Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
1.
PLoS Pathog ; 20(7): e1012348, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-39008518

RESUMEN

Severe fever with thrombocytopenia syndrome (SFTS) virus, a tick-borne bunyavirus, causes a severe/fatal disease termed SFTS; however, the viral virulence is not fully understood. The viral non-structural protein, NSs, is the sole known virulence factor. NSs disturbs host innate immune responses and an NSs-mutant SFTS virus causes no disease in an SFTS animal model. The present study reports a novel determinant of viral tropism as well as virulence in animal models, within the glycoprotein (GP) of SFTS virus and an SFTS-related tick-borne bunyavirus. Infection with mutant SFTS viruses lacking the N-linked glycosylation of GP resulted in negligible usage of calcium-dependent lectins in cells, less efficient infection, high susceptibility to a neutralizing antibody, low cytokine production in macrophage-like cells, and reduced virulence in Ifnar-/- mice, when compared with wildtype virus. Three SFTS virus-related bunyaviruses had N-glycosylation motifs at similar positions within their GP and a glycan-deficient mutant of Heartland virus showed in vitro and in vivo phenotypes like those of the SFTS virus. Thus, N-linked glycosylation of viral GP is a novel determinant for the tropism and virulence of SFTS virus and of a related virus. These findings will help us understand the process of severe/fatal diseases caused by tick-borne bunyaviruses.


Asunto(s)
Glicoproteínas , Phlebovirus , Tropismo Viral , Animales , Glicosilación , Ratones , Virulencia , Phlebovirus/patogenicidad , Phlebovirus/genética , Glicoproteínas/metabolismo , Glicoproteínas/genética , Humanos , Síndrome de Trombocitopenia Febril Grave/virología , Ratones Endogámicos C57BL , Infecciones por Bunyaviridae/virología , Infecciones por Bunyaviridae/metabolismo , Garrapatas/virología , Ratones Noqueados , Orthobunyavirus/patogenicidad , Orthobunyavirus/genética , Orthobunyavirus/metabolismo
2.
Arch Virol ; 169(6): 133, 2024 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-38829449

RESUMEN

Akabane virus (AKAV), Aino virus, Peaton virus, Sathuperi virus, and Shamonda virus are arthropod-borne viruses belonging to the order Elliovirales, family Peribunyaviridae, genus Orthobunyavirus. These viruses cause or may cause congenital malformations in ruminants, including hydranencephaly, poliomyelitis, and arthrogryposis, although their pathogenicity may vary among field cases. AKAV may cause relatively severe congenital lesions such as hydranencephaly in calves. Furthermore, strains of AKAV genogroups I and II exhibit different disease courses. Genogroup I strains predominantly cause postnatal viral encephalomyelitis, while genogroup II strains are primarily detected in cases of congenital malformation. However, the biological properties of AKAV and other orthobunyaviruses are insufficiently investigated in hosts in the field and in vitro. Here, we used an immortalized bovine brain cell line (FBBC-1) to investigate viral replication efficiency, cytopathogenicity, and host innate immune responses. AKAV genogroup II and Shamonda virus replicated to higher titers in FBBC-1 cells compared with the other viruses, and only AKAV caused cytopathic effects. These results may be associated with the severe congenital lesions in the brain caused by AKAV genogroup II. AKAV genogroup II strains replicated to higher titers in FBBC-1 cells than AKAV genogroup I strains, suggesting that genogroup II strains replicated more efficiently in fetal brain cells, accounting for the detection of the latter strains mainly in fetal infection cases. Therefore, FBBC-1 cells may serve as a valuable tool for investigating the virulence and tropism of the orthobunyaviruses for bovine neonatal brain tissues in vitro.


Asunto(s)
Encéfalo , Infecciones por Bunyaviridae , Orthobunyavirus , Replicación Viral , Animales , Bovinos , Orthobunyavirus/patogenicidad , Orthobunyavirus/genética , Orthobunyavirus/fisiología , Orthobunyavirus/clasificación , Encéfalo/virología , Encéfalo/patología , Línea Celular , Infecciones por Bunyaviridae/virología , Infecciones por Bunyaviridae/veterinaria , Infecciones por Bunyaviridae/patología , Enfermedades de los Bovinos/virología , Feto/virología , Efecto Citopatogénico Viral , Inmunidad Innata
3.
Lancet Infect Dis ; 24(7): e439-e452, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38281494

