Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 154
Filtrar
1.
Viruses ; 16(7)2024 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-39066280

RESUMEN

We conducted an integrative analysis to elucidate the spatial epidemiological patterns of the Vesicular Stomatitis New Jersey virus (VSNJV) during the 2014-15 epizootic cycle in the United States (US). Using georeferenced VSNJV genomics data, confirmed vesicular stomatitis (VS) disease cases from surveillance, and a suite of environmental factors, our study assessed environmental and phylogenetic similarity to compare VS cases reported in 2014 and 2015. Despite uncertainties from incomplete virus sampling and cross-scale spatial processes, patterns suggested multiple independent re-invasion events concurrent with potential viral overwintering between sequential seasons. Our findings pointed to a geographically defined southern virus pool at the US-Mexico interface as the source of VSNJV invasions and overwintering sites. Phylodynamic analysis demonstrated an increase in virus diversity before a rise in case numbers and a pronounced reduction in virus diversity during the winter season, indicative of a genetic bottleneck and a significant narrowing of virus variation between the summer outbreak seasons. Environment-vector interactions underscored the central role of meta-population dynamics in driving disease spread. These insights emphasize the necessity for location- and time-specific management practices, including rapid response, movement restrictions, vector control, and other targeted interventions.


Asunto(s)
Brotes de Enfermedades , Genoma Viral , Filogenia , Estaciones del Año , Estomatitis Vesicular , Virus de la Estomatitis Vesicular New Jersey , Animales , Estomatitis Vesicular/virología , Estomatitis Vesicular/epidemiología , Virus de la Estomatitis Vesicular New Jersey/genética , Estados Unidos/epidemiología , Genómica , Geografía , Bovinos , Variación Genética , Enfermedades de los Bovinos/virología , Enfermedades de los Bovinos/epidemiología
2.
mBio ; 15(7): e0056824, 2024 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-38888342

RESUMEN

The interferon (IFN) system protects mammals from diseases caused by virus infections. IFN synthesis is induced by pattern recognition receptor signaling pathways activated by virus infection. IFN is secreted from the infected cells and acts upon neighboring cells by binding cell surface receptors and triggering induction of hundreds of IFN-stimulated genes and proteins, many of which block different steps of virus replication. The IFN-induced tetratricopeptide repeat proteins (IFIT) are a family of RNA-binding proteins. We and others have previously reported that IFIT2 protects mice from many neurotropic RNA viruses; indeed, Ifit2-/- mice are very susceptible to intranasal or subcutaneous infections with vesicular stomatitis virus (VSV). Here, using a newly generated conditional knockout mouse, we report that ablation of Ifit2 expression only in neuronal cells was sufficient to render mice susceptible to neuropathogenesis caused by intranasal, but not subcutaneous, infection of VSV. Another genetically modified mouse line, expressing a mutant IFIT2 that cannot bind RNA, was as susceptible to VSV infection as Ifit2-/- mice. These results demonstrated that IFIT2 RNA-binding activity is essential for protecting mice against neurological diseases caused by intranasal infection of VSV.IMPORTANCEInterferon's (IFN's) antiviral effects are mediated by the proteins encoded by the interferon-stimulated genes. IFN-stimulated genes (IFIT2) is one such protein, which inhibits replication of many RNA viruses in the mouse brain and the resultant neuropathology. Our study sheds light on how IFIT2 works. By ablating Ifit2 expression only in neuronal cells, using a newly generated conditional knockout mouse line, we showed that Ifit2 induction in the neurons of the infected mouse was necessary for antiviral function of interferon. IFIT2 has no known enzyme activity; instead, it functions by binding to cellular or viral proteins or RNAs. We engineered a new mouse line that expressed a mutant IFIT2 that cannot bind RNA. These mice were very susceptible to infection with vesicular stomatitis virus indicating that the RNA-binding property of IFIT2 was essential for its antiviral function in vivo.


Asunto(s)
Ratones Noqueados , Neuronas , Proteínas de Unión al ARN , Estomatitis Vesicular , Animales , Ratones , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Neuronas/virología , Neuronas/metabolismo , Estomatitis Vesicular/virología , Estomatitis Vesicular/inmunología , Estomatitis Vesicular/genética , Replicación Viral , Vesiculovirus/inmunología , Vesiculovirus/genética , Ratones Endogámicos C57BL , Virus de la Estomatitis Vesicular Indiana/inmunología , Virus de la Estomatitis Vesicular Indiana/genética , Proteínas Reguladoras de la Apoptosis
3.
Emerg Infect Dis ; 30(5): 1004-1008, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38666640

RESUMEN

We evaluated the in vitro effects of lyophilization for 2 vesicular stomatitis virus-based vaccines by using 3 stabilizing formulations and demonstrated protective immunity of lyophilized/reconstituted vaccine in guinea pigs. Lyophilization increased stability of the vaccines, but specific vesicular stomatitis virus-based vaccines will each require extensive analysis to optimize stabilizing formulations.


Asunto(s)
Modelos Animales de Enfermedad , Liofilización , Estomatitis Vesicular , Vacunas Virales , Animales , Cobayas , Vacunas Virales/inmunología , Vacunas Virales/administración & dosificación , Estomatitis Vesicular/inmunología , Estomatitis Vesicular/prevención & control , Estomatitis Vesicular/virología , Vesiculovirus/inmunología , Vesiculovirus/genética , Anticuerpos Antivirales/inmunología , Anticuerpos Antivirales/sangre , Eficacia de las Vacunas , Virus de la Estomatitis Vesicular Indiana/inmunología
4.
Am J Vet Res ; 85(5)2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38382199

RESUMEN

OBJECTIVE: To describe an outbreak of vesicular stomatitis virus (VSV) in southern white rhinoceros (SWR; Ceratotherium simum simum) and greater one-horned rhinoceros (GOHR; Rhinoceros unicornis) at a safari park in San Diego, CA, from May to September 2023. ANIMALS: 21 SWR and 5 GOHR in professionally managed care. METHODS: Rhinoceros of both species presented with a range of clinical signs and severities. Lesion locations were categorized as cutaneous (coronary bands, heels and soles, limbs, ventrum, neck folds, and ears) and mucocutaneous (lips, nostrils, mucous membranes of the oral cavity, and vulva). Clinical signs included lethargy, lameness, difficulty with prehension, hyporexia to anorexia, and hypersalivation. Severely affected rhinoceros had clinical pathology findings consistent with systemic inflammation. RESULTS: Vesicular stomatitis New Jersey virus was confirmed via PCR from swabs of lesions in 10/26 (38%) rhinoceros. Of these 10 confirmed cases, 9 (90%) were SWR and 1 (10%) was a GOHR. A further 6/26 (24%) were considered probable cases, and 10/26 (38%) were considered suspect cases based on clinical signs, but the inability to appropriately sample due to the housing environment precluded confirmation. Histopathology samples from 3 rhinoceros were consistent with VSV, and viral RNA was localized in histologic lesions via RNA in situ hybridization for 1 case. All rhinoceros survived infection despite severe systemic illness in 2 animals. CLINICAL RELEVANCE: This case series describes the clinical appearance and progression of VSV in 2 rhinoceros species. To the authors' knowledge, this is the first report of VSV in a rhinoceros.


Asunto(s)
Animales de Zoológico , Perisodáctilos , Animales , Perisodáctilos/virología , California/epidemiología , Femenino , Masculino , Brotes de Enfermedades/veterinaria , Virus de la Estomatitis Vesicular New Jersey/genética , Virus de la Estomatitis Vesicular New Jersey/aislamiento & purificación , Estomatitis Vesicular/virología , Estomatitis Vesicular/patología
5.
Vet Clin North Am Food Anim Pract ; 40(2): 251-259, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38402042

RESUMEN

Vesicular stomatitis (VS) is a vector-borne livestock disease caused by either VS New Jersey virus or VS Indiana virus. The disease circulates endemically in northern South America, Central America, and Mexico and only occasionally causes outbreaks in the United States. During the past 20 years, VS outbreaks in the southwestern and Rocky Mountain regions occurred periodically with incursion years followed by virus overwintering and subsequent expansion outbreak years. Regulatory response by animal health officials prevents spread from lesioned animals and manages trade impacts. Recent US outbreaks highlight potential climate change impacts on insect vectors or other transmission-related variables.


Asunto(s)
Estomatitis Vesicular , Virus de la Estomatitis Vesicular New Jersey , Animales , Estomatitis Vesicular/virología , Brotes de Enfermedades/veterinaria , Brotes de Enfermedades/prevención & control , Virus de la Estomatitis Vesicular Indiana
6.
Anticancer Res ; 43(3): 1185-1191, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36854523

RESUMEN

BACKGROUND/AIM: Attempts have been made to enhance treatment with vesicular stomatitis virus (VSV) for osteosarcoma. We have previously shown that VSV incorporated with miRNA143 enhanced the antitumor effect at some doses; however, the range of the doses was narrow. This has not been evaluated in vivo, and the synergistic effect of this antitumor effect in animals is unknown. The purpose of the study was to evaluate the oncolytic effect of VSV-miRNA on osteosarcoma cells in vivo. MATERIALS AND METHODS: A novel oncolytic VSV was developed by incorporating the tumor-suppressor microRNA143 (rVSV-miR143). In order to compare the antitumor effects of administration methods (intravenous and intratumoral administration) of rVSV-miR143 with those of VSV, a comparative analysis of primary tumor volume, metastatic lesions and survival rate was performed in mouse models of osteosarcoma. RESULTS: Following intratumoral injection, rVSV-miR143 showed a significant reduction in primary tumor volume, but no significant difference was observed in metastatic lesions and survival rate compared to VSV. Following intravenous injection, rVSV-miR143 revealed no significant difference in primary tumor volume, metastatic lesion and survival rate compared to VSV. CONCLUSION: VSV incorporating tumor-suppressor miRNA143 demonstrated a slightly synergistic antitumor effect on osteosarcoma in vivo.


Asunto(s)
Neoplasias Óseas , MicroARNs , Virus Oncolíticos , Osteosarcoma , Vesiculovirus , Animales , Ratones , Neoplasias Óseas/genética , Neoplasias Óseas/terapia , Modelos Animales de Enfermedad , MicroARNs/genética , MicroARNs/uso terapéutico , Osteosarcoma/genética , Osteosarcoma/terapia , Estomatitis Vesicular/virología , Virus Oncolíticos/metabolismo
7.
Proc Natl Acad Sci U S A ; 119(35): e2110105119, 2022 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-35994646

RESUMEN

The spike (S) protein of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the main target for neutralizing antibodies (NAbs). The S protein trimer is anchored in the virion membrane in its prefusion (preS) but metastable form. The preS protein has been stabilized by introducing two or six proline substitutions, to generate stabilized, soluble 2P or HexaPro (6P) preS proteins. Currently, it is not known which form is the most immunogenic. Here, we generated recombinant vesicular stomatitis virus (rVSV) expressing preS-2P, preS-HexaPro, and native full-length S, and compared their immunogenicity in mice and hamsters. The rVSV-preS-HexaPro produced and secreted significantly more preS protein compared to rVSV-preS-2P. Importantly, rVSV-preS-HexaPro triggered significantly more preS-specific serum IgG antibody than rVSV-preS-2P in both mice and hamsters. Antibodies induced by preS-HexaPro neutralized the B.1.1.7, B.1.351, P.1, B.1.427, and B.1.617.2 variants approximately two to four times better than those induced by preS-2P. Furthermore, preS-HexaPro induced a more robust Th1-biased cellular immune response than preS-2P. A single dose (104 pfu) immunization with rVSV-preS-HexaPro and rVSV-preS-2P provided complete protection against challenge with mouse-adapted SARS-CoV-2 and B.1.617.2 variant, whereas rVSV-S only conferred partial protection. When the immunization dose was lowered to 103 pfu, rVSV-preS-HexaPro induced two- to sixfold higher antibody responses than rVSV-preS-2P in hamsters. In addition, rVSV-preS-HexaPro conferred 70% protection against lung infection whereas only 30% protection was observed in the rVSV-preS-2P. Collectively, our data demonstrate that both preS-2P and preS-HexaPro are highly efficacious but preS-HexaPro is more immunogenic and protective, highlighting the advantages of using preS-HexaPro in the next generation of SARS-CoV-2 vaccines.


Asunto(s)
Prolina , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus , Desarrollo de Vacunas , Estomatitis Vesicular , Vacunas Virales , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , COVID-19/genética , COVID-19/inmunología , COVID-19/prevención & control , COVID-19/virología , Vacunas contra la COVID-19/inmunología , Cricetinae , Humanos , Ratones , Prolina/inmunología , SARS-CoV-2/genética , SARS-CoV-2/inmunología , Glicoproteína de la Espiga del Coronavirus/genética , Glicoproteína de la Espiga del Coronavirus/inmunología , Estomatitis Vesicular/inmunología , Estomatitis Vesicular/prevención & control , Estomatitis Vesicular/virología , Vesiculovirus/inmunología , Proteínas Virales/inmunología , Vacunas Virales/inmunología
8.
PLoS One ; 17(2): e0263065, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35108303

RESUMEN

The matrix (M) protein of vesicular stomatitis virus (VSV) has a complex role in infection and immune evasion, particularly with respect to suppression of Type I interferon (IFN). Viral strains bearing the wild-type (wt) M protein are able to suppress Type I IFN responses. We recently reported that the 22-25 strain of VSV encodes a wt M protein, however its sister plaque isolate, strain 22-20, carries a M[MD52G] mutation that perturbs the ability of the M protein to block NFκB, but not M-mediated inhibition of host transcription. Therefore, although NFκB is activated in 22-20 infected murine L929 cells infected, no IFN mRNA or protein is produced. To investigate the impact of the M[D52G] mutation on immune evasion by VSV, we used transcriptomic data from L929 cells infected with wt, 22-25, or 22-20 to define parameters in a family of executable logical models with the aim of discovering direct targets of viruses encoding a wt or mutant M protein. After several generations of pruning or fixing hypothetical regulatory interactions, we identified specific predicted targets of each strain. We predict that wt and 22-25 VSV both have direct inhibitory actions on key elements of the NFκB signaling pathway, while 22-20 fails to inhibit this pathway.


Asunto(s)
Biología Computacional/métodos , Fibroblastos/metabolismo , Proteínas Mutantes/metabolismo , FN-kappa B/metabolismo , Transcriptoma , Estomatitis Vesicular/metabolismo , Proteínas de la Matriz Viral/metabolismo , Animales , Fibroblastos/virología , Interferón Tipo I/metabolismo , Ratones , Proteínas Mutantes/genética , FN-kappa B/genética , Estomatitis Vesicular/genética , Estomatitis Vesicular/virología , Virus de la Estomatitis Vesicular Indiana/fisiología , Proteínas de la Matriz Viral/genética
9.
PLoS Pathog ; 18(2): e1010287, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35108335

RESUMEN

The unconventional mRNA capping enzyme (GDP polyribonucleotidyltransferase, PRNTase) domain of the vesicular stomatitis virus (VSV) L protein possesses a dual-functional "priming-capping loop" that governs terminal de novo initiation for leader RNA synthesis and capping of monocistronic mRNAs during the unique stop-start transcription cycle. Here, we investigated the roles of basic amino acid residues on a helix structure directly connected to the priming-capping loop in viral RNA synthesis and identified single point mutations that cause previously unreported defective phenotypes at different steps of stop-start transcription. Mutations of residue R1183 (R1183A and R1183K) dramatically reduced the leader RNA synthesis activity by hampering early elongation, but not terminal de novo initiation or productive elongation, suggesting that the mutations negatively affect escape from the leader promoter. On the other hand, mutations of residue R1178 (R1178A and R1178K) decreased the efficiency of polyadenylation-coupled termination of mRNA synthesis at the gene junctions, but not termination of leader RNA synthesis at the leader-to-N-gene junction, resulting in the generation of larger amounts of aberrant polycistronic mRNAs. In contrast, both the R1183 and R1178 residues are not essential for cap-forming activities. The R1183K mutation was lethal to VSV, whereas the R1178K mutation attenuated VSV and triggered the production of the polycistronic mRNAs in infected cells. These observations suggest that the PRNTase domain plays multiple roles in conducting accurate stop-start transcription beyond its known role in pre-mRNA capping.


Asunto(s)
Polirribonucleótido Nucleotidiltransferasa/metabolismo , ARN Viral/metabolismo , ARN Polimerasa Dependiente del ARN/genética , ARN Polimerasa Dependiente del ARN/metabolismo , Estomatitis Vesicular/virología , Virus de la Estomatitis Vesicular Indiana/genética , Virus de la Estomatitis Vesicular Indiana/metabolismo , Proteínas Virales/genética , Proteínas Virales/metabolismo , Sustitución de Aminoácidos , Animales , Línea Celular , ARN Polimerasas Dirigidas por ADN/genética , ARN Polimerasas Dirigidas por ADN/metabolismo , Mutación , Nucleotidiltransferasas/metabolismo , Polirribonucleótido Nucleotidiltransferasa/genética , Conformación Proteica , Dominios Proteicos , Precursores del ARN/metabolismo , Transcripción Genética , Replicación Viral
10.
Cell Rep ; 37(5): 109926, 2021 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-34731629

RESUMEN

Interferon regulatory factor 3 (IRF3) is an essential transductor for initiation of many immune responses. Here, we show that lncRNA-ISIR directly binds IRF3 to promote its phosphorylation, dimerization, and nuclear translocation, along with enhanced target gene productions. In vivo lncRNA-ISIR deficiency results in reduced IFN production, uncontrolled viral replication, and increased mortality. The human homolog, AK131315, also binds IRF3 and promotes its activation. More important, AK131315 expression is positively correlated with type I interferon (IFN-I) level and severity in patients with lupus. Mechanistically, in resting cells, IRF3 is bound to suppressor protein Flightless-1 (Fli-1), which keeps its inactive state. Upon infection, IFN-I-induced lncRNA-ISIR binds IRF3 at DNA-binding domain in cytoplasm and removes Fli-1's association from IRF3, consequently facilitating IRF3 activation. Our results demonstrate that IFN-I-inducible lncRNA-ISIR feedback strengthens IRF3 activation by removing suppressive Fli-1 in immune responses, revealing a method of lncRNA-mediated modulation of transcription factor (TF) activation.


Asunto(s)
Factor 3 Regulador del Interferón/metabolismo , Lupus Eritematoso Sistémico/metabolismo , Macrófagos Peritoneales/metabolismo , ARN Largo no Codificante/metabolismo , Estomatitis Vesicular/metabolismo , Animales , Estudios de Casos y Controles , Chlorocebus aethiops , Modelos Animales de Enfermedad , Silenciador del Gen , Células HEK293 , Humanos , Factor 3 Regulador del Interferón/genética , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/inmunología , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/virología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Proto-Oncogénica c-fli-1/genética , Proteína Proto-Oncogénica c-fli-1/metabolismo , Células RAW 264.7 , ARN Largo no Codificante/genética , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/metabolismo , Células Vero , Estomatitis Vesicular/genética , Estomatitis Vesicular/inmunología , Estomatitis Vesicular/virología , Virus de la Estomatitis Vesicular Indiana/inmunología , Virus de la Estomatitis Vesicular Indiana/patogenicidad
11.
Front Immunol ; 12: 730483, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34512666

RESUMEN

The antiviral innate immunity is the first line of host defense against viral infection. Mitochondrial antiviral signaling protein (MAVS, also named Cardif/IPS-1/VISA) is a critical protein in RNA virus-induced antiviral signaling pathways. Our previous research suggested that E3 ubiquitin-protein ligases RING-finger protein (RNF90) negatively regulate cellular antiviral responses by targeting STING for degradation, though its role in RNA virus infection remains unknown. This study demonstrated that RNF90 negatively regulated RNA virus-triggered antiviral innate immune responses in RNF90-silenced PMA-THP1 cells, RNF90-deficient cells (including HaCaTs, MEFs, and BMDMs), and RNF90-deficient mice. However, RNF90 regulated RNA virus-triggered antiviral innate immune responses independent of STING. RNF90 promoted K48-linked ubiquitination of MAVS and its proteasome-dependent degradation, leading to the inhibition of innate immune responses. Altogether, our findings suggested a novel function and mechanism of RNF90 in antiviral innate immunity.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Inmunidad Innata , Proteínas de Motivos Tripartitos/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Estomatitis Vesicular/metabolismo , Vesiculovirus/inmunología , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/inmunología , Animales , Chlorocebus aethiops , Citocinas/genética , Citocinas/inmunología , Citocinas/metabolismo , Células HEK293 , Células HaCaT , Interacciones Huésped-Patógeno , Humanos , Ratones Noqueados , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis , Transducción de Señal , Células THP-1 , Proteínas de Motivos Tripartitos/genética , Proteínas de Motivos Tripartitos/inmunología , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/inmunología , Ubiquitinación , Células Vero , Estomatitis Vesicular/genética , Estomatitis Vesicular/inmunología , Estomatitis Vesicular/virología , Vesiculovirus/patogenicidad
12.
Viruses ; 13(9)2021 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-34578344

RESUMEN

Lassa virus (LASV)-a member of the family Arenaviridae-causes Lassa fever in humans and is endemic in West Africa. Currently, no approved drugs are available. We screened 2480 small compounds for their potential antiviral activity using pseudotyped vesicular stomatitis virus harboring the LASV glycoprotein (VSV-LASVGP) and a related prototypic arenavirus, lymphocytic choriomeningitis virus (LCMV). Follow-up studies confirmed that CP100356 hydrochloride (CP100356), a specific P-glycoprotein (P-gp) inhibitor, suppressed VSV-LASVGP, LCMV, and LASV infection with half maximal inhibitory concentrations of 0.52, 0.54, and 0.062 µM, respectively, without significant cytotoxicity. Although CP100356 did not block receptor binding at the cell surface, it inhibited low-pH-dependent membrane fusion mediated by arenavirus glycoproteins. P-gp downregulation did not cause a significant reduction in either VSV-LASVGP or LCMV infection, suggesting that P-gp itself is unlikely to be involved in arenavirus entry. Finally, our data also indicate that CP100356 inhibits the infection by other mammarenaviruses. Thus, our findings suggest that CP100356 can be considered as an effective virus entry inhibitor for LASV and other highly pathogenic mammarenaviruses.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/efectos de los fármacos , Arenaviridae/metabolismo , Isoquinolinas/farmacología , Virus Lassa/efectos de los fármacos , Quinazolinas/farmacología , Internalización del Virus/efectos de los fármacos , Animales , Antivirales/farmacología , Chlorocebus aethiops , Humanos , Fiebre de Lassa/tratamiento farmacológico , Fiebre de Lassa/virología , Virus de la Coriomeningitis Linfocítica , Receptores Virales , Células Vero , Estomatitis Vesicular/virología , Inhibidores de Proteínas Virales de Fusión/farmacología
13.
mBio ; 12(4): e0046321, 2021 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-34340542

RESUMEN

Oropouche virus (OROV) infection of humans is associated with a debilitating febrile illness that can progress to meningitis or encephalitis. First isolated from a forest worker in Trinidad and Tobago in 1955, the arbovirus OROV has since been detected throughout the Amazon basin with an estimated 500,000 human infections over 60 years. Like other members of the family Peribunyaviridae, the viral genome exists as 3 single-stranded negative-sense RNA segments. The medium-sized segment encodes a viral glycoprotein complex (GPC) that is proteolytically processed into two viral envelope proteins, Gn and Gc, responsible for attachment and membrane fusion. There are no therapeutics or vaccines to combat OROV infection, and we have little understanding of protective immunity to infection. Here, we generated a replication competent chimeric vesicular stomatitis virus (VSV), in which the endogenous glycoprotein was replaced by the GPC of OROV. Serum from mice immunized by intramuscular injection with VSV-OROV specifically neutralized wild-type OROV, and using peptide arrays we mapped multiple epitopes within an N-terminal variable region of Gc recognized by the immune sera. VSV-OROV lacking this variable region of Gc was also immunogenic in mice producing neutralizing sera that recognize additional regions of Gc. Challenge of both sets of immunized mice with wild-type OROV shows that the VSV-OROV chimeras reduce wild-type viral infection and suggest that antibodies that recognize the variable N terminus of Gc afford less protection than those that target more conserved regions of Gc. IMPORTANCE Oropouche virus (OROV), an orthobunyavirus found in Central and South America, is an emerging public health challenge that causes debilitating febrile illness. OROV is transmitted by arthropods, and increasing mobilization has the potential to significantly increase the spread of OROV globally. Despite this, no therapeutics or vaccines have been developed to combat infection. Using vesicular stomatitis (VSV) as a backbone, we developed a chimeric virus bearing the OROV glycoproteins (VSV-OROV) and tested its ability to elicit a neutralizing antibody response. Our results demonstrate that VSV-OROV produces a strong neutralizing antibody response that is at least partially targeted to the N-terminal region of Gc. Importantly, vaccination with VSV-OROV reduces viral loads in mice challenged with wild-type virus. These data provide novel evidence that targeting the OROV glycoproteins may be an effective vaccination strategy to combat OROV infection.


Asunto(s)
Infecciones por Bunyaviridae/prevención & control , Genoma Viral , Orthobunyavirus/genética , Vesiculovirus/genética , Vesiculovirus/inmunología , Proteínas del Envoltorio Viral/genética , Animales , Anticuerpos Neutralizantes , Infecciones por Bunyaviridae/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Estomatitis Vesicular/virología , Replicación Viral
14.
Proc Natl Acad Sci U S A ; 118(29)2021 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-34266951

RESUMEN

Interferons induce cell-intrinsic responses associated with resistance to viral infection. To overcome the suppressive action of interferons and their effectors, viruses have evolved diverse mechanisms. Using vesicular stomatitis virus (VSV), we report that the host cell N6-adenosine messenger RNA (mRNA) cap methylase, phosphorylated C-terminal domain interacting factor 1 (PCIF1), attenuates the antiviral response. We employed cell-based and in vitro biochemical assays to demonstrate that PCIF1 efficiently modifies VSV mRNA cap structures to m7Gpppm6Am and define the substrate requirements for this modification. Functional assays revealed that the PCIF1-dependent modification of VSV mRNA cap structures is inert with regard to mRNA stability, translation, and viral infectivity but attenuates the antiviral effects of the treatment of cells with interferon-ß. Cells lacking PCIF1 or expressing a catalytically inactive PCIF1 exhibit an augmented inhibition of viral replication and gene expression following interferon-ß treatment. We further demonstrate that the mRNA cap structures of rabies and measles viruses are also modified by PCIF1 to m7Gpppm6Am This work identifies a function of PCIF1 and cap-proximal m6Am in attenuation of the host response to VSV infection that likely extends to other viruses.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Interferón beta/inmunología , Proteínas Nucleares/metabolismo , Caperuzas de ARN/metabolismo , ARN Mensajero/metabolismo , ARN Viral/metabolismo , Estomatitis Vesicular/inmunología , Virus de la Estomatitis Vesicular Indiana/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/inmunología , Interacciones Huésped-Patógeno , Humanos , Interferón beta/genética , Metilación , Proteínas Nucleares/genética , Proteínas Nucleares/inmunología , Caperuzas de ARN/genética , Estabilidad del ARN , ARN Mensajero/química , ARN Mensajero/genética , ARN Viral/química , ARN Viral/genética , Estomatitis Vesicular/genética , Estomatitis Vesicular/metabolismo , Estomatitis Vesicular/virología , Virus de la Estomatitis Vesicular Indiana/química , Virus de la Estomatitis Vesicular Indiana/genética , Replicación Viral
15.
Front Immunol ; 12: 595390, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33995343

RESUMEN

Replication competent vesicular stomatitis virus (VSV) is the basis of a vaccine against Ebola and VSV strains are developed as oncolytic viruses. Both functions depend on the ability of VSV to induce adequate amounts of interferon-α/ß. It is therefore important to understand how VSV triggers interferon responses. VSV activates innate immunity via retinoic acid-inducible gene I (RIG-I), a sensor for viral RNA. Our results show that VSV needs to replicate for a robust interferon response. Analysis of RIG-I-associated RNA identified a copy-back defective-interfering (DI) genome and full-length viral genomes as main trigger of RIG-I. VSV stocks depleted of DI genomes lost most of their interferon-stimulating activity. The remaining full-length genome and leader-N-read-through sequences, however, still triggered RIG-I. Awareness for DI genomes as trigger of innate immune responses will help to standardize DI genome content and to purposefully deplete or use DI genomes as natural adjuvants in VSV-based therapeutics.


Asunto(s)
Proteína 58 DEAD Box/metabolismo , Genoma Viral , Mutación , Receptores Inmunológicos/metabolismo , Estomatitis Vesicular/metabolismo , Estomatitis Vesicular/virología , Virus de la Estomatitis Vesicular Indiana/fisiología , Replicación Viral , Animales , Línea Celular , Genoma Viral/genética , Genoma Viral/inmunología , Interacciones Huésped-Patógeno , Humanos , Inmunomodulación , ARN Viral/genética , ARN Viral/inmunología
16.
Rev. bras. ciênc. vet ; 28(2): 97-101, abr./jun. 2021. il.
Artículo en Portugués | LILACS, VETINDEX | ID: biblio-1367729

RESUMEN

Objetivou-se investigar a presença do Vírus da Estomatite Vesicular (VEV) e seus fatores de risco para ocorrência e disseminação da enfermidade em equídeos das mesorregiões Leste e Oeste Potiguar do estado do Rio Grande do Norte, Brasil. Foram analisadas pela técnica de virusneutralização, 809 amostras sanguíneas de equídeos provenientes de noventa propriedades de dezesseis municípios Potiguares durante os meses de julho de 2018 a fevereiro de 2019. Os fatores de riscos associados ao VEV foram avaliados por meio de questionário epidemiológico e os dados submetidos a análise estatística no programa IBM SPSS Statistics versão 21.0 com nível de confiança de 95%. Posteriormente, todas as variáveis estatisticamente significantes foram submetidas a análise de regressão de Poisson. A soroprevalência de anticorpos anti-VEV foi 24,6% (199/809), sendo 3,2% (13/402) de soropositivos na mesorregião Leste e 45,7% (186/407) na do Oeste Potiguar. Com relação aos sorotipos, observou-se uma prevalência de 3,8% (31/809) e 24,5% (198/809) para Indiana 2 e 3 respectivamente, com 15,1% (30/198) de coinfecção. Equídeos criados na mesorregião Oeste, em propriedades que não realizam quarentena e onde os animais enfermos são mantidos no rebanho, foram consideradas fatores predisponentes a infecção pelo VEV. Esses resultados demonstram a circulação do VEV em equídeos no Rio Grande do Norte, com destaque ao Oeste Potiguar, e sendo necessário a aplicação de medidas sanitárias que impeçam introdução e disseminação do vírus ente as espécies susceptíveis, principalmente em condições climáticas favoráveis para a sua manutenção, no ambiente de criação e pastagens.


This study aimed to investigate the presence of Vesicular stomatitis virus (VSV) and risk factors for its occurrence and dissemination in equines from the Eastern and Western mesoregions of the state of Rio Grande do Norte, Brazil. Blood samples were analyzed, by Serum Virus Neutralization Assay, from 809 animals belonging to 90 properties distributed in sixteen municipalities from July 2018 to February 2019. Risk factors were assessed using an epidemiological questionnaire. Data were submitted to statistical analysis using the software IBM SPSS Statistics, version 21.0 with a 95% confidence level. Also, all statistically significant variables were subjected to Poisson regression analysis. The occurrence of anti-VSV antibodies was 24.6% (199/809) with 3.2% (13/402) and 45.7% (186/407) of seropositivity in the Western and Eastern mesoregion, respectively. Regarding serotypes, there were an occurrence of 3.8% (31/809) and 24.5% (198/809) for Indiana 2 and 3, respectively, and 15.1% (30/198) of co-infection for both. Equines kept of the Western mesoregion, on properties that do not quarantine, and where sick animals are kept in the herd, were considered risk factors for LVV infection. These results demonstrate the presence of VSV in equines in Rio Grande do Norte, with emphasis on Oeste Potiguar, and that sanitary measures must be adopted to prevent the introduction and viral spreading among susceptible species, especially due to favorable climatic conditions for the maintenance of VSV in the breeding and pasture environment.


Asunto(s)
Animales , Virus de la Estomatitis Vesicular Indiana , Enfermedades de los Caballos/virología , Factores de Riesgo , Estomatitis Vesicular/virología , Anticuerpos Antivirales/análisis
17.
Rev. bras. ciênc. vet ; 28(2): 97-101, abr./jun. 2021. map, tab
Artículo en Portugués | LILACS, VETINDEX | ID: biblio-1491709

RESUMEN

Objetivou-se investigar a presença do Vírus da Estomatite Vesicular (VEV) e seus fatores de risco para ocorrência e disseminação da enfermidade em equídeos das mesorregiões Leste e Oeste Potiguar do estado do Rio Grande do Norte, Brasil. Foram analisadas pela técnica de virusneutralização, 809 amostras sanguíneas de equídeos provenientes de noventa propriedades de dezesseis municípios Potiguares durante os meses de julho de 2018 a fevereiro de 2019. Os fatores de riscos associados ao VEV foram avaliados por meio de questionário epidemiológico e os dados submetidos a análise estatística no programa IBM SPSS Statistics versão 21.0 com nível de confiança de 95%. Posteriormente, todas as variáveis estatisticamente significantes foram submetidas a análise de regressão de Poisson. A soroprevalência de anticorpos anti-VEV foi 24,6% (199/809), sendo 3,2% (13/402) de soropositivos na mesorregião Leste e 45,7% (186/407) na do Oeste Potiguar. Com relação aos sorotipos, observou-se uma prevalência de 3,8% (31/809) e 24,5% (198/809) para Indiana 2 e 3 respectivamente, com 15,1% (30/198) de coinfecção. Equídeos criados na mesorregião Oeste, em propriedades que não realizam quarentena e onde os animais enfermos são mantidos no rebanho, foram consideradas fatores predisponentes a infecção pelo VEV. Esses resultados demonstram a circulação do VEV em equídeos no Rio Grande do Norte, com destaque ao Oeste Potiguar, e sendo necessário a aplicação de medidas sanitárias que impeçam introdução e disseminação do vírus ente as espécies susceptíveis, principalmente em condições climáticas favoráveis para a sua manutenção, no ambiente de criação e pastagens.


This study aimed to investigate the presence of Vesicular stomatitis virus (VSV) and risk factors for its occurrence and dissemination in equines from the Eastern and Western mesoregions of the state of Rio Grande do Norte, Brazil. Blood samples were analyzed, by Serum Virus Neutralization Assay, from 809 animals belonging to 90 properties distributed in sixteen municipalities from July 2018 to February 2019. Risk factors were assessed using an epidemiological questionnaire. Data were submitted to statistical analysis using the software IBM SPSS Statistics, version 21.0 with a 95% confidence level. Also, all statistically significant variables were subjected to Poisson regression analysis. The occurrence of anti-VSV antibodies was 24.6% (199/809) with 3.2% (13/402) and 45.7% (186/407) of seropositivity in the Western and Eastern mesoregion, respectively. Regarding serotypes, there were an occurrence of 3.8% (31/809) and 24.5% (198/809) for Indiana 2 and 3, respectively, and 15.1% (30/198) of co-infection for both. Equines kept of the Western mesoregion, on properties that do not quarantine, and where sick animals are kept in the herd, were considered risk factors for LVV infection. These results demonstrate the presence of VSV in equines in Rio Grande do Norte, with emphasis on Oeste Potiguar, and that sanitary measures must be adopted to prevent the introduction and viral spreading among susceptible species, especially due to favorable climatic conditions for the maintenance of VSV in the breeding and pasture environment.


Asunto(s)
Animales , Caballos , Caballos/virología , Estomatitis Vesicular/virología , Factores Biológicos/análisis , Factores de Riesgo , Infecciones por Rhabdoviridae/diagnóstico
18.
Emerg Microbes Infect ; 10(1): 651-663, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33719915

RESUMEN

ABSTRACTThe recent impact of Ebola virus disease (EVD) on public health in Africa clearly demonstrates the need for a safe and efficacious vaccine to control outbreaks and mitigate its threat to global health. ERVEBO® is an effective recombinant Vesicular Stomatitis Virus (VSV)-vectored Ebola virus vaccine (VSV-EBOV) that was approved by the FDA and EMA in late 2019 for use in prevention of EVD. Since the parental virus VSV, which was used to construct VSV-EBOV, is pathogenic for livestock and the vaccine virus may be shed at low levels by vaccinated humans, widespread deployment of the vaccine requires investigation into its infectivity and transmissibility in VSV-susceptible livestock species. We therefore performed a comprehensive clinical analysis of the VSV-EBOV vaccine virus in swine to determine its infectivity and potential for transmission. A high dose of VSV-EBOV resulted in VSV-like clinical signs in swine, with a proportion of pigs developing ulcerative vesicular lesions at the nasal injection site and feet. Uninoculated contact control pigs co-mingled with VSV-EBOV-inoculated pigs did not become infected or display any clinical signs of disease, indicating the vaccine is not readily transmissible to naïve pigs during prolonged close contact. In contrast, virulent wild-type VSV Indiana had a shorter incubation period and was transmitted to contact control pigs. These results indicate that the VSV-EBOV vaccine causes vesicular illness in swine when administered at a high dose. Moreover, the study demonstrates the VSV-EBOV vaccine is not readily transmitted to uninfected pigs, encouraging its safe use as an effective human vaccine.


Asunto(s)
Vacunas contra el Virus del Ébola/efectos adversos , Vacunas contra el Virus del Ébola/inmunología , Ebolavirus/inmunología , Estomatitis Vesicular/transmisión , Estomatitis Vesicular/virología , Virus de la Estomatitis Vesicular Indiana/inmunología , Vesiculovirus/inmunología , África , Animales , Chlorocebus aethiops , Ebolavirus/genética , Femenino , Fiebre Hemorrágica Ebola/inmunología , Fiebre Hemorrágica Ebola/virología , Humanos , Masculino , Modelos Animales , ARN Viral , Porcinos , Vacunación/métodos , Vacunas Sintéticas/efectos adversos , Vacunas Sintéticas/inmunología , Células Vero , Vesiculovirus/genética
19.
Cell Rep ; 34(9): 108798, 2021 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-33657363

RESUMEN

Type I interferons (IFNs) induce hundreds of IFN-stimulated genes (ISGs) in response to viral infection. Induction of these ISGs must be regulated for an efficient and controlled antiviral response, but post-transcriptional controls of these genes have not been well defined. Here, we identify a role for the RNA base modification N6-methyladenosine (m6A) in the regulation of ISGs. Using ribosome profiling and quantitative mass spectrometry, coupled with m6A-immunoprecipitation and sequencing, we identify a subset of ISGs, including IFITM1, whose translation is enhanced by m6A and the m6A methyltransferase proteins METTL3 and METTL14. We further determine that the m6A reader YTHDF1 increases the expression of IFITM1 in an m6A-binding-dependent manner. Importantly, we find that the m6A methyltransferase complex promotes the antiviral activity of type I IFN. Thus, these studies identify m6A as having a role in post-transcriptional control of ISG translation during the type I IFN response for antiviral restriction.


Asunto(s)
Adenosina/análogos & derivados , Biosíntesis de Proteínas , Procesamiento Postranscripcional del ARN , Transcripción Genética , Estomatitis Vesicular/genética , Vesiculovirus/patogenicidad , Células A549 , Adenosina/metabolismo , Animales , Antígenos de Diferenciación/biosíntesis , Antígenos de Diferenciación/genética , Antivirales/farmacología , Chlorocebus aethiops , Células HEK293 , Interacciones Huésped-Patógeno , Humanos , Interferón beta/farmacología , Metiltransferasas/biosíntesis , Metiltransferasas/genética , Biosíntesis de Proteínas/efectos de los fármacos , Procesamiento Postranscripcional del ARN/efectos de los fármacos , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Transcripción Genética/efectos de los fármacos , Células Vero , Estomatitis Vesicular/metabolismo , Estomatitis Vesicular/virología , Vesiculovirus/crecimiento & desarrollo , Replicación Viral
20.
Virology ; 556: 124-132, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33561699

RESUMEN

The heartland virus (HRTV) is a novel phlebovirus that causes severe infections in the USA and closely related to the severe fever thrombocytopenia syndrome virus (SFTSV), a causative agent for SFTS in Asia. The entry mechanisms of HRTV remain unclear. Here, we developed the pseudotyped vesicular stomatitis virus bearing the HRTV glycoprotein (GP) (HRTVpv), and the antigenicity and the entry mechanisms of HRTV were analyzed. HRTVpv was neutralized by anti-SFTSV Gc antibody, but not the anti-SFTSV Gn antibodies. Entry of HRTVpv to cells was inhibited by bafilomycin A1 and dynasore, and but it was enhanced in cells overexpressed with C-type lectins. Production of infectious HRTVpv and SFTSVpv was reduced by Nn-DNJ, α-glucosidase inhibitor. The entry of HRTV occurs via pH- and dynamin-dependent endocytosis. Furthermore, Nn-DNJ may be a possible therapeutic agent against HRTV and SFTSV.


Asunto(s)
Infecciones por Bunyaviridae/virología , Phlebovirus/patogenicidad , Estomatitis Vesicular/virología , Vesiculovirus/patogenicidad , Proteínas del Envoltorio Viral/metabolismo , 1-Desoxinojirimicina/análogos & derivados , 1-Desoxinojirimicina/farmacología , Animales , Línea Celular , Cricetinae , Haplorrinos , Humanos , Ratones , Internalización del Virus
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA