Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
1.
Cell Host Microbe ; 32(6): 980-995.e9, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38729153

ABSTRACT

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.


Subject(s)
Capsid Proteins , Neuropilin-1 , Orthoreovirus, Mammalian , Receptors, Virus , Reoviridae Infections , Virus Internalization , Animals , Mice , Capsid Proteins/metabolism , Capsid Proteins/genetics , Neuropilin-1/metabolism , Neuropilin-1/genetics , Orthoreovirus, Mammalian/genetics , Orthoreovirus, Mammalian/physiology , Orthoreovirus, Mammalian/metabolism , Reoviridae Infections/virology , Reoviridae Infections/metabolism , Receptors, Virus/metabolism , Humans , Capsid/metabolism , Cell Line , HEK293 Cells , Protein Binding , Mice, Inbred C57BL
2.
J Virol ; 98(6): e0030524, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38771042

ABSTRACT

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.


Subject(s)
Capsid Proteins , Reoviridae Infections , Virus Replication , Animals , Mice , Capsid Proteins/metabolism , Capsid Proteins/genetics , Reoviridae Infections/virology , Reoviridae Infections/metabolism , Virus Attachment , Polymorphism, Genetic , Orthoreovirus, Mammalian/genetics , Orthoreovirus, Mammalian/physiology , Orthoreovirus, Mammalian/metabolism , Virus Assembly , Cell Line , Capsid/metabolism , Humans
3.
Mol Immunol ; 170: 131-143, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38663254

ABSTRACT

Mammalian reovirus (MRV) is a non-enveloped, gene segmented double-stranded RNA (dsRNA) virus. It is an important zoonotic pathogen that infects many mammals and vertebrates that act as natural hosts and causes respiratory and digestive tract diseases. Studies have reported that RIG-I and MDA5 in the innate immune cytoplasmic RNA-sensing RIG-like receptor (RLR) signaling pathway can recognize dsRNA from MRV and promote antiviral type I interferon (IFN) responses. However, the mechanism by which many MRV-encoded proteins evade the host innate immune response remains unclear. Here, we show that exogenous µ1 protein promoted the proliferation of MRV in vitro, while knockdown of MRV µ1 protein expression by shRNA could impair MRV proliferation. Specifically, µ1 protein inhibited MRV or poly(I:C)-induced IFN-ß expression, and attenuated RIG-I/MDA5-mediated signaling axis transduction during MRV infection. Importantly, we found that µ1 protein significantly decreased IFN-ß mRNA expression induced by MDA5, RIG-I, MAVS, TBK1, IRF3(5D), and degraded the protein expression of exogenous MDA5, RIG-I, MAVS, TBK1 and IRF3 via the proteasomal and lysosomal pathways. Additionally, we show that µ1 protein can physically interact with MDA5, RIG-I, MAVS, TBK1, and IRF3 and attenuate the RIG-I/MDA5-mediated signaling cascades by blocking the phosphorylation and nuclear translocation of IRF3. In conclusion, our findings reveal that MRV outer capsid protein µ1 is a key factor in antagonizing RLRs signaling cascades and provide new strategies for effective prevention and treatment of MRV infection.


Subject(s)
Capsid Proteins , DEAD Box Protein 58 , Interferon Regulatory Factor-3 , Interferon-Induced Helicase, IFIH1 , Orthoreovirus, Mammalian , Receptors, Immunologic , Signal Transduction , Animals , Humans , Active Transport, Cell Nucleus , Cell Nucleus/metabolism , DEAD Box Protein 58/metabolism , HEK293 Cells , Immunity, Innate/immunology , Interferon Regulatory Factor-3/metabolism , Interferon-beta/metabolism , Interferon-beta/immunology , Interferon-Induced Helicase, IFIH1/metabolism , Interferon-Induced Helicase, IFIH1/genetics , Orthoreovirus, Mammalian/immunology , Orthoreovirus, Mammalian/physiology , Phosphorylation , Protein Serine-Threonine Kinases , Reoviridae Infections/immunology , Signal Transduction/immunology , Viral Proteins/metabolism , Capsid Proteins/metabolism
4.
Nat Commun ; 14(1): 2615, 2023 05 05.
Article in English | MEDLINE | ID: mdl-37147336

ABSTRACT

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.


Subject(s)
Orthoreovirus, Mammalian , Receptors, Immunologic , Receptors, Virus , Reoviridae Infections , Animals , Humans , Mice , Antibodies, Viral , Orthoreovirus, Mammalian/physiology , Receptors, Immunologic/metabolism , Reoviridae Infections/metabolism , Receptors, Virus/metabolism
5.
J Virol ; 97(5): e0058523, 2023 05 31.
Article in English | MEDLINE | ID: mdl-37167564

ABSTRACT

Structural metastability of viral capsids is pivotal for viruses to survive in harsh environments and to undergo timely conformational changes required for cell entry. Mammalian orthoreovirus (reovirus) is a model to study capsid metastability. Following initial disassembly of the reovirus particle mediated by proteases, a metastable intermediate called the infectious subvirion particle (ISVP) is generated. Using a σ1 monoreassortant virus, we recently showed that σ1 properties affect its encapsidation on particles and the metastability of ISVPs. How metastability is impacted by σ1 and whether the lower encapsidation level of σ1 is connected to this property is unknown. To define a correlation between encapsidation of σ1 and ISVP stability, we generated mutant viruses with single amino acid polymorphisms in σ1 or those that contain chimeric σ1 molecules composed of σ1 portions from type 1 and type 3 reovirus strains. We found that under most conditions where σ1 encapsidation on the particle was lower, ISVPs displayed lower stability. Characterization of mutant viruses selected for enhanced stability via a forward genetic approach also revealed that in some cases, σ1 properties influence stability without influencing σ1 encapsidation. These data indicate that σ1 can also influence ISVP stability independent of its level of incorporation. Together, our work reveals an underappreciated effect of the σ1 attachment protein on the properties of the reovirus capsid. IMPORTANCE Reovirus particles are comprised of eight proteins. Among them, the reovirus σ1 protein functions engages cellular receptors. σ1 also influences the stability of an entry intermediate called ISVP. Here, we sought to define the basis of the link between σ1 properties and stability of ISVPs. Using variety of mutant strains, we determined that when virus preparations contain particles with a high amount of encapsidated σ1, ISVP stability is higher. Additionally, we identified portions of σ1 that impact its encapsidation and consequently the stability of ISVPs. We also determined that in some cases, σ1 properties alter stability of ISVPs without affecting encapsidation. This work highlights that proteins of these complex particles are arranged in an intricate, interconnected manner such that changing the properties of these proteins has a profound impact on the remainder of the particle.


Subject(s)
Mammalian orthoreovirus 3 , Orthoreovirus, Mammalian , Virus Internalization , Capsid/metabolism , Cell Line , Orthoreovirus, Mammalian/physiology , Mammalian orthoreovirus 3/physiology
6.
Virology ; 558: 38-48, 2021 06.
Article in English | MEDLINE | ID: mdl-33721728

ABSTRACT

Mammalian orthoreovirus (MRV) is a safe and effective cancer killing virus that has completed Phase I-III clinical trials against numerous cancer types. While many patients experience benefit from MRV therapy, pre-defined set points necessary for FDA approval have not been reached. Therefore, additional research into MRV biology and the effect of viral therapy on different tumor genetic subtypes and microenvironments is necessary to identify tumors most amenable to MRV virotherapy. In this work we analyzed the stage of viral infection necessary to inhibit HIF-1α, an aggressive cancer activator induced by hypoxia. We demonstrated that two viral capsid proteins were not necessary and that a step parallel with virus core movement across the endosomal membrane was required for this inhibition. Altogether, this work clarifies the mechanisms of MRV-induced HIF-1α inhibition and provides biological relevance for using MRV to inhibit the devastating effects of tumor hypoxia.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Oncolytic Virotherapy , Orthoreovirus, Mammalian/physiology , Prostatic Neoplasms , Tumor Microenvironment/physiology , Cell Line, Tumor , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/analysis , Male , Tumor Hypoxia
7.
Viruses ; 13(2)2021 02 11.
Article in English | MEDLINE | ID: mdl-33670092

ABSTRACT

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.


