pigs for studies focused on testing the adjuvant effect of RNA-based strategies in combination with veterinarian vaccines.


Subject(s)
Foot-and-Mouth Disease Virus , Foot-and-Mouth Disease , Viral Vaccines , Adjuvants, Immunologic/pharmacology , Adjuvants, Vaccine , Animals , Antibodies, Viral , Foot-and-Mouth Disease/prevention & control , Foot-and-Mouth Disease Virus/genetics , Mice , RNA , Swine , Vaccines, Synthetic , Viral Vaccines/genetics
5.
AIDS Care ; 34(2): 155-162, 2022 02.
Article in English | MEDLINE | ID: mdl-34743624

ABSTRACT

In an ongoing Mediterranean cohort, we compared age-related conditions between 208 HIV-infected persons and 104 matched controls. ≥3 comorbidities were found in 31.0% of HIV-infected patients and 8.7% of controls. Conditions significantly more frequent among the HIV-infected population were: lipid abnormalities, cancer, osteopenia/osteoporosis, liver disease, sexual dysfunction, hearing deficit, sleep disorders, falls, cognitive complaints, being single, living alone, and being elderly at risk. HIV-infected patients aged >70 years had a significantly higher number of cardiovascular risk factors (CVRF) and comorbidities than controls. HIV-infected persons who had never smoked had a higher prevalence of CVRFs, ≥3 comorbidities, liver disease, cancer, and cognitive complaints compared to controls. Factors associated with frailty were being a man who has sex with men, ≥3 CVRFs, nadir CD470 years. The multidisciplinary assessment also revealed concerning findings in social, cognitive, and functional variables among HIV-infected individuals, with a higher prevalence of elderly at risk than among controls.


Subject(s)
HIV Infections , Aged , Aging , Cohort Studies , Comorbidity , HIV Infections/complications , HIV Infections/epidemiology , Humans , Male , Prevalence , Risk Factors
6.
Wiley Interdiscip Rev RNA ; 12(4): e1645, 2021 07.
Article in English | MEDLINE | ID: mdl-33605051

ABSTRACT

RNA viruses have developed specialized mechanisms to subvert host RNA-binding proteins (RBPs) favoring their own gene expression. The Leader (L) protein of foot-and-mouth disease virus, a member of the Picornaviridae family, is a papain-like cysteine protease that self-cleaves from the polyprotein. Early in infection, the L protease cleaves the translation initiation factors eIF4GI and eIF4GII, inducing the shutdown of cap-dependent translation. However, the cleavage sites on the viral polyprotein, eIF4GI, and eIF4GII differ in sequence, challenging the definition of a consensus site for L targets. Identification of Gemin5 and Daxx proteolytic products in infected cells unveiled a motif centered on the RKAR sequence. The RBP Gemin5 is a member of the survival of motor neurons complex, a ribosome interacting protein, and a translation downregulator. Likewise, the Fas-ligand Daxx is a multifunctional adaptor that plays key roles in transcription control, apoptosis, and innate immune antiviral response. Remarkably, the cleavage site on the RNA helicases MDA5 and LGP2, two relevant immune sensors of the retinoic acid-inducible gene-I (RIG-I)-like receptors family, resembles the L target site of Gemin5 and Daxx, and similar cleavage sites have been reported in ISG15 and TBK1, two proteins involved in type I interferon response and signaling pathway, respectively. In this review we dissect the features of the L cleavage sites in essential RBPs, eventually helping in the discovery of novel L targets. This article is categorized under: RNA in Disease and Development > RNA in Disease Translation > Translation Regulation.


Subject(s)
Antiviral Restriction Factors/immunology , Foot-and-Mouth Disease Virus , Immunity, Innate , RNA , Animals , Foot-and-Mouth Disease Virus/enzymology , Foot-and-Mouth Disease Virus/genetics , RNA Helicases
7.
Cell Death Dis ; 11(8): 718, 2020 09 02.
Article in English | MEDLINE | ID: mdl-32879301