RESUMEN

Since its discovery in 1955, the incidence and geographical spread of reported Oropouche virus (OROV) infections have increased. Oropouche fever has been suggested to be one of the most important vector-borne diseases in Latin America. However, both literature on OROV and genomic sequence availability are scarce, with few contributing laboratories worldwide. Three reassortant OROV glycoprotein gene variants termed Iquitos, Madre de Dios, and Perdões virus have been described from humans and non-human primates. OROV predominantly causes acute febrile illness, but severe neurological disease such as meningoencephalitis can occur. Due to unspecific symptoms, laboratory diagnostics are crucial. Several laboratory tests have been developed but robust commercial tests are hardly available. Although OROV is mainly transmitted by biting midges, it has also been detected in several mosquito species and a wide range of vertebrate hosts, which likely facilitates its widespread emergence. However, potential non-human vertebrate reservoirs have not been systematically studied. Robust animal models to investigate pathogenesis and immune responses are not available. Epidemiology, pathogenesis, transmission cycle, cross-protection from infections with OROV reassortants, and the natural history of infection remain unclear. This Review identifies Oropouche fever as a neglected disease and offers recommendations to address existing knowledge gaps, enable risk assessments, and ensure effective public health responses.


Asunto(s)
Infecciones por Bunyaviridae , Humanos , Animales , América Latina/epidemiología , Infecciones por Bunyaviridae/epidemiología , Infecciones por Bunyaviridae/transmisión , Infecciones por Bunyaviridae/diagnóstico , Infecciones por Bunyaviridae/virología , Orthobunyavirus/genética , Orthobunyavirus/patogenicidad , Orthobunyavirus/aislamiento & purificación , Enfermedades Transmisibles Emergentes/epidemiología , Enfermedades Transmisibles Emergentes/virología
4.
BMC Vet Res ; 17(1): 349, 2021 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-34781948

RESUMEN

BACKGROUND: Akabane virus (AKAV) is an important insect-borne virus which is widely distributed throughout the world except the Europe and is considered as a great threat to herbivore health. RESULTS: An AKAV strain defined as TJ2016 was firstly isolated from the bovine sera in China in 2016. Sequence analysis of the S and M segments suggested that the isolated AKAV strain was closely related to the AKAV strains JaGAr39 and JaLAB39, which belonged to AKAV genogroup II. To further study the pathogenic mechanism of AKAV, the full-length cDNA clone of TJ2016 S, M, and L segment was constructed separately into the TVT7R plasmid at the downsteam of T7 promoter and named as TVT7R-S, TVT7R-M, and TVT7R-L, respectively. The above three plasmids were further transfected into the BSR-T7/5 cells simultaneously with a ratio of 1:1:1 to produce the rescued virus AKAV. Compared with the parental wild type AKAV (wtAKAV), the rescued virus (rAKAV) was proved to be with similar cytopathic effects (CPE), plaque sizes and growth kinetics in BHK-21 cells. CONCLUSION: We successfully isolated a AKAV strain TJ2016 from the sera of cattle and established a reverse genetic platform for AKAV genome manipulation. The established reverse genetic system is also a powerful tool for further research on AKAV pathogenesis and even vaccine studies.


Asunto(s)
Infecciones por Bunyaviridae/veterinaria , Orthobunyavirus/genética , Orthobunyavirus/aislamiento & purificación , Animales , Infecciones por Bunyaviridae/virología , Bovinos , Enfermedades de los Bovinos/virología , Línea Celular , Cricetinae , Genotipo , Orthobunyavirus/patogenicidad , Filogenia , Genética Inversa/veterinaria
5.
Viruses ; 13(10)2021 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-34696477

RESUMEN

Severe fever with thrombocytopenia syndrome virus (SFTSV) is an emerging tick-borne bunyavirus in Asia that causes severe disease. Despite its clinical importance, treatment options for SFTSV infection remains limited. The SFTSV glycoprotein Gn plays a major role in mediating virus entry into host cells and is therefore a potential antiviral target. In this study, we employed an in silico structure-based strategy to design novel cyclic antiviral peptides that target the SFTSV glycoprotein Gn. Among the cyclic peptides, HKU-P1 potently neutralizes the SFTSV virion. Combinatorial treatment with HKU-P1 and the broad-spectrum viral RNA-dependent RNA polymerase inhibitor favipiravir exhibited synergistic antiviral effects in vitro. The in silico peptide design platform in this study may facilitate the generation of novel antiviral peptides for other emerging viruses.