Subject(s)
Host-Pathogen Interactions/physiology , Orthoreovirus, Mammalian/genetics , Orthoreovirus, Mammalian/physiology , Protein Biosynthesis/genetics , Virus Replication/physiology , Animals , Humans , RNA, Viral/genetics , Reoviridae Infections/pathology , Ribosomes/metabolism , Viral Proteins/genetics
8.
Viruses ; 13(2)2021 01 28.
Article in English | MEDLINE | ID: mdl-33525628

ABSTRACT

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.


Subject(s)
Oncolytic Viruses/physiology , Orthoreovirus, Mammalian/physiology , Reoviridae Infections/virology , Stress, Physiological , Eukaryotic Initiation Factor-2/metabolism , Humans , Oncolytic Virotherapy , Phosphorylation , Protein Biosynthesis , RNA Caps/metabolism , Reoviridae Infections/metabolism
9.
Viruses ; 13(2)2021 01 21.
Article in English | MEDLINE | ID: mdl-33494426

ABSTRACT

Mammalian orthoreovirus (reovirus), a dsRNA virus with a multilayered capsid, serves as a model system for studying the entry of similar viruses. The outermost layer of this capsid undergoes processing to generate a metastable intermediate. The metastable particle undergoes further remodeling to generate an entry-capable form that delivers the genome-containing inner capsid, or core, into the cytoplasm. In this review, we highlight capsid proteins and the intricacies of their interactions that control the stability of the capsid and consequently impact capsid structural changes that are prerequisites for entry. We also discuss a novel proviral role of host membranes in promoting capsid conformational transitions. Current knowledge gaps in the field that are ripe for future investigation are also outlined.


Subject(s)
Capsid Proteins/metabolism , Orthoreovirus, Mammalian/physiology , Proteolysis , Reoviridae Infections/virology , Virion/physiology , Virus Internalization , Animals , Capsid Proteins/genetics , Cell Line , Mice , Orthoreovirus, Mammalian/genetics , Protein Domains , Reoviridae Infections/genetics , Virion/genetics
10.
J Virol ; 95(4)2021 01 28.
Article in English | MEDLINE | ID: mdl-33208448

ABSTRACT

Mammalian orthoreovirus (reovirus) spreads from the site of infection to every organ system in the body via the blood. However, mechanisms that underlie reovirus hematogenous spread remain undefined. Nonstructural protein σ1s is a critical determinant of reovirus bloodstream dissemination that is required for efficient viral replication in many types of cultured cells. Here, we used the specificity of the σ1s protein for promoting hematogenous spread as a platform to uncover a role for lymphatic type 1 interferon (IFN-1) responses in limiting reovirus systemic dissemination. We found that replication of a σ1s-deficient reovirus was restored to wild-type levels in cells with defective interferon-α receptor (IFNAR1) signaling. Reovirus spreads systemically following oral inoculation of neonatal mice, whereas the σ1s-null virus remains localized to the intestine. We found that σ1s enables reovirus spread in the presence of a functional IFN-1 response, as the σ1s-deficient reovirus disseminated comparably to wild-type virus in IFNAR1-/- mice. Lymphatics are hypothesized to mediate reovirus spread from the intestine to the bloodstream. IFNAR1 deletion from cells expressing lymphatic vessel endothelium receptor 1 (LYVE-1), a marker for lymphatic endothelial cells, enabled the σ1s-deficient reovirus to disseminate systemically. Together, our findings indicate that IFN-1 responses in lymphatics limit reovirus dissemination. Our data further suggest that the lymphatics are an important conduit for reovirus hematogenous spread.IMPORTANCE Type 1 interferons (IFN-1) are critical host responses to viral infection. However, the contribution of IFN-1 responses to control of viruses in specific cell and tissue types is not fully defined. Here, we identify IFN-1 responses in lymphatics as important for limiting reovirus dissemination. We found that nonstructural protein σ1s enhances reovirus resistance to IFN-1 responses, as a reovirus mutant lacking σ1s was more sensitive to IFN-1 than wild-type virus. In neonatal mice, σ1s is required for reovirus systemic spread. We used tissue-specific IFNAR1 deletion in combination with the IFN-1-sensitive σ1s-null reovirus as a tool to test how IFN-1 responses in lymphatics affect reovirus systemic spread. Deletion of IFNAR1 in lymphatic cells using Cre-lox technology enabled dissemination of the IFN-1-sensitive σ1s-deficient reovirus. Together, our results indicate that IFN-1 responses in lymphatics are critical for controlling reovirus systemic spread.


Subject(s)
Endothelial Cells/immunology , Interferon Type I/immunology , Orthoreovirus, Mammalian/physiology , Receptor, Interferon alpha-beta/immunology , Reoviridae Infections , Viral Nonstructural Proteins/immunology , Animals , Animals, Newborn , Endothelial Cells/cytology , Fibroblasts , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Reoviridae Infections/immunology , Reoviridae Infections/virology
11.
Dev Comp Immunol ; 116: 103909, 2021 03.
Article in English | MEDLINE | ID: mdl-33129882

ABSTRACT

Mre11A is considered as a cytosolic DNA receptor in mammals. However, it is rarely known about Mre11A in other vertebrates. Recently, a mammalian ortholog of Mre11A has been identified in grass carp (Ctenopharyngodon idellus) in our lab. Phylogenetic-tree analysis provided evidence for a close genetic relationship between C.idellus Mre11A and Carassius auratus Mre11A. The tissue expression profile of CiMre11A was detected, with a relatively higher level of expression in kidney, intestines, liver and spleen than that in other tissues after grass carp reovirus (GCRV) infection. Similarly, CiMre11A was also up-regulated in CIK cells after treatment with GCRV. Q-PCR and dual-luciferase assays indicated that the transcription levels of IFN1 and ISG15 were inhibited by CiMre11A knockdown, but were gradually augmented after CIK cells were transfected with increasing amounts of CiMre11A. Subcellular localization assays showed that a part of CiMre11A was translocated from the nucleus to the cytoplasm. Co-immunoprecipitation and co-localization assays demonstrated that CiMre11A interacts with CiSTING in response to GCRV infection. In CIK cells, the expressions of both IFN1 and ISG15 were acutely up-regulated by CiMre11A overexpression, as well as by co-overexpression of CiMre11A and CiSTING. CiMre11A and CiSTING induced the phosphorylation and cytoplasmic-to-nuclear translocation of IRF7 in CIK cells. The multiplication of GCRV in CIK cells was inhibited by the overexpression of CiMre11A and CiSTING.


Subject(s)
Carps/immunology , Fish Diseases/immunology , Fish Proteins/immunology , Interferon Type I/immunology , MRE11 Homologue Protein/immunology , Amino Acid Sequence , Animals , Carps/genetics , Carps/virology , Cytokines/genetics , Cytokines/immunology , Cytokines/metabolism , Disease Resistance/genetics , Disease Resistance/immunology , Fish Diseases/genetics , Fish Diseases/virology , Fish Proteins/classification , Fish Proteins/genetics , Gene Expression Profiling , Gene Expression Regulation/immunology , Host-Pathogen Interactions/immunology , Interferon Type I/genetics , Interferon Type I/metabolism , MRE11 Homologue Protein/classification , MRE11 Homologue Protein/genetics , Orthoreovirus, Mammalian/immunology , Orthoreovirus, Mammalian/physiology , Phylogeny , Protein Binding , Sequence Homology, Amino Acid , Ubiquitins/genetics , Ubiquitins/immunology , Ubiquitins/metabolism
12.
PLoS Pathog ; 16(9): e1008803, 2020 09.
Article in English | MEDLINE | ID: mdl-32956403

ABSTRACT

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.


Subject(s)
Capsid Proteins/metabolism , Interferons/metabolism , Polymorphism, Genetic , RNA, Double-Stranded/metabolism , RNA-Binding Proteins/metabolism , Receptors, Retinoic Acid/metabolism , Reoviridae Infections/virology , Virus Replication , Capsid Proteins/genetics , Cytokines , Humans , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/metabolism , Orthoreovirus, Mammalian/physiology , RNA, Double-Stranded/genetics , RNA, Viral/genetics , RNA, Viral/metabolism , RNA-Binding Proteins/genetics , Receptors, Retinoic Acid/genetics , Reoviridae Infections/genetics , Reoviridae Infections/metabolism , Signal Transduction
13.
J Virol ; 92(15)2018 08 01.
Article in English | MEDLINE | ID: mdl-29769334

ABSTRACT

Viral nonstructural proteins, which are not packaged into virions, are essential for the replication of most viruses. Reovirus, a nonenveloped, double-stranded RNA (dsRNA) virus, encodes three nonstructural proteins that are required for viral replication and dissemination in the host. The reovirus nonstructural protein σNS is a single-stranded RNA (ssRNA)-binding protein that must be expressed in infected cells for production of viral progeny. However, the activities of σNS during individual steps of the reovirus replication cycle are poorly understood. We explored the function of σNS by disrupting its expression during infection using cells expressing a small interfering RNA (siRNA) targeting the σNS-encoding S3 gene and found that σNS is required for viral genome replication. Using complementary biochemical assays, we determined that σNS forms complexes with viral and nonviral RNAs. We also discovered, using in vitro and cell-based RNA degradation experiments, that σNS increases the RNA half-life. Cryo-electron microscopy revealed that σNS and ssRNAs organize into long, filamentous structures. Collectively, our findings indicate that σNS functions as an RNA-binding protein that increases the viral RNA half-life. These results suggest that σNS forms RNA-protein complexes in preparation for genome replication.IMPORTANCE Following infection, viruses synthesize nonstructural proteins that mediate viral replication and promote dissemination. Viruses from the family Reoviridae encode nonstructural proteins that are required for the formation of progeny viruses. Although nonstructural proteins of different viruses in the family Reoviridae diverge in primary sequence, they are functionally homologous and appear to facilitate conserved mechanisms of dsRNA virus replication. Using in vitro and cell culture approaches, we found that the mammalian reovirus nonstructural protein σNS binds and stabilizes viral RNA and is required for genome synthesis. This work contributes new knowledge about basic mechanisms of dsRNA virus replication and provides a foundation for future studies to determine how viruses in the family Reoviridae assort and replicate their genomes.


Subject(s)
Genome, Viral , Orthoreovirus, Mammalian/physiology , RNA, Viral/biosynthesis , RNA-Binding Proteins/metabolism , Viral Nonstructural Proteins/metabolism , Virus Replication/physiology , HEK293 Cells , Humans , RNA, Viral/genetics , RNA-Binding Proteins/genetics , Viral Nonstructural Proteins/genetics
14.
Adv Virus Res ; 100: 223-246, 2018.
Article in English | MEDLINE | ID: mdl-29551138

ABSTRACT

Viruses are constantly engaged in a molecular arms race with the host, where efficient and tactical use of cellular receptors benefits critical steps in infection. Receptor use dictates initiation, establishment, and spread of viral infection to new tissues and hosts. Mammalian orthoreoviruses (reoviruses) are pervasive pathogens that use multiple receptors to overcome protective host barriers to disseminate from sites of initial infection and cause disease in young mammals. In particular, reovirus invades the central nervous system (CNS) with serotype-dependent tropism and disease. A single viral gene, encoding the attachment protein σ1, segregates with distinct patterns of CNS injury. Despite the identification and characterization of several reovirus receptors, host factors that dictate tropism via interaction with σ1 remain undefined. Here, we summarize the state of the reovirus receptor field and discuss open questions toward understanding how the reovirus attachment protein dictates CNS tropism.


Subject(s)
Capsid Proteins/metabolism , Orthoreovirus, Mammalian/physiology , Receptors, Virus/metabolism , Reoviridae Infections/virology , Animals , Host-Pathogen Interactions , Humans , Orthoreovirus, Mammalian/pathogenicity , Viral Tropism/physiology , Virus Internalization , Virus Replication
15.
Virology ; 518: 77-86, 2018 05.
Article in English | MEDLINE | ID: mdl-29455064

ABSTRACT

Mammalian reovirus viral factories (VFs) form filamentous or globular structures depending on the viral strain. In this study, we attempt to characterize the dynamics of both filamentous and globular VFs. Here, we present evidence demonstrating that globular VFs are dynamic entities coalescing between them, thereby gaining in size and concomitantly decreasing in numbers during the course of the infection. Additionally, both kinds of VFs condense into a perinuclear position. Our results show that globular VFs rely on an intact MT-network for dynamic motion, structural assembly, and maintenance and for perinuclear condensation. Interestingly, dynein localizes in both kinds of VFs, having a role at least in large globular VFs formation. To study filamentous VF dynamics, we used different transfection ratios of µNS with filamentous µ2. We found a MT-network dependency for VF-like structures perinuclear condensation. Also, µNS promotes VFLSs perinuclear positioning as well as an increase in acetylated tubulin levels.


Subject(s)
Microtubules/physiology , Orthoreovirus, Mammalian/physiology , Virus Replication/physiology , Animals , Cell Line , Gene Expression Regulation, Viral/physiology , Molecular Motor Proteins
16.
Pediatr Res ; 83(5): 1057-1066, 2018 05.
Article in English | MEDLINE | ID: mdl-29364865

ABSTRACT

BackgroundInfants and young children are particularly susceptible to viral encephalitis; however, the mechanisms are unknown. We determined the age-dependent contribution of innate and adaptive immune functions to reovirus-induced encephalitis in mice.MethodsNewborn wild-type mice, 2-20 days of age, were inoculated with reovirus or diluent and monitored for mortality, weight gain, and viral load. Four- and fifteen-day-old IFNAR-/- and RAG2-/- mice were inoculated with reovirus and similarly monitored.ResultsWeight gain was impaired in mice inoculated with reovirus at 8 days of age or less. Clinical signs of encephalitis were detected in mice inoculated at 10 days of age or less. Mortality decreased when mice were inoculated after 6 days of age. Survival was ≤15% in wild type (WT), RAG2-/-, and IFNAR-/- mice inoculated at 4 days of age. All WT mice, 92% of RAG2-/- mice, and only 48% of IFNAR-/- mice survived following inoculation at 15 days of age.ConclusionsSusceptibility of mice to reovirus-induced disease decreases between 6 and 8 days of age. Enhanced reovirus virulence in IFNAR-/- mice relative to WT and RAG2-/- mice inoculated at 15 days of age suggests that maturation of the type-I interferon response contributes to age-related mortality following reovirus infection.


Subject(s)
Age Factors , DNA-Binding Proteins/genetics , Encephalitis, Viral/immunology , Receptor, Interferon alpha-beta/genetics , Reoviridae Infections/immunology , Adaptive Immunity , Animals , Apoptosis , Brain/metabolism , Cell Line , DNA-Binding Proteins/immunology , Gene Expression Regulation, Viral , Immunity, Innate , Interferon Type I/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Orthoreovirus, Mammalian/genetics , Orthoreovirus, Mammalian/physiology , Receptor, Interferon alpha-beta/immunology , Spleen/metabolism , Viral Load , Virus Replication
17.
J Virol ; 91(22)2017 11 15.
Article in English | MEDLINE | ID: mdl-28878073

ABSTRACT

Within infected host cells, mammalian orthoreovirus (MRV) forms viral factories (VFs), which are sites of viral transcription, translation, assembly, and replication. The MRV nonstructural protein µNS comprises the structural matrix of VFs and is involved in recruiting other viral proteins to VF structures. Previous attempts have been made to visualize VF dynamics in live cells, but due to current limitations in recovery of replicating reoviruses carrying large fluorescent protein tags, researchers have been unable to directly assess VF dynamics from virus-produced µNS. We set out to develop a method to overcome this obstacle by utilizing the 6-amino-acid (CCPGCC) tetracysteine (TC) tag and FlAsH-EDT2 reagent. The TC tag was introduced into eight sites throughout µNS, and the capacity of the TC-µNS fusion proteins to form virus factory-like (VFL) structures and colocalize with virus proteins was characterized. Insertion of the TC tag interfered with recombinant virus rescue in six of the eight mutants, likely as a result of loss of VF formation or important virus protein interactions. However, two recombinant (r)TC-µNS viruses were rescued and VF formation, colocalization with associating virus proteins, and characterization of virus replication were subsequently examined. Furthermore, the rTC-µNS viruses were utilized to infect cells and examine VF dynamics using live-cell microscopy. These experiments demonstrate active VF movement with fusion events as well as transient interactions between individual VFs and demonstrate the importance of microtubule stability for VF fusion during MRV infection. This work provides important groundwork for future in-depth studies of VF dynamics and host cell interactions.IMPORTANCE MRV has historically been used as a model to study the double-stranded RNA (dsRNA) Reoviridae family, the members of which infect and cause disease in humans, animals, and plants. During infection, MRV forms VFs that play a critical role in virus infection but remain to be fully characterized. To study VFs, researchers have focused on visualizing the nonstructural protein µNS, which forms the VF matrix. This work provides the first evidence of recovery of replicating reoviruses in which VFs can be labeled in live cells via introduction of a TC tag into the µNS open reading frame. Characterization of each recombinant reovirus sheds light on µNS interactions with viral proteins. Moreover, utilizing the TC-labeling FlAsH-EDT2 biarsenical reagent to visualize VFs, evidence is provided of dynamic VF movement and interactions at least partially dependent on intact microtubules.


Subject(s)
Capsid Proteins/metabolism , Exosomes/virology , Orthoreovirus, Mammalian/physiology , RNA, Viral/metabolism , Virus Replication/physiology , Antibodies, Monoclonal, Murine-Derived/chemistry , Antibodies, Viral/chemistry , Capsid Proteins/genetics , Cell Line , Exosomes/genetics , Exosomes/metabolism , Humans , RNA, Viral/genetics
18.
Oncotarget ; 8(21): 35138-35153, 2017 May 23.
Article in English | MEDLINE | ID: mdl-28422714

ABSTRACT

New therapeutic interventions are essential for improved management of patients with metastatic colorectal cancer (mCRC). This is especially critical for those patients whose tumors harbor a mutation in the KRAS oncogene (40-45% of all patients). This patient cohort is excluded from receiving anti-EGFR monoclonal antibodies that have added a significant therapeutic benefit for KRAS wild type CRC patients. Reovirus, a double stranded (ds) RNA virus is in clinical development for patients with chemotherapy refractory KRAS mutated tumors. Toll Like Receptor (TLR) 3, a member of the toll like receptor family of the host innate immune system is the pattern recognition motif for dsRNA pathogens. Using TLR3 expressing commercial HEK-BlueTM-hTLR3 cells we confirm that TLR3 is the host pattern recognition motif responsible for the detection of reovirus. Further, our investigation with KRAS mutated HCT116 cell line showed that effective expression of host TLR3 dampens the infection potential of reovirus by mounting a robust innate immune response. Down regulation of TLR3 expression with siRNA improves the anticancer activity of reovirus. In vivo experiments using human CRC cells derived xenografts in athymic mice further demonstrate the beneficial effects of TLR3 knock down by improving tumor response rates to reovirus. Strategies to mitigate the TLR3 response pathway can be utilized as a tool towards improved reovirus efficacy to specifically target the dissemination of KRAS mutated CRC.


Subject(s)
Colorectal Neoplasms/therapy , Orthoreovirus, Mammalian/physiology , Proto-Oncogene Proteins p21(ras)/genetics , Toll-Like Receptor 3/genetics , Animals , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Gene Knockdown Techniques , HCT116 Cells , Humans , Mice , Oncolytic Virotherapy , Orthoreovirus, Mammalian/immunology , Signal Transduction , Xenograft Model Antitumor Assays
19.
J Virol ; 91(11)2017 06 01.
Article in English | MEDLINE | ID: mdl-28298603

ABSTRACT

Reoviruses, like many eukaryotic viruses, contain an inverted 7-methylguanosine (m7G) cap linked to the 5' nucleotide of mRNA. The traditional functions of capping are to promote mRNA stability, protein translation, and concealment from cellular proteins that recognize foreign RNA. To address the role of mRNA capping during reovirus replication, we assessed the benefits of adding the African swine fever virus NP868R capping enzyme during reovirus rescue. C3P3, a fusion protein containing T7 RNA polymerase and NP868R, was found to increase protein expression 5- to 10-fold compared to T7 RNA polymerase alone while enhancing reovirus rescue from the current reverse genetics system by 100-fold. Surprisingly, RNA stability was not increased by C3P3, suggesting a direct effect on protein translation. A time course analysis revealed that C3P3 increased protein synthesis within the first 2 days of a reverse genetics transfection. This analysis also revealed that C3P3 enhanced processing of outer capsid µ1 protein to µ1C, a previously described hallmark of reovirus assembly. Finally, to determine the rate of infectious-RNA incorporation into new virions, we developed a new recombinant reovirus S1 gene that expressed the fluorescent protein UnaG. Following transfection of cells with UnaG and infection with wild-type virus, passage of UnaG through progeny was significantly enhanced by C3P3. These data suggest that capping provides nontraditional functions to reovirus, such as promoting assembly and infectious-RNA incorporation.IMPORTANCE Our findings expand our understanding of how viruses utilize capping, suggesting that capping provides nontraditional functions to reovirus, such as promoting assembly and infectious-RNA incorporation, in addition to enhancing protein translation. Beyond providing mechanistic insight into reovirus replication, our findings also show that reovirus reverse genetics rescue is enhanced 100-fold by the NP868R capping enzyme. Since reovirus shows promise as a cancer therapy, efficient reovirus reverse genetics rescue will accelerate production of recombinant reoviruses as candidates to enhance therapeutic potency. NP868R-assisted reovirus rescue will also expedite production of recombinant reovirus for mechanistic insights into reovirus protein function and structure.


Subject(s)
African Swine Fever Virus/enzymology , Nucleotidyltransferases/metabolism , Orthoreovirus, Mammalian/genetics , Orthoreovirus, Mammalian/physiology , RNA, Viral/metabolism , Virion/physiology , Virus Assembly , African Swine Fever Virus/genetics , Cell Line , Recombinant Fusion Proteins/metabolism , Reverse Genetics , Virion/genetics , Virus Replication
20.
Mol Biol (Mosk) ; 51(1): 111-117, 2017.
Article in Russian | MEDLINE | ID: mdl-28251973

ABSTRACT

The experimental study identified the antiviral activity of Bacillus pumilus RNase (binase) against the reovirus of serotype 1/strain Lang. For the first time, it has been found that 50 µg/mL of binase effectively reduced the hemagglutinin and cytocidal activity of reovirus in Vero cell line. The preincubation of the enzyme with reovirus before infection of the cells inhibited the viral replication. To determine the stagedependent effect of reovirus reproduction upon binase inhibition, the infected cells were treated with binase or RNase A at different phases of the infectious cycle. The treatment of virus-infected cells has revealed that both enzymes have a maximal antiviral effect on the reovirus propagation during early phases of the reovirus reproduction cycle, with binase being more effective than RNase A. It has been hypothesized that the combined action of the oncolytic reovirus and binase is promising for the elimination of tumor cells carrying mutated RAS gene.


Subject(s)
Endoribonucleases/pharmacology , Orthoreovirus, Mammalian/physiology , Ribonucleases/pharmacology , Animals , Antiviral Agents/pharmacology , Chlorocebus aethiops , Orthoreovirus, Mammalian/drug effects , Serogroup , Vero Cells , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL