Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 94
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 24(3)2023 Jan 24.
Article in English | MEDLINE | ID: mdl-36768641

ABSTRACT

Reoviruses are used as oncolytic viruses to destroy tumor cells. The concomitant induction of anti-tumor immune responses enhances the efficacy of therapy in tumors with low amounts of immune infiltrates before treatment. The reoviruses should provoke immunogenic cell death (ICD) to stimulate a tumor cell-directed immune response. Necroptosis is considered a major form of ICD, and involves receptor-interacting protein kinase 1 (RIPK1), RIPK3 and phosphorylation of mixed-lineage kinase domain-like protein (MLKL). This leads to cell membrane disintegration and the release of damage-associated molecular patterns that can activate immune responses. Reovirus Type 3 Dearing (T3D) can induce necroptosis in mouse L929 fibroblast cells and mouse embryonic fibroblasts. Most human tumor cell lines have a defect in RIPK3 expression and consequently fail to induce necroptosis as measured by MLKL phosphorylation. We used the human colorectal adenocarcinoma HT29 cell line as a model to study necroptosis in human cells since this cell line has frequently been described in necroptosis-related studies. To stimulate MLKL phosphorylation and induce necroptosis, HT29 cells were treated with a cocktail consisting of TNFα, the SMAC mimetic BV6, and the caspase inhibitor Z-VAD-FMK. While this treatment induced necroptosis, three different reovirus T3D variants, i.e., the plasmid-based reverse genetics generated virus (T3DK), the wild-type reovirus T3D isolate R124, and the junction adhesion molecule-A-independent reovirus mutant (jin-1) failed to induce necroptosis in HT29 cells. In contrast, these viruses induced MLKL phosphorylation in murine L929 cells, albeit with varying efficiencies. Our study shows that while reoviruses efficiently induce necroptosis in L929 cells, this is not a common phenotype in human cell lines. This study emphasizes the difficulties of translating the results of ICD studies from murine cells to human cells.


Subject(s)
Mammalian orthoreovirus 3 , Humans , Animals , Mice , Mammalian orthoreovirus 3/metabolism , Necroptosis/genetics , Protein Kinases/genetics , Protein Kinases/metabolism , Fibroblasts/metabolism , Cell Line, Tumor , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Apoptosis/genetics
2.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 38(2): 171-176, 2022 Feb.
Article in Chinese | MEDLINE | ID: mdl-35356886

ABSTRACT

Objective To prepare T3Dsigma1 polyclonal antibody with purified sigma1 fusion protein of mammalian orthoreovirus (MRV) serotype 3 Dearing (T3D). Methods The recombinant plasmid T3DS1-pET28a was transformed into Rosetta (DE3) competent cells, and the isopropyl-ß-D-thiogalactopyranoside (IPTG) was used to induce a large amount of target proteins which were subjected to purification by histidine-tagged Ni-IDA chromatography column to obtain the T3Dsigma1 fusion protein. New Zealand white rabbits were immunized with the purified protein to prepare polyclonal antibodies specific against sigma1 protein. The titer of polyclonal antibody was detected by indirect ELISA, and the specificity by Western blot analysis and indirect immunofluorescence assay (IFA). Results The relative molecular mass (Mr) of T3Dsigma1 fusion protein, mainly in the form of inclusion body, was about 32 000. The fusion protein was purified by denaturation and renaturation. The polyclonal antibody with the titer greater than 1:106 was prepared by immunizing New Zealand white rabbits and detected by Western blot analysis and IFA. Conclusion The T3Dsigma1 polyclonal antibody with high titer and sensitivity was prepared.


Subject(s)
Mammalian orthoreovirus 3 , Animals , Antibodies , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Mammalian orthoreovirus 3/metabolism , Mammals , Rabbits , Serogroup
3.
J Virol ; 96(2): e0187921, 2022 01 26.
Article in English | MEDLINE | ID: mdl-34757847

ABSTRACT

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.


Subject(s)
Capsid Proteins/metabolism , Mammalian orthoreovirus 3/pathogenicity , Myocarditis/virology , Reoviridae Infections/virology , Animals , Animals, Newborn , Capsid Proteins/genetics , Inflammation , Mammalian orthoreovirus 3/genetics , Mammalian orthoreovirus 3/metabolism , Mice , Mice, Inbred C57BL , Myocarditis/mortality , Myocarditis/pathology , Orthoreovirus, Mammalian/genetics , Orthoreovirus, Mammalian/metabolism , Orthoreovirus, Mammalian/pathogenicity , Reoviridae Infections/mortality , Reoviridae Infections/pathology , Viral Load , Virulence , Virus Replication
4.
Viruses ; 13(2)2021 02 12.
Article in English | MEDLINE | ID: mdl-33673179

