Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.572
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
BMC Genomics ; 25(1): 715, 2024 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-39048939

RESUMEN

BF/C2 is a crucial molecule in the coagulation complement cascade pathway and plays a significant role in the immune response of grass carp through the classical, alternative, and lectin pathways during GCRV infection. In vivo experiments demonstrated that the mRNA expression levels of BF/C2 (A, B) in grass carp positively correlated with GCRV viral replication at various stages of infection. Excessive inflammation leading to death coincided with peak levels of BF/C2 (A, B) mRNA expression and GCRV viral replication. Correspondingly, BF/C2 (A, B) recombinant protein, CIK cells and GCRV co-incubation experiments yielded similar findings. Therefore, 3 h (incubation period) and 9 h (death period) were selected as critical points for this study. Transcriptome sequencing analysis revealed significant differences in the expression of BF/C2A and BF/C2B during different stages of CIK infection with GCRV and compared to the blank control group (PBS). Specifically, the BF/C2A_3 and BF/C2A_9 groups exhibited 2729 and 2228 differentially expressed genes (DEGs), respectively, with 1436 upregulated and 1293 downregulated in the former, and 1324 upregulated and 904 downregulated in the latter. The BF/C2B_3 and BF/C2B_9 groups showed 2303 and 1547 DEGs, respectively, with 1368 upregulated and 935 downregulated in the former, and 818 upregulated and 729 downregulated in the latter. KEGG functional enrichment analysis of these DEGs identified shared pathways between BF/C2A and PBS groups at 3 and 9 h, including the C-type lectin receptor signaling pathway, protein processing in the endoplasmic reticulum, Toll-like receptor signaling pathway, Salmonella infection, apoptosis, tight junction, and adipocytokine signaling pathway. Additionally, the BF/C2B groups at 3 and 9 h shared pathways related to protein processing in the endoplasmic reticulum, glycolysis/gluconeogenesis, and biosynthesis of amino acids. The mRNA levels of these DEGs were validated in cellular models, confirming consistency with the sequencing results. In addition, the mRNA expression levels of these candidate genes (mapk1, il1b, rela, nfkbiab, akt3a, hyou1, hsp90b1, dnajc3a et al.) in the head kidney, kidney, liver and spleen of grass carp immune tissue were significantly different from those of the control group by BF/C2 (A, B) protein injection in vivo. These candidate genes play an important role in the response of BF/C2 (A, B) to GCRV infection and it also further confirmed that BF/C2 (A, B) of grass carp plays an important role in coping with GCRV infection.


Asunto(s)
Carpas , Enfermedades de los Peces , Proteínas de Peces , Infecciones por Reoviridae , Reoviridae , Animales , Carpas/genética , Carpas/virología , Carpas/inmunología , Enfermedades de los Peces/virología , Enfermedades de los Peces/inmunología , Enfermedades de los Peces/genética , Infecciones por Reoviridae/veterinaria , Infecciones por Reoviridae/inmunología , Infecciones por Reoviridae/genética , Infecciones por Reoviridae/virología , Proteínas de Peces/genética , Proteínas de Peces/metabolismo , Reoviridae/fisiología , Perfilación de la Expresión Génica , Transcriptoma , Replicación Viral , Regulación de la Expresión Génica
2.
Viruses ; 16(7)2024 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-39066213

RESUMEN

In this study, we provide a genomic description of the first isolation of the Umattila virus (UMAV) in Brazil. The virus was obtained from the blood of a bird (Turdus fumigatus) and isolated in a C6/36 cell culture. The viral genome contains ten segments, and its organization is characteristic of viruses of the genus Orbivirus (family Sedoreoviridae). The coding region of each segment was sequenced, demonstrating the nucleotide identity with UMAV. The phylogenetic inference results were in line with these findings and demonstrated the formation of two distinct monophyletic clades containing strains isolated around the world, where our isolate, belonging to the same clade as the prototype strain, was allocated to a different subclade, highlighting the genetic divergence between them. This work reports the first isolation of UMAV in Brazil, and due to the scarcity of information on this viral agent in the scientific literature, it is essential to carry out further studies to better understand its epidemiology, dispersion, and, in particular, its interactions with vertebrate hosts, vectors, and the environment.


Asunto(s)
Genoma Viral , Orbivirus , Filogenia , Brasil , Animales , Orbivirus/aislamiento & purificación , Orbivirus/genética , Orbivirus/clasificación , Infecciones por Reoviridae/virología , Infecciones por Reoviridae/veterinaria , Aves/virología , Enfermedades de las Aves/virología , ARN Viral/genética , Línea Celular
3.
Viruses ; 16(7)2024 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-39066218

RESUMEN

Avian orthoreviruses have become a global challenge to the poultry industry, causing significant economic impacts on commercial poultry. Avian reoviruses (ARVs) are resistant to heat, proteolytic enzymes, a wide range of pH values, and disinfectants, so keeping chicken farms free of ARV infections is difficult. This review focuses on the global prevalence of ARVs and associated clinical signs and symptoms. The most common signs and symptoms include tenosynovitis/arthritis, malabsorption syndrome, runting-stunting syndrome, and respiratory diseases. Moreover, this review also focused on the characterization of ARVs in genotypic clusters (I-VI) and their relation to tissue tropism or viral distribution. The prevailing strains of ARV in Africa belong to all genotypic clusters (GCs) except for GC VI, whereas all GCs are present in Asia and the Americas. In addition, all ARV strains are associated with or belong to GC I-VI in Europe. Moreover, in Oceania, only GC V and VI are prevalent. This review also showed that, regardless of the genotypic cluster, tenosynovitis/arthritis was the predominant clinical manifestation, indicating its universal occurrence across all clusters. Globally, most avian reovirus infections can be prevented by vaccination against four major strains: S1133, 1733, 2408, and 2177. Nevertheless, these vaccines may not a provide sufficient defense against field isolates. Due to the increase in the number of ARV variants, classical vaccine approaches are being developed depending on the degree of antigenic similarity between the vaccine and field strains, which determines how successful the vaccination will be. Moreover, there is a need to look more closely at the antigenic and pathogenic properties of reported ARV strains. The information acquired will aid in the selection of more effective vaccine strains in combination with biosecurity and farm management methods to prevent ARV infections.


Asunto(s)
Genotipo , Orthoreovirus Aviar , Enfermedades de las Aves de Corral , Infecciones por Reoviridae , Orthoreovirus Aviar/genética , Orthoreovirus Aviar/clasificación , Animales , Enfermedades de las Aves de Corral/virología , Enfermedades de las Aves de Corral/epidemiología , Infecciones por Reoviridae/veterinaria , Infecciones por Reoviridae/epidemiología , Infecciones por Reoviridae/virología , Filogenia , Pollos/virología , Prevalencia , Aves de Corral/virología
4.
Front Immunol ; 15: 1419321, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39081319

RESUMEN

Similar to other RNA viruses, grass carp reovirus, the causative agent of the hemorrhagic disease, replicates in cytoplasmic viral inclusion bodies (VIBs), orchestrated by host proteins and lipids. The host pathways that facilitate the formation and function of GCRV VIBs are poorly understood. This work demonstrates that GCRV manipulates grass carp oxysterol binding protein 1 (named as gcOSBP1) and vesicle-associated membrane protein-associated protein A/B (named as gcVAP-A/B), 3 components of cholesterol transport pathway, to generate VIBs. By siRNA-mediated knockdown, we demonstrate that gcOSBP1 is an essential host factor for GCRV replication. We reveal that the nonstructural proteins NS80 and NS38 of GCRV interact with gcOSBP1, and that the gcOSBP1 is recruited by NS38 and NS80 for promoting the generation of VIBs. gcOSBP1 increases the expression of gcVAP-A/B and promotes the accumulation of intracellular cholesterol. gcOSBP1 also interacts with gcVAP-A/B for forming gcOSBP1-gcVAP-A/B complexes, which contribute to enhance the accumulation of intracellular cholesterol and gcOSBP1-mediated generation of VIBs. Inhibiting cholesterol accumulation by lovastatin can completely abolish the effects of gcOSBP1 and/or gcVAP-A/B in promoting GCRV infection, suggesting that cholesterol accumulation is vital for gcOSBP1- and/or gcVAP-A/B-mediated GCRV replication. Thus, our results, which highlight that gcOSBP1 functions in the replication of GCRV via its interaction with essential viral proteins for forming VIBs and with host gcVAP-A/B, provide key molecular targets for obtaining anti-hemorrhagic disease grass carp via gene editing technology.


Asunto(s)
Carpas , Colesterol , Cuerpos de Inclusión Viral , Receptores de Esteroides , Reoviridae , Replicación Viral , Animales , Reoviridae/fisiología , Carpas/virología , Carpas/metabolismo , Cuerpos de Inclusión Viral/metabolismo , Colesterol/metabolismo , Receptores de Esteroides/metabolismo , Enfermedades de los Peces/virología , Enfermedades de los Peces/metabolismo , Enfermedades de los Peces/inmunología , Interacciones Huésped-Patógeno , Infecciones por Reoviridae/veterinaria , Infecciones por Reoviridae/metabolismo , Infecciones por Reoviridae/virología , Proteínas de Peces/metabolismo , Proteínas de Peces/genética , Proteínas no Estructurales Virales/metabolismo , Proteínas no Estructurales Virales/genética
5.
Virulence ; 15(1): 2368080, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38899573

RESUMEN

Dendritic cells (DCs) present an ideal target for delivering immunogenic cargo due to their potent antigen-presenting capabilities. This targeting approach holds promise in vaccine development by enhancing the efficiency of antigen recognition and capture by DCs. To identify a high-affinity targeting peptide binding to rabbit DCs, rabbit monocyte-derived DCs (raMoDCs) were isolated and cultured, and a novel peptide, HS (HSLRHDYGYPGH), was identified using a phage-displayed peptide library. Alongside HS, two other DC-targeting peptides, KC1 and MY, previously validated in our laboratory, were employed to construct recombinant Lactgobacillus reuteri fusion-expressed rabbit hemorrhagic disease virus (RHDV) capsid protein VP60. These recombinant Lactobacillus strains were named HS-VP60/L. reuteri, KC1-VP60/L. reuteri, and MY-VP60/L. reuteri. The ability of these recombinant Lactobacillus to bind rabbit DCs was evaluated both in vivo and in vitro. Results demonstrated that the DC-targeting peptide KC1 significantly enhanced the capture efficiency of recombinant Lactobacillus by raMoDCs, promoted DC maturation, and increased cytokine secretion. Furthermore, oral administration of KC1-VP60/L. reuteri effectively induced SIgA and IgG production in rabbits, prolonged rabbit survival post-challenge, and reduced RHDV copies in organs. In summary, the DC-targeting peptide KC1 exhibited robust binding to raMoDCs, and recombinant Lactobacillus expressing KC1-VP60 protein antigens efficiently induced systemic and mucosal immune responses in rabbits, conferring protective efficacy against RHDV. This study offers valuable insights for the development of novel RHDV vaccines.


Asunto(s)
Células Dendríticas , Virus de la Enfermedad Hemorrágica del Conejo , Limosilactobacillus reuteri , Péptidos , Animales , Células Dendríticas/inmunología , Conejos , Virus de la Enfermedad Hemorrágica del Conejo/inmunología , Virus de la Enfermedad Hemorrágica del Conejo/genética , Limosilactobacillus reuteri/genética , Limosilactobacillus reuteri/inmunología , Péptidos/inmunología , Péptidos/genética , Infecciones por Caliciviridae/prevención & control , Infecciones por Caliciviridae/inmunología , Infecciones por Reoviridae/prevención & control , Infecciones por Reoviridae/inmunología , Proteínas de la Cápside/genética , Proteínas de la Cápside/inmunología , Vacunas Virales/inmunología , Vacunas Virales/genética , Lactobacillus/genética , Lactobacillus/inmunología
6.
Cell Host Microbe ; 32(6): 945-946, 2024 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-38870904

RESUMEN

In this issue of Cell Host & Microbe, Shang et al. identify murine neuropilin 1 as a host factor that binds reovirus particles, directing cell entry and contributing to viral dissemination and neurovirulence. This study highlights the reovirus model system to investigate host receptors and their significance in viral pathogenesis.


Asunto(s)
Neuronas , Neuropilina-1 , Reoviridae , Internalización del Virus , Animales , Ratones , Neuronas/virología , Neuropilina-1/metabolismo , Reoviridae/fisiología , Reoviridae/genética , Reoviridae/patogenicidad , Humanos , Interacciones Huésped-Patógeno , Infecciones por Reoviridae/virología , Receptores Virales/metabolismo
7.
Int J Biol Sci ; 20(8): 3076-3093, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38904031

RESUMEN

Epizootic hemorrhagic disease (EHD), caused by Epizootic hemorrhagic disease virus (EHDV), is an emerging and severe livestock disease. Recent incursion and distribution of EHDV in Europe have outlined the emerging character of EHD. Despite its worldwide impact, numerous knowledge gaps exist. A range of inconveniences restricts utilization of natural hosts of EHDV. Here, we show that adult mice deficient in type I IFN receptor (IFNAR(-/-)) are highly susceptible to EHDV-6 and EHDV-8 infection when the virus is administered subcutaneously. Disease was characterized by ruffled hair, reluctance to move, dehydration and conjunctivitis, with viraemia detected from day 5 post-infection. A deeper characterization of EHDV-8 infection showed viral replication in the lung, liver, spleen, kidney, testis and ovaries. Importantly, increased expression levels of pro-inflammatory cytokines IL-1ß, IL-6 and CXCL2 were observed in spleen after EHDV-8 infection. Furthermore, IFNAR(-/-) adult mice immunized with a EHDV-8 inactivated vaccine elicited neutralizing antibodies specific of EHDV-8 and full protection against challenge with a lethal dose of this virus. This study also explores the possibilities of this animal model for study of BTV and EHDV coinfection. In summary, the IFNAR(-/-) mouse model faithfully recapitulates EHD and can be applied for vaccine testing, which can facilitate progress in addressing the animal health challenge posed by this virus.


Asunto(s)
Modelos Animales de Enfermedad , Virus de la Enfermedad Hemorrágica Epizoótica , Receptor de Interferón alfa y beta , Vacunas Virales , Animales , Ratones , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/metabolismo , Virus de la Enfermedad Hemorrágica Epizoótica/inmunología , Virus de la Enfermedad Hemorrágica Epizoótica/genética , Vacunas Virales/inmunología , Infecciones por Reoviridae/inmunología , Femenino , Ratones Noqueados , Anticuerpos Neutralizantes/inmunología , Masculino
8.
Int J Mol Sci ; 25(12)2024 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-38928143

RESUMEN

Grass Carp Reovirus (GCRV) and Aeromonas hydrophila (Ah) are the causative agents of haemorrhagic disease in grass carp. This study aimed to investigate the molecular mechanisms and immune responses at the miRNA, mRNA, and protein levels in grass carp kidney cells (CIK) infected by Grass Carp Reovirus (GCRV, NV) and Aeromonas hydrophilus (Bacteria, NB) to gain insight into their pathogenesis. Within 48 h of infection with Grass Carp Reovirus (GCRV), 99 differentially expressed microRNA (DEMs), 2132 differentially expressed genes (DEGs), and 627 differentially expressed proteins (DEPs) were identified by sequencing; a total of 92 DEMs, 3162 DEGs, and 712 DEPs were identified within 48 h of infection with Aeromonas hydrophila. It is worth noting that most of the DEGs in the NV group were primarily involved in cellular processes, while most of the DEGs in the NB group were associated with metabolic pathways based on KEGG enrichment analysis. This study revealed that the mechanism of a grass carp haemorrhage caused by GCRV infection differs from that caused by the Aeromonas hydrophila infection. An important miRNA-mRNA-protein regulatory network was established based on comprehensive transcriptome and proteome analysis. Furthermore, 14 DEGs and 6 DEMs were randomly selected for the verification of RNA/small RNA-seq data by RT-qPCR. Our study not only contributes to the understanding of the pathogenesis of grass carp CIK cells infected with GCRV and Aeromonas hydrophila, but also serves as a significant reference value for other aquatic animal haemorrhagic diseases.


Asunto(s)
Aeromonas hydrophila , Carpas , MicroARNs , ARN Mensajero , Reoviridae , Transcriptoma , Animales , Carpas/genética , Carpas/microbiología , Carpas/virología , Carpas/inmunología , MicroARNs/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reoviridae/fisiología , Proteómica/métodos , Enfermedades de los Peces/microbiología , Enfermedades de los Peces/inmunología , Enfermedades de los Peces/virología , Enfermedades de los Peces/genética , Perfilación de la Expresión Génica , Infecciones por Bacterias Gramnegativas/inmunología , Infecciones por Bacterias Gramnegativas/veterinaria , Infecciones por Bacterias Gramnegativas/microbiología , Infecciones por Bacterias Gramnegativas/genética , Línea Celular , Infecciones por Reoviridae/veterinaria , Infecciones por Reoviridae/inmunología , Infecciones por Reoviridae/genética , Redes Reguladoras de Genes
9.
Viruses ; 16(6)2024 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-38932213

RESUMEN

The mode and outcome of fish-virus interactions are influenced by many abiotic factors, among which water temperature is especially important in poikilothermic fish. Rare minnow Gobiocypris rarus is a eurythermal small cyprinid fish that is sensitive to infection with genotype II grass carp reovirus (GCRV). HSP70, a conservative and key player in heat shock response, is previously identified as an induced pro-viral factor during GCRV infection in vitro. Here, rare minnow was subjected to heat shock treatment (HST), 1 h treatment at 32 °C followed by reverting to a normal temperature of 24 °C, and subsequently challenged with GCRV-II at a dosage of 1 × LD50. The effect of HST on GCRV virulence in vivo was evaluated by calculating virus-associated mortality and viral load in both dead and survival fish. The results revealed that HST enhanced the mortality of rare minnow infected with GCRV; the fact that viral loads in the tissue samples of HST-treated fish were significantly higher than those in samples of the control group at 6, 8 d p.i. reflected a faster infection process due to HST. Quantitative gene expression analysis was further employed to show that the expression levels of Hsp70 in intestine and liver tissues from the HST group declined faster than muscle tissue after HST. HST W/O GCRV challenge upregulated proinflammatory cytokines such as MyD88 and Nf-κB, which was in consistence with the inflammation observed in histopathological analysis. This study shed light on the complexity of the interaction between fish abiotic and biotic stress response, which suggested that HST, an abiotic stress, could enhance the virulence of GCRV in Gobiocypris rarus that involved modulating the gene expression of host heat shock, as well as a pro-inflammatory response.


Asunto(s)
Cyprinidae , Enfermedades de los Peces , Infecciones por Reoviridae , Reoviridae , Animales , Enfermedades de los Peces/virología , Reoviridae/patogenicidad , Reoviridae/genética , Reoviridae/fisiología , Virulencia , Infecciones por Reoviridae/virología , Infecciones por Reoviridae/veterinaria , Cyprinidae/virología , Carga Viral , Carpas/virología , Respuesta al Choque Térmico , Proteínas HSP70 de Choque Térmico/genética , Proteínas HSP70 de Choque Térmico/metabolismo , Calor
10.
Virus Res ; 346: 199413, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38848818

RESUMEN

The conversion of Adenosine (A) to Inosine (I), by Adenosine Deaminases Acting on RNA or ADARs, is an essential post-transcriptional modification that contributes to proteome diversity and regulation in metazoans including humans. In addition to its transcriptome-regulating role, ADARs also play a major part in immune response to viral infection, where an interferon response activates interferon-stimulated genes, such as ADARp150, in turn dynamically regulating host-virus interactions. A previous report has shown that infection from reoviruses, despite strong activation of ADARp150, does not influence the editing of some of the major known editing targets, while likely editing others, suggesting a potentially nuanced editing pattern that may depend on different factors. However, the results were based on a handful of selected editing sites and did not cover the entire transcriptome. Thus, to determine whether and how reovirus infection specifically affects host ADAR editing patterns, we analyzed a publicly available deep-sequenced RNA-seq dataset, from murine fibroblasts infected with wild-type and mutant reovirus strains that allowed us to examine changes in editing patterns on a transcriptome-wide scale. To the best of our knowledge, this is the first transcriptome-wide report on host editing changes after reovirus infection. Our results demonstrate that reovirus infection induces unique nuanced editing changes in the host, including introducing sites uniquely edited in infected samples. Genes with edited sites are overrepresented in pathways related to immune regulation, cellular signaling, metabolism, and growth. Moreover, a shift in editing targets has also been observed, where the same genes are edited in infection and control conditions but at different sites, or where the editing rate is increased for some and decreased for other differential targets, supporting the hypothesis of dynamic and condition-specific editing by ADARs.


Asunto(s)
Adenosina Desaminasa , Fibroblastos , Inosina , Edición de ARN , Transcriptoma , Animales , Ratones , Fibroblastos/virología , Fibroblastos/metabolismo , Inosina/metabolismo , Inosina/genética , Adenosina Desaminasa/genética , Adenosina Desaminasa/metabolismo , Adenosina/metabolismo , Adenosina/genética , Infecciones por Reoviridae/virología , Infecciones por Reoviridae/genética , Interacciones Huésped-Patógeno , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Reoviridae/genética , Reoviridae/fisiología
11.
J Med Microbiol ; 73(6)2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38935078

RESUMEN

Introduction. Avian reovirus (ARV) is associated with arthritis/tenosynovitis and malabsorption syndrome in chickens. The σC and σB proteins, both exposed to the virus capsid, are highly immunogenic and could form the basis for diagnostic devices designed to assess the immunological status of the flock.Gap Statement. Commercial ARV ELISAs cannot distinguish between vaccinated and infected animals and might not detect circulating ARV strains.Aim. We aimed to develop a customized test to detect the circulating field ARV strains as well as distinguish between vaccinated and unvaccinated animals.Methodology. We developed ELISA assays based on recombinant (r) σB, σC and the nonstructural protein σNS and tested them using antisera of vaccinated and unvaccinated chickens as well as negative controls. Fragments of σB and σC proteins were also used to study regions that could be further exploited in diagnostic tests.Results. Vaccinated and unvaccinated birds were positive by commercial ELISA, with no difference in optical density values. In contrast, samples of unvaccinated animals showed lower absorbance in the rσB and rσC ELISA tests and higher absorbance in the rσNS ELISA test than the vaccinated animals. Negative control samples were negative in all tests. Fragmentation of σB and σC proteins showed that some regions can differentiate between vaccinated and unvaccinated animals. For example, σB amino acids 128-179 (σB-F4) and σC amino acids 121-165 (σC-F4) exhibited 85 and 95% positivity among samples of vaccinated animals but only 5% and zero positivity among samples of unvaccinated animals, respectively.Conclusion. These data suggest that unvaccinated birds might have been exposed to field strains of ARV. The reduction in absorbance in the recombinant tests possibly reflects an increased specificity of our test since unvaccinated samples showed less cross-reactivity with the vaccine proteins immobilized on ELISAs. The discrepant results obtained with the protein fragment tests between vaccinated and unvaccinated animals are discussed in light of the diversity between ARV strains.


Asunto(s)
Pollos , Ensayo de Inmunoadsorción Enzimática , Orthoreovirus Aviar , Enfermedades de las Aves de Corral , Proteínas Recombinantes , Infecciones por Reoviridae , Animales , Orthoreovirus Aviar/inmunología , Orthoreovirus Aviar/genética , Orthoreovirus Aviar/aislamiento & purificación , Ensayo de Inmunoadsorción Enzimática/métodos , Ensayo de Inmunoadsorción Enzimática/veterinaria , Infecciones por Reoviridae/veterinaria , Infecciones por Reoviridae/diagnóstico , Enfermedades de las Aves de Corral/virología , Enfermedades de las Aves de Corral/diagnóstico , Proteínas Recombinantes/inmunología , Anticuerpos Antivirales/sangre , Proteínas de la Cápside/inmunología , Proteínas de la Cápside/genética , Proteínas Virales/inmunología , Proteínas Virales/genética
12.
Virus Res ; 347: 199416, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38897236

RESUMEN

Epizootic haemorrhagic disease (EHD), caused by the EHD virus (EHDV), is a vector-borne viral disease transmitted through Culicoides biting midges. EHDV comprises seven serotypes (1, 2, and 4-8), with EHDV-8 having recently emerged and spread in Europe over the last two years. Such event has raised concerns about the significant threat posed by EHDV-8 to livestock industry. In this study, an inactivated vaccine against EHDV-8 (vEHDV8-IZSAM) was developed. Safety and efficacy of the vaccine were evaluated in calves through clinical, serological, and virological monitoring following experimental challenge. The vaccine was proven safe, with only transient fever and localized reactions observed in a few animals, consistent with adjuvanted vaccine side effects. vEHDV8-IZSAM elicited a robust humoral response, as evidenced by the presence of neutralizing antibodies. After challenge with a virulent isolate, viraemia and clinical signs were evidenced in control animals but in none of the vaccinated animals. This study highlights the potential of vEHDV8-IZSAM as a safe and highly effective vaccine against EHDV-8 in cattle. It offers protection from clinical disease and effectively prevents viraemia. With the recent spread of EHDV-8 in European livestock, the use of an inactivated vaccine could be key in protecting animals from clinical disease and thus to mitigate the economic impact of the disease. Further investigations are warranted to assess the duration of the induced immunity and the applicability of this vaccine in real-world settings. Accordingly, joint efforts between public veterinary institutions and pharmaceutical companies are recommended to scale up vaccine production.


Asunto(s)
Anticuerpos Neutralizantes , Anticuerpos Antivirales , Enfermedades de los Bovinos , Virus de la Enfermedad Hemorrágica Epizoótica , Vacunas de Productos Inactivados , Vacunas Virales , Viremia , Animales , Bovinos , Vacunas de Productos Inactivados/inmunología , Vacunas de Productos Inactivados/administración & dosificación , Viremia/prevención & control , Enfermedades de los Bovinos/prevención & control , Enfermedades de los Bovinos/virología , Enfermedades de los Bovinos/inmunología , Vacunas Virales/inmunología , Vacunas Virales/administración & dosificación , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Anticuerpos Neutralizantes/sangre , Anticuerpos Neutralizantes/inmunología , Virus de la Enfermedad Hemorrágica Epizoótica/inmunología , Infecciones por Reoviridae/prevención & control , Infecciones por Reoviridae/veterinaria , Infecciones por Reoviridae/inmunología , Eficacia de las Vacunas , Vacunación/veterinaria
13.
Fish Shellfish Immunol ; 151: 109712, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38901682

RESUMEN

The grass carp (Ctenopharyngodon idella) constitutes a significant economic resource within the aquaculture sector of our nation, yet it has been chronically afflicted by the Grass Carp Reovirus (GCRV) disease. The complement system, a vital component of fish's innate immunity, plays a crucial role in combating viral infections. This research investigates the potential role of MASP1, a key molecule in the lectin pathway of the complement system, in the GCRV infection in grass carp. An analysis of the molecular characteristics of MASP1 in grass carp revealed that its identity and similarity percentages range from 35.10 to 91.00 % and 35.30-91.00 %, respectively, in comparison to other species. Phylogenetically, MASP1 in C. idella aligns closely with species such as Danio rerio, Cyprinus carpio, and Carassius carassius, exhibiting chromosomal collinearity with the zebrafish. Subsequent tissue analysis in both healthy and GCRV-infected grass carp indicated that MASP1's basal expression was predominantly in the liver. Post-GCRV infection, MASP1 expression in various tissues exhibited temporal variations: peaking in the liver on day 5, spleen on day 7, and kidney on day 14. Furthermore, employing Complement Component 3 (C3) as a benchmark for complement system activation, it was observed that MASP1 could activate and cleave C3 to C3b. MASP1 also demonstrated an inhibitory effect on GCRV replication (compared with the control group, VP2 and VP7 decreased by 6.82-fold and 4.37-fold) and enhanced the expression of antiviral genes, namely IRF3, IRF7 and IFN1 (compared with the control group, increased 2.25-fold, 45.38-fold and 22.37-fold, respectively). In vivo protein injection experiments substantiated MASP1's influence on the relative mRNA expression levels of C3 in various tissues and its protein expression in serum. This study also verified that C3 could modulate the expression of antiviral genes such as IFN1 and IRF3.


Asunto(s)
Carpas , Enfermedades de los Peces , Proteínas de Peces , Inmunidad Innata , Serina Proteasas Asociadas a la Proteína de Unión a la Manosa , Filogenia , Infecciones por Reoviridae , Reoviridae , Animales , Infecciones por Reoviridae/inmunología , Infecciones por Reoviridae/veterinaria , Enfermedades de los Peces/inmunología , Enfermedades de los Peces/virología , Carpas/inmunología , Carpas/genética , Reoviridae/fisiología , Proteínas de Peces/genética , Proteínas de Peces/inmunología , Serina Proteasas Asociadas a la Proteína de Unión a la Manosa/genética , Serina Proteasas Asociadas a la Proteína de Unión a la Manosa/inmunología , Inmunidad Innata/genética , Regulación de la Expresión Génica/inmunología , Perfilación de la Expresión Génica/veterinaria , Proteínas del Sistema Complemento/inmunología , Proteínas del Sistema Complemento/genética , Secuencia de Aminoácidos , Alineación de Secuencia/veterinaria
14.
Vet Microbiol ; 295: 110149, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38909417

RESUMEN

Avian reovirus (ARV) is a significant pathogen that causes various clinical diseases in chickens, including viral arthritis, chronic respiratory diseases, retarded growth, and malabsorption syndrome. These conditions result in substantial economic losses for the global poultry industry. MicroRNAs (miRNAs), a type of small noncoding RNAs that regulate gene expression post transcriptionally by silencing or degrading their RNA targets, play crucial roles in response to pathogenic infections. In this study, transfection of DF-1 cells with gga-miR-200a-3p, an upregulated miRNA observed in ARV-infected cells, significantly suppressed ARV-induced apoptosis by directly targeting GRB2 and impeded ARV replication. Conversely, knockdown of endogenous gga-miR-200a-3p in DF-1 cells using a specific miRNA inhibitor enhanced ARV-induced apoptosis and promoted GRB2 expression, thereby facilitating viral growth within cells. Consistently, inhibition of GRB2 activity through siRNA-mediated knockdown reduced viral titers. Therefore, gga-miR-200a-3p plays a vital antiviral role in the host response to ARV infection by suppressing apoptosis via direct targeting of GRB2 protein. This information enhances our understanding of the mechanisms by which host cells combat against ARV infection through self-encoded small RNA molecules and expands our knowledge regarding the involvement of microRNAs in the host response to pathogenic infections.


Asunto(s)
Apoptosis , Pollos , Proteína Adaptadora GRB2 , MicroARNs , Orthoreovirus Aviar , Replicación Viral , Animales , MicroARNs/genética , MicroARNs/metabolismo , Orthoreovirus Aviar/fisiología , Orthoreovirus Aviar/genética , Proteína Adaptadora GRB2/metabolismo , Proteína Adaptadora GRB2/genética , Línea Celular , Enfermedades de las Aves de Corral/virología , Infecciones por Reoviridae/virología , Infecciones por Reoviridae/veterinaria
15.
Fish Shellfish Immunol ; 151: 109730, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38942250

RESUMEN

RLR helicases RIG-I and MDA5, which are known as pattern recognition receptors to sense cytoplasmic viral RNAs and trigger antiviral immune responses, are DExD/H-box helicases. In teleost, whether and how non-RLR helicases regulate RLR helicases to affect viral infection remains unclear. Here, we report that the non-RLR helicase DHX40 from grass carp (namely gcDHX40) is a negative regulator of grass carp reovirus (GCRV) infection and RLR-mediated type I IFN production. GcDHX40 was a cytoplasmic protein. Ectopic expression of gcDHX40 facilitated GCRV replication and suppressed type I IFN production induced by GCRV infection and by those genes involved the RLR antiviral signaling pathway. Mechanistically, gcDHX40 promoted the generation of viral inclusion bodies (VIBs) by interacting with the NS38 protein of GCRV. Additionally, gcDHX40 interacted with RLR helicase, and impaired the formation of RLR-MAVS functional complexes. Taken together, our results indicate that gcDHX40 is a novel important proviral host factor involving in promoting the generation of GCRV VIBs and inhibiting the production of RLR-mediated type I IFNs.


Asunto(s)
Carpas , ARN Helicasas DEAD-box , Enfermedades de los Peces , Proteínas de Peces , Inmunidad Innata , Infecciones por Reoviridae , Reoviridae , Proteínas no Estructurales Virales , Animales , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/inmunología , Proteínas no Estructurales Virales/metabolismo , Enfermedades de los Peces/inmunología , Enfermedades de los Peces/virología , Proteínas de Peces/genética , Proteínas de Peces/inmunología , Carpas/inmunología , Carpas/genética , Infecciones por Reoviridae/veterinaria , Infecciones por Reoviridae/inmunología , Reoviridae/fisiología , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/inmunología , ARN Helicasas DEAD-box/metabolismo , Inmunidad Innata/genética , ARN Helicasas/genética , ARN Helicasas/metabolismo , ARN Helicasas/inmunología , Regulación de la Expresión Génica/inmunología
16.
Poult Sci ; 103(8): 103940, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38909506

RESUMEN

Migratory wild birds can carry various pathogens, such as influenza A virus, which can spread to globally and cause disease outbreaks and epidemics. Continuous epidemiological surveillance of migratory wild birds is of great significance for the early warning, prevention, and control of epidemics. To investigate the pathogen infection status of migratory wild birds in eastern China, fecal samples were collected from wetlands to conduct pathogen surveillance. The results showed that duck orthoreovirus (DRV) and goose parvovirus (GPV) nucleic acid were detected positive in the fecal samples collected from wild ducks, egrets, and swan. Phylogenetic analysis of the amplified viral genes reveals that the isolates were closely related to the prevalent strains in the regions involved in East Asian-Australasian (EAA) migratory flyway. Phylogenetic analysis of the amplified viral genes confirmed that they were closely related to circulating strains in the regions involved in the EAA migration pathway. The findings of this study have expanded the host range of the orthoreovirus and parvovirus, and revealed possible virus transmission between wild migratory birds and poultry.


Asunto(s)
Animales Salvajes , Enfermedades de las Aves , Orthoreovirus Aviar , Infecciones por Parvoviridae , Parvovirus , Filogenia , Infecciones por Reoviridae , Animales , Infecciones por Reoviridae/veterinaria , Infecciones por Reoviridae/epidemiología , Infecciones por Reoviridae/virología , Orthoreovirus Aviar/aislamiento & purificación , Orthoreovirus Aviar/genética , Infecciones por Parvoviridae/veterinaria , Infecciones por Parvoviridae/virología , Infecciones por Parvoviridae/epidemiología , China/epidemiología , Enfermedades de las Aves/virología , Enfermedades de las Aves/epidemiología , Animales Salvajes/virología , Parvovirus/genética , Parvovirus/aislamiento & purificación , Heces/virología , Reservorios de Enfermedades/veterinaria , Reservorios de Enfermedades/virología , Patos/virología , Anseriformes/virología , Monitoreo Epidemiológico/veterinaria
17.
J Vet Diagn Invest ; 36(3): 338-345, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38693675

RESUMEN

We performed a diagnostic disease investigation on a cohort of coho salmon (Oncorhynchus kisutch) fingerlings in Alaska exhibiting anorexia, gaping mouths, anemia, and increased mortality. Histologic examination revealed mild-to-severe myocardial degeneration and lymphohistiocytic and neutrophilic myocarditis, moderate splenic histiocytosis, and mild renal histiocytosis. Piscine orthoreoviruses 1 and 3 were not detected by molecular methods, and no other viruses could be cultured on 3 common diagnostic fish cell lines. De novo assembly produced a viral genome of 10 linear segments with >80% homology to piscine orthoreovirus 2 (PRV2) encoding all 11 PRV2 proteins. An in situ hybridization probe using RNAscope was developed against 697 viral nucleotides identified by sequencing, which revealed viral genome in heart, spleen, gill, kidney, liver, blood, and the lamina propria of the intestines. Our findings are supportive of a novel piscine orthoreovirus most closely related to PRV2 associated with morbidity and mortality of coho salmon in the northeastern Pacific.


Asunto(s)
Enfermedades de los Peces , Oncorhynchus kisutch , Orthoreovirus , Infecciones por Reoviridae , Animales , Enfermedades de los Peces/virología , Enfermedades de los Peces/patología , Infecciones por Reoviridae/veterinaria , Infecciones por Reoviridae/virología , Orthoreovirus/genética , Orthoreovirus/aislamiento & purificación , Alaska , Oncorhynchus kisutch/virología , Filogenia , Genoma Viral , Distribución Tisular
18.
Dev Comp Immunol ; 157: 105189, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38692524

RESUMEN

Grass carp, one of the major freshwater aquaculture species in China, is susceptible to grass carp reovirus (GCRV). GCRV is a non-enveloped RNA virus and has a double-layered capsid, causing hemorrhagic disease and high mortalities in infected fish. However, the tropism of GCRV infection has not been investigated. In this study, monoclonal antibodies against recombinant VP35 protein were generated in mice and characterized. The antibodies exhibited specific binding to the N terminal region (1-155 aa) of the recombinant VP35 protein expressed in the HEK293 cells, and native VP35 protein in the GCRV-II infected CIK cells. Immunofluorescent staining revealed that viruses aggregated in the cytoplasm of infected cells. In vivo challenge experiments showed that high levels of GCRV-II viruses were present in the gills, intestine, spleen and liver, indicating that they are the major sites for virus infection. Our study showed that the VP35 antibodies generated in this study exhibited high specificity, and are valuable for the development of diagnostic tools for GCRV-II infection.


Asunto(s)
Anticuerpos Monoclonales , Anticuerpos Antivirales , Carpas , Enfermedades de los Peces , Infecciones por Reoviridae , Reoviridae , Animales , Carpas/inmunología , Carpas/virología , Infecciones por Reoviridae/inmunología , Infecciones por Reoviridae/veterinaria , Infecciones por Reoviridae/virología , Reoviridae/inmunología , Reoviridae/fisiología , Enfermedades de los Peces/inmunología , Enfermedades de los Peces/virología , Ratones , Humanos , Células HEK293 , Anticuerpos Monoclonales/inmunología , Anticuerpos Antivirales/inmunología , Tropismo Viral , Proteínas de la Cápside/inmunología , Proteínas de la Cápside/metabolismo , Ratones Endogámicos BALB C , China
19.
Fish Shellfish Immunol ; 150: 109647, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38797335

RESUMEN

NIK (NF-κB inducing kinase) belongs to the mitogen-activated protein kinase family, which activates NF-κB and plays a vital role in immunology, inflammation, apoptosis, and a series of pathological responses. In NF-κB noncanonical pathway, NIK and IKKα have been often studied in mammals and zebrafish. However, few have explored the relationship between NIK and other subunits of the IKK complex. As a classic kinase in the NF-κB canonical pathway, IKKß has never been researched with NIK in fish. In this paper, the full-length cDNA sequence of grass carp (Ctenopharyngodon idella) NIK (CiNIK) was first cloned and identified. The expression level of CiNIK in grass carp cells was increased under GCRV stimuli. Under the stimulation of GCRV, poly (I:C), and LPS, the expression of NIK in various tissues of grass carp was also increased. This suggests that CiNIK responds to viral stimuli. To study the relationship between CiNIK and CiIKKß, we co-transfected CiNIK-FLAG and CiIKKB-GFP into grass carp cells in coimmunoprecipitation and immunofluorescence experiments. The results revealed that CiNIK interacts with CiIKKß. Besides, the degree of autophosphorylation of CiNIK was enhanced under poly (I:C) stimulation. CiIKKß was phosphorylated by CiNIK and then activated the activity of p65. The activity change of p65 indicates that NF-κB downstream inflammatory genes will be functioning. CiNIK or CiIKKß up-regulated the expression of IL-8. It got higher when CiNIK and CiIKKß coexisted. This paper revealed that NF-κB canonical pathway and noncanonical pathway are not completely separated in generating benefits.


Asunto(s)
Secuencia de Aminoácidos , Carpas , Proteínas de Peces , Interleucina-8 , FN-kappa B , Proteínas Serina-Treonina Quinasas , Regulación hacia Arriba , Animales , Carpas/genética , Carpas/inmunología , Proteínas de Peces/genética , Proteínas de Peces/inmunología , Proteínas de Peces/química , FN-kappa B/genética , FN-kappa B/metabolismo , Interleucina-8/genética , Interleucina-8/metabolismo , Interleucina-8/inmunología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/inmunología , Proteínas Serina-Treonina Quinasas/metabolismo , Enfermedades de los Peces/inmunología , Transducción de Señal , Reoviridae/fisiología , Filogenia , Quinasa de Factor Nuclear kappa B , Regulación de la Expresión Génica/inmunología , Poli I-C/farmacología , Lipopolisacáridos/farmacología , Infecciones por Reoviridae/inmunología , Infecciones por Reoviridae/veterinaria , Alineación de Secuencia/veterinaria , Inmunidad Innata/genética , Secuencia de Bases , Perfilación de la Expresión Génica/veterinaria
20.
J Virol ; 98(6): e0030524, 2024 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-38771042

RESUMEN

Many functions of viral attachment proteins are established, but less is known about the biological importance of viral attachment protein encapsidation efficiency. The mammalian orthoreovirus (reovirus) σ1 attachment protein forms filamentous trimers that incorporate into pentamers of the λ2 capsid protein. Reovirus strains vary in the efficiency of σ1 encapsidation onto progeny virions, which influences viral stability during entry into cells and the efficacy of tumor cell lysis. While the role of σ1 encapsidation has been evaluated in studies using cultured cells, the contribution of attachment protein encapsidation efficiency to viral infection in animals is less clear. Polymorphisms in reovirus σ1 at residues 22 and 249 have been implicated in viral dissemination in mice and susceptibility to proteolysis in the murine intestine, respectively. To determine whether these residues contribute to σ1 encapsidation efficiency, we engineered σ1 mutant viruses with single- and double-residue substitutions at sites 22 and 249. We found that substitutions at these sites alter the encapsidation of σ1 and that reoviruses encapsidating higher amounts of σ1 bind cells more avidly and have a modest replication advantage in a cell-type-specific manner relative to low σ1-encapsidating reoviruses. Furthermore, we found that a high σ1-encapsidating reovirus replicates and disseminates more efficiently in mice relative to a low σ1-encapsidating reovirus. These findings provide evidence of a relationship between viral attachment protein encapsidation efficiency and viral replication in cell culture and animal hosts. IMPORTANCE: Viral attachment proteins can serve multiple functions during viral replication, including attachment to host cells, cell entry and disassembly, and modulation of host immune responses. The relationship between viral attachment protein encapsidation efficiency and viral replication in cells and animals is poorly understood. We engineered and characterized a panel of reoviruses that differ in the capacity to encapsidate the σ1 attachment protein. We found that strains encapsidating σ1 with higher efficiency bind cells more avidly and replicate and spread more efficiently in mice relative to those encapsidating σ1 with lower efficiency. These results highlight a function for σ1 attachment protein capsid abundance in viral replication in cells and animals, which may inform future use of reovirus as an oncolytic therapeutic.


Asunto(s)
Proteínas de la Cápside , Infecciones por Reoviridae , Replicación Viral , Animales , Ratones , Proteínas de la Cápside/metabolismo , Proteínas de la Cápside/genética , Infecciones por Reoviridae/virología , Infecciones por Reoviridae/metabolismo , Acoplamiento Viral , Polimorfismo Genético , Orthoreovirus de los Mamíferos/genética , Orthoreovirus de los Mamíferos/fisiología , Orthoreovirus de los Mamíferos/metabolismo , Ensamble de Virus , Línea Celular , Cápside/metabolismo , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA