48 results on '"Shmulevitz M"'
Search Results
2. Oncolytic reovirus induces intracellular redistribution of Ras to promote apoptosis and progeny virus release
- Author
-
Garant, K A, Shmulevitz, M, Pan, L, Daigle, R M, Ahn, D-G, Gujar, S A, and Lee, P W K
- Published
- 2016
- Full Text
- View/download PDF
3. Activated Ras signaling significantly enhances reovirus replication and spread
- Author
-
Shmulevitz, M, Marcato, P, and Lee, P WK
- Published
- 2010
- Full Text
- View/download PDF
4. The design, development and qualification of a lightweight antenna pointing mechanism
- Author
-
Shmulevitz, M and Halsband, A
- Subjects
Mechanical Engineering - Abstract
This paper describes the design, development, and qualification of a new lightweight and compact Antenna Pointing Mechanism (APM). The APM was specially designed to meet the stringent mass, envelope, and environmental requirements of OFFEQ experimental satellite. During the development phase, some problems were encountered with the brushless DC motors, slip ring contact resistance, and bearing drag torque. All of these problems were resolved, and two APM units have been operating successfully in orbit since April, 1995.
- Published
- 1996
5. Oncolytic reovirus induces intracellular redistribution of Ras to promote apoptosis and progeny virus release
- Author
-
Garant, K A, primary, Shmulevitz, M, additional, Pan, L, additional, Daigle, R M, additional, Ahn, D-G, additional, Gujar, S A, additional, and Lee, P W K, additional
- Published
- 2015
- Full Text
- View/download PDF
6. Reovirus Variants with Mutations in Genome Segments S1 and L2 Exhibit Enhanced Virion Infectivity and Superior Oncolysis
- Author
-
Shmulevitz, M., primary, Gujar, S. A., additional, Ahn, D.-G., additional, Mohamed, A., additional, and Lee, P. W. K., additional
- Published
- 2012
- Full Text
- View/download PDF
7. Stabilisation of p53 enhances reovirus-induced apoptosis and virus spread through p53-dependent NF-κB activation
- Author
-
Pan, D, primary, Pan, L-Z, additional, Hill, R, additional, Marcato, P, additional, Shmulevitz, M, additional, Vassilev, L T, additional, and Lee, P W K, additional
- Published
- 2011
- Full Text
- View/download PDF
8. Activated Ras signaling significantly enhances reovirus replication and spread
- Author
-
Shmulevitz, M, primary, Marcato, P, additional, and Lee, P W K, additional
- Published
- 2009
- Full Text
- View/download PDF
9. Membrane metabolism mediated by Sec14 family members influences Arf GTPase activating protein activity for transport from the trans-Golgi
- Author
-
Wong, T. A., primary, Fairn, G. D., additional, Poon, P. P., additional, Shmulevitz, M., additional, McMaster, C. R., additional, Singer, R. A., additional, and Johnston, G. C., additional
- Published
- 2005
- Full Text
- View/download PDF
10. Erratum: Improved oncolytic activity of a reovirus mutant that displays enhanced virus spread due to reduced cell attachment.
- Author
-
Cristi F, Walters M, Narayan N, Agopsowicz K, Hitt MM, and Shmulevitz M
- Abstract
[This corrects the article DOI: 10.1016/j.omto.2023.100743.]., (© 2024 The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
11. Rational design of an artificial tethered enzyme for non-templated post-transcriptional mRNA polyadenylation by the second generation of the C3P3 system.
- Author
-
Le Boulch M, Jacquet E, Nhiri N, Shmulevitz M, and Jaïs PH
- Subjects
- Animals, Humans, Mice, RNA, Messenger metabolism, Polynucleotide Adenylyltransferase genetics, Polynucleotide Adenylyltransferase metabolism, Poly A genetics, Poly A metabolism, Polyadenylation, DNA-Directed RNA Polymerases genetics
- Abstract
We have previously introduced the first generation of C3P3, an artificial system that allows the autonomous in-vivo production of mRNA with
m7 GpppN-cap. While C3P3-G1 synthesized much larger amounts of capped mRNA in human cells than conventional nuclear expression systems, it produced a proportionately much smaller amount of the corresponding proteins, indicating a clear defect of mRNA translatability. A possible mechanism for this poor translatability could be the rudimentary polyadenylation of the mRNA produced by the C3P3-G1 system. We therefore sought to develop the C3P3-G2 system using an artificial enzyme to post-transcriptionally lengthen the poly(A) tail. This system is based on the mutant mouse poly(A) polymerase alpha fused at its N terminus with an N peptide from the λ virus, which binds to BoxBr sequences placed in the 3'UTR region of the mRNA of interest. The resulting system selectively brings mPAPαm7 to the target mRNA to elongate its poly(A)-tail to a length of few hundred adenosine. Such elongation of the poly(A) tail leads to an increase in protein expression levels of about 2.5-3 times in cultured human cells compared to the C3P3-G1 system. Finally, the coding sequence of the tethered mutant poly(A) polymerase can be efficiently fused to that of the C3P3-G1 enzyme via an F2A sequence, thus constituting the single-ORF C3P3-G2 enzyme. These technical developments constitute an important milestone in improving the performance of the C3P3 system, paving the way for its applications in bioproduction and non-viral human gene therapy., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
12. Improved oncolytic activity of a reovirus mutant that displays enhanced virus spread due to reduced cell attachment.
- Author
-
Cristi F, Walters M, Narayan N, Agopsowicz K, Hitt MM, and Shmulevitz M
- Abstract
Wild-type reovirus serotype 3 Dearing (T3wt), a non-pathogenic intestinal virus, has shown promise as a cancer therapy in clinical trials, but it would benefit from an increased potency. Given that T3wt is naturally adapted to the intestinal environment (rather than tumors), we genetically modified reovirus to improve its infectivity in cancer cells. Various reovirus mutants were created, and their oncolytic potency was evaluated in vitro using plaque size as a measure of virus fitness in cancer cells. Notably, Super Virus 5 (SV5), carrying five oncolytic mutations, displayed the largest plaques in breast cancer cells among the mutants tested, indicating the potential for enhancing oncolytic potency through the combination of mutations. Furthermore, in a HER2+ murine breast cancer model, mice treated with SV5 exhibited superior tumor reduction and increased survival compared with those treated with PBS or T3wt. Intriguingly, SV5 did not replicate faster than T3wt in cultured cells but demonstrated a farther spread relative to T3wt, attributed to its reduced attachment to cancer cells. These findings highlight the significance of increased virus spread as a crucial mechanism for improving oncolytic virus activity. Thus, genetic modifications of reovirus hold the potential for augmenting its efficacy in cancer therapy., Competing Interests: The authors declare no competing interests., (© 2023 The Authors.)
- Published
- 2023
- Full Text
- View/download PDF
13. Reovirus genomic diversity confers plasticity for protease utility during adaptation to intracellular uncoating.
- Author
-
Lin QF, Wong CXL, Eaton HE, Pang X, and Shmulevitz M
- Subjects
- Animals, Mice, Genomics, L Cells, Serial Passage, Sewage virology, Genetic Fitness, Genome, Viral genetics, Peptide Hydrolases metabolism, Reoviridae classification, Reoviridae genetics, Reoviridae metabolism, Adaptation, Physiological, Virus Uncoating, Host Microbial Interactions
- Abstract
Importance: Reoviruses infect many mammals and are widely studied as a model system for enteric viruses. However, most of our reovirus knowledge comes from laboratory strains maintained on immortalized L929 cells. Herein, we asked whether naturally circulating reoviruses possess the same genetic and phenotypic characteristics as laboratory strains. Naturally circulating reoviruses obtained from sewage were extremely diverse genetically. Moreover, sewage reoviruses exhibited poor fitness on L929 cells and relied heavily on gut proteases for viral uncoating and productive infection compared to laboratory strains. We then examined how naturally circulating reoviruses might adapt to cell culture conditions. Within three passages, virus isolates from the parental sewage population were selected, displaying improved fitness and intracellular uncoating in L929 cells. Remarkably, selected progeny clones were present at 0.01% of the parental population. Altogether, using reovirus as a model, our study demonstrates how the high genetic diversity of naturally circulating viruses results in rapid adaptation to new environments., Competing Interests: The authors declare no conflict of interest.
- Published
- 2023
- Full Text
- View/download PDF
14. Special Issue of the 4th Symposium of the Canadian Society for Virology 2022.
- Author
-
Meier-Stephenson V, Evans D, Hobman T, Marchant D, Noyce R, and Shmulevitz M
- Subjects
- Canada, Virology, Viruses genetics
- Abstract
In this Special Issue of Viruses , we showcase some of the fascinating and diverse virology being undertaken in Canada that was presented at the 4th Symposium of the Canadian Society for Virology 2022 [...].
- Published
- 2023
- Full Text
- View/download PDF
15. p38 Mitogen-Activated Protein Kinase Signaling Enhances Reovirus Replication by Facilitating Efficient Virus Entry, Capsid Uncoating, and Postuncoating Steps.
- Author
-
Mohamed A, Lin QF, Eaton HE, and Shmulevitz M
- Subjects
- Female, Humans, Capsid metabolism, Capsid Proteins genetics, Capsid Proteins metabolism, Virus Replication, Cell Line, Tumor, Breast Neoplasms, Mammalian orthoreovirus 3 physiology, p38 Mitogen-Activated Protein Kinases genetics, p38 Mitogen-Activated Protein Kinases metabolism, Reoviridae Infections, Virus Internalization
- Abstract
Mammalian orthoreovirus serotype 3 Dearing is an oncolytic virus currently undergoing multiple clinical trials as a potential cancer therapy. Previous clinical trials have emphasized the importance of prescreening patients for prognostic markers to improve therapeutic success. However, only generic cancer markers such as epidermal growth factor receptor (EGFR), Hras, Kras, Nras, Braf, and p53 are currently utilized, with limited benefit in predicting therapeutic efficacy. This study aimed to investigate the role of p38 mitogen-activated protein kinase (MAPK) signaling during reovirus infection. Using a panel of specific p38 MAPK inhibitors and an inactive inhibitor analogue, p38 MAPK signaling was found to be essential for establishment of reovirus infection by enhancing reovirus endocytosis, facilitating efficient reovirus uncoating at the endo-lysosomal stage, and augmenting postuncoating replication steps. Using a broad panel of human breast cancer cell lines, susceptibility to reovirus infection corresponded with virus binding and uncoating efficiency, which strongly correlated with status of the p38β isoform. Together, results suggest p38β isoform as a potential prognostic marker for early stages of reovirus infection that are crucial to successful reovirus infection. IMPORTANCE The use of Pelareorep (mammalian orthoreovirus) as a therapy for metastatic breast cancer has shown promising results in recent clinical trials. However, the selection of prognostic markers to stratify patients has had limited success due to the fact that these markers are upstream receptors and signaling pathways that are present in a high percentage of cancers. This study demonstrates that the mechanism of action of p38 MAPK signaling plays a key role in establishment of reovirus infection at both early entry and late replication steps. Using a panel of breast cancer cell lines, we found that the expression levels of the MAPK11 (p38β) isoform are a strong determinant of reovirus uncoating and infection establishment. Our findings suggest that selecting prognostic markers that target key steps in reovirus replication may improve patient stratification during oncolytic reovirus therapy.
- Published
- 2023
- Full Text
- View/download PDF
16. Reovirus uses temporospatial compartmentalization to orchestrate core versus outercapsid assembly.
- Author
-
Kniert J, Dos Santos T, Eaton HE, Jung Cho W, Plummer G, and Shmulevitz M
- Subjects
- Animals, Capsid Proteins metabolism, Cell Line, Cycloheximide, Kinetics, Mammals, RNA, Viral genetics, Viral Proteins, Virus Assembly, Reoviridae genetics, Reoviridae metabolism
- Abstract
Reoviridae virus family members, such as mammalian orthoreovirus (reovirus), encounter a unique challenge during replication. To hide the dsRNA from host recognition, the genome remains encapsidated in transcriptionally active proteinaceous core capsids that transcribe and release +RNA. De novo +RNAs and core proteins must repeatedly assemble into new progeny cores in order to logarithmically amplify replication. Reoviruses also produce outercapsid (OC) proteins μ1, σ3 and σ1 that assemble onto cores to create highly stable infectious full virions. Current models of reovirus replication position amplification of transcriptionally-active cores and assembly of infectious virions in shared factories, but we hypothesized that since assembly of OC proteins would halt core amplification, OC assembly is somehow regulated. Kinetic analysis of virus +RNA production, core versus OC protein expression, and core particles versus whole virus particle accumulation, indicated that assembly of OC proteins onto core particles was temporally delayed. All viral RNAs and proteins were made simultaneously, eliminating the possibility that delayed OC RNAs or proteins account for delayed OC assembly. High resolution fluorescence and electron microscopy revealed that core amplification occurred early during infection at peripheral core-only factories, while all OC proteins associated with lipid droplets (LDs) that coalesced near the nucleus in a μ1-dependent manner. Core-only factories transitioned towards the nucleus despite cycloheximide-mediated halting of new protein expression, while new core-only factories developed in the periphery. As infection progressed, OC assembly occurred at LD-and nuclear-proximal factories. Silencing of OC μ1 expression with siRNAs led to large factories that remained further from the nucleus, implicating μ1 in the transition to perinuclear factories. Moreover, late during infection, +RNA pools largely contributed to the production of de-novo viral proteins and fully-assembled infectious viruses. Altogether the results suggest an advanced model of reovirus replication with spatiotemporal segregation of core amplification, OC complexes and fully assembled virions., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2022
- Full Text
- View/download PDF
17. Editor's Note: Oncogenic Ras Promotes Reovirus Spread by Suppressing IFNβ Production through Negative Regulation of RIG-I Signaling.
- Author
-
Shmulevitz M, Pan LZ, Garant K, Pan D, and Lee PWK
- Published
- 2022
- Full Text
- View/download PDF
18. Genetic Modifications That Expand Oncolytic Virus Potency.
- Author
-
Cristi F, Gutiérrez T, Hitt MM, and Shmulevitz M
- Abstract
Oncolytic viruses (OVs) are a promising type of cancer therapy since they selectively replicate in tumor cells without damaging healthy cells. Many oncolytic viruses have progressed to human clinical trials, however, their performance as monotherapy has not been as successful as expected. Importantly, recent literature suggests that the oncolytic potential of these viruses can be further increased by genetically modifying the viruses. In this review, we describe genetic modifications to OVs that improve their ability to kill tumor cells directly, to dismantle the tumor microenvironment, or to alter tumor cell signaling and enhance anti-tumor immunity. These advances are particularly important to increase virus spread and reduce metastasis, as demonstrated in animal models. Since metastasis is the principal cause of mortality in cancer patients, having OVs designed to target metastases could transform cancer therapy. The genetic alterations reported to date are only the beginning of all possible improvements to OVs. Modifications described here could be combined together, targeting multiple processes, or with other non-viral therapies with potential to provide a strong and lasting anti-tumor response in cancer patients., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Cristi, Gutiérrez, Hitt and Shmulevitz.)
- Published
- 2022
- Full Text
- View/download PDF
19. The reovirus μ2 C-terminal loop inversely regulates NTPase and transcription functions versus binding to factory-forming μNS and promotes replication in tumorigenic cells.
- Author
-
Yip WKW, Cristi F, Trifonov G, Narayan N, Kubanski M, and Shmulevitz M
- Abstract
Wild type reovirus serotype 3 'Dearing PL strain' (T3wt) is being heavily evaluated as an oncolytic and immunotherapeutic treatment for cancers. Mutations that promote reovirus entry into tumor cells were previously reported to enhance oncolysis; herein we aimed to discover mutations that enhance the post-entry steps of reovirus infection in tumor cells. Using directed evolution, we identified that reovirus variant T3v10
M1 exhibited enhanced replication relative to T3wt on a panel of cancer cells. T3v10M1 contains an alanine-to-valine substitution (A612V) in the core-associated μ2, which was previously found to have NTPase activities in virions and to facilitate virus factory formation by association with μNS. Paradoxically, the A612V mutation in μ2 from T3v10M1 was discovered to impair NTPase activities and RNA synthesis, leading to five-fold higher probability of abortive infection for T3v10M1 relative to T3wt. The A612V mutation resides in a previously uncharacterized C-terminal region that juxtaposes the template entry site of the polymerase μ2; our findings thus support an important role for this domain during virus transcription. Despite crippled onset of infection, T3v10M1 exhibited greater accumulation of viral proteins and progeny during replication, leading to increased overall virus burst size. Both Far-Western and co-immunoprecipitation approaches corroborated that the A612V mutation in μ2 increased association with the non-structural virus protein μNS and enhances burst size. Altogether the data supports that mutations in the C-terminal loop domain of μ2 inversely regulate NTPase and RNA synthesis versus interactions with μNS, but with a net gain of replication in tumorigenic cells. SIGNIFICANCE Reovirus is a model system for understanding virus replication but also a clinically relevant virus for cancer therapy. We identified the first mutation that increases reovirus infection in tumorigenic cells by enhancing post-entry stages of reovirus replication. The mutation is in a previously uncharacterized c-terminal region of the M1-derived μ2 protein, which we demonstrated affects multiple functions of μ2; NTPase, RNA synthesis, inhibition of antiviral immune response and association with the virus replication factory-forming μNS protein. These findings promote a mechanistic understanding of viral protein functions. In the future, the benefits of μ2 mutations may be useful for enhancing reovirus potency in tumors., (Copyright © 2021 American Society for Microbiology.)- Published