ABSTRACT

Studies of conditionally lethal mutants can help delineate the structure-function relationships of biomolecules. Temperature-sensitive (ts) mammalian reovirus (MRV) mutants were isolated and characterized many years ago. Two of the most well-defined MRV ts mutants are tsC447, which contains mutations in the S2 gene encoding viral core protein σ2, and tsG453, which contains mutations in the S4 gene encoding major outer-capsid protein σ3. Because many MRV ts mutants, including both tsC447 and tsG453, encode multiple amino acid substitutions, the specific amino acid substitutions responsible for the ts phenotype are unknown. We used reverse genetics to recover recombinant reoviruses containing the single amino acid polymorphisms present in ts mutants tsC447 and tsG453 and assessed the recombinant viruses for temperature-sensitivity by efficiency-of-plating assays. Of the three amino acid substitutions in the tsG453 S4 gene, Asn16-Lys was solely responsible for the tsG453ts phenotype. Additionally, the mutant tsC447 Ala188-Val mutation did not induce a temperature-sensitive phenotype. This study is the first to employ reverse genetics to identify the dominant amino acid substitutions responsible for the tsC447 and tsG453 mutations and relate these substitutions to respective phenotypes. Further studies of other MRV ts mutants are warranted to define the sequence polymorphisms responsible for temperature sensitivity.


Subject(s)
Capsid Proteins/genetics , Mammalian orthoreovirus 3/metabolism , Point Mutation , Reoviridae Infections/virology , Amino Acid Substitution , Capsid/metabolism , Capsid Proteins/metabolism , Humans , Mammalian orthoreovirus 3/chemistry , Mammalian orthoreovirus 3/genetics , Phenotype , Temperature
5.
Sci Rep ; 11(1): 3508, 2021 02 10.
Article in English | MEDLINE | ID: mdl-33568687

ABSTRACT

Mouse reovirus type 3 (Reo-3) infection is a viral disease that is harmful for laboratory mice. No rapid and accurate detection methods are currently available for this infection. In this study, we describe a rapid, simple, closed-tube, one step, reverse transcription-loop-mediated isothermal amplification (RT-LAMP) assay for Reo-3 and compare our assay with indirect enzyme-linked immunosorbent assay (ELISA). Three sets of RT-LAMP primers were designed by sequence analysis of a specific conserved sequence of the Reo-3 S1 gene. Using RS2 primer set, the RT-LAMP assay required 60 min at 65 °C to amplify the S1 gene in one step by using Reo-3 RNA template and had no cross-reactivity with the other related pathogens, such as Sendai virus (SV), pneumonia virus of mice (PVM), mouse hepatitis virus (MHV), Ectromelia virus (Ect), minute virus of mice (MVM), P. pneumotropica, B. bronchiseptica, K. pneumonia and P. aeruginosa. in our LAMP reaction system. The limit of detection (LOD) of our RT-LAMP assay is 4 fg/µL. The established RT-LAMP assay enabled visual detection when fluorescence detection reagents were added, and was demonstrated to be effective and efficient. We tested 30 clinical blood samples and five artificial positive samples from SPF mice, the concordance between the two methods for blood samples was 100% compared with indirect ELISA and RT-PCR. Considering its performance, specificity, sensitivity, and repeatability, the developed RT-LAMP could be a valuable tool to supply a more effective Reo-3 detection method in laboratory animal quality monitoring.


Subject(s)
Mammalian orthoreovirus 3/genetics , Mammalian orthoreovirus 3/metabolism , Molecular Diagnostic Techniques , Nucleic Acid Amplification Techniques , RNA, Viral/genetics , Reverse Transcription/physiology , Animals , Mice , Molecular Diagnostic Techniques/methods , Nucleic Acid Amplification Techniques/methods , Sensitivity and Specificity
6.
J Virol ; 94(23)2020 11 09.
Article in English | MEDLINE | ID: mdl-32907973

ABSTRACT

Mammalian reovirus (MRV) strain type 3 Dearing (T3D) is a naturally occurring oncolytic virus that has been developed as a potential cancer therapeutic. However, MRV treatment cannot be applied to cancer cells expressing low levels of junctional adhesion molecule A (JAM-A), which is the entry receptor of MRV. In this study, we developed a reverse genetics system for MRV strain T3D-L, which showed high oncolytic potency. To modify the cell tropism of MRV, an arginine-glycine-aspartic acid (RGD) peptide with an affinity to integrin was inserted at the C terminus or loop structures of the viral cell attachment protein σ1. The recombinant RGD σ1-modified viruses induced remarkable cell lysis in human cancer cell lines with marginal JAM-A expression and in JAM-A knockout cancer cell lines generated by a CRISPR/Cas9 system. Pretreatment of cells with anti-integrin antibody decreased cell death caused by the RGD σ1-modified virus, suggesting the infection to the cells was via a specific interaction with integrin αV. By using mouse models, we assessed virulence of the RGD σ1-modified viruses in vivo This system will open new avenues for the use of genetically modified oncolytic MRV for use as a cancer therapy.IMPORTANCE Oncolytic viruses kill tumors without affecting normal cells. A variety of oncolytic viruses are used as cancer therapeutics. Mammalian reovirus (MRV), which belongs to the genus Orthoreovirus, family Reoviridae, is one such natural oncolytic virus. The anticancer effects of MRV are being evaluated in clinical trials. Unlike other oncolytic viruses, MRV has not been genetically modified for use as a cancer therapeutic in clinical trials. Here, we used a reverse genetic approach to introduce an integrin-affinity peptide sequence into the MRV cell attachment protein σ1 to alter the natural tropism of the virus. The recombinant viruses were able to infect cancer cell lines expressing very low levels of the MRV entry receptor, junctional adhesion molecule A (JAM-A), and cause tumor cell death while maintaining its original tropism via JAM-A. This is a novel report of a genetically modified oncolytic MRV by introducing a peptide sequence into σ1.


Subject(s)
Junctional Adhesion Molecule A/genetics , Junctional Adhesion Molecule A/metabolism , Oligopeptides/metabolism , Reoviridae/genetics , Reoviridae/metabolism , Amino Acid Sequence , Animals , CRISPR-Cas Systems , Cell Adhesion Molecules , Cell Line, Tumor , Gene Knockout Techniques , Humans , Mammalian orthoreovirus 3/genetics , Mammalian orthoreovirus 3/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Nude , Oncolytic Virotherapy , Oncolytic Viruses/genetics , Orthoreovirus/genetics , Orthoreovirus/metabolism , Receptors, Cell Surface , Virus Replication
7.
J Virol ; 94(4)2020 01 31.
Article in English | MEDLINE | ID: mdl-31776267

ABSTRACT

Reovirus serotype 3 Dearing (T3D) replicates preferentially in transformed cells and is in clinical trials as a cancer therapy. Laboratory strains of T3D, however, exhibit differences in plaque size on cancer cells and differences in oncolytic activity in vivo This study aimed to determine why the most oncolytic T3D reovirus lab strain, the Patrick Lee laboratory strain (T3DPL), replicates more efficiently in cancer cells than other commonly used laboratory strains, the Kevin Coombs laboratory strain (T3DKC) and Terence Dermody laboratory (T3DTD) strain. In single-step growth curves, T3DPL titers increased at higher rates and produced ∼9-fold higher burst size. Furthermore, the number of reovirus antigen-positive cells increased more rapidly for T3DPL than for T3DTD In conclusion, the most oncolytic T3DPL possesses replication advantages in a single round of infection. Two specific mechanisms for enhanced infection by T3DPL were identified. First, T3DPL exhibited higher cell attachment, which was attributed to a higher proportion of virus particles with insufficient (≤3) σ1 cell attachment proteins. Second, T3DPL transcribed RNA at rates superior to those of the less oncolytic T3D strains, which is attributed to polymorphisms in M1-encoding µ2 protein, as confirmed in an in vitro transcription assay, and which thus demonstrates that T3DPL has an inherent transcription advantage that is cell type independent. Accordingly, T3DPL established rapid onset of viral RNA and protein synthesis, leading to more rapid kinetics of progeny virus production, larger virus burst size, and higher levels of cell death. Together, these results emphasize the importance of paying close attention to genomic divergence between virus laboratory strains and, mechanistically, reveal the importance of the rapid onset of infection for reovirus oncolysis.IMPORTANCE Reovirus serotype 3 Dearing (T3D) is in clinical trials for cancer therapy. Recently, it was discovered that highly related laboratory strains of T3D exhibit large differences in their abilities to replicate in cancer cells in vitro, which correlates with oncolytic activity in a murine model of melanoma. The current study reveals two mechanisms for the enhanced efficiency of T3DPL in cancer cells. Due to polymorphisms in two viral genes, within the first round of reovirus infection, T3DPL binds to cells more efficiency and more rapidly produces viral RNAs; this increased rate of infection relative to that of the less oncolytic strains gives T3DPL a strong inherent advantage that culminates in higher virus production, more cell death, and higher virus spread.


Subject(s)
Mammalian orthoreovirus 3/genetics , Oncolytic Viruses/genetics , Animals , Capsid Proteins/genetics , Cell Adhesion/genetics , Cell Line , Genes, Viral/genetics , Humans , Kinetics , Mammalian orthoreovirus 3/metabolism , Mice , Oncolytic Virotherapy/methods , Polymorphism, Genetic/genetics , Reoviridae/genetics , Reoviridae Infections/genetics , Transcription, Genetic/genetics , Viral Proteins/metabolism , Virion/metabolism , Virus Replication/genetics
8.
J Virol ; 94(4)2020 01 31.
Article in English | MEDLINE | ID: mdl-31748391

ABSTRACT

Little is known about how genetic variations in viruses affect their success as therapeutic agents. The type 3 Dearing strain of Mammalian orthoreovirus (T3D) is undergoing clinical trials as an oncolytic virotherapy. Worldwide, studies on reovirus oncolysis use T3D stocks propagated in different laboratories. Here, we report that genetic diversification among T3D stocks from various sources extensively impacts oncolytic activity. The T3D strain from the Patrick Lee laboratory strain (TD3PL) showed significantly stronger oncolytic activities in a murine model of melanoma than the strain from the Terence Dermody laboratory (T3DTD). Overall in vitro replication and cytolytic properties of T3D laboratory strains were assessed by measuring virus plaque size on a panel of human and mouse tumor cells, and results were found to correlate with in vivo oncolytic potency in a melanoma model. T3DPL produced larger plaques than T3DTD and than the T3D strain from the ATCC (T3DATCC) and from the Kevin Coombs laboratory (T3DKC). Reassortant and reverse genetics analyses were used to decipher key genes and polymorphisms that govern enhanced plaque size of T3DPL Five single amino acid changes in the S4, M1, and L3 genome segments of reovirus were each partially correlated with plaque size and when combined were able to fully account for differences between T3DPL and T3DTD Moreover, polymorphisms were discovered in T3DTD that promoted virus replication and spread in tumors, and a new T3DPL/T3DTD hybrid was generated with enhanced plaque size compared to that of T3DPL Altogether, single amino acid changes acquired during laboratory virus propagation can have a large impact on reovirus therapeutic potency and warrant consideration as possible confounding variables between studies.IMPORTANCE The reovirus serotype 3 Dearing (T3D) strain is in clinical trials for cancer therapy. We find that closely related laboratory strains of T3D exhibit large differences in their abilities to replicate in cancer cells in vitro, which correlates with oncolytic activity in a in a murine model of melanoma. The study reveals that five single amino acid changes among three reovirus genes strongly impact reovirus therapeutic potency. In general, the findings suggest that attention should be given to genomic divergence of virus strains during research and optimization for cancer therapy.


Subject(s)
Mammalian orthoreovirus 3/genetics , Oncolytic Virotherapy/methods , Virus Replication/genetics , Amino Acids/genetics , Animals , Cell Line , Cell Line, Tumor , Female , Genetic Variation/genetics , Humans , Mammalian orthoreovirus 3/metabolism , Mice , Mice, Inbred C57BL , Orthoreovirus, Mammalian/genetics , Orthoreovirus, Mammalian/metabolism , Phylogeny , Reoviridae/genetics , Viral Proteins/metabolism
9.
J Virol ; 93(9)2019 05 01.
Article in English | MEDLINE | ID: mdl-30787157

ABSTRACT

The environment represents a significant barrier to infection. Physical stressors (heat) or chemical agents (ethanol) can render virions noninfectious. As such, discrete proteins are necessary to stabilize the dual-layered structure of mammalian orthoreovirus (reovirus). The outer capsid participates in cell entry: (i) σ3 is degraded to generate the infectious subviral particle, and (ii) µ1 facilitates membrane penetration and subsequent core delivery. µ1-σ3 interactions also prevent inactivation; however, this activity is not fully characterized. Using forward and reverse genetic approaches, we identified two mutations (µ1 M258I and σ3 S344P) within heat-resistant strains. σ3 S344P was sufficient to enhance capsid integrity and to reduce protease sensitivity. Moreover, these changes impaired replicative fitness in a reassortant background. This work reveals new details regarding the determinants of reovirus stability.IMPORTANCE Nonenveloped viruses rely on protein-protein interactions to shield their genomes from the environment. The capsid, or protective shell, must also disassemble during cell entry. In this work, we identified a determinant within mammalian orthoreovirus that regulates heat resistance, disassembly kinetics, and replicative fitness. Together, these findings show capsid function is balanced for optimal replication and for spread to a new host.


Subject(s)
Capsid Proteins , Capsid/metabolism , Hot Temperature , Mammalian orthoreovirus 3 , Mutation , Animals , Capsid Proteins/genetics , Capsid Proteins/metabolism , Cell Line , Mammalian orthoreovirus 3/genetics , Mammalian orthoreovirus 3/metabolism , Mice , Protein Stability
10.
Gene Ther ; 25(5): 331-344, 2018 08.
Article in English | MEDLINE | ID: mdl-30013187

ABSTRACT

While the mammalian orthoreovirus type 3 dearing (reovirus T3D) infects many different tumour cells, various cell lines resist the induction of reovirus-mediated cell death. In an effort to increase the oncolytic potency, we introduced transgenes into the S1 segment of reovirus T3D. The adenovirus E4orf4 gene was selected as transgene since the encoded E4orf4 protein induces cell death in transformed cells. The induction of cell death by E4orf4 depends in part on its binding to phosphatase 2A (PP2A). In addition to the S1-E4orf4 reovirus, two other reoviruses were employed in our studies. The reovirus rS1-RFA encodes an E4orf4 double-mutant protein that cannot interact with PP2A and the rS1-iLOV virus encoding the fluorescent marker iLOV as a reporter. The replacement of the codons for the junction adhesion molecule-A (JAM-A) binding head domain of the truncated spike protein blocks the entry of these recombinant viruses via the reovirus receptor JAM-A. Instead these viruses rely on internalization via binding to sialic acids on the cell surface. This expands their tropism and allows infection of JAM-A-deficient tumour cells. Here we not only demonstrate the feasibility of this approach but also established that the cytolytic activity of these recombinant viruses is largely transgene independent.


Subject(s)
Mammalian orthoreovirus 3/physiology , Viral Proteins/physiology , Viral Tropism/genetics , Cell Line , Humans , Mammalian orthoreovirus 3/genetics , Mammalian orthoreovirus 3/metabolism , Reoviridae Infections/virology , Viral Proteins/genetics , Viral Proteins/metabolism
11.
Cancer Gene Ther ; 25(5-6): 148-160, 2018 06.
Article in English | MEDLINE | ID: mdl-29720674

ABSTRACT

There are currently numerous oncolytic viruses undergoing clinical trial evaluation in cancer patients and one agent, Talimogene laherparepvec, has been approved for the treatment of malignant melanoma. This progress highlights the huge clinical potential of this treatment modality, and the focus is now combining these agents with conventional anticancer treatments or agents that enhance viral replication, and thereby oncolysis, in the tumour microenvironment. We evaluated the combination of reovirus with rapamycin in B16F10 cell, a murine model of malignant melanoma, based on potential mechanisms by which mTOR inhibitors might enhance viral oncolysis. Rapamycin was not immunomodulatory in that it had no effect on the generation of an antireovirus-neutralising antibody response in C57/black 6 mice. The cell cycle effects of reovirus (increase G0/G1 fraction) were unaffected by concomitant or sequential exposure of rapamycin. However, rapamycin attenuated viral replication if given prior or concomitantly with reovirus and similarly reduced reovirus-induced apoptotic cell death Annexin V/PI and caspase 3/7 activation studies. We found clear evidence of synergistic antitumour effects of the combination both in vitro and in vivo, which was sequence dependent only in the in vitro setting. In conclusion, we have demonstrated synergistic antitumour efficacy of reovirus and rapamycin combination.


Subject(s)
Mammalian orthoreovirus 3/metabolism , Melanoma/therapy , Oncolytic Virotherapy/methods , Oncolytic Viruses/metabolism , Sirolimus/pharmacology , Animals , Cell Line, Tumor , Melanoma/microbiology , Mice
12.
J Virol ; 91(19)2017 10 01.
Article in English | MEDLINE | ID: mdl-28724773

ABSTRACT

The recent approval of oncolytic virus for therapy of melanoma patients has increased the need for precise evaluation of the mechanisms by which oncolytic viruses affect tumor growth. Here we show that the human NK cell-activating receptor NKp46 and the orthologous mouse protein NCR1 recognize the reovirus sigma1 protein in a sialic-acid-dependent manner. We identify sites of NKp46/NCR1 binding to sigma1 and show that sigma1 binding by NKp46/NCR1 leads to NK cell activation in vitro Finally, we demonstrate that NCR1 activation is essential for reovirus-based therapy in vivo Collectively, we have identified sigma1 as a novel ligand for NKp46/NCR1 and demonstrated that NKp46/NCR1 is needed both for clearance of reovirus infection and for reovirus-based tumor therapy.IMPORTANCE Reovirus infects much of the population during childhood, causing mild disease, and hence is considered to be efficiently controlled by the immune system. Reovirus also specifically infects tumor cells, leading to tumor death, and is currently being tested in human clinical trials for cancer therapy. The mechanisms by which our immune system controls reovirus infection and tumor killing are not well understood. We report here that natural killer (NK) cells recognize a viral protein named sigma1 through the NK cell-activating receptor NKp46. Using several mouse tumor models, we demonstrate the importance of NK cells in protection from reovirus infection and in reovirus killing of tumors in vivo Collectively, we identify a new ligand for the NKp46 receptor and provide evidence for the importance of NKp46 in the control of reovirus infections and in reovirus-based cancer therapy.


Subject(s)
Antigens, Ly/metabolism , Killer Cells, Natural/immunology , Mammalian orthoreovirus 3/metabolism , Natural Cytotoxicity Triggering Receptor 1/metabolism , Oncolytic Virotherapy/methods , Oncolytic Viruses/metabolism , Viral Proteins/metabolism , Animals , Binding Sites , Chlorocebus aethiops , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Humans , Lymphocyte Activation/immunology , Melanoma/therapy , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , N-Acetylneuraminic Acid/metabolism , Vero Cells , Viral Proteins/genetics
13.
Virology ; 448: 133-45, 2014 Jan 05.
Article in English | MEDLINE | ID: mdl-24314644

ABSTRACT

At early times in Mammalian Orthoreovirus (MRV) infection, cytoplasmic inclusions termed stress granules (SGs) are formed as a component of the innate immune response, however, at later times they are no longer present despite continued immune signaling. To investigate the roles of MRV proteins in SG modulation we examined non-structural protein µNS localization relative to SGs in infected and transfected cells. Using a series of mutant plasmids, we mapped the necessary µNS residues for SG localization to amino acids 78 and 79. We examined the capacity of a µNS(78-79) mutant to associate with known viral protein binding partners of µNS and found that it loses association with viral core protein λ2. Finally, we show that while this mutant cannot support de novo viral replication, it is able to rescue replication following siRNA knockdown of µNS. These data suggest that µNS association with SGs, λ2, or both play roles in MRV replication.


Subject(s)
Cytoplasmic Granules/virology , Mammalian orthoreovirus 3/metabolism , Reoviridae Infections/virology , Viral Core Proteins/metabolism , Viral Nonstructural Proteins/administration & dosage , Viral Nonstructural Proteins/chemistry , Virus Replication , Amino Acid Motifs , Animals , Cell Line , Humans , Mammalian orthoreovirus 3/chemistry , Mammalian orthoreovirus 3/genetics , Protein Binding , Viral Core Proteins/genetics , Viral Nonstructural Proteins/genetics
14.
J Virol ; 87(23): 12967-79, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24067959

ABSTRACT

Reovirus nonstructural protein σ1s is implicated in cell cycle arrest at the G2/M boundary and induction of apoptosis. However, the contribution of σ1s to these effects in an otherwise isogenic viral background has not been defined. To evaluate the role of σ1s in cell cycle arrest and apoptosis, we used reverse genetics to generate a σ1s-null reovirus. Following infection with wild-type virus, we observed an increase in the percentage of cells in G2/M, whereas the proportion of cells in G2/M following infection with the σ1s-null mutant was unaffected. Similarly, we found that the wild-type virus induced substantially greater levels of apoptosis than the σ1s-null mutant. These data indicate that σ1s is required for both reovirus-induced cell cycle arrest and apoptosis. To define sequences in σ1s that mediate these effects, we engineered viruses encoding C-terminal σ1s truncations by introducing stop codons in the σ1s open reading frame. We also generated viruses in which charged residues near the σ1s amino terminus were replaced individually or as a cluster with nonpolar residues. Analysis of these mutants revealed that amino acids 1 to 59 and the amino-terminal basic cluster are required for induction of both cell cycle arrest and apoptosis. Remarkably, viruses that fail to induce cell cycle arrest and apoptosis also are attenuated in vivo. Thus, identical sequences in σ1s are required for reovirus-induced cell cycle arrest, apoptosis, and pathogenesis. Collectively, these findings provide evidence that the σ1s-mediated properties are genetically linked and suggest that these effects are mechanistically related.


Subject(s)
Apoptosis , Cell Cycle Checkpoints , Mammalian orthoreovirus 3/metabolism , Reoviridae Infections/physiopathology , Reoviridae Infections/virology , Viral Nonstructural Proteins/metabolism , Amino Acid Motifs , Animals , Cell Line , Humans , Mammalian orthoreovirus 3/chemistry , Mammalian orthoreovirus 3/genetics , Mice , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/genetics , Virus Replication
15.
PLoS One ; 7(10): e48064, 2012.
Article in English | MEDLINE | ID: mdl-23110175

ABSTRACT

Mammalian Reovirus is a double-stranded RNA virus with a distinctive preference to replicate in and lyse transformed cells. On that account, Reovirus type 3 Dearing (T3D) is clinically evaluated as oncolytic agent. The therapeutic efficacy of this approach depends in part on the accessibility of the reovirus receptor Junction Adhesion Molecule-A (JAM-A) on the target cells. Here, we describe the isolation and characterization of reovirus T3D mutants that can infect human tumor cells independent of JAM-A. The JAM-A-independent (jin) mutants were isolated on human U118MG glioblastoma cells, which do not express JAM-A. All jin mutants harbour mutations in the S1 segments close to the region that encodes the sialic acid-binding pocket in the shaft of the spike protein. In addition, two of the jin mutants encode spike proteins with a Q336R substitution in their head domain. The jin mutants can productively infect a wide range of cell lines that resist wt reovirus T3D infection, including chicken LMH cells, hamster CHO cells, murine endothelioma cells, human U2OS and STA-ET2.1 cells, but not primary human fibroblasts. The jin-mutants rely on the presence of sialic-acid residues on the cell surface for productive infection, as is evident from wheat germ agglutinin (WGA) inhibition experiments, and from the jin-reovirus resistance of CHO-Lec2 cells, which have a deficiency of sialic-acids on their glycoproteins. The jin mutants may be useful as oncolytic agents for use in tumors in which JAM-A is absent or inaccessible.


Subject(s)
Cell Adhesion Molecules/genetics , Mammalian orthoreovirus 3/genetics , Mutation , Receptors, Cell Surface/genetics , Animals , CHO Cells , Cell Adhesion Molecules/metabolism , Cell Line , Cell Line, Tumor , Cricetinae , Cricetulus , Cysteine Proteinase Inhibitors/pharmacology , Glioblastoma/genetics , Glioblastoma/pathology , Glioblastoma/virology , Host Specificity/genetics , Host-Pathogen Interactions/genetics , Humans , Leucine/analogs & derivatives , Leucine/pharmacology , Mammalian orthoreovirus 3/metabolism , Mammalian orthoreovirus 3/physiology , Molecular Sequence Data , Neoplasms/genetics , Neoplasms/pathology , Neoplasms/virology , Oncolytic Virotherapy/methods , Protein Multimerization , Receptors, Cell Surface/metabolism , Receptors, Virus/genetics , Receptors, Virus/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA , Viral Proteins/chemistry , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Internalization/drug effects
16.
J Virol ; 86(24): 13164-73, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23035227

ABSTRACT

Mammalian reoviruses display serotype-specific patterns of tropism and disease in the murine central nervous system (CNS) attributable to polymorphisms in viral attachment protein σ1. While all reovirus serotypes use junctional adhesion molecule-A as a cellular receptor, they differ in their utilization of carbohydrate coreceptors. This observation raises the possibility that carbohydrate binding by σ1 influences reovirus pathology in the CNS. In this study, we sought to define the function of carbohydrate binding in reovirus neuropathogenesis. Newborn mice were inoculated intramuscularly with wild-type strain type 3 Dearing (T3D) and T3D-σ1R202W, a point mutant T3D derivative that does not bind sialic acid (SA). Infected mice were monitored for survival, and viral loads at the sites of primary and secondary replication were quantified. Fewer mice inoculated with the wild-type virus survived in comparison to those inoculated with the mutant virus. The wild-type virus also produced higher titers in the spinal cord and brain at late times postinoculation but lower titers in the liver in comparison to those produced by the mutant virus. In addition, the wild-type virus was more virulent and produced higher titers in the brain than the mutant following intracranial inoculation. These animal infectivity studies suggest that T3D-σ1R202W harbors a defect in neural growth. Concordantly, compared with the wild-type virus, the mutant virus displayed a decreased capacity to infect and replicate in primary cultures of cortical neurons, a property dependent on cell surface SA. These results suggest that SA binding enhances the kinetics of reovirus replication in neural tissues and highlight a functional role for sialylated glycans as reovirus coreceptors in the CNS.


Subject(s)
Central Nervous System/virology , Mammalian orthoreovirus 3/pathogenicity , N-Acetylneuraminic Acid/metabolism , Polysaccharides/metabolism , Receptors, Virus/metabolism , Animals , Base Sequence , Cell Line , DNA Primers , Immunohistochemistry , Mammalian orthoreovirus 3/isolation & purification , Mammalian orthoreovirus 3/metabolism , Mice , Mice, Inbred C57BL , Polymerase Chain Reaction , Viral Load , Virulence , Virus Replication
17.
J Virol ; 86(4): 2302-11, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22156521

ABSTRACT

Myocarditis is indicated as the second leading cause of sudden death in young adults. Reovirus induces myocarditis in neonatal mice, providing a tractable model system for investigation of this important disease. Alpha/beta-interferon (IFN-α/ß) treatment improves cardiac function and inhibits viral replication in patients with chronic myocarditis, and the host IFN-α/ß response is a determinant of reovirus strain-specific differences in induction of myocarditis. Virus-induced IFN-ß stimulates a signaling cascade that establishes an antiviral state and further induces IFN-α/ß through an amplification loop. Reovirus strain-specific differences in induction of and sensitivity to IFN-α/ß are associated with the viral M1, L2, and S2 genes. The reovirus M1 gene-encoded µ2 protein is a strain-specific repressor of IFN-ß signaling, providing one possible mechanism for the variation in resistance to IFN and induction of myocarditis between different reovirus strains. We report here that µ2 amino acid 208 determines repression of IFN-ß signaling and modulates reovirus induction of IFN-ß in cardiac myocytes. Moreover, µ2 amino acid 208 determines reovirus replication, both in initially infected cardiac myocytes and after viral spread, by regulating the IFN-ß response. Amino acid 208 of µ2 also influences the cytopathic effect in cardiac myocytes after spread. Finally, µ2 amino acid 208 modulates myocarditis in neonatal mice. Thus, repression of IFN-ß signaling mediated by reovirus µ2 amino acid 208 is a determinant of the IFN-ß response, viral replication and damage in cardiac myocytes, and myocarditis. These results demonstrate that a single amino acid difference between viruses can dictate virus strain-specific differences in suppression of the host IFN-ß response and, consequently, damage to the heart.


Subject(s)
Down-Regulation , Interferon-alpha/metabolism , Interferon-beta/metabolism , Myocarditis/metabolism , Orthoreovirus, Mammalian/genetics , Polymorphism, Single Nucleotide , Reoviridae Infections/metabolism , Signal Transduction , Viral Proteins/genetics , Animals , Cell Line , Cells, Cultured , Humans , Interferon-alpha/genetics , Interferon-beta/genetics , Mammalian orthoreovirus 3/genetics , Mammalian orthoreovirus 3/metabolism , Mice , Myocarditis/genetics , Myocarditis/virology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/virology , Orthoreovirus, Mammalian/metabolism , Reoviridae Infections/genetics , Reoviridae Infections/virology
18.
J Virol ; 83(5): 2178-87, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19109390

ABSTRACT

The secreted cytokine alpha/beta interferon (IFN-alpha/beta) binds its receptor to activate the Jak-STAT signal transduction pathway, leading to formation of the heterotrimeric IFN-stimulated gene factor 3 (ISGF3) transcription complex for induction of IFN-stimulated genes (ISGs) and establishment of an antiviral state. Many viruses have evolved countermeasures to inhibit the IFN pathway, thereby subverting the innate antiviral response. Here, we demonstrate that the mildly myocarditic reovirus type 1 Lang (T1L), but not the nonmyocarditic reovirus type 3 Dearing, represses IFN induction of a subset of ISGs and that this repressor function segregates with the T1L M1 gene. Concordantly, the T1L M1 gene product, mu2, dramatically inhibits IFN-beta-induced reporter gene expression. Surprisingly, T1L infection does not degrade components of the ISGF3 complex or interfere with STAT1 or STAT2 nuclear translocation as has been observed for other viruses. Instead, infection with T1L or reassortant or recombinant viruses containing the T1L M1 gene results in accumulation of interferon regulatory factor 9 (IRF9) in the nucleus. This effect has not been previously described for any virus and suggests that mu2 modulates IRF9 interactions with STATs for both ISGF3 function and nuclear export. The M1 gene is a determinant of virus strain-specific differences in the IFN response, which are linked to virus strain-specific differences in induction of murine myocarditis. We find that virus-induced myocarditis is associated with repression of IFN function, providing new insights into the pathophysiology of this disease. Together, these data provide the first report of an increase in IRF9 nuclear accumulation associated with viral subversion of the IFN response and couple virus strain-specific differences in IFN antagonism to the pathogenesis of viral myocarditis.


Subject(s)
Interferon-Stimulated Gene Factor 3, gamma Subunit/metabolism , Interferon-beta/pharmacology , Mammalian orthoreovirus 3/pathogenicity , Orthoreovirus, Mammalian/pathogenicity , Viral Proteins/metabolism , Animals , Cell Nucleus/metabolism , Cells, Cultured , Gene Expression Regulation , Humans , Interferon-Stimulated Gene Factor 3/metabolism , Interferon-alpha/pharmacology , Mammalian orthoreovirus 3/genetics , Mammalian orthoreovirus 3/metabolism , Mice , Myocarditis/virology , Orthoreovirus, Mammalian/genetics , Orthoreovirus, Mammalian/metabolism , Reassortant Viruses/genetics , Reassortant Viruses/metabolism , Reassortant Viruses/pathogenicity , STAT1 Transcription Factor/metabolism , STAT2 Transcription Factor/metabolism , Transcriptional Activation , Viral Proteins/genetics
19.
J Virol ; 81(13): 6984-92, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17475657

ABSTRACT

Viral encephalitis is a major cause of morbidity and mortality worldwide, yet there is no proven efficacious therapy for most viral infections of the central nervous system (CNS). Many of the viruses that cause encephalitis induce apoptosis and activate c-Jun N-terminal kinase (JNK) following infection. We have previously shown that reovirus infection of epithelial cell lines activates JNK-dependent apoptosis. We now show that reovirus infection resulted in activation of JNK and caspase-3 in the CNS. Treatment of reovirus-infected mice with a cell-permeating peptide that competitively inhibits JNK activity resulted in significantly prolonged survival of intracerebrally infected mice following an otherwise lethal challenge with T3D (100 x 50% lethal dose). Protection correlated with reduced CNS injury, reduced neuronal apoptosis, and reduced c-Jun activation without altering the viral titer or viral antigen distribution. Given the efficacy of the inhibitor in protecting mice from viral encephalitis, JNK inhibition represents a promising and novel treatment strategy for viral encephalitis.


Subject(s)
Central Nervous System/enzymology , Encephalitis, Viral/prevention & control , Enzyme Inhibitors/pharmacology , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mammalian orthoreovirus 3/metabolism , Peptides/pharmacology , Reoviridae Infections/prevention & control , Animals , Apoptosis/drug effects , Caspase 3/metabolism , Cell Line , Central Nervous System/pathology , Central Nervous System/virology , Encephalitis, Viral/enzymology , Encephalitis, Viral/mortality , Encephalitis, Viral/pathology , Enzyme Inhibitors/therapeutic use , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Mice , Neurons/enzymology , Neurons/pathology , Neurons/virology , Peptides/therapeutic use , Reoviridae Infections/enzymology , Reoviridae Infections/mortality , Reoviridae Infections/pathology
20.
J Virol ; 81(3): 1360-71, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17121808

ABSTRACT

Reoviruses induce apoptosis both in cultured cells and in vivo. Apoptosis plays a major role in the pathogenesis of reovirus encephalitis and myocarditis in infected mice. Reovirus-induced apoptosis is dependent on the activation of transcription factor NF-kappaB and downstream cellular genes. To better understand the mechanism of NF-kappaB activation by reovirus, NF-kappaB signaling intermediates under reovirus control were investigated at the level of Rel, IkappaB, and IkappaB kinase (IKK) proteins. We found that reovirus infection leads initially to nuclear translocation of p50 and RelA, followed by delayed mobilization of c-Rel and p52. This biphasic pattern of Rel protein activation is associated with the degradation of the NF-kappaB inhibitor IkappaBalpha but not the structurally related inhibitors IkappaBbeta or IkappaBepsilon. Using IKK subunit-specific small interfering RNAs and cells deficient in individual IKK subunits, we demonstrate that IKKalpha but not IKKbeta is required for reovirus-induced NF-kappaB activation and apoptosis. Despite the preferential usage of IKKalpha, both NF-kappaB activation and apoptosis were attenuated in cells lacking IKKgamma/Nemo, an essential regulatory subunit of IKKbeta. Moreover, deletion of the gene encoding NF-kappaB-inducing kinase, which is known to modulate IKKalpha function, had no inhibitory effect on either response in reovirus-infected cells. Collectively, these findings indicate a novel pathway of NF-kappaB/Rel activation involving IKKalpha and IKKgamma/Nemo, which together mediate the expression of downstream proapoptotic genes in reovirus-infected cells.


Subject(s)
Apoptosis/physiology , I-kappa B Kinase/physiology , Mammalian orthoreovirus 3/physiology , NF-kappa B/metabolism , Gene Expression Regulation/physiology , HeLa Cells , Humans , I-kappa B Kinase/metabolism , Mammalian orthoreovirus 3/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...