Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 304
Filtrar
1.
J Fish Dis ; 47(6): e13939, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38481093

RESUMO

The relationship of histopathological changes and the infection of Piscine orthoreovirus 2 (PRV-2) was investigated in coho salmon that were suffering from the erythrocytic inclusion body syndrome (EIBS). Immunohistochemical observations revealed abundant σ1 protein of PRV-2 in the spongy layer of the ventricle of the heart, where severe myocarditis was observed. In the spleen, the virus protein was detected in many erythrocytes, some of which were spherical-shaped and apparently dead. The number of erythrocytes was decreased in the spleen compared to the apparently healthy fish. The virus protein was also detected in some erythrocytes in blood vessels. The viral protein was often detected in many macrophages ingesting erythrocytes or dead cell debris in the spleen or in the kidney sinusoids. Large amounts of the viral genomic segment L2 were also detected in these organs by RT-qPCR. Many necrotic foci were found in the liver, although the virus protein was not detected in the hepatocytes. These results suggest that the primary targets of PRV-2 are myocardial cells and erythrocytes and that clinical symptoms such as anaemia or jaundice and histopathological changes such as myocarditis in EIBS-affected coho salmon are caused by PRV-2 infection.


Assuntos
Doenças dos Peixes , Oncorhynchus kisutch , Orthoreovirus , Infecções por Reoviridae , Animais , Doenças dos Peixes/virologia , Doenças dos Peixes/patologia , Infecções por Reoviridae/veterinária , Infecções por Reoviridae/virologia , Infecções por Reoviridae/patologia , Orthoreovirus/fisiologia , Oncorhynchus kisutch/virologia , Eritrócitos/virologia , Eritrócitos/patologia , Baço/virologia , Baço/patologia
2.
J Virol ; 98(3): e0146923, 2024 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-38345385

RESUMO

Grass carp reovirus (GCRV), particularly the highly prevalent type II GCRV (GCRV-II), causes huge losses in the aquaculture industry. However, little is known about the mechanisms by which GCRV-II invades grass carp and further disseminates among tissues. In the present study, monocytes/macrophages (Mo/Mφs) were isolated from the peripheral blood of grass carp and infected with GCRV-II. The results of indirect immunofluorescent microscopy, transmission electron microscopy, real-time quantitative RT-PCR (qRT-PCR), western blot (WB), and flow cytometry analysis collectively demonstrated that GCRV-II invaded Mo/Mφs and replicated in them. Additionally, we observed that GCRV-II induced different types (M1 and M2) of polarization of Mo/Mφs in multiple tissues, especially in the brain, head kidney, and intestine. To assess the impact of different types of polarization on GCRV-II replication, we recombinantly expressed and purified the intact cytokines CiIFN-γ2, CiIL-4/13A, and CiIL-4/13B and successfully induced M1 and M2 type polarization of macrophages using these cytokines through in vitro experiments. qRT-PCR, WB, and flow cytometry analyses showed that M2 macrophages had higher susceptibility to GCRV-II infection than other types of Mo/Mφs. In addition, we found GCRV-II induced apoptosis of Mo/Mφs to facilitate virus replication and dissemination and also detected the presence of GCRV-II virus in plasma. Collectively, our findings indicated that GCRV-II could invade immune cells Mo/Mφs and induce apoptosis and polarization of Mo/Mφs for efficient infection and dissemination, emphasizing the crucial role of Mo/Mφs as a vector for GCRV-II infection.IMPORTANCEType II grass carp reovirus (GCRV) is a prevalent viral strain and causes huge losses in aquaculture. However, the related dissemination pathway and mechanism remain largely unclear. Here, our study focused on phagocytic immune cells, monocytes/macrophages (Mo/Mφs) in blood and tissues, and explored whether GCRV-II can invade Mo/Mφs and replicate and disseminate via Mo/Mφs with their differentiated type M1 and M2 macrophages. Our findings demonstrated that GCRV-II infected Mo/Mφs and replicated in them. Furthermore, GCRV-II infection induces an increased number of M1 and M2 macrophages in grass carp tissues and a higher viral load in M2 macrophages. Furthermore, GCRV-II induced Mo/Mφs apoptosis to release viruses, eventually infecting more cells. Our study identified Mo/Mφs as crucial components in the pathway of GCRV-II dissemination and provides a solid foundation for the development of treatment strategies for GCRV-II infection.


Assuntos
Carpas , Doenças dos Peixes , Orthoreovirus , Infecções por Reoviridae , Animais , Apoptose , Citocinas , Doenças dos Peixes/metabolismo , Doenças dos Peixes/patologia , Doenças dos Peixes/virologia , Macrófagos/metabolismo , Macrófagos/patologia , Macrófagos/virologia , Monócitos/metabolismo , Infecções por Reoviridae/metabolismo , Infecções por Reoviridae/patologia , Infecções por Reoviridae/veterinária , Replicação Viral
3.
Microbes Infect ; 26(1-2): 105243, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38380604

RESUMO

Pteropine orthoreovirus (PRV) causes respiratory tract infections in humans. Despite its emergence as a zoonotic and respiratory virus, little is known about its cell tropism, which hampers progress in fully understanding its pathogenesis in humans. Hek293 cells are most susceptible to PRV infection, while HeLa cells are the least. Human cytokeratin 1 (CK1) was identified as the protein that interacts with PRV. The immunofluorescence assay and qPCR results revealed prior treatment with anti-CK1 may provide Hek293 cells protection against PRV. The KRT1-knockout Hek293 cells were less susceptible to PRV infection. Further study into the pathogenesis of PRV in humans is needed.


Assuntos
Doenças dos Peixes , Orthoreovirus , Infecções por Reoviridae , Animais , Humanos , Células HEK293 , Células HeLa , Queratinas , Infecções por Reoviridae/patologia
4.
Cell Tissue Res ; 395(2): 199-210, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38087072

RESUMO

Spatial transcriptomics is a technique that provides insight into gene expression profiles in tissue sections while retaining structural information. We have employed this method to study the pathological conditions related to red and melanized focal changes in farmed Atlantic salmon (Salmo salar). Our findings support a model where similar molecular mechanisms are involved in both red and melanized filet discolorations and genes associated with several relevant pathways show distinct expression patterns in both sample types. Interestingly, there appears to be significant cellular heterogeneity in the foci investigated when looking at gene expression patterns. Some of the genes that show differential spatial expression are involved in cellular processes such as hypoxia and immune responses, providing new insight into the nature of muscle melanization in Atlantic salmon.


Assuntos
Doenças dos Peixes , Infecções por Reoviridae , Salmo salar , Animais , Infecções por Reoviridae/patologia , Salmo salar/genética , Músculo Esquelético/patologia , Perfilação da Expressão Gênica , Transcriptoma/genética , Doenças dos Peixes/patologia
5.
J Immunol ; 208(3): 707-719, 2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-35022273

RESUMO

Grass carp reovirus (GCRV) is a highly virulent RNA virus that mainly infects grass carp and causes hemorrhagic disease. The roles of nonstructural proteins NS38 and NS80 of GCRV-873 in the viral replication cycle and viral inclusion bodies have been established. However, the strategies that NS38 and NS80 used to avoid host antiviral immune response are still unknown. In this study, we report the negative regulations of NS38 and NS80 on the RIG-I-like receptors (RLRs) antiviral signaling pathway and the production of IFNs and IFN-stimulated genes. First, both in the case of overexpression and GCRV infection, NS38 and NS80 inhibited the IFN promoter activation induced by RIG-I, MDA5, MAVS, TBK1, IRF3, and IRF7 and mRNA abundance of key antiviral genes involved in the RLR-mediated signaling. Second, both in the case of overexpression and GCRV infection, NS38 interacted with piscine TBK1 and IRF3, but not with piscine RIG-I, MDA5, MAVS, and TNF receptor-associated factor (TRAF) 3. Whereas NS80 interacted with piscine MAVS, TRAF3, and TBK1, but not with piscine RIG-I, MDA5, and IRF3. Finally, both in the case of overexpression and GCRV infection, NS38 inhibited the formation of the TBK1-IRF3 complex, but NS80 inhibited the formation of the TBK1-TRAF3 complex. Most importantly, NS38 and NS80 could hijack piscine TBK1 and IRF3 into the cytoplasmic viral inclusion bodies and inhibit the translocation of IRF3 into the nucleus. Collectively, all of these data demonstrate that GCRV nonstructural proteins can avoid host antiviral immune response by targeting the RLR signaling pathway, which prevents IFN-stimulated gene production and facilitates GCRV replication.


Assuntos
Carpas/virologia , RNA Helicases DEAD-box/metabolismo , Evasão da Resposta Imune/imunologia , Infecções por Reoviridae/veterinária , Reoviridae/imunologia , Proteínas não Estruturais Virais/imunologia , Animais , Células Cultivadas , Doenças dos Peixes/imunologia , Doenças dos Peixes/virologia , Fatores Reguladores de Interferon/metabolismo , Interferons/imunologia , Proteínas Serina-Treonina Quinases/metabolismo , Infecções por Reoviridae/imunologia , Infecções por Reoviridae/patologia , Fator 3 Associado a Receptor de TNF/metabolismo , Replicação Viral/fisiologia
6.
J Virol ; 96(2): e0187921, 2022 01 26.
Artigo em Inglês | MEDLINE | ID: mdl-34757847

RESUMO

Although a broad range of viruses cause myocarditis, the mechanisms that underlie viral myocarditis are poorly understood. Here, we report that the M2 gene is a determinant of reovirus myocarditis. The M2 gene encodes outer capsid protein µ1, which mediates host membrane penetration during reovirus entry. We infected newborn C57BL/6 mice with reovirus strain type 1 Lang (T1L) or a reassortant reovirus in which the M2 gene from strain type 3 Dearing (T3D) was substituted into the T1L genetic background (T1L/T3DM2). T1L was nonlethal in wild-type mice, whereas more than 90% of mice succumbed to T1L/T3DM2 infection. T1L/T3DM2 produced higher viral loads than T1L at the site of inoculation. In secondary organs, T1L/T3DM2 was detected with more rapid kinetics and reached higher peak titers than T1L. We found that hearts from T1L/T3DM2-infected mice were grossly abnormal, with large lesions indicative of substantial inflammatory infiltrate. Lesions in T1L/T3DM2-infected mice contained necrotic cardiomyocytes with pyknotic debris, as well as extensive lymphocyte and histiocyte infiltration. In contrast, T1L induced the formation of small purulent lesions in a small subset of animals, consistent with T1L being mildly myocarditic. Finally, more activated caspase-3-positive cells were observed in hearts from animals infected with T1L/T3DM2 than T1L. Together, our findings indicate that substitution of the T3D M2 allele into an otherwise T1L genetic background is sufficient to change a nonlethal infection into a lethal infection. Our results further indicate that T3D M2 enhances T1L replication and dissemination in vivo, which potentiates the capacity of reovirus to cause myocarditis. IMPORTANCE Reovirus is a nonenveloped virus with a segmented double-stranded RNA genome that serves as a model for studying viral myocarditis. The mechanisms by which reovirus drives myocarditis development are not fully elucidated. We found that substituting the M2 gene from strain type 3 Dearing (T3D) into an otherwise type 1 Lang (T1L) genetic background (T1L/T3DM2) was sufficient to convert the nonlethal T1L strain into a lethal infection in neonatal C57BL/6 mice. T1L/T3DM2 disseminated more efficiently and reached higher maximum titers than T1L in all organs tested, including the heart. T1L is mildly myocarditic and induced small areas of cardiac inflammation in a subset of mice. In contrast, hearts from mice infected with T1L/T3DM2 contained extensive cardiac inflammatory infiltration and more activated caspase-3-positive cells, which is indicative of apoptosis. Together, our findings identify the reovirus M2 gene as a new determinant of reovirus-induced myocarditis.


Assuntos
Proteínas do Capsídeo/metabolismo , Orthoreovirus Mamífero 3/patogenicidade , Miocardite/virologia , Infecções por Reoviridae/virologia , Animais , Animais Recém-Nascidos , Proteínas do Capsídeo/genética , Inflamação , Orthoreovirus Mamífero 3/genética , Orthoreovirus Mamífero 3/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Miocardite/mortalidade , Miocardite/patologia , Orthoreovirus de Mamíferos/genética , Orthoreovirus de Mamíferos/metabolismo , Orthoreovirus de Mamíferos/patogenicidade , Infecções por Reoviridae/mortalidade , Infecções por Reoviridae/patologia , Carga Viral , Virulência , Replicação Viral
7.
Int J Mol Sci ; 22(21)2021 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-34769442

RESUMO

Complement factor D (Df) is a serine protease well known for activating the alternative pathway (AP) in mammals by promoting the cleavage of complement component 3 (C3), thus becoming involved in innate defense. In teleost fish, however, the functional mechanisms of Df in the AP and against pathogen infection are far from clear. In the present study, we cloned and characterized the Df gene, CiDf, from grass carp (Ctenopharyngodon idella) and analyzed its function in promoting C3 cleavage and expression changes after grass carp reovirus (GCRV) infection. The open reading frame of CiDf was found to be 753 bp, encoding 250 amino acids with a molecular mass of 27.06 kDa. CiDf harbors a conserved Tryp_SPc domain, with three conserved residues representing the catalytic triad and three conserved binding sites in the substrate specificity pocket. Pairwise alignment showed that CiDf shares the highest identity (96%) and similarity (98%) with Df from Anabarilius grahami. Phylogenetic analysis indicated that CiDf and other fish Dfs formed a distinct evolutionary branch. Similar to most Dfs from other vertebrates, the CiDf gene structure is characterized by four introns and five exons. The incubation of recombinant CiDf protein with grass carp serum significantly increased the C3b content, demonstrating the conserved function of CiDf in the AP in promoting C3 cleavage, similar to Dfs in mammals. CiDf mRNA expression was widely detected in various tissues and levels were relatively higher in the liver, spleen, and intestine of grass carp. During GCRV infection over a 168-hour period, a high level of CiDf mRNA expression in the liver, spleen, and intestine was maintained at 144 and 168 h, suggesting AP activity at the late stage of GCRV infection. Collectively, the above results reveal the conserved structure and function of CiDf and its distinct expression patterns after GCRV infection, which provide a key basis for studying the roles of Df and AP during GCRV infection in the grass carp C. idella.


Assuntos
Carpas/metabolismo , Fator D do Complemento/metabolismo , Proteínas de Peixes/metabolismo , Infecções por Reoviridae/metabolismo , Reoviridae/fisiologia , Sequência de Aminoácidos , Animais , Carpas/genética , Carpas/virologia , Clonagem Molecular/métodos , Fator D do Complemento/genética , Doenças dos Peixes/genética , Doenças dos Peixes/patologia , Proteínas de Peixes/genética , Filogenia , Infecções por Reoviridae/genética , Infecções por Reoviridae/patologia , Infecções por Reoviridae/virologia , Análise de Sequência de DNA/métodos , Homologia de Sequência de Aminoácidos
8.
Infect Genet Evol ; 92: 104847, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33823307

RESUMO

Emerging variant novel duck reovirus (NDRV) strains that cause spleen swelling and necrosis have seriously threatened the waterfowl industry since 2017. However, there is no report about the complete genomic sequence of emerging variant strains isolated from Cherry valley ducks. In this study, we acquired the complete genome sequences of two variant NDRV strains, SD19/6201 and SD19/6202, and analyzed their genetic and evolutionary relationship with other orthoreoviruses. The phylogenetic analysis of σC showed that all the Chinese NDRVs were clustered into two distinct branches. The SD19/6201 strain located in branch I with most of the Chinese NDRVs, while SD19/6202 was clustered in branch II with significantly different from the existing strains. Within the branch I, the NDRVs isolated in 2017 and thereafter clustered in a new subgroup. Comparison analysis of σC amino acid sequences indicated that ten amino acid differences were found between SD19/6201 and SD19/6202. Apart from the SD19/6201 and SD19/6202 strains, isolates in 2017 and thereafter had specific mutations at residues 132A, 138R, 158H, and 258A. These two NDRV strains showed different pathogenicity in SPF duck embryos and ducks. The viral loads in the spleen of infected ducks were significantly higher than those of other organs, which might be the reason why NDRV could cause obvious spleen necrosis in ducks. This study will help us to formulate effective prevention and control strategies against NDRV and enrich our understanding of the intra- and inter-species relationships of orthoreoviruses.


Assuntos
Patos/virologia , Genoma Viral/genética , Necrose/virologia , Orthoreovirus Aviário/genética , Baço/virologia , Sequência de Aminoácidos , Animais , Células Cultivadas , Genômica/métodos , Mutação/genética , Necrose/patologia , Doenças das Aves Domésticas/patologia , Doenças das Aves Domésticas/virologia , Infecções por Reoviridae/patologia , Infecções por Reoviridae/virologia , Análise de Sequência de DNA/métodos , Baço/patologia , Sequenciamento Completo do Genoma/métodos
9.
Viruses ; 13(2)2021 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-33670092

RESUMO

De novo viral protein synthesis following entry into host cells is essential for viral replication. As a consequence, viruses have evolved mechanisms to engage the host translational machinery while at the same time avoiding or counteracting host defenses that act to repress translation. Mammalian orthoreoviruses are dsRNA-containing viruses whose mRNAs were used as models for early investigations into the mechanisms that underpin the recognition and engagement of eukaryotic mRNAs by host cell ribosomes. However, there remain many unanswered questions and paradoxes regarding translation of reoviral mRNAs in the context of infection. This review summarizes the current state of knowledge about reovirus translation, identifies key unanswered questions, and proposes possible pathways toward a better understanding of reovirus translation.


Assuntos
Interações Hospedeiro-Patógeno/fisiologia , Orthoreovirus de Mamíferos/genética , Orthoreovirus de Mamíferos/fisiologia , Biossíntese de Proteínas/genética , Replicação Viral/fisiologia , Animais , Humanos , RNA Viral/genética , Infecções por Reoviridae/patologia , Ribossomos/metabolismo , Proteínas Virais/genética
10.
Microb Pathog ; 139: 103859, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31707078

RESUMO

Grass carp hemorrhagic disease caused by grass carp reovirus (GCRV) is the most important disease for grass carp aquaculture. Its typical clinical symptom is haemorrhaging, although the mechanism was remained unclear. In this study, we investigated the differences in blood parameters and histopathological features between grass carp infected with a virulent and avirulent isolates of genotype II GCRV. Infection with the virulent isolate resulted in increases in 8 routine blood and 2 serum biochemical parameters (P < 0.05); while 9 routine blood and 5 biochemical parameters were significantly decreased (P < 0.05) compared with fish infected with the avirulent isolate. The majority of these alterations were related to hemorrhage, inflammatory reactions and organic damage. The histopathologic changes were primarily vasodilation and hyperaemia in multiple organs, lymphocyte and macrophage infiltration as well as severe vacuolar degeneration in spleen, kidney and liver. The histopathology changes in fish infected with the avirulent isolate were minimal. These results indicated that the pathogenicity of GCRV was primarily reflected in destruction of the blood circulatory system and parenchymatous organs. This study lays the foundation for further research on the pathogenesis of bleeding caused by GCRV infection and the use of blood parameters and histopathology as tools for disease diagnosis.


Assuntos
Carpas/virologia , Doenças dos Peixes/sangue , Doenças dos Peixes/patologia , Doenças dos Peixes/virologia , Infecções por Reoviridae/patologia , Infecções por Reoviridae/veterinária , Infecções por Reoviridae/virologia , Reoviridae/isolamento & purificação , Animais , Linhagem Celular , Modelos Animais de Doenças , Genótipo , Hemorragia , Rim/patologia , Fígado/patologia , Reoviridae/genética , Reoviridae/patogenicidade , Baço/patologia
11.
J S Afr Vet Assoc ; 90(0): e1-e5, 2019 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-31793309

RESUMO

Infectious arthritis or tenosynovitis in broiler and breeder chickens results in major loss of productivity because of reduced growth and downgrading at processing plants. The most common causative agents of avian infectious arthritis are the bacterium Mycoplasma synoviae and avian reoviruses (ARVs) (family Reoviridae, genus Orthoreovirus). In this study, we evaluated the occurrence of these two pathogens in arthritis or tenosynovitis lesions of broilers and breeder flocks in southern Brazil using molecular detection. Tissue sections from tibiotarsal joints with visible lesions from 719 broilers and 505 breeders were analysed using pathogen-specific polymerase chain reaction (PCR) assays. In breeders, 41.2% (n = 296) of lesions were positive for M. synoviae, 26.4% (n = 190) were positive for ARV, while co-infection was present in 12.2% (n = 88) of the samples. In broilers, 20.8% (n = 105) of lesions were positive for M. synoviae, 11.9% (n = 60) for ARV and 7.7% (n = 39) of these cases were positive for both pathogens. Post-mortem examination revealed lesions with varying degrees of gross pathological severity. Histopathological examination showed intense, diffuse lymphohistiocytic inflammatory infiltrates with heterophil accumulation, primarily in the synovial capsule and digital flexor tendon, in all samples. Improved strategies for early detection and control of these major avian pathogens are highly desirable for preventing the spread of infection and reducing economic losses in the poultry industry.


Assuntos
Artrite/veterinária , Infecções por Mycoplasma/veterinária , Doenças das Aves Domésticas/microbiologia , Infecções por Reoviridae/veterinária , Tenossinovite/veterinária , Animais , Artrite/epidemiologia , Artrite/microbiologia , Artrite/patologia , Autopsia/veterinária , Brasil , Galinhas , Infecções por Mycoplasma/epidemiologia , Infecções por Mycoplasma/patologia , Mycoplasma synoviae/isolamento & purificação , Orthoreovirus Aviário/isolamento & purificação , Reação em Cadeia da Polimerase/veterinária , Doenças das Aves Domésticas/epidemiologia , Doenças das Aves Domésticas/virologia , Infecções por Reoviridae/epidemiologia , Infecções por Reoviridae/patologia , Tenossinovite/epidemiologia , Tenossinovite/microbiologia , Tenossinovite/patologia
12.
Mater Sci Eng C Mater Biol Appl ; 105: 110052, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31546360

RESUMO

Novel duck reovirus (NDRV) disease is a serious infectious disease for poultry, for which no effective therapy has been established. Therefore, development of novel antivirals against NDRV is urgently needed. In present study, we developed a complex wherein hypericin (HY), which shows broad-spectrum antiviral activity, was loaded onto graphene oxide (GO), which has a high drug-loading capacity and low cytotoxicity. The antiviral activity of the complex (GO/HY) was studied in DF-1 cells and in ducklings infected with the NDRV TH11 strain. GO/HY showed a dose-dependent inhibition of NDRV replication, which may be attributed to direct virus inactivation or inhibition of virus attachment. Western blotting and indirect immunofluorescence assay (IFA) showed markedly suppressed protein expression in GO/HY-treated NDRV-infected DF-1 cells. Moreover, GO/HY prolonged the survival time of the ducklings by reducing pathological lesions caused by the infection and inhibiting viral replication in the liver and lungs. These results suggest that GO/HY has antiviral activity against NDRV both in vitro and in vivo.


Assuntos
Portadores de Fármacos , Patos , Grafite , Orthoreovirus Aviário/metabolismo , Perileno/análogos & derivados , Doenças das Aves Domésticas , Infecções por Reoviridae , Animais , Antracenos , Linhagem Celular , Portadores de Fármacos/química , Portadores de Fármacos/farmacologia , Patos/metabolismo , Patos/virologia , Grafite/química , Grafite/farmacologia , Perileno/química , Perileno/farmacologia , Doenças das Aves Domésticas/tratamento farmacológico , Doenças das Aves Domésticas/metabolismo , Doenças das Aves Domésticas/patologia , Doenças das Aves Domésticas/virologia , Infecções por Reoviridae/tratamento farmacológico , Infecções por Reoviridae/metabolismo , Infecções por Reoviridae/patologia , Infecções por Reoviridae/veterinária
13.
Vet Microbiol ; 233: 85-92, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-31176417

RESUMO

Muscovy duck reovirus (MDRV) causes serious immunodeficiency in the intestinal mucosa, although the underlying histopathological mechanisms remain unclear. Thus, we investigated the impact of MDRV infection on intestinal morphology using hematoxylin and eosin staining. Immune-related cells were also quantified by staining with hematoxylin and eosin, toluidine blue, and periodic acid-Schiff stain, or by immunohistochemistry and cytochemistry for lectin. Similarly, CD4+ and CD8+ cells were quantified by flow cytometry, and the expression of several immune-related molecules was quantified by radioimmunoassay. We found that MDRV clearly damaged the intestinal mucosa, based on tissue morphology, villus length, villus width, intestinal thickness, villus height/crypt depth ratio, and villus surface area. MDRV also altered the density or distribution of lymphocytes, mastocytes, and goblet cells in the small intestinal mucosa, as well as microfold cells in Peyer's patches. In addition, MDRV markedly depleted CD4+ cells from the intestinal mucosa and lowered the CD4+:CD8+ ratio in peripheral blood. Moreover, MDRV diminished the levels of secretory IgA and mucosal addressin cell adhesion molecule-1 (p < 0.01), but elevated those of histamine and nitric oxide (p < 0.01 or p < 0.05). Finally, MDRV significantly suppressed IL-1ß, IL-4, IL-5, and IL-8 levels (p < 0.01 or p < 0.05) mid-infection. Collectively, our data suggest that MDRV severely damages the structure and function of the intestinal mucosa by modulating immune cells and immune-related factors, thus leading to local immunodeficiency. Our findings lay the foundation for further research on the pathogenesis of MDRV.


Assuntos
Mucosa Intestinal/imunologia , Mucosa Intestinal/patologia , Intestino Delgado/virologia , Orthoreovirus Aviário/imunologia , Infecções por Reoviridae/imunologia , Fatores Etários , Animais , Contagem de Linfócito CD4 , Citocinas/imunologia , Patos/virologia , Duodeno , Fibroblastos/virologia , Histamina/análise , Imunoglobulina A Secretora/análise , Intestino Delgado/imunologia , Óxido Nítrico/análise , Orthoreovirus Aviário/patogenicidade , Doenças das Aves Domésticas/virologia , Infecções por Reoviridae/patologia , Carga Viral
14.
J Fish Dis ; 42(6): 935-945, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30972792

RESUMO

Melanized focal changes in skeletal muscle of farmed Atlantic salmon (Salmo salar) are a major quality problem. The aetiology is unknown, but infection with Piscine orthoreovirus (PRV) has been associated with the condition. Here, we addressed the pathogenesis of red and melanized focal changes and their association with PRV. First, a population of farmed fish (PRV-negative prior to sea transfer) was sequentially investigated throughout the seawater period. The fish were autopsied and tested for PRV infection. Muscular changes were described by macroscopy and histology, and a classification system was established. Second, in an experimental infection trial, PRV was injected intramuscularly to induce changes. The farmed fish was gradually infected with PRV. Red focal changes occurred throughout the observation period with a low prevalence regardless of PRV status. Melanized changes were highly diverse and their prevalence increased during the trial. Changes of low macroscopic grade and histological category were more prevalent in PRV-negative fish. Diffuse granulomatous melanized changes only occurred after PRV infection. No muscular changes were observed in the experimentally challenged fish. Our studies do not indicate that PRV infection causes red focal changes, but seems important in the development of granulomatous melanized changes.


Assuntos
Doenças dos Peixes/virologia , Músculo Esquelético/patologia , Orthoreovirus/patogenicidade , Infecções por Reoviridae/veterinária , Salmo salar/virologia , Animais , Aquicultura , Doenças dos Peixes/patologia , Melaninas , Músculo Esquelético/virologia , Noruega , RNA Viral/genética , Infecções por Reoviridae/patologia
15.
J Biol Chem ; 293(45): 17387-17401, 2018 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-30237170

RESUMO

Autophagy plays many physiological and pathophysiological roles. However, the roles and the regulatory mechanisms of autophagy in response to viral infections are poorly defined in teleost fish, such as grass carp (Ctenopharyngodon idella), which is one of the most important aquaculture species in China. In this study, we found that both grass carp reovirus (GCRV) infection and hydrogen peroxide (H2O2) treatment induced the accumulation of reactive oxygen species (ROS) in C. idella kidney cells and stimulate autophagy. Suppressing ROS accumulation with N-acetyl-l-cysteine significantly inhibited GCRV-induced autophagy activation and enhanced GCRV replication. Although ROS-induced autophagy, in turn, restricted GCRV replication, further investigation revealed that the multifunctional cellular protein high-mobility group box 1b (HMGB1b) serves as a heat shock protein 70 (HSP70)-dependent, pro-autophagic protein in grass carp. Upon H2O2 treatment, cytoplasmic HSP70 translocated to the nucleus, where it interacted with HMGB1b and promoted cytoplasmic translocation of HMGB1b. Overexpression and siRNA-mediated knockdown assays indicated that HSP70 and HMGB1b synergistically enhance ROS-induced autophagic activation in the cytoplasm. Moreover, HSP70 reinforced an association of HMGB1b with the C. idella ortholog of Beclin 1 (a mammalian ortholog of the autophagy-associated yeast protein ATG6) by directly interacting with C. idella Beclin 1. In summary, this study highlights the antiviral function of ROS-induced autophagy in response to GCRV infection and reveals the positive role of HSP70 in HMGB1b-mediated autophagy initiation in teleost fish.


Assuntos
Autofagia , Cipriniformes , Doenças dos Peixes , Proteínas de Peixes/metabolismo , Proteína HMGB1/metabolismo , Proteínas de Choque Térmico HSP70/metabolismo , Rim/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Infecções por Reoviridae , Reoviridae/metabolismo , Animais , Células Cultivadas , Cipriniformes/metabolismo , Cipriniformes/virologia , Doenças dos Peixes/metabolismo , Doenças dos Peixes/patologia , Doenças dos Peixes/virologia , Rim/patologia , Rim/virologia , Infecções por Reoviridae/metabolismo , Infecções por Reoviridae/patologia , Infecções por Reoviridae/veterinária
16.
J Virol ; 92(10)2018 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-29514905

RESUMO

Several viruses induce intestinal epithelial cell death during enteric infection. However, it is unclear whether proapoptotic capacity promotes or inhibits replication in this tissue. We infected mice with two reovirus strains that infect the intestine but differ in the capacity to alter immunological tolerance to new food antigen. Infection with reovirus strain T1L, which induces an inflammatory immune response to fed antigen, is prolonged in the intestine, whereas T3D-RV, which does not induce this response, is rapidly cleared from the intestine. Compared with T1L, T3D-RV infection triggered apoptosis of intestinal epithelial cells and subsequent sloughing of dead cells into the intestinal lumen. We conclude that the infection advantage of T1L derives from its capacity to subvert host restriction by epithelial cell apoptosis, providing a possible mechanism by which T1L enhances inflammatory signals during antigen feeding. Using a panel of T1L × T3D-RV reassortant viruses, we identified the viral M1 and M2 gene segments as determinants of reovirus-induced apoptosis in the intestine. Expression of the T1L M1 and M2 genes in a T3D-RV background was sufficient to limit epithelial cell apoptosis and enhance viral infection to levels displayed by T1L. These findings define additional reovirus gene segments required for enteric infection of mice and illuminate the antiviral effect of intestinal epithelial cell apoptosis in limiting enteric viral infection. Viral strain-specific differences in the capacity to infect the intestine may be useful in identifying viruses capable of ameliorating tolerance to fed antigen in autoimmune conditions like celiac disease.IMPORTANCE Acute viral infections are thought to be cleared by the host with few lasting consequences. However, there may be much broader and long-lasting effects of viruses on immune homeostasis. Infection with reovirus, a common, nonpathogenic virus, triggers inflammation against innocuous food antigens, implicating this virus in the development of celiac disease, an autoimmune intestinal disorder triggered by exposure to dietary gluten. Using two reovirus strains that differ in the capacity to abrogate oral tolerance, we found that strain-specific differences in the capacity to replicate in the intestine inversely correlate with the capacity to induce apoptotic death of intestinal epithelial cells, providing a host-mediated process to restrict intestinal infection. This work contributes new knowledge about virus-host interactions in the intestine and establishes a foundation for future studies to define mechanisms by which viruses break oral tolerance in celiac disease.


Assuntos
Apoptose/imunologia , Células Epiteliais/imunologia , Mucosa Intestinal/imunologia , Orthoreovirus Mamífero 3/imunologia , Orthoreovirus de Mamíferos/imunologia , Infecções por Reoviridae/imunologia , Animais , Antígenos Virais/imunologia , Linhagem Celular , Cricetinae , Células Epiteliais/patologia , Células Epiteliais/virologia , Mucosa Intestinal/patologia , Mucosa Intestinal/virologia , Camundongos , Infecções por Reoviridae/patologia
17.
J Virol ; 92(6)2018 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-29298891

RESUMO

The mammalian orthoreovirus (reovirus) outer capsid, which is composed of 200 µ1/σ3 heterohexamers and a maximum of 12 σ1 trimers, contains all of the proteins that are necessary for attaching to and entering host cells. Following attachment, reovirus is internalized by receptor-mediated endocytosis and acid-dependent cathepsin proteases degrade the σ3 protein. This process generates a metastable intermediate, called infectious subviral particle (ISVP), in which the µ1 membrane penetration protein is exposed. ISVPs undergo a second structural rearrangement to deposit the genome-containing core into the host cytoplasm. The conformationally altered particle is called ISVP*. ISVP-to-ISVP* conversion culminates in the release of µ1 N- and C-terminal fragments, µ1N and Φ, respectively. Released µ1N is thought to facilitate core delivery by generating size-selective pores within the endosomal membrane, whereas the precise role of Φ, particularly in the context of viral entry, is undefined. In this report, we characterize a recombinant reovirus that fails to cleave Φ from µ1 in vitro Φ cleavage, which is not required for ISVP-to-ISVP* conversion, enhances the disruption of liposomal membranes and facilitates the recruitment of ISVP*s to the site of pore formation. Moreover, the Φ cleavage-deficient strain initiates infection of host cells less efficiently than the parental strain. These results indicate that µ1N and Φ contribute to reovirus pore forming activity.IMPORTANCE Host membranes represent a physical barrier that prevents infection. To overcome this barrier, viruses utilize diverse strategies, such as membrane fusion or membrane disruption, to access internal components of the cell. These strategies are characterized by discrete protein-protein and protein-lipid interactions. The mammalian orthoreovirus (reovirus) outer capsid undergoes a series of well-defined conformational changes, which conclude with pore formation and delivery of the viral genetic material. In this report, we characterize the role of the small, reovirus-derived Φ peptide in pore formation. Φ cleavage from the outer capsid enhances membrane disruption and facilitates the recruitment of virions to membrane-associated pores. Moreover, Φ cleavage promotes the initiation of infection. Together, these results reveal an additional component of the reovirus pore forming apparatus and highlight a strategy for penetrating host membranes.


Assuntos
Proteínas do Capsídeo/metabolismo , Orthoreovirus de Mamíferos/metabolismo , Proteólise , Infecções por Reoviridae/metabolismo , Vírion/metabolismo , Animais , Proteínas do Capsídeo/genética , Linhagem Celular , Camundongos , Orthoreovirus de Mamíferos/genética , Domínios Proteicos , Infecções por Reoviridae/genética , Infecções por Reoviridae/patologia , Vírion/genética
18.
Avian Pathol ; 47(2): 127-139, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28911249

RESUMO

The aim of this work was to clarify the molecular mechanism underlying the fatty degeneration of livers infected with Muscovy duck reovirus (MDRV), which produces obvious white necrotic foci in the liver. Transcriptome data for MDRV-infected Muscovy duck livers and control livers were sequenced, assembled, and annotated with Illumina® HiSeq 2000. The differentially expressed genes were screened and their functions were analysed. We also determined and confirmed the molecular mechanism of the hepatic fat metabolism disorder caused by MDRV infection. The expression of 4190 genes was higher in the infected livers than in the control livers, and the expression of 1113 genes was reduced. A Gene Ontology analysis showed that these genes were involved in 48 biological functions, and were significantly enriched in 237 Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways. The free fatty acid content was significantly higher in the livers of infected Muscovy ducks than in the control livers (P < 0.01). The KEGG analysis showed that MDRV infection inhibited the cholesterol efflux from hepatic cells and reduced the expression of key enzymes involved in fatty acid degradation (scavenger receptor class b type 1, ABCG8, and APOA4), leading to the accumulation of fatty acids and cholesterol in the liver cells. In this study, we have identified the genes differentially expressed in livers infected by MDRV, from which we inferred the genes associated with lipodystrophia, and elucidated the molecular mechanism of the hepatic steatosis induced by MDRV. ABBREVIATIONS: ABC: ATP binding cassette transport; ACADVL: acyl-CoA dehydrogenase, very long chain; ACAT: mitochondrial-like acetyl-CoA acetyltransferase A; ACAT2: acetyl-CoA acyltransferase 2; ACNAT2: acyl-coenzyme A amino acid N-acyltransferase 2-like; ACOT1: acyl-CoA thioesterase 1; ACOT7: acyl-CoA thioesterase 7; ACOX1: acyl-CoA oxidase 1, palmitoyl; ACSBG2: acyl-CoA synthetase bubblegum family member 2; ACSL1: acyl-CoA synthetase long-chain family member 1; ADH1: alcohol dehydrogenase 1; APOA4: apolipoprotein A-IV; ARV: avian reovirus; cDNA: complementary deoxyribonucleic acid; COG: Clusters of Orthologous Groups; DEG: differentially expressed gene; DGAT: diacylgycerol acyltransferase; DNA: deoxyribonucleic acid; ECI2: enoyl-CoA delta isomerase 2; EHHADH: enoyl-CoA hydratase/3-hydroxyacyl-CoA dehydrogenase; FDR: false discovery rate; GCDH: Pseudopodoces humilis glutaryl-CoA dehydrogenase; GO: Gene Ontology; HADHA: hydroxyacyl-CoA dehydrogenase/3-ketoacyl-CoA thiolase/enoyl-CoA hydratase (trifunctional protein), alpha subunit; I-FABP: intestinal fatty acid binding protein; KEGG: Kyoto Encyclopedia of Genes and Genomes; L-FABP: liver fatty acid binding protein; MDRV: Muscovy duck reovirus; MOI: multiplicity of infection; NPC1L1: Niemann-Pick C1-like 1; qPCR: real-time quantitative polymerase chain reaction; RNA: ribonucleic acid; RNase: ribonuclease; RNA-seq: RNA sequencing technology; RPKM: reads per kilobase per million mapped reads; SR-B1: scavenger receptor class b type 1.


Assuntos
Patos , Transtornos do Metabolismo dos Lipídeos/veterinária , Fígado/metabolismo , Orthoreovirus Aviário , Doenças das Aves Domésticas/virologia , Infecções por Reoviridae/veterinária , Transcriptoma , Animais , Regulação da Expressão Gênica , Transtornos do Metabolismo dos Lipídeos/metabolismo , Transtornos do Metabolismo dos Lipídeos/virologia , Doenças das Aves Domésticas/metabolismo , Doenças das Aves Domésticas/patologia , Infecções por Reoviridae/metabolismo , Infecções por Reoviridae/patologia , Infecções por Reoviridae/virologia
19.
Virology ; 514: 57-65, 2018 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-29128757

RESUMO

Pteropine orthoreovirus (PRV) is an emerging bat-borne human pathogen causing severe respiratory illness. To date, however, the evaluation of PRV virulence has largely depended on the limited numbers of clinical cases owing to the lack of animal models. To develop an in vivo model of PRV infection, an inbred C3H mouse strain was infected intranasally with pathogenic PRV strain Miyazaki-Bali/2007. C3H mice suffered severe lung infection with significant body weight reduction and died within 7 days after intranasal infection. Infectious viruses were isolated mainly from the lungs and trachea. Histopathological examination revealed interstitial pneumonia with monocytes infiltration. Following repeated intranasal infection, mice developed antibodies to particular structural and non-structural proteins of PRV. The results of these immunological assays will help to develop laboratory protocols for sero-epidemiological studies. Our small rodent model of lethal respiratory infection will further allow investigation of the molecular mechanisms underlying the high pathogenicity of PRV.


Assuntos
Orthoreovirus/fisiologia , Infecções por Reoviridae/virologia , Infecções Respiratórias/virologia , Animais , Anticorpos Antivirais/sangue , Modelos Animais de Doenças , Humanos , Pulmão/patologia , Pulmão/virologia , Masculino , Camundongos , Camundongos Endogâmicos C3H , Orthoreovirus/genética , Infecções por Reoviridae/sangue , Infecções por Reoviridae/mortalidade , Infecções por Reoviridae/patologia , Infecções Respiratórias/sangue , Infecções Respiratórias/mortalidade , Infecções Respiratórias/patologia
20.
PLoS Pathog ; 13(12): e1006768, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29211815

RESUMO

Enteric viruses encounter diverse environments as they migrate through the gastrointestinal tract to infect their hosts. The interaction of eukaryotic viruses with members of the host microbiota can greatly impact various aspects of virus biology, including the efficiency with which viruses can infect their hosts. Mammalian orthoreovirus, a human enteric virus that infects most humans during childhood, is negatively affected by antibiotic treatment prior to infection. However, it is not known how components of the host microbiota affect reovirus infectivity. In this study, we show that reovirus virions directly interact with Gram positive and Gram negative bacteria. Reovirus interaction with bacterial cells conveys enhanced virion thermostability that translates into enhanced attachment and infection of cells following an environmental insult. Enhanced virion thermostability was also conveyed by bacterial envelope components lipopolysaccharide (LPS) and peptidoglycan (PG). Lipoteichoic acid and N-acetylglucosamine-containing polysaccharides enhanced virion stability in a serotype-dependent manner. LPS and PG also enhanced the thermostability of an intermediate reovirus particle (ISVP) that is associated with primary infection in the gut. Although LPS and PG alter reovirus thermostability, these bacterial envelope components did not affect reovirus utilization of its proteinaceous cellular receptor junctional adhesion molecule-A or cell entry kinetics. LPS and PG also did not affect the overall number of reovirus capsid proteins σ1 and σ3, suggesting their effect on virion thermostability is not mediated through altering the overall number of major capsid proteins on the virus. Incubation of reovirus with LPS and PG did not significantly affect the neutralizing efficiency of reovirus-specific antibodies. These data suggest that bacteria enhance reovirus infection of the intestinal tract by enhancing the thermal stability of the reovirus particle at a variety of temperatures through interactions between the viral particle and bacterial envelope components.


Assuntos
Bacillus subtilis/fisiologia , Enterócitos/virologia , Escherichia coli K12/fisiologia , Infecções por Reoviridae/virologia , Reoviridae/fisiologia , Acetilglucosamina/análogos & derivados , Acetilglucosamina/metabolismo , Acetilglucosamina/toxicidade , Bacillus subtilis/metabolismo , Bacillus subtilis/ultraestrutura , Bacillus subtilis/virologia , Células CACO-2 , Endotoxinas/metabolismo , Endotoxinas/toxicidade , Enterócitos/efeitos dos fármacos , Enterócitos/microbiologia , Enterócitos/patologia , Escherichia coli K12/metabolismo , Escherichia coli K12/ultraestrutura , Escherichia coli K12/virologia , Microbioma Gastrointestinal , Células HeLa , Temperatura Alta , Humanos , Lipopolissacarídeos/metabolismo , Lipopolissacarídeos/toxicidade , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Microscopia Eletrônica de Transmissão , Peptidoglicano/metabolismo , Peptidoglicano/toxicidade , RNA/metabolismo , Estabilidade de RNA/efeitos dos fármacos , Proteínas Recombinantes/metabolismo , Reoviridae/química , Reoviridae/efeitos dos fármacos , Reoviridae/patogenicidade , Infecções por Reoviridae/metabolismo , Infecções por Reoviridae/microbiologia , Infecções por Reoviridae/patologia , Ácidos Teicoicos/metabolismo , Ácidos Teicoicos/toxicidade , Vírion/química , Vírion/patogenicidade , Vírion/fisiologia , Ligação Viral/efeitos dos fármacos , Internalização do Vírus/efeitos dos fármacos , Proteína Vermelha Fluorescente
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...