- 2021
- Full Text
- View/download PDF
20. Captivating Perplexities of Spinareovirinae 5' RNA Caps.
- Author
-
Kniert J, Lin QF, and Shmulevitz M
- Subjects
- Animals, Humans, RNA Caps chemistry, RNA Caps genetics, RNA Processing, Post-Transcriptional, RNA, Viral chemistry, RNA, Viral genetics, Reoviridae chemistry, Reoviridae metabolism, RNA Caps metabolism, RNA, Viral metabolism, Reoviridae genetics, Reoviridae Infections virology
- Abstract
RNAs with methylated cap structures are present throughout multiple domains of life. Given that cap structures play a myriad of important roles beyond translation, such as stability and immune recognition, it is not surprising that viruses have adopted RNA capping processes for their own benefit throughout co-evolution with their hosts. In fact, that RNAs are capped was first discovered in a member of the Spinareovirinae family, Cypovirus , before these findings were translated to other domains of life. This review revisits long-past knowledge and recent studies on RNA capping among members of Spinareovirinae to help elucidate the perplex processes of RNA capping and functions of RNA cap structures during Spinareovirinae infection. The review brings to light the many uncertainties that remain about the precise capping status, enzymes that facilitate specific steps of capping, and the functions of RNA caps during Spinareovirinae replication.
- Published
- 2021
- Full Text
- View/download PDF
21. Closely related reovirus lab strains induce opposite expression of RIG-I/IFN-dependent versus -independent host genes, via mechanisms of slow replication versus polymorphisms in dsRNA binding σ3 respectively.
- Author
-
Mohamed A, Konda P, Eaton HE, Gujar S, Smiley JR, and Shmulevitz M
- Subjects
- 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, Capsid Proteins metabolism, Interferons metabolism, Polymorphism, Genetic, RNA, Double-Stranded metabolism, RNA-Binding Proteins metabolism, Receptors, Retinoic Acid metabolism, Reoviridae Infections virology, Virus Replication
- 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., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2020
- Full Text
- View/download PDF
22. Single Amino Acid Differences between Closely Related Reovirus T3D Lab Strains Alter Oncolytic Potency In Vitro and In Vivo .
- Author
-
Mohamed A, Clements DR, Gujar SA, Lee PW, Smiley JR, and Shmulevitz M
- Subjects
- 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, Mammalian orthoreovirus 3 genetics, Oncolytic Virotherapy methods, Virus Replication genetics
- 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 (TD3
PL ) 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., (Copyright © 2020 American Society for Microbiology.)- Published
- 2020
- Full Text
- View/download PDF
23. Polymorphisms in the Most Oncolytic Reovirus Strain Confer Enhanced Cell Attachment, Transcription, and Single-Step Replication Kinetics.
- Author
-
Mohamed A, Smiley JR, and Shmulevitz M
- Subjects
- 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, Mammalian orthoreovirus 3 genetics, Oncolytic Viruses genetics
- 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 (T3D
PL ), 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., (Copyright © 2020 American Society for Microbiology.)- Published
- 2020
- Full Text
- View/download PDF
24. Generation of Recombinant Rotavirus Expressing NSP3-UnaG Fusion Protein by a Simplified Reverse Genetics System.
- Author
-
Philip AA, Perry JL, Eaton HE, Shmulevitz M, Hyser JM, and Patton JT
- Subjects
- Animals, Cell Line, Gene Expression Regulation, Viral, Genes, Reporter, Genes, Viral, Green Fluorescent Proteins genetics, Green Fluorescent Proteins metabolism, Models, Molecular, Open Reading Frames, Plasmids, RNA, Double-Stranded genetics, RNA, Viral genetics, Rotavirus Infections virology, Virus Replication, Recombinant Fusion Proteins genetics, Recombinant Fusion Proteins metabolism, Reverse Genetics methods, Rotavirus genetics, Viral Nonstructural Proteins genetics, Viral Nonstructural Proteins metabolism
- Abstract
Rotavirus is a segmented double-stranded RNA (dsRNA) virus that causes severe gastroenteritis in young children. We have established an efficient simplified rotavirus reverse genetics (RG) system that uses 11 T7 plasmids, each expressing a unique simian SA11 (+)RNA, and a cytomegalovirus support plasmid for the African swine fever virus NP868R capping enzyme. With the NP868R-based system, we generated recombinant rotavirus (rSA11/NSP3-FL-UnaG) with a genetically modified 1.5-kb segment 7 dsRNA encoding full-length nonstructural protein 3 (NSP3) fused to UnaG, a 139-amino-acid green fluorescent protein (FP). Analysis of rSA11/NSP3-FL-UnaG showed that the virus replicated efficiently and was genetically stable over 10 rounds of serial passaging. The NSP3-UnaG fusion product was well expressed in rSA11/NSP3-FL-UnaG-infected cells, reaching levels similar to NSP3 levels in wild-type recombinant SA11-infected cells. Moreover, the NSP3-UnaG protein, like functional wild-type NSP3, formed dimers in vivo Notably, the NSP3-UnaG protein was readily detected in infected cells via live-cell imaging, with intensity levels ∼3-fold greater than those of the NSP1-UnaG fusion product of rSA11/NSP1-FL-UnaG. Our results indicate that FP-expressing recombinant rotaviruses can be made through manipulation of the segment 7 dsRNA without deletion or interruption of any of the 12 open reading frames (ORFs) of the virus. Because NSP3 is expressed at higher levels than NSP1 in infected cells, rotaviruses expressing NSP3-based FPs may be more sensitive tools for studying rotavirus biology than rotaviruses expressing NSP1-based FPs. This is the first report of a recombinant rotavirus containing a genetically engineered segment 7 dsRNA. IMPORTANCE Previous studies generated recombinant rotaviruses that express FPs by inserting reporter genes into the NSP1 ORF of genome segment 5. Unfortunately, NSP1 is expressed at low levels in infected cells, making viruses expressing FP-fused NSP1 less than ideal probes of rotavirus biology. Moreover, FPs were inserted into segment 5 in such a way as to compromise NSP1, an interferon antagonist affecting viral growth and pathogenesis. We have identified an alternative approach for generating rotaviruses expressing FPs, one relying on fusing the reporter gene to the NSP3 ORF of genome segment 7. This was accomplished without interrupting any of the viral ORFs, yielding recombinant viruses that likely express the complete set of functional viral proteins. Given that NSP3 is made at moderate levels in infected cells, rotaviruses encoding NSP3-based FPs should be more sensitive probes of viral infection than rotaviruses encoding NSP1-based FPs., (Copyright © 2019 American Society for Microbiology.)
- Published
- 2019
- Full Text
- View/download PDF
25. Breast Tumor-Associated Metalloproteases Restrict Reovirus Oncolysis by Cleaving the σ1 Cell Attachment Protein and Can Be Overcome by Mutation of σ1.
- Author
-
Fernandes JP, Cristi F, Eaton HE, Chen P, Haeflinger S, Bernard I, Hitt MM, and Shmulevitz M
- Subjects
- A549 Cells, Animals, Breast Neoplasms therapy, Breast Neoplasms virology, Capsid Proteins genetics, Cell Adhesion Molecules metabolism, Cell Line, Female, HeLa Cells, Humans, Metalloproteases metabolism, Mice, Mutation, N-Acetylneuraminic Acid metabolism, Oncolytic Virotherapy methods, Receptors, Virus metabolism, Reoviridae metabolism, Reoviridae pathogenicity, Reoviridae Infections immunology, Tumor Microenvironment physiology, Viral Proteins metabolism, Virus Attachment, Virus Replication physiology, Capsid Proteins metabolism, Reoviridae Infections metabolism, Virus Replication genetics
- Abstract
Reovirus is undergoing clinical testing as an oncolytic therapy for breast cancer. Given that reovirus naturally evolved to thrive in enteric environments, we sought to better understand how breast tumor microenvironments impinge on reovirus infection. Reovirus was treated with extracellular extracts generated from polyomavirus middle T-antigen-derived mouse breast tumors. Unexpectedly, these breast tumor extracellular extracts inactivated reovirus, reducing infectivity of reovirus particles by 100-fold. Mechanistically, inactivation was attributed to proteolytic cleavage of the viral cell attachment protein σ1, which diminished virus binding to sialic acid (SA)-low tumor cells. Among various specific protease class inhibitors and metal ions, EDTA and ZnCl
2 effectively modulated σ1 cleavage, indicating that breast tumor-associated zinc-dependent metalloproteases are responsible for reovirus inactivation. Moreover, media from MCF7, MB468, MD-MB-231, and HS578T breast cancer cell lines recapitulated σ1 cleavage and reovirus inactivation, suggesting that inactivation of reovirus is shared among mouse and human breast cancers and that breast cancer cells by themselves can be a source of reovirus-inactivating proteases. Binding assays and quantification of SA levels on a panel of cancer cells showed that truncated σ1 reduced virus binding to cells with low surface SA. To overcome this restriction, we generated a reovirus mutant with a mutation (T249I) in σ1 that prevents σ1 cleavage and inactivation by breast tumor-associated proteases. The mutant reovirus showed similar replication kinetics in tumorigenic cells, toxicity equivalent to that of wild-type reovirus in a severely compromised mouse model, and increased tumor titers. Overall, the data show that tumor microenvironments have the potential to reduce infectivity of reovirus. IMPORTANCE We demonstrate that metalloproteases in breast tumor microenvironments can inactivate reovirus. Our findings expose that tumor microenvironment proteases could have a negative impact on proteinaceous cancer therapies, such as reovirus, and that modification of such therapies to circumvent inactivation by tumor metalloproteases merits consideration., (Copyright © 2019 American Society for Microbiology.)- Published
- 2019
- Full Text
- View/download PDF
26. C3P3-G1: first generation of a eukaryotic artificial cytoplasmic expression system.
- Author
-
Jaïs PH, Decroly E, Jacquet E, Le Boulch M, Jaïs A, Jean-Jean O, Eaton H, Ponien P, Verdier F, Canard B, Goncalves S, Chiron S, Le Gall M, Mayeux P, and Shmulevitz M
- Subjects
- Animals, CHO Cells, Cricetulus, Cytoplasm chemistry, DNA-Directed RNA Polymerases chemistry, Eukaryotic Cells chemistry, Eukaryotic Cells metabolism, Humans, Poly A genetics, Polyadenylation genetics, Cell Nucleus genetics, Cytoplasm genetics, DNA-Directed RNA Polymerases genetics, Transcription, Genetic
- Abstract
Most eukaryotic expression systems make use of host-cell nuclear transcriptional and post-transcriptional machineries. Here, we present the first generation of the chimeric cytoplasmic capping-prone phage polymerase (C3P3-G1) expression system developed by biological engineering, which generates capped and polyadenylated transcripts in host-cell cytoplasm by means of two components. First, an artificial single-unit chimeric enzyme made by fusing an mRNA capping enzyme and a DNA-dependent RNA polymerase. Second, specific DNA templates designed to operate with the C3P3-G1 enzyme, which encode for the transcripts and their artificial polyadenylation. This system, which can potentially be adapted to any in cellulo or in vivo eukaryotic expression applications, was optimized for transient expression in mammalian cells. C3P3-G1 shows promising results for protein production in Chinese Hamster Ovary (CHO-K1) cells. This work also provides avenues for enhancing the performances for next generation C3P3 systems., (© The Author(s) 2019. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2019
- Full Text
- View/download PDF
27. Going (Reo)Viral: Factors Promoting Successful Reoviral Oncolytic Infection.
- Author
-
Bourhill T, Mori Y, Rancourt DE, Shmulevitz M, and Johnston RN
- Subjects
- Animals, Clinical Trials as Topic, Genetic Vectors, Humans, Melanoma therapy, Mice, Neoplasms virology, Orphan Drug Production, United States, United States Food and Drug Administration, Virus Replication, Neoplasms therapy, Oncolytic Virotherapy, Oncolytic Viruses physiology, Reoviridae physiology
- Abstract
Oncolytic viruses show intriguing potential as cancer therapeutic agents. These viruses are capable of selectively targeting and killing cancerous cells while leaving healthy cells largely unaffected. The use of oncolytic viruses for cancer treatments in selected circumstances has recently been approved by the Food and Drug Administration (FDA) of the US and work is progressing on engineering viral vectors for enhanced selectivity, efficacy and safety. However, a better fundamental understanding of tumour and viral biology is essential for the continued advancement of the oncolytic field. This knowledge will not only help to engineer more potent and effective viruses but may also contribute to the identification of biomarkers that can determine which patients will benefit most from this treatment. A mechanistic understanding of the overlapping activity of viral and standard chemotherapeutics will enable the development of better combinational approaches to improve patient outcomes. In this review, we will examine each of the factors that contribute to productive viral infections in cancerous cells versus healthy cells. Special attention will be paid to reovirus as it is a well-studied virus and the only wild-type virus to have received orphan drug designation by the FDA. Although considerable insight into reoviral biology exists, there remain numerous deficiencies in our understanding of the factors regulating its successful oncolytic infection. Here we will discuss what is known to regulate infection as well as speculate about potential new mechanisms that may enhance successful replication. A joint appreciation of both tumour and viral biology will drive innovation for the next generation of reoviral mediated oncolytic therapy.
- Published
- 2018
- Full Text
- View/download PDF
28. African Swine Fever Virus NP868R Capping Enzyme Promotes Reovirus Rescue during Reverse Genetics by Promoting Reovirus Protein Expression, Virion Assembly, and RNA Incorporation into Infectious Virions.
- Author
-
Eaton HE, Kobayashi T, Dermody TS, Johnston RN, Jais PH, and Shmulevitz M
- Subjects
- African Swine Fever Virus genetics, Cell Line, Recombinant Fusion Proteins metabolism, Reverse Genetics, Virion genetics, Virus Replication, mRNA Guanylyltransferases, African Swine Fever Virus enzymology, Nucleotidyltransferases metabolism, Orthoreovirus, Mammalian genetics, Orthoreovirus, Mammalian physiology, RNA, Viral metabolism, Virion physiology, Virus Assembly
- 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., (Copyright © 2017 American Society for Microbiology.)
- Published
- 2017
- Full Text
- View/download PDF
29. Novel High-throughput Approach for Purification of Infectious Virions.
- Author
-
James KT, Cooney B, Agopsowicz K, Trevors MA, Mohamed A, Stoltz D, Hitt M, and Shmulevitz M
- Subjects
- Adenoviridae isolation & purification, Centrifugation, Density Gradient, Microscopy, Electron, Reoviridae isolation & purification, Chromatography methods, Resins, Synthetic metabolism, Virion isolation & purification
- Abstract
Viruses are extensively studied as pathogens and exploited as molecular tools and therapeutic agents. Existing methods to purify viruses such as gradient ultracentrifugation or chromatography have limitations, for example demand for technical expertise or specialized equipment, high time consumption, and restricted capacity. Our laboratory explores mutations in oncolytic reovirus that could improve oncolytic activity, and makes routine use of numerous virus variants, genome reassortants, and reverse engineered mutants. Our research pace was limited by the lack of high-throughput virus purification methods that efficiently remove confounding cellular contaminants such as cytokines and proteases. To overcome this shortcoming, we evaluated a commercially available resin (Capto Core 700) that captures molecules smaller than 700 kDa. Capto. Core 700 chromatography produced virion purity and infectivity indistinguishable from CsCl density gradient ultracentrifugation as determined by electron microscopy, gel electrophoresis analysis and plaque titration. Capto Core 700 resin was then effectively adapted to a rapid in-slurry pull-out approach for high-throughput purification of reovirus and adenovirus. The in-slurry purification approach offered substantially increased virus purity over crude cell lysates, media, or high-spin preparations and would be especially useful for high-throughput virus screening applications where density gradient ultracentrifugation is not feasible., Competing Interests: K.T. James, M. Hitt, K. Agopsowicz, M.A. Trevors, D. Stoltz and M. Shmulevitz declare no conflicts of interest. B. Cooney is a Field Application Specialist affiliated with GE Healthcare Canada and was involved in conceptual design of approach, provided GE Healthcare reagents at no cost for this project, and may benefit indirectly from increased sales of associated reagents and equipment. B. Cooney remained at arms-length during experimentation and analysis.
- Published
- 2016
- Full Text
- View/download PDF
30. Potential for Improving Potency and Specificity of Reovirus Oncolysis with Next-Generation Reovirus Variants.
- Author
-
Mohamed A, Johnston RN, and Shmulevitz M
- Subjects
- Clinical Trials as Topic, Drug Discovery methods, Oncolytic Virotherapy methods, Oncolytic Viruses isolation & purification, Oncolytic Viruses physiology, Reoviridae isolation & purification, Reoviridae physiology
- Abstract
Viruses that specifically replicate in tumor over normal cells offer promising cancer therapies. Oncolytic viruses (OV) not only kill the tumor cells directly; they also promote anti-tumor immunotherapeutic responses. Other major advantages of OVs are that they dose-escalate in tumors and can be genetically engineered to enhance potency and specificity. Unmodified wild type reovirus is a propitious OV currently in phase I-III clinical trials. This review summarizes modifications to reovirus that may improve potency and/or specificity during oncolysis. Classical genetics approaches have revealed reovirus variants with improved adaptation towards tumors or with enhanced ability to establish specific steps of virus replication and cell killing among transformed cells. The recent emergence of a reverse genetics system for reovirus has provided novel strategies to fine-tune reovirus proteins or introduce exogenous genes that could promote oncolytic activity. Over the next decade, these findings are likely to generate better-optimized second-generation reovirus vectors and improve the efficacy of oncolytic reotherapy.
- Published
- 2015
- Full Text
- View/download PDF
31. Nogo to IFN by Ras.
- Author
-
Shmulevitz M and Mohamed A
- Subjects
- Animals, Humans, Interferons metabolism, Receptors, Cell Surface metabolism, ras Proteins metabolism
- Published
- 2015
- Full Text
- View/download PDF
32. Reduction of virion-associated σ1 fibers on oncolytic reovirus variants promotes adaptation toward tumorigenic cells.
- Author
-
Mohamed A, Teicher C, Haefliger S, and Shmulevitz M
- Subjects
- Animals, Blotting, Western, Capsid Proteins genetics, Cell Line, Tumor, Chymotrypsin, Flow Cytometry, Gene Knockdown Techniques, Humans, Immunohistochemistry, Immunoprecipitation, Mammalian orthoreovirus 3 pathogenicity, Mice, Mutation genetics, Oncolytic Viruses pathogenicity, RNA Interference, Adaptation, Biological genetics, Capsid Proteins metabolism, Mammalian orthoreovirus 3 genetics, Oncolytic Viruses genetics, Virion metabolism, Virus Internalization
- Abstract
Unlabelled: Wild-type mammalian orthoreovirus serotype 3 Dearing (T3wt) is nonpathogenic in humans but preferentially infects and kills cancer cells in culture and demonstrates promising antitumor activity in vivo. Using forward genetics, we previously isolated two variants of reovirus, T3v1 and T3v2, with increased infectivity toward a panel of cancer cell lines and improved in vivo oncolysis in a murine melanoma model relative to that of T3wt. Our current study explored how mutations in T3v1 and T3v2 promote infectivity. Reovirions contain trimers of σ1, the reovirus cell attachment protein, at icosahedral capsid vertices. Quantitative Western blot analysis showed that purified T3v1 and T3v2 virions had ∼ 2- and 4-fold-lower levels of σ1 fiber than did T3wt virions. Importantly, using RNA interference to reduce σ1 levels during T3wt production, we were able to generate wild-type reovirus with reduced levels of σ1 per virion. As σ1 levels were reduced, virion infectivity increased by 2- to 5-fold per cell-bound particle, demonstrating a causal relationship between virion σ1 levels and the infectivity of incoming virions. During infection of tumorigenic L929 cells, T3wt, T3v1, and T3v2 uncoated the outer capsid proteins σ3 and μ1C at similar rates. However, having started with fewer σ1 molecules, a complete loss of σ1 was achieved sooner for T3v1 and T3v2. Distinct from intracellular uncoating, chymotrypsin digestion, as a mimic of natural enteric infection, resulted in more rapid σ3 and μ1C removal, unique disassembly intermediates, and a rapid loss of infectivity for T3v1 and T3v2 compared to T3wt. Optimal infectivity toward natural versus therapeutic niches may therefore require distinct reovirus structures and σ1 levels., Importance: Wild-type reovirus is currently in clinical trials as a potential cancer therapy. Our molecular studies on variants of reovirus with enhanced oncolytic activity in vitro and in vivo now show that distinct reovirus structures promote adaptation toward cancer cells and away from conditions that mimic natural routes of infection. Specifically, we found that reovirus particles with fewer molecules of the cell attachment protein σ1 became more infectious toward transformed cells. Reduced σ1 levels conferred a benefit to incoming particles only, resulting in an earlier depletion of σ1 and a higher probability of establishing productive infection. Conversely, reovirus variants with fewer σ1 molecules showed reduced stability and infectivity and distinct disassembly when exposed to conditions that mimic natural intestinal proteolysis. These findings support a model where the mode of infection dictates the precise optimum of reovirus structure and provide a molecular rationale for considering alternative reovirus structures during oncolytic therapy., (Copyright © 2015, American Society for Microbiology. All Rights Reserved.)
- Published
- 2015
- Full Text
- View/download PDF
33. Oncolytic activity of reovirus in HPV positive and negative head and neck squamous cell carcinoma.
- Author
-
Cooper T, Biron VL, Fast D, Tam R, Carey T, Shmulevitz M, and Seikaly H
- Subjects
- Carcinoma, Squamous Cell pathology, Cell Death, Humans, Otorhinolaryngologic Neoplasms pathology, Papillomavirus Infections pathology, Carcinoma, Squamous Cell therapy, Carcinoma, Squamous Cell virology, Human papillomavirus 16, Mammalian orthoreovirus 3, Oncolytic Virotherapy methods, Otorhinolaryngologic Neoplasms therapy, Otorhinolaryngologic Neoplasms virology, Papillomavirus Infections therapy, Papillomavirus Infections virology
- Abstract
Background: The management of patients with advanced stages of head and neck cancer requires a multidisciplinary and multimodality treatment approach which includes a combination of surgery, radiation, and chemotherapy. These toxic treatment protocols have significantly improved survival outcomes in a distinct population of human papillomavirus (HPV) associated oropharyngeal cancer. HPV negative head and neck squamous cell carcinoma (HNSCC) remains a challenge to treat because there is only a modest improvement in survival with the present treatment regimens, requiring innovative and new treatment approaches. Oncolytic viruses used as low toxicity adjunct cancer therapies are novel, potentially effective treatments for HNSCC. One such oncolytic virus is Respiratory Orphan Enteric virus or reovirus. Susceptibility of HNSCC cells towards reovirus infection and reovirus-induced cell death has been previously demonstrated but has not been compared in HPV positive and negative HNSCC cell lines., Objectives: To compare the infectivity and oncolytic activity of reovirus in HPV positive and negative HNSCC cell lines., Methods: Seven HNSCC cell lines were infected with serial dilutions of reovirus. Two cell lines (UM-SCC-47 and UM-SCC-104) were positive for type 16 HPV. Infectivity was measured using a cell-based ELISA assay 18 h after infection. Oncolytic activity was determined using an alamar blue viability assay 96 h after infection. Non-linear regression models were used to calculate the amounts of virus required to infect and to cause cell death in 50% of a given cell line (EC50). EC50 values were compared., Results: HPV negative cells were more susceptible to viral infection and oncolysis compared to HPV positive cell lines. EC50 for infectivity at 18 h ranged from multiplicity of infection (MOI) values (PFU/cell) of 18.6 (SCC-9) to 3133 (UM-SCC 104). EC50 for cell death at 96 h ranged from a MOI (PFU/cell) of 1.02×10(2) (UM-SCC-14A) to 3.19×10(8) (UM-SCC-47). There was a 3×10(6) fold difference between the least susceptible cell line (UM-SCC-47) and the most susceptible line (UM-SCC 14A) EC50 for cell death at 96 h., Conclusions: HPV negative HNSCC cell lines appear to demonstrate greater reovirus infectivity and virus-mediated oncolysis compared to HPV positive HNSCC. Reovirus shows promise as a novel therapy in HNSCC, and may be of particular benefit in HPV negative patients.
- Published
- 2015
- Full Text
- View/download PDF
34. Multifaceted therapeutic targeting of ovarian peritoneal carcinomatosis through virus-induced immunomodulation.
- Author
-
Gujar S, Dielschneider R, Clements D, Helson E, Shmulevitz M, Marcato P, Pan D, Pan LZ, Ahn DG, Alawadhi A, and Lee PW
- Subjects
- ATP Binding Cassette Transporter, Subfamily B, Member 2, ATP Binding Cassette Transporter, Subfamily B, Member 3, ATP-Binding Cassette Transporters genetics, ATP-Binding Cassette Transporters metabolism, Animals, Antigen Presentation immunology, CD8-Positive T-Lymphocytes immunology, Cell Line, Tumor, Cellular Microenvironment, Cytokines immunology, Dendritic Cells immunology, Dendritic Cells pathology, Dendritic Cells virology, Female, Genetic Vectors, Humans, Immunotherapy, Mice, Mice, Inbred C57BL, Phenotype, Real-Time Polymerase Chain Reaction, Reoviridae immunology, Carcinoma therapy, Immunomodulation, Oncolytic Virotherapy methods, Ovarian Neoplasms therapy, Peritoneal Neoplasms therapy, Reoviridae genetics
- Abstract
Immunosuppression associated with ovarian cancer (OC) and resultant peritoneal carcinomatosis (PC) hampers the efficacy of many promising treatment options, including immunotherapies. It is hypothesized that oncolytic virus-based therapies can simultaneously kill OC and mitigate immunosuppression. Currently, reovirus-based anticancer therapy is undergoing phase I/II clinical trials for the treatment of OC. Hence, this study was focused on characterizing the effects of reovirus therapy on OC and associated immune microenvironment. Our data shows that reovirus efficiently killed OC cells and induced higher expression of the molecules involved in antigen presentation including major histocompatibility complex (MHC) class I, β2-microglobulin (β2M), TAP-1, and TAP-2. In addition, in the presence of reovirus, dendritic cells (DCs) overcame the OC-mediated phenotypic suppression and successfully stimulated tumor-specific CD8+ T cells. In animal studies, reovirus targeted local and distal OC, alleviated the severity of PC and significantly prolonged survival. These therapeutic effects were accompanied by decreased frequency of suppressive cells, e.g., Gr1.1+, CD11b+ myeloid derived suppressor cells (MDSCs), and CD4+, CD25+, FOXP3+ Tregs, tumor-infiltration of CD3+ cells and higher expression of Th1 cytokines. Finally, reovirus therapy during early stages of OC also resulted in the postponement of PC development. This report elucidates timely information on a therapeutic approach that can target OC through clinically desired multifaceted mechanisms to better the outcomes.
- Published
- 2013
- Full Text
- View/download PDF
35. Activation of p53 by chemotherapeutic agents enhances reovirus oncolysis.
- Author
-
Pan D, Marcato P, Ahn DG, Gujar S, Pan LZ, Shmulevitz M, and Lee PW
- Subjects
- Apoptosis drug effects, Cyclin-Dependent Kinase Inhibitor p21 metabolism, Dactinomycin pharmacology, Dose-Response Relationship, Drug, Etoposide pharmacology, Gene Expression Regulation, Neoplastic drug effects, HCT116 Cells, Humans, NF-kappa B metabolism, bcl-2-Associated X Protein metabolism, Antineoplastic Agents pharmacology, Combined Modality Therapy methods, Oncolytic Viruses physiology, Reoviridae physiology, Tumor Suppressor Protein p53 metabolism
- Abstract
Mammalian reovirus is a benign virus that possesses the natural ability to preferentially infect and kill cancer cells (reovirus oncolysis). Reovirus exploits aberrant Ras signalling in many human cancers to promote its own replication and spread. In vitro and in vivo studies using reovirus either singly or in combination with anti-cancer drugs have shown very encouraging results. Presently, a number of reovirus combination therapies are undergoing clinical trials for a variety of cancers. Previously we showed that accumulation of the tumor suppressor protein p53 by Nutlin-3a (a specific p53 stabilizer) enhanced reovirus-induced apoptosis, and resulted in significantly higher levels of reovirus dissemination. In this study, we examined the role of p53 in combination therapies involving reovirus and chemotherapeutic drugs. We showed that sub-lethal concentrations of traditional chemotherapy drugs actinomycin D or etoposide, but not doxorubicin, enhanced reovirus-induced apoptosis in a p53-dependent manner. Furthermore, NF-κB activation and expression of p53-target genes (p21 and bax) were important for the p53-dependent enhancement of cell death. Our results show that p53 status affects the efficacy of combination therapy involving reovirus. Choosing the right combination partner for reovirus and a low dosage of the drug may help to both enhance reovirus-induced cancer elimination and reduce drug toxicity.
- Published
- 2013
- Full Text
- View/download PDF
36. Exploring host factors that impact reovirus replication, dissemination, and reovirus-induced cell death in cancer versus normal cells in culture.
- Author
-
Shmulevitz M and Lee PW
- Subjects
- Apoptosis, Cell Line, Tumor, Cell Transformation, Neoplastic, Humans, MAP Kinase Signaling System genetics, Neoplasms metabolism, Oncolytic Viruses genetics, Oncolytic Viruses isolation & purification, Reoviridae genetics, Reoviridae isolation & purification, ras Proteins genetics, Neoplasms therapy, Oncolytic Virotherapy methods, Oncolytic Viruses physiology, Reoviridae physiology, Virus Replication genetics, ras Proteins metabolism
- Abstract
Oncolytic viruses, such as reovirus, offer a promising approach to cancer treatment. Concurrently, oncolytic viruses provide a valuable tool for deciphering unique attributes of cancer cells that support superior virus replication, cell death, or virus dissemination. Through our studies on various cancer cell lines, as well as isogenic cells with and without transformation by oncogenic Ras, we have identified at least four steps of virus replication that can be augmented in transformed cells. Ras transformation can support efficient reovirus uncoating during entry, production of progeny with high infectious capacity, and reovirus-induced apoptosis. Furthermore, Ras transformation also precludes interferon production following reovirus infection, permitting enhanced cell-to-cell virus spread. Methods that measure the efficiency of reovirus replication and dissemination described in this chapter can be used in combination with assorted cell culture systems to better understand the host factors that regulate reovirus oncolysis.
- Published
- 2012
- Full Text
- View/download PDF
37. Oncogenic Ras promotes reovirus spread by suppressing IFN-beta production through negative regulation of RIG-I signaling.
- Author
-
Shmulevitz M, Pan LZ, Garant K, Pan D, and Lee PW
- Subjects
- Animals, Blotting, Western, Cell Transformation, Neoplastic, Cells, Cultured, DEAD Box Protein 58, DEAD-box RNA Helicases metabolism, Flow Cytometry, Gene Expression Regulation, Gene Expression Regulation, Viral, Humans, Immunoenzyme Techniques, Interferon-beta antagonists & inhibitors, Mice, Mitogen-Activated Protein Kinase 1 genetics, Mitogen-Activated Protein Kinase 1 metabolism, Mitogen-Activated Protein Kinase 3 genetics, Mitogen-Activated Protein Kinase 3 metabolism, Mitogen-Activated Protein Kinase Kinases genetics, Mitogen-Activated Protein Kinase Kinases metabolism, NIH 3T3 Cells, Oncogenes physiology, RNA, Messenger genetics, RNA, Messenger metabolism, RNA, Viral genetics, RNA, Viral metabolism, Receptors, Immunologic, Reoviridae Infections genetics, Reoviridae Infections pathology, Reverse Transcriptase Polymerase Chain Reaction, Virion genetics, raf Kinases genetics, raf Kinases metabolism, DEAD-box RNA Helicases genetics, Genes, ras physiology, Interferon-beta metabolism, Reoviridae physiology, Reoviridae Infections metabolism, Virion metabolism
- Abstract
Reovirus is the first naturally occurring human virus reported to exploit activated Ras signaling in the host cell for infection, and is currently undergoing clinical trials as a cancer therapeutic. Recent evidence suggests that Ras transformation promotes three reoviral replication steps during the first round of infection: uncoating of the incoming virion, generation of progeny viruses with enhanced infectivity, and virus release through enhanced apoptosis. Whether oncogenic Ras also enhances reovirus spread in subsequent rounds of infection through other mechanisms has not been examined. Here, we show that compared with nontransformed cells, Ras-transformed cells are severely compromised not only in their response to IFN-beta, but also in the induction of IFN-beta mRNA following reovirus infection. Defects in both IFN-beta production and response allow for efficient virus spread in Ras-transformed cells. We show that the MEK/ERK pathway downstream of Ras is responsible for inhibiting IFN-beta expression by blocking signaling from the retinoic acid-inducible gene I (RIG-I) which recognizes viral RNAs. Overexpression of wild-type RIG-I restores INF-beta expression in reovirus-infected Ras-transformed cells. In vitro-synthesized viral mRNAs also invoke robust RIG-I-mediated IFN-beta production in transfected nontransformed cells, but not in Ras-transformed cells. Collectively, our data suggest that oncogenic Ras promotes virus spread by suppressing viral RNA-induced IFN-beta production through negative regulation of RIG-I signaling.
- Published
- 2010
- Full Text
- View/download PDF
38. Human mast cell activation with virus-associated stimuli leads to the selective chemotaxis of natural killer cells by a CXCL8-dependent mechanism.
- Author
-
Burke SM, Issekutz TB, Mohan K, Lee PW, Shmulevitz M, and Marshall JS
- Subjects
- Antiviral Agents pharmacology, CD56 Antigen metabolism, Cell Communication immunology, Cells, Cultured, Culture Media, Conditioned, Fibroblasts cytology, Humans, Interleukin-8 metabolism, Keratinocytes cytology, Killer Cells, Natural immunology, Killer Cells, Natural metabolism, Mast Cells cytology, Mast Cells immunology, Poly I-C pharmacology, Receptors, CXCR3 metabolism, T-Lymphocyte Subsets cytology, T-Lymphocyte Subsets immunology, T-Lymphocyte Subsets metabolism, Chemotaxis, Leukocyte immunology, Interleukin-8 immunology, Killer Cells, Natural cytology, Mammalian orthoreovirus 3, Mast Cells virology, Reoviridae Infections immunology
- Abstract
Human mast cells are found in skin and mucosal surfaces and next to blood vessels. They play a sentinel cell role in immunity, recognizing invading pathogens and producing proinflammatory mediators. Mast cells can recruit granulocytes, and monocytes in allergic disease and bacterial infection, but their ability to recruit antiviral effector cells such as natural killer (NK) cells and T cells has not been fully elucidated. To investigate the role of human mast cells in response to virus-associated stimuli, human cord blood-derived mast cells (CBMCs) were stimulated with polyinosinic.polycytidylic acid, a double-stranded RNA analog, or infected with the double-stranded RNA virus, reovirus serotype 3 Dearing for 24 hours. CBMCs responded to stimulation with polyinosinic.polycytidylic acid by producing a distinct chemokine profile, including CCL4, CXCL8, and CXCL10. CBMCs produced significant amounts of CXCL8 in response to low levels of reovirus infection, while both skin- and lung-derived fibroblasts were unresponsive unless higher doses of reovirus were used. Supernatants from CBMCs infected with reovirus induced substantial NK cell chemotaxis that was highly dependent on CXCL8 and CXCR1. These results suggest a novel role for mast cells in the recruitment of human NK cells to sites of early viral infection via CXCL8.
- Published
- 2008
- Full Text
- View/download PDF
39. Leaky scanning and scanning-independent ribosome migration on the tricistronic S1 mRNA of avian reovirus.
- Author
-
Racine T, Barry C, Roy K, Dawe SJ, Shmulevitz M, and Duncan R
- Subjects
- Animals, Cell Line, Codon, Initiator genetics, Eukaryotic Initiation Factor-4G genetics, Eukaryotic Initiation Factor-4G metabolism, Genome, Viral, Models, Biological, Open Reading Frames physiology, Orthoreovirus, Avian genetics, Poliovirus genetics, Poliovirus metabolism, Quail, RNA, Viral genetics, Codon, Initiator metabolism, Genes physiology, Orthoreovirus, Avian metabolism, Peptide Chain Initiation, Translational physiology, RNA, Viral metabolism, Ribosomes metabolism, Viral Proteins biosynthesis
- Abstract
The S1 genome segments of avian and Nelson Bay reovirus encode tricistronic mRNAs containing three sequential partially overlapping open reading frames (ORFs). The translation start site of the 3'-proximal ORF encoding the sigmaC protein lies downstream of two ORFs encoding the unrelated p10 and p17 proteins and more than 600 nucleotides distal from the 5'-end of the mRNA. It is unclear how translation of this remarkable tricistronic mRNA is regulated. We now show that the p10 and p17 ORFs are coordinately expressed by leaky scanning. Translation initiation events at these 5'-proximal ORFs, however, have little to no effect on translation of the 3'-proximal sigmaC ORF. Northern blotting, insertion of upstream stop codons or optimized translation start sites, 5'-truncation analysis, and poliovirus 2A protease-mediated cleavage of eIF4G indicated sigmaC translation derives from a full-length tricistronic mRNA using a mechanism that is eIF4G-dependent but leaky scanning- and translation reinitiation-independent. Further analysis of artificial bicistronic mRNAs failed to provide any evidence that sigmaC translation derives from an internal ribosome entry site. Additional features of the S1 mRNA and the mechanism of sigmaC translation also differ from current models of ribosomal shunting. Translation of the tricistronic reovirus S1 mRNA, therefore, is dependent both on leaky scanning and on a novel scanning-independent mechanism that allows translation initiation complexes to efficiently bypass two functional upstream ORFs.
- Published
- 2007
- Full Text
- View/download PDF
40. Ras transformation mediates reovirus oncolysis by enhancing virus uncoating, particle infectivity, and apoptosis-dependent release.
- Author
-
Marcato P, Shmulevitz M, Pan D, Stoltz D, and Lee PW
- Subjects
- Animals, Cell Line, Transformed, Gene Expression Regulation, Viral, Mice, Oncolytic Virotherapy, Oncolytic Viruses pathogenicity, Protein Biosynthesis genetics, Reoviridae pathogenicity, Transcription, Genetic genetics, Virion genetics, ras Proteins genetics, Apoptosis, Cell Transformation, Neoplastic, Oncolytic Viruses physiology, Reoviridae physiology, Virion metabolism, ras Proteins metabolism
- Abstract
Reovirus, a potential cancer therapy, replicates more efficiently in Ras-transformed cells than in non-transformed cells. It was presumed that increased translation was the mechanistic basis of reovirus oncolysis. Analyses of each step of the reovirus life cycle now show that cellular processes deregulated by Ras transformation promote not one but three viral replication steps. First, in Ras-transformed cells, proteolytic disassembly (uncoating) of the incoming virions, required for onset of infection, occurs more efficiently. Consequently, threefold more Ras-transformed cells become productively infected with reovirus than non-transformed cells, which accounts for the observed increase of reovirus proteins in Ras-transformed cells. Second, Ras transformation increases the infectious-to-noninfectious virus particle ratio, as virions purified from Ras-transformed cells are fourfold more infectious than those purified from non-transformed cells. Progeny assembled in non- and Ras-transformed cells appear similar by electron microscopy and sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) analysis, suggesting that Ras transformation introduces a subtle change necessary for virus infectivity. Finally, reovirus release, mediated by caspase-induced apoptosis, is ninefold more efficient in Ras-transformed cells. The combined effects of enhanced virus uncoating, infectivity, and release result in >100-fold differences in virus titers within one round of replication. Our analysis reveals previously unrecognized mechanisms by which Ras transformation mediates selective viral oncolysis.
- Published
- 2007
- Full Text
- View/download PDF
41. Unshackling the links between reovirus oncolysis, Ras signaling, translational control and cancer.
- Author
-
Shmulevitz M, Marcato P, and Lee PW
- Subjects
- Cell Transformation, Neoplastic, Neoplasms therapy, Neoplasms virology, Oncolytic Virotherapy, Oncolytic Viruses physiology, Reoviridae genetics, Reoviridae Infections, Signal Transduction, eIF-2 Kinase metabolism, Protein Biosynthesis, Reoviridae physiology, Virus Replication, ras Proteins physiology
- Abstract
Reovirus has an inherent preference for replicating in cells with dysregulated growth factor signaling cascades that comprise Ras activation. Precisely how reovirus exploits the host cell Ras pathway is unclear, but there is evidence suggesting that activated Ras signaling is important for efficient viral protein synthesis. Defining the molecular mechanism of reovirus oncolysis will shed light on reovirus replication and important aspects of cellular transformation, Ras signaling cascades and regulation of protein translation.
- Published
- 2005
- Full Text
- View/download PDF
42. A newly identified interaction between IVa2 and pVIII proteins during porcine adenovirus type 3 infection.
- Author
-
Singh M, Shmulevitz M, and Tikoo SK
- Subjects
- Animals, Cell Line, Immunoprecipitation, Microscopy, Confocal, Protein Binding, Swine, Two-Hybrid System Techniques, Adenoviruses, Porcine physiology, Viral Proteins metabolism
- Abstract
The adenovirus IVa2 is an intermediate viral gene product that appears to perform multiple essential roles in viral infection. Using IVa2 as bait in the yeast two-hybrid system, we screened selected open reading frames (ORFs) of porcine adenovirus (PAdV)-3 for potential interaction with IVa2. Interestingly, pVIII showed specific interaction with IVa2. The yeast two-hybrid findings were validated by GST pull-down assays, in vitro binding studies employing cell-free coupled transcription-translation products and in vitro co-immunoprecipitations using protein-specific antibodies. Finally, we demonstrated that IVa2 specifically interacts with pVIII during PAdV-3 infection.
- Published
- 2005
- Full Text
- View/download PDF
43. Connecting reovirus oncolysis and Ras signaling.
- Author
-
Marcato P, Shmulevitz M, and Lee PW
- Subjects
- Animals, Cell Line, Tumor, Humans, Mutation, Protein Biosynthesis, Signal Transduction, Virus Replication, Genetic Therapy methods, Neoplasms therapy, Oncolytic Viruses metabolism, Reoviridae metabolism, ras Proteins metabolism
- Abstract
Reovirus is a benign virus with innate oncolytic activity and is a potential novel therapeutic for a number of cancers. Reovirus can replicate in, and induce death of cancerous cells having an activated Ras pathway. Ras activation leads to the inactivation of cellular antiviral mechanisms, specifically removing the translation block on reovirus transcripts. This review outlines recent progress towards elucidating the molecular connection between the Ras-signaling pathway and reovirus replication.
- Published
- 2005
- Full Text
- View/download PDF
44. Cell-cell fusion induced by the avian reovirus membrane fusion protein is regulated by protein degradation.
- Author
-
Shmulevitz M, Corcoran J, Salsman J, and Duncan R
- Subjects
- Amino Acid Sequence, Animals, Cell Line, Molecular Sequence Data, Viral Fusion Proteins chemistry, Virus Replication, Membrane Fusion, Orthoreovirus, Avian physiology, Viral Fusion Proteins physiology
- Abstract
The p10 fusion-associated small transmembrane protein of avian reovirus induces extensive syncytium formation in transfected cells. Here we show that p10-induced cell-cell fusion is restricted by rapid degradation of the majority of newly synthesized p10. The small ectodomain of p10 targets the protein for degradation following p10 insertion into an early membrane compartment. Paradoxically, conservative amino acid substitutions in the p10 ectodomain hydrophobic patch that eliminate fusion activity also increase p10 stability. The small amount of p10 that escapes intracellular degradation accumulates at the cell surface in a relatively stable form, where it mediates cell-cell fusion as a late-stage event in the virus replication cycle. The unusual relationship between a nonstructural viral membrane fusion protein and the replication cycle of a nonenveloped virus has apparently contributed to the evolution of a novel mechanism for restricting the extent of virus-induced cell-cell fusion.
- Published
- 2004
- Full Text
- View/download PDF
45. Structural and functional properties of an unusual internal fusion peptide in a nonenveloped virus membrane fusion protein.
- Author
-
Shmulevitz M, Epand RF, Epand RM, and Duncan R
- Subjects
- Amino Acid Sequence, Animals, Cell Fusion, Cell Line, Hydrophobic and Hydrophilic Interactions, Liposomes metabolism, Molecular Sequence Data, Mutation, Peptides genetics, Peptides metabolism, Quail, Structure-Activity Relationship, Transfection, Viral Fusion Proteins genetics, Viral Fusion Proteins metabolism, Membrane Fusion, Orthoreovirus physiology, Orthoreovirus, Avian physiology, Peptides chemistry, Viral Fusion Proteins chemistry
- Abstract
The avian and Nelson Bay reoviruses are two of only a limited number of nonenveloped viruses capable of inducing cell-cell membrane fusion. These viruses encode the smallest known membrane fusion proteins (p10). We now show that a region of moderate hydrophobicity we call the hydrophobic patch (HP), present in the small N-terminal ectodomain of p10, shares the following characteristics with the fusion peptides of enveloped virus fusion proteins: (i) an abundance of glycine and alanine residues, (ii) a potential amphipathic secondary structure, (iii) membrane-seeking characteristics that correspond to the degree of hydrophobicity, and (iv) the ability to induce lipid mixing in a liposome fusion assay. The p10 HP is therefore predicted to provide a function in the mechanism of membrane fusion similar to those of the fusion peptides of enveloped virus fusion peptides, namely, association with and destabilization of opposing lipid bilayers. Mutational and biophysical analysis suggested that the internal fusion peptide of p10 lacks alpha-helical content and exists as a disulfide-stabilized loop structure. Similar kinked structures have been reported in the fusion peptides of several enveloped virus fusion proteins. The preservation of a predicted loop structure in the fusion peptide of this unusual nonenveloped virus membrane fusion protein supports an imperative role for a kinked fusion peptide motif in biological membrane fusion.
- Published
- 2004
- Full Text
- View/download PDF
46. Palmitoylation, membrane-proximal basic residues, and transmembrane glycine residues in the reovirus p10 protein are essential for syncytium formation.
- Author
-
Shmulevitz M, Salsman J, and Duncan R
- Subjects
- Amino Acid Motifs, Animals, Glycine, Membrane Fusion, Membrane Proteins chemistry, Palmitates metabolism, Quail, Structure-Activity Relationship, Viral Nonstructural Proteins physiology, Virus Replication, Giant Cells physiology, Reoviridae chemistry, Viral Nonstructural Proteins chemistry
- Abstract
Avian reovirus and Nelson Bay reovirus are two unusual nonenveloped viruses that induce extensive cell-cell fusion via expression of a small nonstructural protein, termed p10. We investigated the importance of the transmembrane domain, a conserved membrane-proximal dicysteine motif, and an endodomain basic region in the membrane fusion activity of p10. We now show that the p10 dicysteine motif is palmitoylated and that loss of palmitoylation correlates with a loss of fusion activity. Mutational and functional analyses also revealed that a triglycine motif within the transmembrane domain and the membrane-proximal basic region were essential for p10-mediated membrane fusion. Mutations in any of these three motifs did not influence events upstream of syncytium formation, such as p10 membrane association, protein topology, or surface expression, suggesting that these motifs are more intimately associated with the membrane fusion reaction. These results suggest that the rudimentary p10 fusion protein has evolved a mechanism of inducing membrane merger that is highly dependent on the specific interaction of several different motifs with donor membranes. In addition, cross-linking, coimmunoprecipitation, and complementation assays provided no evidence for p10 homo- or heteromultimer formation, suggesting that p10 may be the first example of a membrane fusion protein that does not form stable, higher-order multimers.
- Published
- 2003
- Full Text
- View/download PDF
47. Sequential partially overlapping gene arrangement in the tricistronic S1 genome segments of avian reovirus and Nelson Bay reovirus: implications for translation initiation.
- Author
-
Shmulevitz M, Yameen Z, Dawe S, Shou J, O'Hara D, Holmes I, and Duncan R
- Subjects
- Amino Acid Sequence, Animals, Birds virology, Cell Line, Conserved Sequence genetics, Molecular Sequence Data, Molecular Weight, Open Reading Frames genetics, Sequence Alignment, Sequence Analysis, DNA, Viral Proteins biosynthesis, Viral Proteins chemistry, Viral Proteins genetics, Genes genetics, Genes, Overlapping genetics, Genes, Viral genetics, Genome, Viral, Orthoreovirus genetics, Orthoreovirus, Avian genetics, Peptide Chain Initiation, Translational genetics
- Abstract
Previous studies of the avian reovirus strain S1133 (ARV-S1133) S1 genome segment revealed that the open reading frame (ORF) encoding the final sigmaC viral cell attachment protein initiates over 600 nucleotides distal from the 5' end of the S1 mRNA and is preceded by two predicted small nonoverlapping ORFs. To more clearly define the translational properties of this unusual polycistronic RNA, we pursued a comparative analysis of the S1 genome segment of the related Nelson Bay reovirus (NBV). Sequence analysis indicated that the 3'-proximal ORF present on the NBV S1 genome segment also encodes a final sigmaC homolog, as evidenced by the presence of an extended N-terminal heptad repeat characteristic of the coiled-coil region common to the cell attachment proteins of reoviruses. Most importantly, the NBV S1 genome segment contains two conserved ORFs upstream of the final sigmaC coding region that are extended relative to the predicted ORFs of ARV-S1133 and are arranged in a sequential, partially overlapping fashion. Sequence analysis of the S1 genome segments of two additional strains of ARV indicated a similar overlapping tricistronic gene arrangement as predicted for the NBV S1 genome segment. Expression analysis of the ARV S1 genome segment indicated that all three ORFs are functional in vitro and in virus-infected cells. In addition to the previously described p10 and final sigmaC gene products, the S1 genome segment encodes from the central ORF a 17-kDa basic protein (p17) of no known function. Optimizing the translation start site of the ARV p10 ORF lead to an approximately 15-fold increase in p10 expression with little or no effect on translation of the downstream final sigmaC ORF. These results suggest that translation initiation complexes can bypass over 600 nucleotides and two functional overlapping upstream ORFs in order to access the distal final sigmaC start site.
- Published
- 2002
- Full Text
- View/download PDF
48. A new class of fusion-associated small transmembrane (FAST) proteins encoded by the non-enveloped fusogenic reoviruses.
- Author
-
Shmulevitz M and Duncan R
- Subjects
- Amino Acid Sequence, Animals, Membrane Proteins genetics, Membrane Proteins metabolism, Molecular Sequence Data, Virus Replication, Reoviridae physiology, Viral Fusion Proteins genetics, Viral Fusion Proteins metabolism, Viral Proteins genetics, Viral Proteins metabolism
- Abstract
The non-enveloped fusogenic avian and Nelson Bay reoviruses encode homologous 10 kDa non-structural transmembrane proteins. The p10 proteins localize to the cell surface of transfected cells in a type I orientation and induce efficient cell-cell fusion. Mutagenic studies revealed the importance of conserved sequence-predicted structural motifs in the membrane association and fusogenic properties of p10. These motifs included a centrally located transmembrane domain, a conserved cytoplasmic basic region, a small hydrophobic motif in the N-terminal domain and four conserved cysteine residues. Functional analysis indicated that the extreme C-terminus of p10 functions in a sequence-independent manner to effect p10 membrane localization, while the N-terminal domain displays a sequence-dependent effect on the fusogenic property of p10. The small size, unusual arrangement of structural motifs and lack of any homologues in previously described membrane fusion proteins suggest that the fusion-associated small transmembrane (FAST) proteins of reovirus represent a new class of membrane fusion proteins.
- Published
- 2000
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.