ABSTRACT

The RIG-I-like receptor (RLR) melanoma differentiation-associated gene 5 (MDA5) plays a key role in triggering innate antiviral response during infection by RNA viruses. MDA5 activation leads to transcription induction of type-I interferon (IFN) and proinflammatory cytokines. MDA5 has also been associated with autoimmune and autoinflammatory diseases by dysfunctional activation of innate immune response in the absence of infection. Here, we show how foot-and-mouth disease virus (FMDV) counteracts the specific antiviral effect exerted by MDA5 targeting the protein for cleavage by the viral Leader protease (Lpro). MDA5 overexpression had an inhibitory effect on FMDV infection in IFN-competent cells. Remarkably, immunostimulatory viral RNA co-immunoprecipitated with MDA5 in infected cells. Moreover, specific cleavage of MDA5 by Lpro was detected in co-transfected cells, as well as during the course of FMDV infection. A significant reduction in IFN induction associated with MDA5 cleavage was detected by comparison with a non-cleavable MDA5 mutant protein with preserved antiviral activity. The Lpro cleavage site in MDA5 was identified as the RGRAR sequence in the conserved helicase motif VI, coinciding with that recently reported for Lpro in LGP2, another member of the RLRs family involved in antiviral defenses. Interestingly, specific mutations within the MDA5 Lpro target sequence have been associated with immune disease in mice and humans. Our results reveal a pleiotropic strategy for immune evasion based on a viral protease targeting phylogenetically conserved domains of immune sensors. Identification of viral strategies aimed to disrupt MDA5 functionality may also contribute to develop new treatment tools for MDA5-related disorders.


Subject(s)
Endopeptidases/metabolism , Foot-and-Mouth Disease Virus/metabolism , Interferon-Induced Helicase, IFIH1/metabolism , Animals , Cell Line , DEAD Box Protein 58/metabolism , Endopeptidases/genetics , Foot-and-Mouth Disease Virus/genetics , Genetic Pleiotropy/genetics , HEK293 Cells , Humans , Immunity, Innate , Interferon Type I/metabolism , Interferon-Induced Helicase, IFIH1/physiology , Proteolysis , RNA, Viral/immunology , Receptors, Immunologic/metabolism , Signal Transduction , Swine
8.
Front Vet Sci ; 7: 495, 2020.
Article in English | MEDLINE | ID: mdl-32851049

ABSTRACT

Foot-and-mouth disease virus (FMDV) is the causative agent of a highly contagious viral disease that affects multiple cloven-hooved hosts including important livestock (pigs, cattle, sheep and goats) as well as several wild animal species. Crossover of FMDV between domestic and wildlife populations may prolong virus circulation during outbreaks. The wild boar (Sus scrofa) is considered a reservoir of various pathogens that can infect other wildlife, domestic animals, and humans. As wild boar and domestic pigs are susceptible to the same pathogens and can infect each other, infected wild boar populations may represent a threat to the pig industry and to international trade. The ncRNAs are synthetic non-coding RNA transcripts, mimicking structural domains in the FMDV genome, known to exert a broad-spectrum antiviral and immunomodulatory effect in swine, bovine and mice cells. Here, we show the type I interferon-dependent, robust and broad range antiviral activity induced by the ncRNAs in a cell line derived from wild boar lung cells (WSL). Transfection of WSL cells with the ncRNAs exerted a protective effect against infection with FMDV, vesicular stomatitis virus (VSV), swine vesicular disease virus (SVDV) and African swine fever virus (ASFV). Our results prove the biological activity of the ncRNAs in cells of an FMDV wild animal host species against a variety of viruses affecting pigs, including relevant viral pathogens of epizootic risk.

9.
Transbound Emerg Dis ; 67(4): 1614-1622, 2020 Jul.
Article in English | MEDLINE | ID: mdl-31994334

ABSTRACT

Foot-and-mouth disease virus (FMDV) causes a widely extended contagious disease of livestock. We have previously reported that a synthetic dendrimeric peptide, termed B2 T(mal), consisting of two copies of a B-cell epitope [VP1(140-158)] linked through maleimide groups to a T-cell epitope [3A(21-35)] of FMDV, elicits potent B- and T-cell-specific responses and confers solid protection in pigs to type O FMDV challenge. Longer duration of the protective response and the possibility of inducing protection after a single dose are important requirements for an efficient FMD vaccine. Herein, we show that administration of two doses of B2 T(mal) elicited high levels of specific total IgGs and neutralizing antibodies that lasted 4-5 months after the peptide boost. Additionally, concomitant levels of IFN-γ-producing specific T cells were observed. Immunization with two doses of B2 T(mal) conferred a long-lasting reduced susceptibility to FMDV infection, up to 136 days (19/20 weeks) post-boost. Remarkably, a similar duration of the protective response was achieved by a single dose of B2 T(mal). The effect on the B2 T(mal) vaccine of RNA transcripts derived from non-coding regions in the FMDV genome, known to enhance the immune response and protection induced by a conventional inactivated vaccine, was also analysed. The contribution of our results to the development of FMD dendrimeric vaccines is discussed.


Subject(s)
Epitopes, B-Lymphocyte/immunology , Foot-and-Mouth Disease Virus/immunology , Foot-and-Mouth Disease/prevention & control , Peptides/immunology , Swine Diseases/prevention & control , Viral Vaccines/administration & dosage , Animals , Antibodies, Neutralizing , Antibodies, Viral/immunology , Dendrimers , Epitopes, T-Lymphocyte/immunology , Female , Foot-and-Mouth Disease/immunology , Foot-and-Mouth Disease/virology , Immunity , Neutralization Tests , Swine , Swine Diseases/immunology , Swine Diseases/virology , T-Lymphocytes/immunology , Viral Vaccines/immunology
10.
Vet Microbiol ; 221: 8-12, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29981712

ABSTRACT

Foot-and-mouth disease virus (FMDV) is the causative agent of a highly transmissible disease affecting wild and domestic animals including pigs, cattle and sheep. The ability of synthetic RNA transcripts mimicking distinct domains in the non-coding regions of the FMDV genome (ncRNAs) to induce a potent innate immune response in swine cultured cells and mice has been previously described, as well as their enhancing effect on conventional inactivated FMD vaccines. Here, we provide evidence of the activation of interferon regulatory factor 3 (IRF3), a key transcriptional regulator of type I interferon (IFN)-dependent immune responses after transfection of swine and bovine cells with transcripts corresponding to the FMDV 3´ non-coding region (3´NCR). Induction of IFN-ß and Mx1expression, concomitantly with antiviral activity and IRF3 activation was observed in bovine MDBK cells transfected with the 3´NCR. Our results link the stimulation of the innate immune response observed in 3´NCR-transfected cells to the intracellular type I IFN signaling pathway and suggest the potential use of these molecules for antiviral strategies in cattle.


Subject(s)
Foot-and-Mouth Disease Virus/genetics , Interferon Regulatory Factor-3/metabolism , RNA, Viral/chemical synthesis , RNA, Viral/immunology , Animals , Cattle , Cell Line , Immunity, Innate , Swine
11.
PLoS Pathog ; 14(6): e1007135, 2018 06.
Article in English | MEDLINE | ID: mdl-29958302

ABSTRACT

The RNA helicase LGP2 (Laboratory of Genetics and Physiology 2) is a non-signaling member of the retinoic acid-inducible gene-I (RIG-I)-like receptors (RLRs), whose pivotal role on innate immune responses against RNA viruses is being increasingly uncovered. LGP2 is known to work in synergy with melanoma differentiation-associated gene 5 (MDA5) to promote the antiviral response induced by picornavirus infection. Here, we describe the activity of the foot-and-mouth disease virus (FMDV) Leader protease (Lpro) targeting LGP2 for cleavage. When LGP2 and Lpro were co-expressed, cleavage products were observed in an Lpro dose-dependent manner while co-expression with a catalytically inactive Lpro mutant had no effect on LGP2 levels or pattern. We further show that Lpro localizes and immunoprecipitates with LGP2 in transfected cells supporting their interaction within the cytoplasm. Evidence of LGP2 proteolysis was also detected during FMDV infection. Moreover, the inhibitory effect of LGP2 overexpression on FMDV growth observed was reverted when Lpro was co-expressed, concomitant with lower levels of IFN-ß mRNA and antiviral activity in those cells. The Lpro target site in LGP2 was identified as an RGRAR sequence in a conserved helicase motif whose replacement to EGEAE abrogated LGP2 cleavage by Lpro. Taken together, these data suggest that LGP2 cleavage by the Leader protease of aphthoviruses may represent a novel antagonistic mechanism for immune evasion.


