Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 144
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
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
2.
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
3.
Cell Host Microbe ; 32(6): 980-995.e9, 2024 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-38729153

RESUMEN

Mammalian orthoreovirus (reovirus) is a nonenveloped virus that establishes primary infection in the intestine and disseminates to sites of secondary infection, including the CNS. Reovirus entry involves multiple engagement factors, but how the virus disseminates systemically and targets neurons remains unclear. In this study, we identified murine neuropilin 1 (mNRP1) as a receptor for reovirus. mNRP1 binds reovirus with nanomolar affinity using a unique mechanism of virus-receptor interaction, which is coordinated by multiple interactions between distinct reovirus capsid subunits and multiple NRP1 extracellular domains. By exchanging essential capsid protein-encoding gene segments, we determined that the multivalent interaction is mediated by outer-capsid protein σ3 and capsid turret protein λ2. Using capsid mutants incapable of binding NRP1, we found that NRP1 contributes to reovirus dissemination and neurovirulence in mice. Collectively, our results demonstrate that NRP1 is an entry receptor for reovirus and uncover mechanisms by which NRPs promote viral entry and pathogenesis.


Asunto(s)
Proteínas de la Cápside , Neuropilina-1 , Orthoreovirus de los Mamíferos , Receptores Virales , Infecciones por Reoviridae , Internalización del Virus , Animales , Ratones , Proteínas de la Cápside/metabolismo , Proteínas de la Cápside/genética , Neuropilina-1/metabolismo , Neuropilina-1/genética , Orthoreovirus de los Mamíferos/genética , Orthoreovirus de los Mamíferos/fisiología , Orthoreovirus de los Mamíferos/metabolismo , Infecciones por Reoviridae/virología , Infecciones por Reoviridae/metabolismo , Receptores Virales/metabolismo , Humanos , Cápside/metabolismo , Línea Celular , Células HEK293 , Unión Proteica , Ratones Endogámicos C57BL
4.
J Virol ; 98(3): e0146923, 2024 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-38345385

RESUMEN

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.


Asunto(s)
Carpas , Enfermedades de los Peces , Orthoreovirus , Infecciones por Reoviridae , Animales , Apoptosis , Citocinas , Enfermedades de los Peces/metabolismo , Enfermedades de los Peces/patología , Enfermedades de los Peces/virología , Macrófagos/metabolismo , Macrófagos/patología , Macrófagos/virología , Monocitos/metabolismo , Infecciones por Reoviridae/metabolismo , Infecciones por Reoviridae/patología , Infecciones por Reoviridae/veterinaria , Replicación Viral
5.
Front Immunol ; 15: 1359552, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38420125

RESUMEN

Piscine red blood cells (RBC) are nucleated and have been characterized as mediators of immune responses in addition to their role in gas exchange. Salmonid RBC are major target cells of Piscine orthoreovirus-1 (PRV-1), the etiological agent of heart and skeletal muscle inflammation (HSMI) in farmed Atlantic salmon (Salmo salar). PRV-1 replicates in RBC ex vivo, but no viral amplification has been possible in available A. salmon cell lines. To compare RBC basal transcripts and transcriptional responses to PRV-1 in the early phase of infection with non-susceptible cells, we exposed A. salmon RBC, Atlantic salmon kidney cells (ASK) and Salmon head kidney cells (SHK-1) to PRV-1 for 24 h. The RNA-seq analysis of RBC supported their previous characterization as pluripotent cells, as they expressed a wide repertoire of genes encoding pattern recognition receptors (PRRs), cytokine receptors, and genes implicated in antiviral activities. The comparison of RBC to ASK and SHK-1 revealed immune cell features exclusively expressed in RBC, such as genes involved in chemotactic activity in response to inflammation. Differential expression analysis of RBC exposed to PRV-1 showed 46 significantly induced genes (≥ 2-fold upregulation) linked to the antiviral response pathway, including RNA-specific PRRs and interferon (IFN) response factors. In SHK-1, PRV induced a more potent or faster antiviral response (213 genes induced). ASK cells showed a differential response pattern (12 genes induced, 18 suppressed) less characterized by the dsRNA-induced antiviral pathway. Despite these differences, the RIG-I-like receptor 3 (RLR3) in the family of cytosolic dsRNA receptors was significantly induced in all PRV-1 exposed cells. IFN regulatory factor 1 (IRF1) was significantly induced in RBC only, in contrast to IRF3/IRF7 induced in SHK-1. Differences in IRF expression and activity may potentially affect viral propagation.


Asunto(s)
Orthoreovirus , Infecciones por Reoviridae , Salmo salar , Animales , Salmo salar/genética , Infecciones por Reoviridae/metabolismo , Inflamación/metabolismo , Eritrocitos/metabolismo , Perfilación de la Expresión Génica , Antivirales/metabolismo
6.
Nat Commun ; 14(1): 2615, 2023 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-37147336

RESUMEN

Mammalian orthoreovirus (reovirus) infects most mammals and is associated with celiac disease in humans. In mice, reovirus infects the intestine and disseminates systemically to cause serotype-specific patterns of disease in the brain. To identify receptors conferring reovirus serotype-dependent neuropathogenesis, we conducted a genome-wide CRISPRa screen and identified paired immunoglobulin-like receptor B (PirB) as a receptor candidate. Ectopic expression of PirB allowed reovirus binding and infection. PirB extracelluar D3D4 region is required for reovirus attachment and infectivity. Reovirus binds to PirB with nM affinity as determined by single molecule force spectroscopy. Efficient reovirus endocytosis requires PirB signaling motifs. In inoculated mice, PirB is required for maximal replication in the brain and full neuropathogenicity of neurotropic serotype 3 (T3) reovirus. In primary cortical neurons, PirB expression contributes to T3 reovirus infectivity. Thus, PirB is an entry receptor for reovirus and contributes to T3 reovirus replication and pathogenesis in the murine brain.


Asunto(s)
Orthoreovirus de los Mamíferos , Receptores Inmunológicos , Receptores Virales , Infecciones por Reoviridae , Animales , Humanos , Ratones , Anticuerpos Antivirales , Orthoreovirus de los Mamíferos/fisiología , Receptores Inmunológicos/metabolismo , Infecciones por Reoviridae/metabolismo , Receptores Virales/metabolismo
7.
PLoS Pathog ; 19(4): e1011320, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-37099596

RESUMEN

Viral seasonality in the aquaculture industry is an important scientific issue for decades. While the molecular mechanisms underpinning the temperature-dependent pathogenesis of aquatic viral diseases remain largely unknown. Here we report that temperature-dependent activation of IL6-STAT3 signaling was exploited by grass carp reovirus (GCRV) to promote viral entry via increasing the expression of heat shock protein 90 (HSP90). Deploying GCRV infection as a model system, we discovered that GCRV induces the IL6-STAT3-HSP90 signaling activation to achieve temperature-dependent viral entry. Further biochemical and microscopic analyses revealed that the major capsid protein VP7 of GCRV interacted with HSP90 and relevant membrane-associated proteins to boost viral entry. Accordingly, exogenous expression of either IL6, HSP90, or VP7 in cells increased GCRV entry in a dose-dependent manner. Interestingly, other viruses (e.g., koi herpesvirus, Rhabdovirus carpio, Chinese giant salamander iridovirus) infecting ectothermic vertebrates have evolved a similar mechanism to promote their infection. This work delineates a molecular mechanism by which an aquatic viral pathogen exploits the host temperature-related immune response to promote its entry and replication, instructing us on new ways to develop targeted preventives and therapeutics for aquaculture viral diseases.


Asunto(s)
Carpas , Enfermedades de los Peces , Orthoreovirus , Infecciones por Reoviridae , Reoviridae , Animales , Internalización del Virus , Interleucina-6/metabolismo , Infecciones por Reoviridae/metabolismo , Proteínas de la Cápside/metabolismo , Anticuerpos Antivirales/metabolismo
8.
Fish Shellfish Immunol ; 128: 148-156, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35921937