Asunto(s)
Péptidos/farmacología , Phlebovirus/efectos de los fármacos , Síndrome de Trombocitopenia Febril Grave/tratamiento farmacológico , Antivirales/farmacología , Infecciones por Bunyaviridae/virología , Línea Celular , Línea Celular Tumoral , Simulación por Computador , Hong Kong , Humanos , Orthobunyavirus/patogenicidad , Phlebovirus/patogenicidad , Síndrome de Trombocitopenia Febril Grave/metabolismo , Síndrome de Trombocitopenia Febril Grave/virología , Trombocitopenia/virología , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/metabolismo , Internalización del Virus/efectos de los fármacos
6.
Commun Biol ; 4(1): 345, 2021 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-33753850

RESUMEN

Bunyaviruses have a genome that is divided over multiple segments. Genome segmentation complicates the generation of progeny virus, since each newly formed virus particle should preferably contain a full set of genome segments in order to disseminate efficiently within and between hosts. Here, we combine immunofluorescence and fluorescence in situ hybridization techniques to simultaneously visualize bunyavirus progeny virions and their genomic content at single-molecule resolution in the context of singly infected cells. Using Rift Valley fever virus and Schmallenberg virus as prototype tri-segmented bunyaviruses, we show that bunyavirus genome packaging is influenced by the intracellular viral genome content of individual cells, which results in greatly variable packaging efficiencies within a cell population. We further show that bunyavirus genome packaging is more efficient in insect cells compared to mammalian cells and provide new insights on the possibility that incomplete particles may contribute to bunyavirus spread as well.


Asunto(s)
Insectos/virología , Orthobunyavirus/genética , Ribonucleoproteínas/genética , Empaquetamiento del Genoma Viral , Proteínas Virales/genética , Virión/metabolismo , Animales , Chlorocebus aethiops , Técnica del Anticuerpo Fluorescente , Hibridación Fluorescente in Situ , Orthobunyavirus/metabolismo , Orthobunyavirus/patogenicidad , Ribonucleoproteínas/metabolismo , Virus de la Fiebre del Valle del Rift/genética , Virus de la Fiebre del Valle del Rift/metabolismo , Virus de la Fiebre del Valle del Rift/patogenicidad , Células Vero , Proteínas Virales/metabolismo , Virión/genética
7.
Sci Rep ; 10(1): 18725, 2020 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-33230115

RESUMEN

Schmallenberg virus (SBV), an arthropod-transmitted pathogenic bunyavirus, continues to be a threat to the European livestock industry, causing morbidity and mortality among young ruminant livestock. Here, we describe a novel SBV subunit vaccine, based on bacterially expressed SBV nucleoprotein (SBV-N) administered with a veterinary-grade Saponin adjuvant. When assayed in an IFNAR-/- mouse model, SBV-N with Saponin induced strong non-neutralizing broadly virus-reactive antibodies, decreased clinical signs, as well as significantly reduced viremia. Vaccination assays also suggest that this level of immune protection is cell mediated, as evidenced by the lack of neutralizing antibodies, as well as interferon-γ secretion observed in vitro. Therefore, based on these results, bacterially expressed SBV-N, co-administered with veterinary-grade Saponin adjuvant may serve as a promising economical alternative to current SBV vaccines, and warrant further evaluation in large ruminant animal models. Moreover, we propose that this strategy may be applicable to other bunyaviruses.


Asunto(s)
Orthobunyavirus/inmunología , Orthobunyavirus/patogenicidad , Vacunas Virales/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Animales , Anticuerpos Antivirales/biosíntesis , Especificidad de Anticuerpos , Anticuerpos ampliamente neutralizantes/biosíntesis , Infecciones por Bunyaviridae/inmunología , Infecciones por Bunyaviridae/prevención & control , Infecciones por Bunyaviridae/veterinaria , Técnicas In Vitro , Interferón gamma/metabolismo , Activación de Linfocitos , Ratones , Ratones Noqueados , Orthobunyavirus/genética , Receptor de Interferón alfa y beta/deficiencia , Receptor de Interferón alfa y beta/genética , Rumiantes , Saponinas/administración & dosificación , Vacunas de Subunidad/administración & dosificación , Vacunas de Subunidad/genética , Vacunas de Subunidad/inmunología , Vacunas Virales/administración & dosificación , Vacunas Virales/genética
8.
Curr Opin Virol ; 44: 16-25, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32619950

RESUMEN

The Bunyavirales order is the largest group of RNA viruses, which includes important human and animal pathogens, that cause serious diseases. Licensed vaccines are often not available for many of these pathogens. The establishment of bunyavirus reverse genetics systems has facilitated the generation of recombinant infectious viruses, which have been employed as powerful tools for understanding bunyavirus biology and identifying important virulence factors. Technological advances in this area have enabled the development of novel strategies, including codon-deoptimization, viral genome rearrangement and single-cycle replicable viruses, for the generation of live-attenuated vaccine candidates. In this review, we have summarized the current knowledge of the bunyavirus reverse genetics approaches for the generation of live-attenuated vaccine candidates and their evaluation in animal models.


Asunto(s)
Orthobunyavirus/genética , Orthobunyavirus/inmunología , Genética Inversa/métodos , Vacunas Virales/genética , Animales , Modelos Animales de Enfermedad , Genoma Viral , Humanos , Ratones , Orthobunyavirus/patogenicidad , Vacunas Atenuadas/genética , Vacunas Atenuadas/inmunología , Vacunas Virales/inmunología , Factores de Virulencia/genética , Replicación Viral
9.
J Virol ; 94(17)2020 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-32522852

RESUMEN

Schmallenberg virus (SBV) is an insect-transmitted orthobunyavirus that can cause abortions and congenital malformations in the offspring of ruminants. Even though the two viral surface glycoproteins Gn and Gc are involved in host cell entry, the specific cellular receptors of SBV are currently unknown. Using genome-wide CRISPR-Cas9 forward screening, we identified 3'-phosphoadenosine 5'-phosphosulfate (PAPS) transporter 1 (PAPST1) as an essential factor for SBV infection. PAPST1 is a sulfotransferase involved in heparan sulfate proteoglycan synthesis encoded by the solute carrier family 35 member B2 gene (SLC35B2). SBV cell surface attachment and entry were largely reduced upon the knockout of SLC35B2, whereas the reconstitution of SLC35B2 in these cells fully restored their susceptibility to SBV infection. Furthermore, treatment of cells with heparinase diminished infection with SBV, confirming that heparan sulfate plays an important role in cell attachment and entry, although to various degrees, heparan sulfate was also found to be important to initiate infection by two other bunyaviruses, La Crosse virus and Rift Valley fever virus. Thus, PAPST1-triggered synthesis of cell surface heparan sulfate is required for the efficient replication of SBV and other bunyaviruses.IMPORTANCE SBV is a newly emerging orthobunyavirus (family Peribunyaviridae) that has spread rapidly across Europe since 2011, resulting in substantial economic losses in livestock farming. In this study, we performed unbiased genome-wide CRISPR-Cas9 screening and identified PAPST1, a sulfotransferase encoded by SLC35B2, as a host entry factor for SBV. Consistent with its role in the synthesis of heparan sulfate, we show that this activity is required for efficient infection by SBV. A comparable dependency on heparan sulfate was also observed for La Crosse virus and Rift Valley fever virus, highlighting the importance of heparan sulfate for host cell infection by bunyaviruses. Thus, the present work provides crucial insights into virus-host interactions of important animal and human pathogens.


Asunto(s)
Infecciones por Bunyaviridae/genética , Infecciones por Bunyaviridae/virología , Sistemas CRISPR-Cas , Orthobunyavirus/genética , Orthobunyavirus/fisiología , Animales , Bunyaviridae , Chlorocebus aethiops , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Europa (Continente) , Técnicas de Inactivación de Genes , Células HEK293 , Heparitina Sulfato/metabolismo , Humanos , Ganado , Glicoproteínas de Membrana/genética , Orthobunyavirus/patogenicidad , Virus de la Fiebre del Valle del Rift , Transportadores de Sulfato/metabolismo , Sulfotransferasas/metabolismo , Células Vero , Acoplamiento Viral
10.
PLoS One ; 15(6): e0233722, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32479526