Subject(s)
Endopeptidases/metabolism , Foot-and-Mouth Disease Virus/immunology , Foot-and-Mouth Disease/virology , Immune Evasion/immunology , Immunity, Innate/immunology , RNA Helicases/metabolism , Animals , Cells, Cultured , Chlorocebus aethiops , Cricetinae , Endopeptidases/genetics , Foot-and-Mouth Disease/immunology , Foot-and-Mouth Disease/pathology , Foot-and-Mouth Disease Virus/enzymology , HEK293 Cells , Humans , RNA Helicases/genetics , RNA Helicases/immunology , Vero Cells
14.
Article in English | MEDLINE | ID: mdl-28660175

ABSTRACT

Foot-and-mouth disease virus (FMDV) is the causative agent of an acute vesicular disease affecting pigs, cattle and other domestic, and wild animals worldwide. The aim of the host interferon (IFN) response is to limit viral replication and spread. Detection of the viral genome and products by specialized cellular sensors initiates a signaling cascade that leads to a rapid antiviral response involving the secretion of type I- and type III-IFNs and other antiviral cytokines with antiproliferative and immunomodulatory functions. During co-evolution with their hosts, viruses have acquired strategies to actively counteract host antiviral responses and the balance between innate response and viral antagonism may determine the outcome of disease and pathogenesis. FMDV proteases Lpro and 3C have been found to antagonize the host IFN response by a repertoire of mechanisms. Moreover, the putative role of other viral proteins in IFN antagonism is being recently unveiled, uncovering sophisticated immune evasion strategies different to those reported to date for other members of the Picornaviridae family. Here, we review the interplay between antiviral responses induced by FMDV infection and viral countermeasures to block them. Research on strategies used by viruses to modulate immunity will provide insights into the function of host pathways involved in defense against pathogens and will also lead to development of new therapeutic strategies to fight virus infections.


Subject(s)
Antiviral Agents/pharmacology , Foot-and-Mouth Disease Virus/drug effects , Foot-and-Mouth Disease Virus/immunology , Foot-and-Mouth Disease/drug therapy , Host-Pathogen Interactions/immunology , Immunity, Innate , Animals , Cytokines/pharmacology , Foot-and-Mouth Disease/immunology , Foot-and-Mouth Disease/metabolism , Foot-and-Mouth Disease/virology , Foot-and-Mouth Disease Virus/pathogenicity , Genome, Viral , Immune Evasion , Interferons/pharmacology , Peptide Hydrolases/immunology , RNA, Viral/immunology , Signal Transduction , Viral Proteins/immunology , Viral Proteins/metabolism , Virus Replication/drug effects
15.
Antiviral Res ; 142: 30-36, 2017 06.
Article in English | MEDLINE | ID: mdl-28315707

ABSTRACT

Foot-and-mouth disease virus (FMDV) is the causative agent of a highly contagious disease and a major concern in animal health worldwide. We have previously reported the use of RNA transcripts mimicking structural domains in the non-coding regions of the FMDV RNA as potent type-I interferon (IFN) inducers showing antiviral effect in vivo, as well as their immunomodulatory properties in combination with an FMD vaccine in mice. Here, we describe the enhancing effect of RNA delivery on the immunogenicity and protection induced by a suboptimal dose of a conventional FMD vaccine in pigs. Animals receiving the RNA developed earlier and higher levels of neutralizing antibodies against homologous and heterologous isolates, compared to those immunized with the vaccine alone, and had higher anti-FMDV titers at late times post-vaccination. RNA delivery also induced higher specific T-cell response and protection levels against FMDV challenge. Peripheral blood mononuclear cells from pigs inoculated with RNA and the vaccine had a higher IFN-γ specific response than those from pigs receiving the vaccine alone. When challenged with FMDV, all three animals immunized with the conventional vaccine developed antibodies to the non-structural viral proteins 3ABC and two of them developed severe signs of disease. In the group receiving the vaccine together with the RNA, two pigs were fully protected while one showed delayed and mild signs of disease. Our results support the immunomodulatory effect of these RNA molecules in natural hosts and suggest their potential use for improvement of FMD vaccines strategies.


Subject(s)
Foot-and-Mouth Disease Virus/immunology , Foot-and-Mouth Disease/immunology , Foot-and-Mouth Disease/prevention & control , RNA/administration & dosage , RNA/immunology , Viral Vaccines/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Female , Foot-and-Mouth Disease Virus/genetics , Immunoglobulin G/blood , Interferon Type I/immunology , Interferon-gamma/immunology , Kinetics , Leukocytes, Mononuclear/immunology , RNA/chemical synthesis , RNA, Small Untranslated , Swine , Swine Diseases/prevention & control , T-Lymphocytes/immunology , Vaccination , Viral Nonstructural Proteins/immunology
16.
Viruses ; 7(7): 3954-73, 2015 Jul 17.
Article in English | MEDLINE | ID: mdl-26193305

ABSTRACT

The innate immune system is the first line of defense against viral infections. Exploiting innate responses for antiviral, therapeutic and vaccine adjuvation strategies is being extensively explored. We have previously described, the ability of small in vitro RNA transcripts, mimicking the sequence and structure of different domains in the non-coding regions of the foot-and-mouth disease virus (FMDV) genome (ncRNAs), to trigger a potent and rapid innate immune response. These synthetic non-infectious molecules have proved to have a broad-range antiviral activity and to enhance the immunogenicity of an FMD inactivated vaccine in mice. Here, we have studied the involvement of pattern-recognition receptors (PRRs) in the ncRNA-induced innate response and analyzed the antiviral and cytokine profiles elicited in swine cultured cells, as well as peripheral blood mononuclear cells (PBMCs).


Subject(s)
DEAD-box RNA Helicases/immunology , Foot-and-Mouth Disease Virus/immunology , Foot-and-Mouth Disease/immunology , Immunity, Innate , RNA, Viral/immunology , Toll-Like Receptors/immunology , Viral Vaccines/immunology , Animals , Antibodies, Viral/immunology , DEAD-box RNA Helicases/genetics , Female , Foot-and-Mouth Disease/genetics , Foot-and-Mouth Disease/prevention & control , Foot-and-Mouth Disease/virology , Foot-and-Mouth Disease Virus/genetics , Leukocytes, Mononuclear/immunology , Male , Mice , RNA, Viral/administration & dosage , RNA, Viral/chemical synthesis , RNA, Viral/genetics , Swine , Toll-Like Receptors/genetics , Viral Vaccines/administration & dosage , Viral Vaccines/chemical synthesis , Viral Vaccines/genetics
17.
Antiviral Res ; 109: 64-7, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24973761

ABSTRACT

In this work we have addressed the effect of synthetic, non-infectious, RNA transcripts, mimicking structural domains of the non-coding regions (NCRs) of the foot-and-mouth disease virus (FMDV) genome on the infection of mice with Rift Valley fever virus (RVFV). Groups of 5 mice were inoculated intraperitoneally (i.p.) with 200 µg of synthetic RNA resembling the 5'-terminal S region, the internal ribosome entry site (IRES) or the 3'-NCR of the FMDV genome. RNA inoculation was performed 24h before (-24 h), 24 h after (+24 h) or simultaneously to the challenge with a lethal dose of RVFV. Administration of the IRES RNA afforded higher survival rates than administration of S or 3'NCR transcripts either at -24h or +24h after challenge. In contrast, when RNA inoculation and viral challenge were performed simultaneously, all mice survived in both IRES- and 3'NCR-inoculated groups, with an 80% survival in mice receiving the S RNA. Among survivors, a complete correlation between significant anti-RVFV circulating antibody titers and resistance to a second lethal challenge with the virus was observed, supporting a limited viral replication in the RNA-inoculated animals upon the first challenge. All three RNA transcripts were able to induce the production of systemic antiviral and pro-inflammatory cytokines. These data show that triggering of intracellular pathogen sensing pathways constitutes a promising approach towards development of novel RVF preventive or therapeutic strategies.


Subject(s)
Foot-and-Mouth Disease Virus/genetics , Interferons/administration & dosage , RNA, Viral/immunology , Rift Valley Fever/prevention & control , Rift Valley fever virus/immunology , Viral Vaccines/immunology , Animals , Antibodies, Viral/immunology , Cross Protection , Foot-and-Mouth Disease Virus/immunology , Genome, Viral , Humans , Mice , Mice, Inbred BALB C , RNA, Viral/administration & dosage , RNA, Viral/chemical synthesis , RNA, Viral/genetics , Rift Valley Fever/immunology , Rift Valley Fever/virology , Rift Valley fever virus/physiology , Vaccination , Viral Vaccines/administration & dosage , Viral Vaccines/chemical synthesis , Viral Vaccines/genetics , Virus Replication
18.
Vaccine ; 31(40): 4375-81, 2013 Sep 13.
Article in English | MEDLINE | ID: mdl-23859841

ABSTRACT

We have recently described the antiviral effect in mice of in vitro-transcribed RNAs mimicking structural domains in the non-coding regions of the foot-and-mouth disease virus (FMDV) genome RNA. These small, synthetic and non-infectious RNA molecules (ncRNAs) are potent type-I interferon (IFN) inducers in vivo. In this work, the immunomodulatory effect of the ncRNA corresponding to the internal ribosome entry site (IRES) on immunization with two different FMD vaccine formulations, both based on inactivated virus, including or not a commercial adjuvant, was analyzed in the mice model. The effect of the time interval between RNA inoculation and immunization was also studied. RNA delivery consistently increased the titers of specific anti-FMDV antibodies, including neutralizing antibodies, elicited after vaccination. Moreover, at day 2 after immunization, significant differences in mean antibody titers could be detected between the groups of mice receiving either vaccine co-administered with the RNA and the control group, unlike those immunized with the vaccine alone. When vaccinated mice were challenged with FMDV, the mean values of viral load were lower in the groups receiving the RNA together with the vaccine. Our results show the enhancing effect of the IRES RNA on the immune response elicited after vaccination and suggest the potential of this molecule as an adjuvant for new FMD vaccine design.


Subject(s)
Foot-and-Mouth Disease Virus/immunology , Foot-and-Mouth Disease/immunology , RNA/administration & dosage , Viral Vaccines/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Animals, Domestic/immunology , Antibodies, Neutralizing , Antibodies, Viral/blood , Foot-and-Mouth Disease/prevention & control , Interferon Type I/biosynthesis , Interferon Type I/immunology , Mice , Mice, Inbred ICR , RNA/immunology , Vaccination , Viral Load/immunology
19.
PLoS One ; 7(11): e49494, 2012.
Article in English | MEDLINE | ID: mdl-23166685

ABSTRACT

West Nile virus (WNV) is a neurovirulent single stranded RNA mosquito-borne flavivirus, whose main natural hosts are birds, but it also infects humans and horses. Nowadays, no human vaccine is commercially available and clinical treatment is only supportive. Recently, it has been shown that RNA transcripts, mimicking structural domains in the non-coding regions (NCRs) of the foot-and mouth disease virus (FMDV) induce a potent IFN response and antiviral activity in transfected cultured cells, and also reduced mice susceptibility to FMDV. By using different transcripts combinations, administration schedules, and infecting routes and doses, we have demonstrated that these FMDV RNA transcripts protect suckling and adult mice against lethal challenge with WNV. The protective activity induced by the transcripts was systemic and dependent on the infection route and dose. These results confirm the antiviral potential of these synthetic RNAs for fighting viruses of different families relevant for human and animal health.


Subject(s)
Interferon Type I/blood , RNA, Viral/genetics , West Nile Fever/immunology , West Nile Fever/prevention & control , West Nile virus/genetics , West Nile virus/immunology , Animals , Gene Expression Regulation, Viral , Immunity, Innate , Interferon Type I/immunology , Mice , RNA, Untranslated/genetics , West Nile Fever/mortality
20.
J Gen Virol ; 93(Pt 11): 2382-2386, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22875255

ABSTRACT

The role of cellular Rab GTPases that govern traffic between different endosome populations was analysed on foot-and-mouth disease virus (FMDV) infection. Changes of viral receptor specificity did not alter Rab5 requirement for infection. However, a correlation between uncoating pH and requirement of Rab5 for infection was observed. A mutant FMDV with less acidic uncoating pH threshold was less sensitive to inhibition of Rab5, whereas another mutant with more acidic requirements was more sensitive to inhibition of Rab5. On the contrary, opposed correlations between uncoating pH and dependence of Rab function were observed upon expression of dominant-negative forms of Rab7 or 11. Modulation of uncoating pH also reduced FMDV virulence in suckling mice. These results are consistent with FMDV uncoating inside early endosomes and indicate that displacements from optimum pH for uncoating reduce viral fitness in vivo.


Subject(s)
Foot-and-Mouth Disease Virus/physiology , Receptors, Virus/physiology , rab5 GTP-Binding Proteins/antagonists & inhibitors , Animals , Buffers , Cell Line , Cricetinae , Endosomes , Gene Expression Regulation , Green Fluorescent Proteins , Hydrogen-Ion Concentration , Plasmids/genetics , Plasmids/metabolism , Virus Internalization , rab5 GTP-Binding Proteins/genetics , rab5 GTP-Binding Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...