RESUMEN

Autophagy impacts the replication cycle of many viruses. Grass Carp Reovirus (GCRV) is an agent that seriously affects the development of the grass carp aquaculture industry. The role of autophagy in GCRV infection is not clearly understood. In this study, we identified that GCRV infection triggered autophagy in CIK cells, which was demonstrated by transmission electron microscopy, the conversion of LC3B I to LC3B II and the level of autophagy substrate p62. Furthermore, we found that GCRV infection activated Akt-mTOR signaling pathway, and the conversion of LC3B I to LC3B II was increased by inhibiting mTOR with rapamycin (Rap) but decreased by activating Akt with insulin. We then assessed the effects of autophagy on GCRV replication. We found that inducing autophagy with Rap promoted GCRV proliferation but inhibiting autophagy with 3 MA or CQ inhibited GCRV replication in CIK cells. Moreover, it was found that enhancing Akt-mTOR activity by insulin, GCRV VP7 protein and viral titers of GCRV were decreased. Collectively, these results indicated that GCRV infection induced autophagy involved in GCRV replication via the Akt-mTOR signal pathway. Thus, new insights into GCRV pathogenesis and antiviral treatment strategies are provided.


Asunto(s)
Carpas , Enfermedades de los Peces , Insulinas , Orthoreovirus , Infecciones por Reoviridae , Reoviridae , Animales , Antivirales/farmacología , Autofagia , Insulinas/farmacología , Insulinas/uso terapéutico , Proteínas Proto-Oncogénicas c-akt , Infecciones por Reoviridae/metabolismo , Infecciones por Reoviridae/veterinaria , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/genética , Replicación Viral
9.
J Virol ; 96(9): e0051522, 2022 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-35416720

RESUMEN

Viral antagonism of innate immune pathways is a common mechanism by which viruses evade immune surveillance. Infection of host cells with reovirus leads to the blockade of NF-κB, a key transcriptional regulator of the hosts' innate immune response. One mechanism by which reovirus infection results in inhibition of NF-κB is through a diminishment in levels of upstream activators, IKKß and NEMO. Here, we demonstrate a second, distinct mechanism by which reovirus blocks NF-κB. We report that expression of a single viral protein, σ3, is sufficient to inhibit expression of NF-κB target genes. Further, σ3-mediated blockade of NF-κB occurs without changes to IκB kinase (IKK) levels or activity. Among NF-κB targets, the expression of type I interferon is significantly diminished by σ3 expression. Expression of NF-κB target genes varies following infection with closely related reovirus strains. Our genetic analysis identifies that these differences are controlled by polymorphisms in the amino acid sequence of σ3. This work identifies a new role for reovirus σ3 as a viral antagonist of NF-κB-dependent antiviral pathways. IMPORTANCE Host cells mount a response to curb virus replication in infected cells and prevent spread of virus to neighboring, as yet uninfected, cells. The NF-κB family of proteins is important for the cell to mediate this response. In this study, we show that a single protein, σ3, produced by mammalian reovirus, impairs the function of NF-κB. We demonstrate that by blocking NF-κB, σ3 diminishes the hosts' response to infection to promote viral replication. This work identifies a second, previously unknown, mechanism by which reovirus blocks this aspect of the host cell response.


Asunto(s)
Orthoreovirus , Infecciones por Reoviridae , Reoviridae , Animales , Antivirales , Mamíferos , FN-kappa B/metabolismo , Orthoreovirus/metabolismo , Reoviridae/fisiología , Infecciones por Reoviridae/metabolismo , Transducción de Señal
10.
PLoS Pathog ; 18(3): e1010322, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35263388

RESUMEN

Cholesterol homeostasis is required for the replication of many viruses, including Ebola virus, hepatitis C virus, and human immunodeficiency virus-1. Niemann-Pick C1 (NPC1) is an endosomal-lysosomal membrane protein involved in cholesterol trafficking from late endosomes and lysosomes to the endoplasmic reticulum. We identified NPC1 in CRISPR and RNA interference screens as a putative host factor for infection by mammalian orthoreovirus (reovirus). Following internalization via clathrin-mediated endocytosis, the reovirus outer capsid is proteolytically removed, the endosomal membrane is disrupted, and the viral core is released into the cytoplasm where viral transcription, genome replication, and assembly take place. We found that reovirus infection is significantly impaired in cells lacking NPC1, but infection is restored by treatment of cells with hydroxypropyl-ß-cyclodextrin, which binds and solubilizes cholesterol. Absence of NPC1 did not dampen infection by infectious subvirion particles, which are reovirus disassembly intermediates that bypass the endocytic pathway for infection of target cells. NPC1 is not required for reovirus attachment to the plasma membrane, internalization into cells, or uncoating within endosomes. Instead, NPC1 is required for delivery of transcriptionally active reovirus core particles from endosomes into the cytoplasm. These findings suggest that cholesterol homeostasis, ensured by NPC1 transport activity, is required for reovirus penetration into the cytoplasm, pointing to a new function for NPC1 and cholesterol homeostasis in viral infection.


Asunto(s)
Infecciones por Reoviridae , Reoviridae , Animales , Colesterol/metabolismo , Endosomas/metabolismo , Homeostasis , Humanos , Mamíferos , Proteína Niemann-Pick C1/metabolismo , Reoviridae/metabolismo , Infecciones por Reoviridae/metabolismo
11.
PLoS Pathog ; 18(3): e1010398, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35320319

RESUMEN

Successful initiation of infection by many different viruses requires their uptake into the endosomal compartment. While some viruses exit this compartment early, others must reach the degradative, acidic environment of the late endosome. Mammalian orthoreovirus (reovirus) is one such late penetrating virus. To identify host factors that are important for reovirus infection, we performed a CRISPR-Cas9 knockout (KO) screen that targets over 20,000 genes in fibroblasts derived from the embryos of C57/BL6 mice. We identified seven genes (WDR81, WDR91, RAB7, CCZ1, CTSL, GNPTAB, and SLC35A1) that were required for the induction of cell death by reovirus. Notably, CRISPR-mediated KO of WD repeat-containing protein 81 (WDR81) rendered cells resistant to reovirus infection. Susceptibility to reovirus infection was restored by complementing KO cells with human WDR81. Although the absence of WDR81 did not affect viral attachment efficiency or uptake into the endosomal compartments for initial disassembly, it reduced viral gene expression and diminished infectious virus production. Consistent with the role of WDR81 in impacting the maturation of endosomes, WDR81-deficiency led to the accumulation of reovirus particles in dead-end compartments. Though WDR81 was dispensable for infection by VSV (vesicular stomatitis virus), which exits the endosomal system at an early stage, it was required for VSV-EBO GP (VSV that expresses the Ebolavirus glycoprotein), which must reach the late endosome to initiate infection. These results reveal a previously unappreciated role for WDR81 in promoting the replication of viruses that transit through late endosomes.


Asunto(s)
Infecciones por Reoviridae , Reoviridae , Animales , Sistemas CRISPR-Cas , Endosomas/metabolismo , Mamíferos , Ratones , Reoviridae/genética , Infecciones por Reoviridae/metabolismo , Repeticiones WD40
12.
Int J Mol Sci ; 22(21)2021 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-34769442

RESUMEN

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.


Asunto(s)
Carpas/metabolismo , Factor D del Complemento/metabolismo , Proteínas de Peces/metabolismo , Infecciones por Reoviridae/metabolismo , Reoviridae/fisiología , Secuencia de Aminoácidos , Animales , Carpas/genética , Carpas/virología , Clonación Molecular/métodos , Factor D del Complemento/genética , Enfermedades de los Peces/genética , Enfermedades de los Peces/patología , Proteínas de Peces/genética , Filogenia , Infecciones por Reoviridae/genética , Infecciones por Reoviridae/patología , Infecciones por Reoviridae/virología , Análisis de Secuencia de ADN/métodos , Homología de Secuencia de Aminoácido
13.
PLoS Pathog ; 17(7): e1009494, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34237110

RESUMEN

The mammalian orthoreovirus double-stranded (ds) RNA-binding protein σ3 is a multifunctional protein that promotes viral protein synthesis and facilitates viral entry and assembly. The dsRNA-binding capacity of σ3 correlates with its capacity to prevent dsRNA-mediated activation of protein kinase R (PKR). However, the effect of σ3 binding to dsRNA during viral infection is largely unknown. To identify functions of σ3 dsRNA-binding activity during reovirus infection, we engineered a panel of thirteen σ3 mutants and screened them for the capacity to bind dsRNA. Six mutants were defective in dsRNA binding, and mutations in these constructs cluster in a putative dsRNA-binding region on the surface of σ3. Two recombinant viruses expressing these σ3 dsRNA-binding mutants, K287T and R296T, display strikingly different phenotypes. In a cell-type dependent manner, K287T, but not R296T, replicates less efficiently than wild-type (WT) virus. In cells in which K287T virus demonstrates a replication deficit, PKR activation occurs and abundant stress granules (SGs) are formed at late times post-infection. In contrast, the R296T virus retains the capacity to suppress activation of PKR and does not mediate formation of SGs at late times post-infection. These findings indicate that σ3 inhibits PKR independently of its capacity to bind dsRNA. In infected mice, K287T produces lower viral titers in the spleen, liver, lungs, and heart relative to WT or R296T. Moreover, mice inoculated with WT or R296T viruses develop myocarditis, whereas those inoculated with K287T do not. Overall, our results indicate that σ3 functions to suppress PKR activation and subsequent SG formation during viral infection and that these functions correlate with virulence in mice.


Asunto(s)
Miocarditis/virología , Proteínas de Unión al ARN/metabolismo , Infecciones por Reoviridae/metabolismo , Proteínas Virales/metabolismo , Factores de Virulencia/metabolismo , Células A549 , Animales , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Miocarditis/metabolismo , eIF-2 Quinasa/metabolismo
14.
Front Immunol ; 12: 694965, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34220856

RESUMEN

Grass carp (Ctenopharyngodon idellus) is an important aquaculture species in China that is affected by serious diseases, especially hemorrhagic disease caused by grass carp reovirus (GCRV). Grass carp have previously shown age-dependent susceptibility to GCRV, however, the mechanism by which this occurs remains poorly understood. Therefore, we performed transcriptome and metabolome sequencing on five-month-old (FMO) and three-year-old (TYO) grass carp to identify the potential mechanism. Viral challenge experiments showed that FMO fish were susceptible, whereas TYO fish were resistant to GCRV. RNA-seq showed that the genes involved in immune response, antigen presentation, and phagocytosis were significantly upregulated in TYO fish before the GCRV infection and at the early stage of infection. Metabolome sequencing showed that most metabolites were upregulated in TYO fish and downregulated in FMO fish after virus infection. Intragroup analysis showed that arachidonic acid metabolism was the most significantly upregulated pathway in TYO fish, whereas choline metabolism in cancer and glycerophospholispid metabolism were significantly downregulated in FMO fish after virus infection. Intergroup comparison revealed that metabolites from carbohydrate, amino acid, glycerophospholipid, and nucleotide metabolism were upregulated in TYO fish when compared with FMO fish. Moreover, the significantly differentially expressed metabolites showed antiviral effects both in vivo and in vitro. Based on these results, we concluded that the immune system and host biosynthesis and metabolism, can explain the age-dependent viral susceptibility in grass carp.


Asunto(s)
Carpas/virología , Enfermedades de los Peces/virología , Genómica , Metaboloma , Metabolómica , Infecciones por Reoviridae/veterinaria , Reoviridae/patogenicidad , Transcriptoma , Factores de Edad , Animales , Carpas/genética , Carpas/metabolismo , Células Cultivadas , Cromatografía Liquida/veterinaria , Metabolismo Energético , Enfermedades de los Peces/genética , Enfermedades de los Peces/metabolismo , Perfilación de la Expresión Génica/veterinaria , Interacciones Huésped-Patógeno , RNA-Seq/veterinaria , Infecciones por Reoviridae/genética , Infecciones por Reoviridae/metabolismo , Infecciones por Reoviridae/virología , Espectrometría de Masas en Tándem/veterinaria
15.
Fish Physiol Biochem ; 47(4): 857-867, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-33745109

RESUMEN

Exosomes are small membrane-enclosed vesicles secreted by various types of cells. Exosomes not only participate in different physiological processes in cells, but also involve in the cellular responses to viral infection. Grass carp reovirus (GCRV) is a non-enveloped virus with segmented, double-stranded RNA genome. Nowadays, the exact role of exosomes in regulating the life cycle of GCRV infection is still unclear. In this study, the exosomes secreted from Ctenopharyngodon idellus kidney (CIK) cells infected or uninfected with GCRV were isolated, and the differential protein expression profiles were analyzed by proteomic technologies. A total of 1297 proteins were identified in the isolated exosomes. The differentially abundant proteins were further analyzed with functional categories, and numerous important pathways were regulated by exosomes in GCRV-infected CIK cells. These exosomal proteins were estimated to interact with the genes (proteins) of the top 10 most enriched signaling pathways. Furthermore, GW4869 exosome inhibitor suppressed the expression level of VP7 in GCRV-infected cells, suggesting that exosomes play a crucial role in the life cycle of GCRV infection. These findings could shed new lights on understanding the functional roles of exosomes in the cellular responses to GCRV infection.


Asunto(s)
Exosomas/metabolismo , Enfermedades de los Peces/metabolismo , Proteínas de Peces/metabolismo , Riñón/citología , Infecciones por Reoviridae/metabolismo , Compuestos de Anilina/farmacología , Animales , Compuestos de Bencilideno/farmacología , Carpas , Células Cultivadas , Exosomas/efectos de los fármacos , Exosomas/virología , Enfermedades de los Peces/virología , Riñón/virología , Proteómica , Reoviridae , Infecciones por Reoviridae/veterinaria , Infecciones por Reoviridae/virología
16.
Viruses ; 13(2)2021 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-33525628

RESUMEN

Cells are continually exposed to stressful events, which are overcome by the activation of a number of genetic pathways. The integrated stress response (ISR) is a large component of the overall cellular response to stress, which ultimately functions through the phosphorylation of the alpha subunit of eukaryotic initiation factor-2 (eIF2α) to inhibit the energy-taxing process of translation. This response is instrumental in the inhibition of viral infection and contributes to evolution in viruses. Mammalian orthoreovirus (MRV), an oncolytic virus that has shown promise in over 30 phase I-III clinical trials, has been shown to induce multiple arms within the ISR pathway, but it successfully evades, modulates, or subverts each cellular attempt to inhibit viral translation. MRV has not yet received Food and Drug Administration (FDA) approval for general use in the clinic; therefore, researchers continue to study virus interactions with host cells to identify circumstances where MRV effectiveness in tumor killing can be improved. In this review, we will discuss the ISR, MRV modulation of the ISR, and discuss ways in which MRV interaction with the ISR may increase the effectiveness of cancer therapeutics whose modes of action are altered by the ISR.


Asunto(s)
Virus Oncolíticos/fisiología , Orthoreovirus de los Mamíferos/fisiología , Infecciones por Reoviridae/virología , Estrés Fisiológico , Factor 2 Eucariótico de Iniciación/metabolismo , Humanos , Viroterapia Oncolítica , Fosforilación , Biosíntesis de Proteínas , Caperuzas de ARN/metabolismo , Infecciones por Reoviridae/metabolismo
17.
Dev Comp Immunol ; 114: 103791, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32784010

RESUMEN

CD3 is an essential component of the TCR-CD3 complex which plays a key role in adaptive immunity. Non-mammalian CD3 complex consists of CD3γ/δ, CD3ε and CD3ζ subunits. In this study, homologues of CD3γ/δ and CD3ε (termed CiCD3γ/δ and CiCD3ε) have been identified in grass carp (Ctenopharyngodon idella). Like their counterparts from other vertebrates, the CiCD3γ/δ and CiCD3ε are clustered in the same locus in the genome and encode proteins which are structurally conserved, comprising a signal peptide, an extracellular domain, a transmembrane domain and a cytoplasmic tail containing two ITAM motifs. Sequence analyses identified two novel conserved motifs in the cytoplasmic tail of CiCD3γ/δ and CiCD3ε, one is composed of an arginine and lysine motif (RK or RR) at the C terminus of CiCD3γ/δ and a proline rich domain (PxxPxP/Q) located at the N terminus of ITAM motifs of CiCD3ε. Both genes were highly expressed at the mRNA level in the spleen and gills of healthy fish and could be modulated by infection of Flavobacterium columnare and grass carp reovirus. A monoclonal antibody against the CiCD3γ/δ (GC38T) was produced and showed good reactivity with the native molecule in Western blotting analysis and flow cytometry. The CiCD3γ/δ+ cells were analysed in the primary leucocytes, accounting for 5.5% of lymphocytes isolated from spleen, 4.5% from head kidney and 2.8% from peripheral blood. The CiCD3γ/δ+ cells were localized in the gills and head kidney by fluorescent confocal microscopy.


Asunto(s)
Complejo CD3/genética , Carpas/inmunología , Proteínas de Peces/genética , Infecciones por Flavobacteriaceae/metabolismo , Flavobacterium/fisiología , Linfocitos/metabolismo , Complejos Multiproteicos/metabolismo , Subunidades de Proteína/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Infecciones por Reoviridae/metabolismo , Reoviridae/fisiología , Bazo/metabolismo , Inmunidad Adaptativa , Secuencias de Aminoácidos/genética , Animales , Complejo CD3/metabolismo , Células Cultivadas , Clonación Molecular , Proteínas de Peces/metabolismo , Infecciones por Flavobacteriaceae/inmunología , Subunidades de Proteína/genética , Transcriptoma
18.
J Virol ; 95(2)2020 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-33087464

RESUMEN

Engagement of cell surface receptors by viruses is a critical determinant of viral tropism and disease. The reovirus attachment protein σ1 binds sialylated glycans and proteinaceous receptors to mediate infection, but the specific requirements for different cell types are not entirely known. To identify host factors required for reovirus-induced cell death, we conducted a CRISPR-knockout screen targeting over 20,000 genes in murine microglial BV2 cells. Candidate genes required for reovirus to cause cell death were highly enriched for sialic acid synthesis and transport. Two of the top candidates identified, CMP N-acetylneuraminic acid synthetase (Cmas) and solute carrier family 35 member A1 (Slc35a1), promote sialic acid expression on the cell surface. Two reovirus strains that differ in the capacity to bind sialic acid, T3SA+ and T3SA-, were used to evaluate Cmas and Slc35a1 as potential host genes required for reovirus infection. Following CRISPR-Cas9 disruption of either gene, cell surface expression of sialic acid was diminished. These results correlated with decreased binding of strain T3SA+, which is capable of engaging sialic acid. Disruption of either gene did not alter the low-level binding of T3SA-, which does not engage sialic acid. Furthermore, infectivity of T3SA+ was diminished to levels similar to those of T3SA- in cells lacking Cmas and Slc35a1 by CRISPR ablation. However, exogenous expression of Cmas and Slc35a1 into the respective null cells restored sialic acid expression and T3SA+ binding and infectivity. These results demonstrate that Cmas and Slc35a1, which mediate cell surface expression of sialic acid, are required in murine microglial cells for efficient reovirus binding and infection.IMPORTANCE Attachment factors and receptors are important determinants of dissemination and tropism during reovirus-induced disease. In a CRISPR cell survival screen, we discovered two genes, Cmas and Slc35a1, which encode proteins required for sialic acid expression on the cell surface and mediate reovirus infection of microglial cells. This work elucidates host genes that render microglial cells susceptible to reovirus infection and expands current understanding of the receptors on microglial cells that are engaged by reovirus. Such knowledge may lead to new strategies to selectively target microglial cells for oncolytic applications.


Asunto(s)
N-Acilneuraminato Citidililtransferasa/metabolismo , Proteínas de Transporte de Nucleótidos/metabolismo , Infecciones por Reoviridae/virología , Reoviridae/fisiología , Animales , Proteínas de la Cápside/genética , Proteínas de la Cápside/metabolismo , Línea Celular , Membrana Celular/metabolismo , Supervivencia Celular , Ratones , Ácido N-Acetilneuramínico/metabolismo , N-Acilneuraminato Citidililtransferasa/genética , Proteínas de Transporte de Nucleótidos/genética , Receptores Virales/metabolismo , Reoviridae/genética , Reoviridae/metabolismo , Infecciones por Reoviridae/metabolismo , Acoplamiento Viral , Replicación Viral
19.
Sci Rep ; 10(1): 15315, 2020 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-32943705

RESUMEN

Duck reovirus (DRV) is a fatal member of the genus Orthoreovirus in the family Reoviridae. The disease caused by DRV leads to huge economic losses to the duck industry. Post-translational modification is an efficient strategy to enhance the immune responses to virus infection. However, the roles of protein phosphorylation in the responses of ducklings to Classic/Novel DRV (C/NDRV) infections are largely unknown. Using a high-resolution LC-MS/MS integrated to highly sensitive immune-affinity antibody method, phosphoproteomes of Cairna moschata spleen tissues under the C/NDRV infections were analyzed, producing a total of 8,504 phosphorylation sites on 2,853 proteins. After normalization with proteomic data, 392 sites on 288 proteins and 484 sites on 342 proteins were significantly changed under the C/NDRV infections, respectively. To characterize the differentially phosphorylated proteins (DPPs), a systematic bioinformatics analyses including Gene Ontology annotation, domain annotation, subcellular localization, and Kyoto Encyclopedia of Genes and Genomes pathway annotation were performed. Two important serine protease system-related proteins, coagulation factor X and fibrinogen α-chain, were identified as phosphorylated proteins, suggesting an involvement of blood coagulation under the C/NDRV infections. Furthermore, 16 proteins involving the intracellular signaling pathways of pattern-recognition receptors were identified as phosphorylated proteins. Changes in the phosphorylation levels of MyD88, NF-κB, RIP1, MDA5 and IRF7 suggested a crucial role of protein phosphorylation in host immune responses of C. moschata. Our study provides new insights into the responses of ducklings to the C/NDRV infections at PTM level.


Asunto(s)
Patos/metabolismo , Patos/virología , Orthoreovirus Aviar/patogenicidad , Infecciones por Reoviridae/metabolismo , Infecciones por Reoviridae/virología , Bazo/metabolismo , Bazo/virología , Animales , Anticuerpos Antivirales/metabolismo , Cromatografía Liquida/métodos , FN-kappa B/metabolismo , Enfermedades de las Aves de Corral/metabolismo , Enfermedades de las Aves de Corral/virología , Proteómica/métodos , Transducción de Señal/fisiología , Espectrometría de Masas en Tándem/métodos
20.
PLoS Pathog ; 16(9): e1008803, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32956403

RESUMEN

The Dearing isolate of Mammalian orthoreovirus (T3D) is a prominent model of virus-host relationships and a candidate oncolytic virotherapy. Closely related laboratory strains of T3D, originating from the same ancestral T3D isolate, were recently found to exhibit significantly different oncolytic properties. Specifically, the T3DPL strain had faster replication kinetics in a panel of cancer cells and improved tumor regression in an in vivo melanoma model, relative to T3DTD. In this study, we discover that T3DPL and T3DTD also differentially activate host signalling pathways and downstream gene transcription. At equivalent infectious dose, T3DTD induces higher IRF3 phosphorylation and expression of type I IFNs and IFN-stimulated genes (ISGs) than T3DPL. Using mono-reassortants with intermediate replication kinetics and pharmacological inhibitors of reovirus replication, IFN responses were found to inversely correlate with kinetics of virus replication. In other words, slow-replicating T3D strains induce more IFN signalling than fast-replicating T3D strains. Paradoxically, during co-infections by T3DPL and T3DTD, there was still high IRF3 phosphorylation indicating a phenodominant effect by the slow-replicating T3DTD. Using silencing and knock-out of RIG-I to impede IFN, we found that IFN induction does not affect the first round of reovirus replication but does prevent cell-cell spread in a paracrine fashion. Accordingly, during co-infections, T3DPL continues to replicate robustly despite activation of IFN by T3DTD. Using gene expression analysis, we discovered that reovirus can also induce a subset of genes in a RIG-I and IFN-independent manner; these genes were induced more by T3DPL than T3DTD. Polymorphisms in reovirus σ3 viral protein were found to control activation of RIG-I/ IFN-independent genes. Altogether, the study reveals that single amino acid polymorphisms in reovirus genomes can have large impact on host gene expression, by both changing replication kinetics and by modifying viral protein activity, such that two closely related T3D strains can induce opposite cytokine landscapes.


Asunto(s)
Proteínas de la Cápside/metabolismo , Interferones/metabolismo , Polimorfismo Genético , ARN Bicatenario/metabolismo , Proteínas de Unión al ARN/metabolismo , Receptores de Ácido Retinoico/metabolismo , Infecciones por Reoviridae/virología , Replicación Viral , Proteínas de la Cápside/genética , Citocinas , Humanos , Factor 3 Regulador del Interferón/genética , Factor 3 Regulador del Interferón/metabolismo , Orthoreovirus de los Mamíferos/fisiología , ARN Bicatenario/genética , ARN Viral/genética , ARN Viral/metabolismo , Proteínas de Unión al ARN/genética , Receptores de Ácido Retinoico/genética , Infecciones por Reoviridae/genética , Infecciones por Reoviridae/metabolismo , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...