RESUMEN

Melon yellow spot orthotospovirus (MYSV), a member of the genus Orthotospovirus, is an important virus in cucurbits. Thrips palmi is considered the most serious pest of cucurbits because it directly damages and indirectly transmits MYSV to the plant. The effects of MYSV-infected plants on the development time, fecundity, and preference of the thrips were analyzed in this study. Our results showed that the development time of male and female thrips did not differ significantly between MYSV-infected and non-infected cucumbers. The survival rate of thrips in non-infected and MYSV-infected cucumbers were not significantly different. In a non-choice assay, T. palmi adults were released on non-infected and MYSV-infected cucumbers and allowed to lay eggs. The number of hatched larvae did not significantly differ between non-infected and MYSV-infected cucumbers. In a choice assay, MYSV had no detectable effect on the number of adult thrips and preceding hatched larvae. In a pull assay, the settling rate of thrips on the released plant did not differ significantly when the adult thrips were released to non-infected or MYSV infected cucumbers for any cucumber cultivar. Based on our results, we propose that the effects of MYSV-infected cucumbers on the development time, fecundity, or preference of T. palmi may not be an important factor in MYSV spread between cucumbers.


Asunto(s)
Cucumis sativus/parasitología , Orthobunyavirus/patogenicidad , Thysanoptera/fisiología , Animales , Femenino , Fertilidad , Especificidad del Huésped , Masculino , Thysanoptera/crecimiento & desarrollo , Thysanoptera/patogenicidad , Thysanoptera/virología
11.
Virus Genes ; 56(2): 150-167, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32076918

RESUMEN

The viruses historically implicated or currently considered as candidates for misuse in bioterrorist events are poxviruses, filoviruses, bunyaviruses, orthomyxoviruses, paramyxoviruses and a number of arboviruses causing encephalitis, including alpha- and flaviviruses. All these viruses are of concern for public health services when they occur in natural outbreaks or emerge in unvaccinated populations. Recent events and intelligence reports point to a growing risk of dangerous biological agents being used for nefarious purposes. Public health responses effective in natural outbreaks of infectious disease may not be sufficient to deal with the severe consequences of a deliberate release of such agents. One important aspect of countermeasures against viral biothreat agents are the antiviral treatment options available for use in post-exposure prophylaxis. These issues were adressed by the organizers of the 16th Medical Biodefense Conference, held in Munich in 2018, in a special session on the development of drugs to treat infections with viruses currently perceived as a threat to societies or associated with a potential for misuse as biothreat agents. This review will outline the state-of-the-art methods in antivirals research discussed and provide an overview of antiviral compounds in the pipeline that are already approved for use or still under development.


Asunto(s)
Antivirales/uso terapéutico , Arbovirus/efectos de los fármacos , Bioterrorismo/prevención & control , Virosis/tratamiento farmacológico , Arbovirus/patogenicidad , Filoviridae/efectos de los fármacos , Filoviridae/patogenicidad , Humanos , Orthobunyavirus/efectos de los fármacos , Orthobunyavirus/patogenicidad , Orthomyxoviridae/efectos de los fármacos , Orthomyxoviridae/patogenicidad , Paramyxovirinae/efectos de los fármacos , Paramyxovirinae/patogenicidad , Poxviridae/efectos de los fármacos , Poxviridae/patogenicidad , Virosis/virología
12.
BMC Res Notes ; 13(1): 67, 2020 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-32041646

RESUMEN

OBJECTIVE: To evaluate the frequency of infection caused by the Oropouche virus (OROV) in 496 patients with acute febrile disease (AFI), whose samples were obtained for the analysis of endemic arboviruses in a previous investigation carried out in 2016. RESULTS: OROV was detected in 26.4% (131/496) of serum samples from patients with AFI. Co-infections with Dengue virus (7.3%), Zika virus (1.8%) and Chikungunya (0.2%) were observed. The most common clinical symptoms reported among the patients with OROV infections were headache 85.5% (112/131), myalgia 80.9% (106/131), arthralgia 72.5% (95/131) and loss of appetite 67.9% (89/131). Headache and myalgia were predominant in all age groups. Both OROV infections and co-infections were more frequent in May, June and July corresponding to the dry season of the region.


Asunto(s)
Infecciones por Bunyaviridae/epidemiología , Infecciones por Bunyaviridae/fisiopatología , Fiebre Chikungunya/epidemiología , Coinfección , Dengue/epidemiología , Orthobunyavirus/patogenicidad , Infección por el Virus Zika/epidemiología , Adolescente , Adulto , Anciano , Infecciones por Bunyaviridae/sangre , Infecciones por Bunyaviridae/complicaciones , Fiebre Chikungunya/sangre , Niño , Preescolar , Comorbilidad , Dengue/sangre , Femenino , Fiebre/epidemiología , Fiebre/etiología , Humanos , Masculino , Persona de Mediana Edad , Perú/epidemiología , Adulto Joven , Infección por el Virus Zika/sangre
13.
Sci Transl Med ; 12(527)2020 01 22.
Artículo en Inglés | MEDLINE | ID: mdl-31969486

RESUMEN

Arthropod-borne viruses (arboviruses) are important human pathogens for which there are no specific antiviral medicines. The abundance of genetically distinct arbovirus species, coupled with the unpredictable nature of their outbreaks, has made the development of virus-specific treatments challenging. Instead, we have defined and targeted a key aspect of the host innate immune response to virus at the arthropod bite that is common to all arbovirus infections, potentially circumventing the need for virus-specific therapies. Using mouse models and human skin explants, we identify innate immune responses by dermal macrophages in the skin as a key determinant of disease severity. Post-exposure treatment of the inoculation site by a topical TLR7 agonist suppressed both the local and subsequent systemic course of infection with a variety of arboviruses from the Alphavirus, Flavivirus, and Orthobunyavirus genera. Clinical outcome was improved in mice after infection with a model alphavirus. In the absence of treatment, antiviral interferon expression to virus in the skin was restricted to dermal dendritic cells. In contrast, stimulating the more populous skin-resident macrophages with a TLR7 agonist elicited protective responses in key cellular targets of virus that otherwise proficiently replicated virus. By defining and targeting a key aspect of the innate immune response to virus at the mosquito bite site, we have identified a putative new strategy for limiting disease after infection with a variety of genetically distinct arboviruses.


Asunto(s)
Infecciones por Arbovirus/inmunología , Infecciones por Arbovirus/metabolismo , Arbovirus/inmunología , Arbovirus/patogenicidad , Macrófagos/metabolismo , Piel/citología , Alphavirus/inmunología , Alphavirus/patogenicidad , Animales , Flavivirus/inmunología , Flavivirus/patogenicidad , Humanos , Glicoproteínas de Membrana/metabolismo , Ratones , Orthobunyavirus/inmunología , Orthobunyavirus/patogenicidad , Receptor Toll-Like 7/metabolismo
14.
Viruses ; 11(8)2019 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-31443153

RESUMEN

Starting in 2006, bluetongue virus serotype 8 (BTV8) was responsible for a major epizootic in Western and Northern Europe. The magnitude and spread of the disease were surprisingly high and the control of BTV improved significantly with the marketing of BTV8 inactivated vaccines in 2008. During late summer of 2011, a first cluster of reduced milk yield, fever, and diarrhoea was reported in the Netherlands. Congenital malformations appeared in March 2012 and Schmallenberg virus (SBV) was identified, becoming one of the very few orthobunyaviruses distributed in Europe. At the start of both epizootics, little was known about the pathogenesis and epidemiology of these viruses in the European context and most assumptions were extrapolated based on other related viruses and/or other regions of the World. Standardized and repeatable models potentially mimicking clinical signs observed in the field are required to study the pathogenesis of these infections, and to clarify their ability to cross the placental barrier. This review presents some of the latest experimental designs for infectious disease challenges with BTV or SBV. Infectious doses, routes of infection, inoculum preparation, and origin are discussed. Particular emphasis is given to the placental crossing associated with these two viruses.


Asunto(s)
Virus de la Lengua Azul/patogenicidad , Lengua Azul/virología , Infecciones por Bunyaviridae/veterinaria , Modelos Animales de Enfermedad , Orthobunyavirus/patogenicidad , Placenta/virología , Rumiantes/virología , Animales , Virus de la Lengua Azul/genética , Virus de la Lengua Azul/fisiología , Infecciones por Bunyaviridae/virología , Femenino , Orthobunyavirus/genética , Orthobunyavirus/fisiología , Embarazo , Rumiantes/clasificación , Virulencia
15.
Microb Pathog ; 135: 103627, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31326560

RESUMEN

The Guama virus (GMAV) is a member of Peribunyaviridae family, Orthobunyavirus genus. Several strains of the virus were isolated in South and Central Americas from several hosts, such as humans, wild animals, including nonhuman primates, wild rodents and mosquitoes as well as mice used as sentinels. The virus is able to cause febrile disease in humans. Here we describe for the first time pathologic and biochemical findings in golden hamsters (Mesocricetus auratus) infected with the prototype GMAV. Blood and organs of infected and control animals were collected every 24 h after infection from the 1st to the 7th day post infection (dpi) and at 21 dpi when experiment was ended. The tissues were processed for histopathology and immunohistochemistry. The blood and serum were used to determine viremia and biochemical markers plus to detect anti-GMAV antibodies. The viremia was early detected already on the 1st dpi and it was no longer detected on the 3rd dpi. Total anti-GMAV antibodies were detected from the 6th dpi. Hepatic markers as ALT of infected animals were increased and showed statistically significant difference in comparison with control animals, indicating damage of the liver; indeed the liver was the most affected organ, but other organs presented lesions and positive GMAV immunostaining as brain, lung, liver, spleen, and kidney. Our findings indicate that golden hamsters are a good animal model for experimental infection of the GMAV.


Asunto(s)
Infecciones por Bunyaviridae/virología , Modelos Animales de Enfermedad , Orthobunyavirus/patogenicidad , Animales , Anticuerpos Antivirales/sangre , Infecciones por Bunyaviridae/sangre , Infecciones por Bunyaviridae/patología , Riñón/patología , Hígado/patología , Masculino , Mesocricetus , Bazo/patología , Viremia
16.
Virus Genes ; 55(2): 123-126, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30600431

RESUMEN

In recent reports about the molecular epidemiology of Schmallenberg virus (SBV), an orthobunyavirus affecting ruminants, it was proposed that the observed sequence variability within the viral M-segment might be higher in sheep than in cattle. However, these analyses are highly biased by the sample material from which the publicly available sequences were generated. While from cattle predominantly blood samples from acutely infected animals were studied, the vast majority of ovine samples originate from malformed fetuses or newborn lambs. Therefore, the observed sequence variability is misinterpreted since the samples from malformed fetuses and lambs do not reflect circulating SBV.


Asunto(s)
Infecciones por Bunyaviridae/virología , Orthobunyavirus/genética , Proteínas de la Matriz Viral/genética , Animales , Animales Recién Nacidos/virología , Infecciones por Bunyaviridae/genética , Infecciones por Bunyaviridae/veterinaria , Bovinos , Orthobunyavirus/patogenicidad , Rumiantes/virología , Ovinos/virología , Proteínas de la Matriz Viral/química
17.
Transbound Emerg Dis ; 66(1): 454-462, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30354028

RESUMEN

Since its first appearance in 2011, Schmallenberg virus (SBV) has been repeatedly detected in aborted ruminant foetuses or severely malformed newborns whose mothers were naturally infected during pregnancy. However, especially the knowledge about dynamics of foetal infection in cattle is still scarce. Therefore, a total of 36 pregnant heifers were experimentally infected during two animal trials with SBV between days 60 and 150 of gestation. The foetuses were collected between 10 and 35 days after infection and virologically and pathologically investigated. Overall, 33 heifers yielded normally developed, macroscopically inconspicuous foetuses, but abundant virus replication was evident at the maternal/foetal interface and viral genome was detectable in at least one organ system of 18 out of 35 foetuses. One heifer was found to be not pregnant at autopsy. One of the animals aborted at day 4 after infection, viral RNA was detectable in the lymphatic tissue of the dam, in the maternal and foetal placenta, and in organs and lymphatic tissue of the foetus. In another foetus, SBV typical malformations like torticollis and arthrogryposis were observed. The corresponding dam was infected at day 90 of pregnancy and viral genome was detectable in the cerebellum of the unborn. Interestingly, no common patterns of infected foetal organs or maternal/foetal placentas could be identified, and both, sites of virus replication and genome loads, varied to a high degree in the individual foetuses. It is therefore concluded, that SBV infects in many cases also the bovine foetus of naïve pregnant cattle, however, the experimentally observed low abortion/malformation rate is in concordance to the reported low rates in the field during the first outbreak wave following the introduction of SBV. This observation speaks for a natural resistance of most bovine foetuses even during the vulnerable phase of early pregnancy, which has to be further studied in the future.


Asunto(s)
Infecciones por Bunyaviridae/veterinaria , Enfermedades de los Bovinos/transmisión , Transmisión Vertical de Enfermedad Infecciosa/veterinaria , Orthobunyavirus/patogenicidad , Complicaciones Infecciosas del Embarazo/veterinaria , Feto Abortado/virología , Aborto Veterinario/virología , Animales , Infecciones por Bunyaviridae/transmisión , Infecciones por Bunyaviridae/virología , Bovinos , Enfermedades de los Bovinos/virología , Brotes de Enfermedades , Femenino , Feto/virología , Embarazo , Complicaciones Infecciosas del Embarazo/virología , Preñez , Rumiantes
18.
Braz. J. Vet. Res. Anim. Sci. (Online) ; 56(3): e154854, out. 2019. ilus
Artículo en Inglés | LILACS, VETINDEX | ID: biblio-1046878

RESUMEN

Akabane virus, a member of the Orthobunyavirus genus in the family Bunyaviridae, causes congenital abnormalities and arthrogryposis with hydrocephalus or hydroencephaly in ruminants. This study intends to describe the clinical signs, gross and histopathological features seen in 25 affected lambs in an outbreak of congenital arthrogryposis with hydrocephalus or hydranencephaly in Al-Muthanna governorate, Iraq after a large number of stillbirths and musculoskeletal deformities from October 2017 to May 2018. Skeletal muscle hypoplasia was seen in the limbs of the affected lambs accompanied with severe arthrogryposis and gross visible brain malformations. In addition, fetal mummifications, stillbirths, and dead lambs were also seen. The most histopathological features in muscle fibers were degenerative lesions and absences of cross-striation accompanied with mild infiltration of neutrophils and mononuclear cells in severely affected lambs. The meninges of affected lambs revealed fused membranes with focal areas of fibrous thickenings and necrotic debris. In conclusion, according to clinical signs, gross and histopathological investigations, Akabane virus, a member of the Orthobunyavirus genus in the family Bunyaviridae, causes congenital abnormalities and arthrogryposis with hydrocephalus or hydroencephaly in ruminants and could be the cause of this outbreak, although future studies must be performed to confirm the etiology of this outbreak. Moreover, other causes of hydrocephalus or cerebellar malformation, such as Schmallenberg virus, bluetongue virus and border disease virus and teratogenic plants that lead to arthrogryposis, have to be investigated. Also, the authorities should take prevention and control measurements to stop the replication of arthropod vectors.(AU)


O presente trabalho descreve os sinais clínicos, as lesões macroscópicas e os aspectos histológicos observados em 25 cordeiros acometidos em um surto de artrogripose congênita com hidrocéfalo ou hidrocefalia registrado no Iraque, governadoria Al-Muthama após a ocorrência de nascimentos prematuros e deformidades músculo-esqueléticas no período compreendido entre outubro de 2017 e maio de 2018. A hipoplasia músculo-esquelética foi observada nos membros dos cordeiros afetados, acompanhada de severa artrogripose e malformações cerebrais, grosseiras visíveis, além de mumificações fetais, nascimentos prematuros e morte de cordeiros. Os principais aspectos histopatológicos nas fibras musculares foram lesões degenerativas e ausências da estriação cruzada acompanhada de leve infiltração de neutrófilos e células mononucleares dos cordeiros severamente afetados. As meninges dos cordeiros afetados apresentaram fusão de membranas com áreas focais de espessamento fibroso e debris necróticos. O vírus Akabane, um membro do gênero Orthobunyavirus, da família Bunyaviridae, causa anormalidades congênitas e artrogripose com hidrocéfalo e hidrocefalia em ruminantes e poderá vir a ser a causa do presente surto. Os autores recomendam a realização de novos estudos com investigações epidemiológicas e isolamento do agente causal. Contudo, outras causas de hidrocéfalo ou malformações cerebrais como as determinadas pelo vírus Schmallenberg, vírus da língua azul e vírus da doença de border, bem como de plantas teratogênicas que determinam a artrogripose, também deverão ser investigadas. As autoridades sanitárias deverão tomar medidas de prevenção e controle para bloquear a replicação do vírus em artrópodes vetores.(AU)


Asunto(s)
Animales , Artrogriposis/veterinaria , Ovinos/anomalías , Orthobunyavirus/patogenicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA