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










Publication year range
1.
Viruses ; 16(4)2024 04 22.
Article in English | MEDLINE | ID: mdl-38675990

ABSTRACT

Infectious hematopoietic necrosis virus (IHNV) and viral hemorrhagic septicemia virus (VHSV) are rhabdoviruses in two different species belonging to the Novirhabdovirus genus. IHNV has a narrow host range restricted to trout and salmon species, and viruses in the M genogroup of IHNV have high virulence in rainbow trout (Oncorhynchus mykiss). In contrast, the VHSV genotype IVb that invaded the Great Lakes in the United States has a broad host range, with high virulence in yellow perch (Perca flavescens), but not in rainbow trout. By using reverse-genetic systems of IHNV-M and VHSV-IVb strains, we generated six IHNV:VHSV chimeric viruses in which the glycoprotein (G), non-virion-protein (NV), or both G and NV genes of IHNV-M were replaced with the analogous genes from VHSV-IVb, and vice versa. These chimeric viruses were used to challenge groups of rainbow trout and yellow perch. The parental recombinants rIHNV-M and rVHSV-IVb were highly virulent in rainbow trout and yellow perch, respectively. Parental rIHNV-M was avirulent in yellow perch, and chimeric rIHNV carrying G, NV, or G and NV genes from VHSV-IVb remained low in virulence in yellow perch. Similarly, the parental rVHSV-IVb exhibited low virulence in rainbow trout, and chimeric rVHSV with substituted G, NV, or G and NV genes from IHNV-M remained avirulent in rainbow trout. Thus, the G and NV genes of either virus were not sufficient to confer high host-specific virulence when exchanged into a heterologous species genome. Some exchanges of G and/or NV genes caused a loss of host-specific virulence, providing insights into possible roles in viral virulence or fitness, and interactions between viral proteins.


Subject(s)
Fish Diseases , Novirhabdovirus , Oncorhynchus mykiss , Perches , Rhabdoviridae Infections , Animals , Oncorhynchus mykiss/virology , Perches/virology , Virulence , Novirhabdovirus/genetics , Novirhabdovirus/pathogenicity , Fish Diseases/virology , Rhabdoviridae Infections/veterinary , Rhabdoviridae Infections/virology , Glycoproteins/genetics , Infectious hematopoietic necrosis virus/genetics , Infectious hematopoietic necrosis virus/pathogenicity , Viral Proteins/genetics , Viral Proteins/metabolism , Host Specificity
2.
Mol Ther Methods Clin Dev ; 26: 532-546, 2022 Sep 08.
Article in English | MEDLINE | ID: mdl-36092362

ABSTRACT

Despite recent therapeutic advances, metastatic breast cancer (MBC) remains incurable. Engineered measles virus (MV) constructs based on the attenuated MV Edmonston vaccine platform have demonstrated significant oncolytic activity against solid tumors. The Helicobacter pylori neutrophil-activating protein (NAP) is responsible for the robust inflammatory reaction in gastroduodenal mucosa during bacterial infection. NAP attracts and activates immune cells at the site of infection, inducing expression of pro-inflammatory mediators. We engineered an MV strain to express the secretory form of NAP (MV-s-NAP) and showed that it exhibits anti-tumor and immunostimulatory activity in human breast cancer xenograft models. In this study, we utilized a measles-infection-permissive mouse model (transgenic IFNAR KO-CD46Ge) to evaluate the biodistribution and safety of MV-s-NAP. The primary objective was to identify potential toxic side effects and confirm the safety of the proposed clinical doses of MV-s-NAP prior to a phase I clinical trial of intratumoral administration of MV-s-NAP in patients with MBC. Both subcutaneous delivery (corresponding to the clinical trial intratumoral administration route) and intravenous (worst case scenario) delivery of MV-s-NAP were well tolerated: no significant clinical, laboratory or histologic toxicity was observed. This outcome supports the safety of MV-s-NAP for oncolytic virotherapy of MBC. The first-in-human clinical trial of MV-s-NAP in patients with MBC (ClinicalTrials.gov: NCT04521764) was subsequently activated.

3.
J Clin Invest ; 131(13)2021 07 01.
Article in English | MEDLINE | ID: mdl-34196308

ABSTRACT

Clinical immunotherapy approaches are lacking efficacy in the treatment of glioblastoma (GBM). In this study, we sought to reverse local and systemic GBM-induced immunosuppression using the Helicobacter pylori neutrophil-activating protein (NAP), a potent TLR2 agonist, as an immunostimulatory transgene expressed in an oncolytic measles virus (MV) platform, retargeted to allow viral entry through the urokinase-type plasminogen activator receptor (uPAR). While single-agent murine anti-PD1 treatment or repeat in situ immunization with MV-s-NAP-uPA provided modest survival benefit in MV-resistant syngeneic GBM models, the combination treatment led to synergy with a cure rate of 80% in mice bearing intracranial GL261 tumors and 72% in mice with CT-2A tumors. Combination NAP-immunovirotherapy induced massive influx of lymphoid cells in mouse brain, with CD8+ T cell predominance; therapeutic efficacy was CD8+ T cell dependent. Inhibition of the IFN response pathway using the JAK1/JAK2 inhibitor ruxolitinib decreased PD-L1 expression on myeloid-derived suppressor cells in the brain and further potentiated the therapeutic effect of MV-s-NAP-uPA and anti-PD1. Our findings support the notion that MV strains armed with bacterial immunostimulatory antigens represent an effective strategy to overcome the limited efficacy of immune checkpoint inhibitor-based therapies in GBM, creating a promising translational strategy for this lethal brain tumor.


Subject(s)
Antigens, Bacterial/therapeutic use , Brain Neoplasms/therapy , Glioblastoma/therapy , Oncolytic Virotherapy/methods , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Animals , Antigens, Bacterial/administration & dosage , Antigens, Bacterial/genetics , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Bacterial Proteins/therapeutic use , Brain Neoplasms/immunology , Brain Neoplasms/pathology , CD8-Positive T-Lymphocytes/immunology , Cell Death/immunology , Cell Line, Tumor , Combined Modality Therapy , Cytokines/metabolism , Cytopathogenic Effect, Viral , Female , Glioblastoma/immunology , Glioblastoma/pathology , Humans , Lymphocytes, Tumor-Infiltrating/immunology , Measles virus/genetics , Measles virus/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Oncolytic Viruses/genetics , Oncolytic Viruses/immunology , Receptors, Urokinase Plasminogen Activator/immunology , Translational Research, Biomedical , Virus Internalization
4.
Mol Ther Oncolytics ; 19: 136-148, 2020 Dec 16.
Article in English | MEDLINE | ID: mdl-33145397

ABSTRACT

Measles virus (MV) Edmonston derivative strains are attractive vector platforms in vaccine development and oncolytic virotherapy. Helicobacter pylori heat shock protein A (HspA) is a bacterial heat shock chaperone with essential function as a Ni-ion scavenging protein. We generated and characterized the immunogenicity of an attenuated MV strain encoding the HspA transgene (MV-HspA). MV-HspA showed faster replication within 48 h of infection with >10-fold higher titers and faster accumulation of the MV proteins. It also demonstrated a superior tumor-killing effect in vitro against a variety of human solid tumor cell lines, including sarcoma, ovarian and breast cancer. Two intraperitoneal (i.p.) doses of 106 50% tissue culture infectious dose (TCID50) MV-HspA significantly improved survival in an ovarian cancer xenograft model: 63.5 days versus 27 days for the control group. The HspA transgene induced a humoral immune response in measles-permissive Ifnarko-CD46Ge transgenic mice. Eight of nine animals developed a long-term anti-HspA antibody response with titers of 1:400 to 1:12,800 without any negative impact on development of protective anti-MV immune memory. MV-HspA triggered an immunogenic cytopathic effect as measured by an HMGB1 assay. The absence of significant elevation of PD-L1 expression indicated that vector-encoded HspA could act as an immunomodulator on the immune check point axis. These data demonstrate that MV-HspA is a potent oncolytic agent and vaccine candidate for clinical translation in cancer treatment and immunoprophylaxis against H. pylori.

5.
Future Virol ; 12(4): 193-213, 2017 Apr.
Article in English | MEDLINE | ID: mdl-29387140

ABSTRACT

Oncolytic viruses (OVs) are engineered and/or evolved to propagate selectively in cancerous tissues. They have a dual mechanism of action; direct killing of infected cancer cells cross-primes anticancer immunity to boost the killing of uninfected cancer cells. The goal of the field is to develop OVs that are easily manufactured, efficiently delivered to disseminated sites of cancer growth, undergo rapid intratumoral spread, selectively kill tumor cells, cause no collateral damage and pose no risk of transmission in the population. Here we discuss the many virus engineering strategies that are being pursued to optimize delivery, intratumoral spread and safety of OVs derived from different virus families. With continued progress, OVs have the potential to transform the paradigm of cancer care.

6.
Mol Ther Oncolytics ; 3: 16019, 2016.
Article in English | MEDLINE | ID: mdl-27556105

ABSTRACT

Mumps virus belongs to the family of Paramyxoviridae and has the potential to be an oncolytic agent. Mumps virus Urabe strain had been tested in the clinical setting as a treatment for human cancer four decades ago in Japan. These clinical studies demonstrated that mumps virus could be a promising cancer therapeutic agent that showed significant antitumor activity against various types of cancers. Since oncolytic virotherapy was not in the limelight until the beginning of the 21(st) century, the interest to pursue mumps virus for cancer treatment slowly faded away. Recent success stories of oncolytic clinical trials prompted us to resurrect the mumps virus and to explore its potential for cancer treatment. We have obtained the Urabe strain of mumps virus from Osaka University, Japan, which was used in the earlier human clinical trials. In this report we describe the development of a reverse genetics system from a major isolate of this Urabe strain mumps virus stock, and the construction and characterization of several recombinant mumps viruses with additional transgenes. We present initial data demonstrating these recombinant mumps viruses have oncolytic activity against tumor cell lines in vitro and some efficacy in preliminary pilot animal tumor models.

7.
Dev Comp Immunol ; 55: 241-252, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26429411

ABSTRACT

The infectious hematopoietic necrosis virus (IHNV; Rhabdoviridae, Novirhabdovirus) infects teleost fish, such as salmon and trout, and is responsible for significant losses in the aquaculture industry and in wild fish populations. Although IHNV enters the host through the skin at the base of the fins, the viral adhesion and entry mechanisms are not fully understood. In recent years, evidence has accumulated in support of the key roles played by protein-carbohydrate interactions between host lectins secreted to the extracellular space and virion envelope glycoproteins in modulating viral adhesion and infectivity. In this study, we assessed in vitro the potential role(s) of zebrafish (Danio rerio) proto type galectin-1 (Drgal1-L2) and a chimera galectin-3 (Drgal3-L1) in IHNV adhesion to epithelial cells. Our results suggest that the extracellular Drgal1-L2 and Drgal3-L1 interact directly and in a carbohydrate-dependent manner with the IHNV glycosylated envelope and glycans on the epithelial cell surface, significantly reducing viral adhesion.


Subject(s)
Epithelial Cells/physiology , Galectins/metabolism , Infectious hematopoietic necrosis virus/immunology , Recombinant Fusion Proteins/metabolism , Rhabdoviridae Infections/immunology , Viral Envelope Proteins/metabolism , Zebrafish Proteins/metabolism , Zebrafish/immunology , Animals , Cells, Cultured , Epithelial Cells/virology , Galectins/genetics , Host-Pathogen Interactions , Infectious hematopoietic necrosis virus/pathogenicity , Recombinant Fusion Proteins/genetics , Rhabdoviridae Infections/transmission , Virulence , Virus Attachment , Zebrafish Proteins/genetics
8.
J Virol ; 88(11): 6148-57, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24648451

ABSTRACT

UNLABELLED: Because of its very low human seroprevalence, vesicular stomatitis virus (VSV) has promise as a systemic oncolytic agent for human cancer therapy. However, as demonstrated in this report, the VSV infectious titer drops by 4 log units during the first hour of exposure to nonimmune human serum. This neutralization occurs relatively slowly and is mediated by the concerted actions of natural IgM and complement. Maraba virus, whose G protein is about 80% homologous to that of VSV, is relatively resistant to the neutralizing activity of nonimmune human serum. We therefore constructed and rescued a recombinant VSV whose G gene was replaced by the corresponding gene from Maraba virus. Comparison of the parental VSV and VSV with Maraba G substituted revealed nearly identical host range properties and replication kinetics on a panel of tumor cell lines. Moreover, in contrast to the parental VSV, the VSV with Maraba G substituted was resistant to nonimmune human serum. Overall, our data suggest that VSV with Maraba G substituted should be further investigated as a candidate for human systemic oncolytic virotherapy applications. IMPORTANCE: Oncolytic virotherapy is a promising approach for the treatment of disseminated cancers, but antibody neutralization of circulating oncolytic virus particles remains a formidable barrier. In this work, we developed a pseudotyped vesicular stomatitis virus (VSV) with a glycoprotein of Maraba virus, a closely related but serologically distinct member of the family Rhabdoviridae, which demonstrated greatly diminished susceptibility to both nonimmune and VSV-immune serum neutralization. VSV with Maraba G substituted or lentiviral vectors should therefore be further investigated as candidates for human systemic oncolytic virotherapy and gene therapy applications.


Subject(s)
Complement System Proteins/immunology , Immunoglobulin M/immunology , Vesiculovirus/immunology , Animals , Antibodies, Monoclonal , Cell Line, Tumor , Chlorocebus aethiops , DNA Primers/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Neutralization Tests , Oncolytic Virotherapy/methods , Vero Cells , Vesiculovirus/genetics , Vesiculovirus/pathogenicity , Viral Envelope Proteins/genetics
9.
J Virol ; 87(24): 13543-55, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24089573

ABSTRACT

We sought proof of principle that tumor-targeting ligands can be displayed on the surface of vesicular stomatitis virus (VSV) by engineering its glycoprotein. Here, we successfully rescued VSVs displaying tumor vasculature-targeting ligands. By using a rational approach, we investigated various feasible insertion sites on the G protein of VSV (VSV-G) for display of tumor vasculature-targeting ligands, cyclic RGD (cRGD) and echistatin. We found seven sites on VSV-G that tolerated insertion of the 9-residue cRGD peptide, two of which could tolerate insertion of the 49-amino acid echistatin domain. All of the ligand-displaying viruses replicated as well as the parental virus. In vitro studies demonstrated that the VSV-echistatin viruses specifically bound to targeted integrins. Since the low-density lipoprotein receptor (LDLR) was recently identified as a major receptor for VSV, we investigated the entry of ligand-displaying viruses after masking LDLR. The experiment showed that the modified viruses can enter the cell independently of LDLR, whereas entry of unmodified virus is significantly blocked by a specific monoclonal antibody against LDLR. Both parental and ligand-displaying viruses displayed equal oncolytic efficacies in a syngeneic mouse myeloma model. We further demonstrated that single-chain antibody fragments against tumor-specific antigens can be inserted at the N terminus of the G protein and that corresponding replication-competent VSVs can be rescued efficiently. Overall, we demonstrated that functional tumor-targeting ligands can be displayed on replication-competent VSVs without perturbing viral growth and oncolytic efficacy. This study provides a rational foundation for the future development of fully retargeted oncolytic VSVs.


Subject(s)
Neoplasms/therapy , Oncolytic Virotherapy , Oncolytic Viruses/genetics , Peptides, Cyclic/genetics , Peptides/genetics , Vesicular stomatitis Indiana virus/genetics , Animals , Cell Line, Tumor , Female , Humans , Intercellular Signaling Peptides and Proteins , Ligands , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred BALB C , Neoplasms/virology , Oncolytic Viruses/physiology , Peptides/metabolism , Peptides, Cyclic/metabolism , Protein Engineering , Vesicular stomatitis Indiana virus/physiology , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , Virus Internalization
10.
J Virol ; 87(6): 3217-28, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23283963

ABSTRACT

Vesicular stomatitis virus (VSV) is potent and a highly promising agent for the treatment of cancer. However, translation of VSV oncolytic virotherapy into the clinic is being hindered by its inherent neurotoxicity. It has been demonstrated that selected picornaviral internal ribosome entry site (IRES) elements possess restricted activity in neuronal tissues. We therefore sought to determine whether the picornavirus IRES could be engineered into VSV to attenuate its neuropathogenicity. We have used IRES elements from human rhinovirus type 2 (HRV2) and foot-and-mouth disease virus (FMDV) to control the translation of the matrix gene (M), which plays a major role in VSV virulence. In vitro studies revealed slowed growth kinetics of IRES-controlled VSVs in most of the cell lines tested. However, in vivo studies explicitly demonstrated that IRES elements of HRV2 and FMDV severely attenuated the neurovirulence of VSV without perturbing its oncolytic potency.


Subject(s)
Gene Expression Regulation, Viral , Protein Biosynthesis , Vesiculovirus/genetics , Vesiculovirus/pathogenicity , Animals , Cell Line , Foot-and-Mouth Disease Virus/genetics , Humans , Oncolytic Viruses/genetics , Oncolytic Viruses/growth & development , Oncolytic Viruses/pathogenicity , Recombination, Genetic , Rhinovirus/genetics , Vesiculovirus/growth & development , Viral Matrix Proteins/biosynthesis , Viral Matrix Proteins/genetics
11.
J Virol ; 85(16): 8393-402, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21653667

ABSTRACT

Viral hemorrhagic septicemia virus (VHSV) and infectious hematopoietic necrosis virus (IHNV) are members of the genus Novirhabdovirus within the Rhabdoviridae family, which can cause severe hemorrhagic disease in fresh- and saltwater fish worldwide. These viruses carry an additional nonvirion (NV) gene, which codes for the nonstructural NV protein that has been implicated to play a role in viral pathogenesis. To determine the precise biological function of this NV gene and its gene product, we generated NV-deficient and NV knockout recombinant VHSVs, using reverse genetics. Comparisons of the replication kinetics and markers for virus-induced apoptosis indicated that the NV-deficient and NV knockout mutant viruses induce apoptosis earlier in cell culture than the wild-type recombinant VHSV. These results suggest that the NV protein has an antiapoptotic function at the early stage of virus infection. Furthermore, we created a chimeric VHSV, in which the NV gene of VHSV was replaced by the IHNV NV gene, which was capable of suppressing apoptosis in cell culture. These results show that the NV protein of other members of Novirhabdovirus can restore the NV protein function. In this study, we also investigated the kinetics of VHSV replication during a single round of viral replication and examined the mechanism of VHSV-induced apoptosis. Our results show that VHSV infection induced caspases 3, 8 and 9 in cell culture.


Subject(s)
Apoptosis , Novirhabdovirus/physiology , Viral Nonstructural Proteins/genetics , Animals , Caspase 3/biosynthesis , Caspase 3/metabolism , Caspase 8/biosynthesis , Caspase 8/metabolism , Caspase 9/biosynthesis , Caspase 9/metabolism , Cell Line , DNA Fragmentation , Fishes , Novirhabdovirus/genetics , Novirhabdovirus/pathogenicity , Sequence Deletion , Virus Replication
12.
Mar Biotechnol (NY) ; 13(4): 672-83, 2011 Aug.
Article in English | MEDLINE | ID: mdl-20936318

ABSTRACT

Viral hemorrhagic septicemia virus (VHSV), belonging to the genus Novirhabdovirus in the family of Rhabdoviridae, causes a highly contagious disease of fresh and saltwater fish worldwide. Recently, a novel genotype of VHSV, designated IVb, has invaded the Great Lakes in North America, causing large-scale epidemics in wild fish. An efficient reverse genetics system was developed to generate a recombinant VHSV of genotype IVb from cloned cDNA. The recombinant VHSV (rVHSV) was comparable to the parental wild-type strain both in vitro and in vivo, causing high mortality in yellow perch (Perca flavescens). A modified recombinant VHSV was generated in which the NV gene was substituted with an enhanced green fluorescent protein gene (rVHSV-ΔNV-EGFP), and another recombinant was made by inserting the EGFP gene into the full-length viral clone between the P and M genes (rVHSV-EGFP). The in vitro replication kinetics of rVHSV-EGFP was similar to rVHSV; however, the rVHSV-ΔNV-EGFP grew 2 logs lower. In yellow perch challenges, wtVHSV and rVHSV induced 82-100% cumulative per cent mortality (CPM), respectively, whereas rVHSV-EGFP produced 62% CPM and rVHSV-ΔNV-EGFP caused only 15% CPM. No reversion of mutation was detected in the recovered viruses and the recombinant viruses stably maintained the foreign gene after several passages. These results indicate that the NV gene of VHSV is not essential for viral replication in vitro and in vivo, but it plays an important role in viral replication efficiency and pathogenicity. This system will facilitate studies of VHSV replication, virulence, and production of viral vectored vaccines.


Subject(s)
DNA, Recombinant/genetics , Genome, Viral/genetics , Novirhabdovirus/genetics , Novirhabdovirus/pathogenicity , Perches/virology , Reverse Genetics/methods , Viral Proteins/genetics , Animals , DNA, Complementary/genetics , Genome Components , Great Lakes Region , Green Fluorescent Proteins/genetics , Oligonucleotides/genetics , Plasmids/genetics , Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Virus Replication/genetics
13.
J Virol ; 85(3): 1408-14, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21106739

ABSTRACT

The delivery of foreign epitopes by a replicating nonpathogenic avian infectious bursal disease virus (IBDV) was explored. The aim of the study was to identify regions in the IBDV genome that are amenable to the introduction of a sequence encoding a foreign peptide. By using a cDNA-based reverse genetics system, insertions or substitutions of sequences encoding epitope tags (FLAG, c-Myc, or hepatitis C virus epitopes) were engineered in the open reading frames of a nonstructural protein (VP5) and the capsid protein (VP2). Attempts were also made to generate recombinant IBDV that displayed foreign epitopes in the exposed loops (P(BC) and P(HI)) of the VP2 trimer. We successfully recovered recombinant IBDVs expressing c-Myc and two different virus-neutralizing epitopes of human hepatitis C virus (HCV) envelope glycoprotein E in the VP5 region. Western blot analyses with anti-c-Myc and anti-HCV antibodies provided positive identification of both the c-Myc and HCV epitopes that were fused to the N terminus of VP5. Genetic analysis showed that the recombinants carrying the c-Myc/HCV epitopes maintained the foreign gene sequences and were stable after several passages in Vero and 293T cells. This is the first report describing efficient expression of foreign peptides from a replication-competent IBDV and demonstrates the potential of this virus as a vector.


Subject(s)
Antigens, Viral/genetics , Drug Carriers , Epitopes/genetics , Genetic Vectors , Hepacivirus/genetics , Infectious bursal disease virus/genetics , Viral Hepatitis Vaccines/genetics , Animals , Antigens, Viral/immunology , Cell Line , Chlorocebus aethiops , Epitopes/immunology , Hepacivirus/immunology , Humans , Iris Diseases , Mutagenesis, Insertional , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Sequence Deletion , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Viral Hepatitis Vaccines/immunology
14.
J Virol Methods ; 167(2): 132-9, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20362010

ABSTRACT

Reverse genetics system is a powerful tool to study the function of a particular gene. The currently available reverse genetics system for Novirhabdovirus is based on vaccinia-driven T7 RNA polymerase expression. An improved system for the recovery of infectious hematopoietic necrosis virus (IHNV) was developed which utilizes cellular RNA polymerase II machinery for transcription. A full-length cDNA clone of IHNV, flanked by hammerhead ribozyme and hepatitis delta ribozyme sequences, was assembled in an expression plasmid under the control of a cytomegalovirus promoter. Transfection of this full-length plasmid along with the supporting plasmids (N, P, NV and L) into epithelioma papulosum cyprini cells resulted in the recovery of a viable recombinant IHNV (rIHNV). The authenticity of rIHNV was confirmed by the presence of restriction sites introduced artificially in the genome. A recombinant IHNV expressing a foreign gene - enhanced green fluorescent protein - was also recovered. The recovered IHNVs showed similar growth characteristics as the parental IHNV in cell cultures. Challenge of susceptible rainbow trout with wild-type IHNV and rIHNV induced clinical disease signs indistinguishable from the parental strain and produced mortality. Thus, a vaccinia-virus-free reverse genetics system described for IHNV is applicable for the recovery of any Novirhabdovirus, which will facilitate studies on viral pathogenesis and the design of new generation of viral vectored vaccines.


Subject(s)
Fish Diseases/virology , Genetic Engineering/methods , Infectious hematopoietic necrosis virus/genetics , Infectious hematopoietic necrosis virus/pathogenicity , Rhabdoviridae Infections/veterinary , Vaccinia virus/genetics , Virology/methods , Animals , Cell Line , Cytomegalovirus/genetics , Fish Diseases/pathology , Genes, Reporter , Green Fluorescent Proteins/genetics , Hepatitis Delta Virus/genetics , Oncorhynchus mykiss , Promoter Regions, Genetic , RNA, Catalytic/genetics , Rhabdoviridae Infections/pathology , Rhabdoviridae Infections/virology , Virulence
15.
Virol J ; 7: 10, 2010 Jan 19.
Article in English | MEDLINE | ID: mdl-20085652

ABSTRACT

BACKGROUND: Infectious hematopoietic necrosis virus (IHNV) is the type species of the genus Novirhabdovirus, within the family Rhabdoviridae, infecting several species of wild and hatchery reared salmonids. Similar to other rhabdoviruses, IHNV has a linear single-stranded, negative-sense RNA genome of approximately 11,000 nucleotides. The IHNV genome encodes six genes; the nucleocapsid, phosphoprotein, matrix protein, glycoprotein, non-virion protein and polymerase protein genes, respectively. This study describes molecular characterization of the virulent IHNV strain 220-90, belonging to the M genogroup, and its phylogenetic relationships with available sequences of IHNV isolates worldwide. RESULTS: The complete genomic sequence of IHNV strain 220-90 was determined from the DNA of six overlapping clones obtained by RT-PCR amplification of genomic RNA. The complete genome sequence of 220-90 comprises 11,133 nucleotides (GenBank GQ413939) with the gene order of 3'-N-P-M-G-NV-L-5'. These genes are separated by conserved gene junctions, with di-nucleotide gene spacers. An additional uracil nucleotide was found at the end of the 5'-trailer region, which was not reported before in other IHNV strains. The first 15 of the 16 nucleotides at the 3'- and 5'-termini of the genome are complementary, and the first 4 nucleotides at 3'-ends of the IHNV are identical to other novirhadoviruses. Sequence homology and phylogenetic analysis of the glycoprotein genes show that 220-90 strain is 97% identical to most of the IHNV strains. Comparison of the virulent 220-90 genomic sequences with less virulent WRAC isolate shows more than 300 nucleotides changes in the genome, which doesn't allow one to speculate putative residues involved in the virulence of IHNV. CONCLUSION: We have molecularly characterized one of the well studied IHNV isolates, 220-90 of genogroup M, which is virulent for rainbow trout, and compared phylogenetic relationship with North American and other strains. Determination of the complete nucleotide sequence is essential for future studies on pathogenesis of IHNV using a reverse genetics approach and developing efficient control strategies.


Subject(s)
Genome, Viral , Infectious hematopoietic necrosis virus/genetics , RNA, Viral/genetics , Animals , Cluster Analysis , Fish Diseases/virology , Genotype , Molecular Sequence Data , Oncorhynchus mykiss/virology , Phylogeny , Rhabdoviridae Infections/veterinary , Rhabdoviridae Infections/virology , Sequence Analysis, DNA , Sequence Homology
16.
Virol J ; 6: 171, 2009 Oct 25.
Article in English | MEDLINE | ID: mdl-19852863

ABSTRACT

BACKGROUND: Viral hemorrhagic septicemia virus (VHSV) is a highly contagious viral disease of fresh and saltwater fish worldwide. VHSV caused several large scale fish kills in the Great Lakes area and has been found in 28 different host species. The emergence of VHS in the Great Lakes began with the isolation of VHSV from a diseased muskellunge (Esox masquinongy) caught from Lake St. Clair in 2003. VHSV is a member of the genus Novirhabdovirus, within the family Rhabdoviridae. It has a linear single-stranded, negative-sense RNA genome of approximately 11 kbp, with six genes. VHSV replicates in the cytoplasm and produces six monocistronic mRNAs. The gene order of VHSV is 3'-N-P-M-G-NV-L-5'. This study describes molecular characterization of the Great Lakes VHSV strain (MI03GL), and its phylogenetic relationships with selected European and North American isolates. RESULTS: The complete genomic sequences of VHSV-MI03GL strain was determined from cloned cDNA of six overlapping fragments, obtained by RT-PCR amplification of genomic RNA. The complete genome sequence of MI03GL comprises 11,184 nucleotides (GenBank GQ385941) with the gene order of 3'-N-P-M-G-NV-L-5'. These genes are separated by conserved gene junctions, with di-nucleotide gene spacers. The first 4 nucleotides at the termini of the VHSV genome are complementary and identical to other novirhadoviruses genomic termini. Sequence homology and phylogenetic analysis show that the Great Lakes virus is closely related to the Japanese strains JF00Ehi1 (96%) and KRRV9822 (95%). Among other novirhabdoviruses, VHSV shares highest sequence homology (62%) with snakehead rhabdovirus. CONCLUSION: Phylogenetic tree obtained by comparing 48 glycoprotein gene sequences of different VHSV strains demonstrate that the Great Lakes VHSV is closely related to the North American and Japanese genotype IVa, but forms a distinct genotype IVb, which is clearly different from the three European genotypes. Molecular characterization of the Great Lakes isolate will be helpful in studying the pathogenesis of VHSV using a reverse genetics approach and developing efficient control strategies.


Subject(s)
Fish Diseases/virology , Genome, Viral , Novirhabdovirus/genetics , Novirhabdovirus/isolation & purification , RNA, Viral/genetics , Sequence Analysis, DNA , Animals , Cluster Analysis , Fishes , Gene Order , Genes, Viral , Great Lakes Region , Molecular Sequence Data , Novirhabdovirus/classification , Phylogeny , Sequence Homology
17.
Virol J ; 6: 158, 2009 Oct 07.
Article in English | MEDLINE | ID: mdl-19811655

ABSTRACT

BACKGROUND: Avian influenza viruses, belonging to the family Orthomyxoviridae, possess distinct combinations of hemagglutinin (H) and the neuraminidase (N) surface glycoproteins. Typing of both H and N antigens is essential for the epidemiological and surveillance studies. Therefore, it is important to find a rapid, sensitive, and specific method for their assay, and ELISA can be useful for this purpose, by using recombinant proteins. RESULTS: The nucleoprotein (NP) and truncated neuraminidase subtype 3 and 7 of avian influenza virus (AIV) were expressed in Saccharomyces cerevisiae and used to develop an indirect enzyme-linked immunosorbent assay for antibody detection. The developed assays were evaluated with a panel of 64 chicken serum samples. The performance of NP-ELISA was compared with the commercially available ProFlok AIV ELISA kit. The results showed comparable agreement and sensitivity between the two tests, indicating that NP-ELISA assay can be used for screening the influenza type A antibody in AIV infected birds. The N3 and N7- ELISAs also reacted specifically to their type specific sera and did not exhibit any cross-reaction with heterologous neuraminidase subtype specific sera. CONCLUSION: The study demonstrates the expression of the NP, N3, and N7 proteins of AIV in yeast (S. cerevisiae) and their application in developing an indirect ELISA for detecting NP, N3 and N7 antibodies from AIV-infected chicken sera. The described indirect ELISAs are rapid, sensitive, specific and can be used as promising tests during serological surveillance.


Subject(s)
Antibodies, Viral/blood , Antigens, Viral/immunology , Enzyme-Linked Immunosorbent Assay/methods , Influenza in Birds/diagnosis , Neuraminidase/immunology , RNA-Binding Proteins/immunology , Viral Core Proteins/immunology , Viral Proteins/immunology , Animals , Chickens , Influenza in Birds/virology , Nucleocapsid Proteins , Poultry Diseases/diagnosis , Poultry Diseases/virology , Recombinant Proteins/immunology , Saccharomyces cerevisiae/genetics , Sensitivity and Specificity
18.
Arch Virol ; 154(3): 495-9, 2009.
Article in English | MEDLINE | ID: mdl-19219402

ABSTRACT

Infectious bronchitis virus (IBV) is the causal agent of infectious bronchitis, which still remains one of the most important poultry diseases worldwide because of numerous serotypes and variants. A virulent strain of IBV, isolated from Arkansas (Ark), was propagated in embryonated eggs (Ark DPI 11). Following 101 serial passages in embryonated eggs, an attenuated strain of IBV was established (Ark DPI 101) that does not induce histopathological lesions in the tracheae of infected chicks. To identify sequence changes responsible for the attenuation of IBV, complete genome sequences of both virulent and attenuated Ark DPI viruses were obtained. Comparison of the genome sequences of the virulent and attenuated Ark DPI viruses reveals that these viruses are similar and differ only by 21 nucleotides, resulting in 17 amino acids changes. Most of those substitutions are located in the replicase 1a and spike genes. No differences were observed in gene 3, M or 5a, and only one nucleotide substitution each was present in 5b, N and 3'UTR. By comparing the deduced amino acid sequences of virulent and attenuated viruses, we identified sequence changes responsible for the adaptation and attenuation of the IBV-Ark DPI strain.


Subject(s)
Coronavirus Infections/veterinary , Infectious bronchitis virus/genetics , Infectious bronchitis virus/pathogenicity , Poultry Diseases/virology , Adaptation, Biological , Amino Acid Substitution , Animals , Coronavirus Infections/virology , Molecular Sequence Data , Mutation, Missense , RNA, Viral/genetics , Sequence Analysis, DNA , Trachea/pathology , Trachea/virology , Viral Proteins/genetics
19.
Viruses ; 1(3): 1166-77, 2009 12.
Article in English | MEDLINE | ID: mdl-21994587

ABSTRACT

Infectious bronchitis virus (IBV) causes highly contagious respiratory or urogenital tract diseases in chickens. The Maryland 27(Md27) strain was first isolated in 1976 from diseased chicken flocks in the Delmarva Peninsula region. To understand the genetic diversity and phylogenetic relationship of existing strains with Md27, the complete nucleotide sequence of the 3'end coding region (∼7.2 kb) of Md27 was determined and compared with other IBV strains and coronaviruses. It has the same S-3-M-5-N-3' gene order, as is the case of other IBV strains. The spike gene of Md27 exhibits 97% identity with the SE17 strain. There are deletions at the spike gene, non-coding region between M and 5 genes, and at the 3' untranslated region (UTR), which is different from Ark-like strains. Phylogenetic analysis and sequence alignments demonstrate that Md27 is a chimera containing different gene segments that are most closely related to the SE17, Conn and JMK strains. This current study provides evidence for genomic mutations and intergenic recombination that have taken place in the evolution of IBV strain Md27.

20.
Virol J ; 5: 157, 2008 Dec 22.
Article in English | MEDLINE | ID: mdl-19102764

ABSTRACT

An infectious bronchitis virus Arkansas DPI (Ark DPI) virulent strain was sequenced, analyzed and compared with many different IBV strains and coronaviruses. The genome of Ark DPI consists of 27,620 nucleotides, excluding poly (A) tail, and comprises ten open reading frames. Comparative sequence analysis of Ark DPI with other IBV strains shows striking similarity to the Conn, Gray, JMK, and Ark 99, which were circulating during that time period. Furthermore, comparison of the Ark genome with other coronaviruses demonstrates a close relationship to turkey coronavirus. Among non-structural genes, the 5'untranslated region (UTR), 3C-like proteinase (3CLpro) and the polymerase (RdRp) sequences are 100% identical to the Gray strain. Among structural genes, S1 has 97% identity with Ark 99; S2 has 100% identity with JMK and 96% to Conn; 3b 99%, and 3C to N is 100% identical to Conn strain. Possible recombination sites were found at the intergenic region of spike gene, 3'end of S1 and 3a gene. Independent recombination events may have occurred in the entire genome of Ark DPI, involving four different IBV strains, suggesting that genomic RNA recombination may occur in any part of the genome at number of sites. Hence, we speculate that the Ark DPI strain originated from the Conn strain, but diverged and evolved independently by point mutations and recombination between field strains.


Subject(s)
Chickens/virology , Coronavirus Infections/veterinary , Evolution, Molecular , Genome, Viral , Infectious bronchitis virus/genetics , Poultry Diseases/virology , 5' Untranslated Regions/genetics , Animals , Coronavirus 3C Proteases , Coronavirus Infections/virology , Cysteine Endopeptidases/genetics , DNA-Directed RNA Polymerases/genetics , Infectious bronchitis virus/classification , Membrane Glycoproteins/genetics , Molecular Sequence Data , Phylogeny , Recombination, Genetic , Sequence Analysis, RNA , Sequence Homology , Spike Glycoprotein, Coronavirus , Viral Envelope Proteins/genetics , Viral Proteins/genetics , Viral Structural Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...