Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Viruses ; 15(1)2022 12 31.
Article in English | MEDLINE | ID: mdl-36680176

ABSTRACT

Old world alphaviruses, such as Ross River virus (RRV), cause debilitating arthralgia during acute and chronic stages of the disease. RRV-induced cartilage degradation has been implicated as a cause of joint pain felt by RRV patients. Chondrocytes are a major cell type of cartilage and are involved in the production and maintenance of the cartilage matrix. It is thought that these cells may play a vital role in RRV disease pathogenesis. In this study, we used RNA-sequencing (RNA-Seq) to examine the transcriptomes of RRV-infected and bystander chondrocytes in the same environment. RRV containing green fluorescent protein (GFP) allowed for the separation of RRV-infected (GFP+) and bystander uninfected cells (GFP-). We found that whereas GFP+ and GFP- populations commonly presented similar gene expression profiles during infection, there were also unique signatures. For example, RIMS2 and FOXJ1 were unique to GFP+ cells, whilst Aim2 and CCL8 were only found in bystander chondrocytes. This indicates that careful selection of potential therapeutic targets is important to minimise adverse effects to the neighbouring uninfected cell populations. Our study serves as a resource to provide more information about the pathways and responses elicited by RRV in cells which are both infected and stimulated because of neighbouring infected cells.


Subject(s)
Alphavirus Infections , Alphavirus , Humans , Chondrocytes/metabolism , Alphavirus/genetics , Ross River virus/genetics , Ross River virus/metabolism
2.
Viruses ; 13(8)2021 07 31.
Article in English | MEDLINE | ID: mdl-34452382

ABSTRACT

Alphaviruses are small enveloped viruses with positive-sense RNA genomes. During infection, the alphavirus capsid protein (Cp) selectively packages and assembles with the viral genomic RNA to form the nucleocapsid core, a process critical to the production of infectious virus. Prior studies of the alphavirus Semliki Forest virus (SFV) showed that packaging and assembly are promoted by Cp binding to multiple high affinity sites on the genomic RNA. Here, we developed an in vitro Cp binding assay based on fluorescently labeled RNA oligos. We used this assay to explore the RNA sequence and structure requirements for Cp binding to site #1, the top binding site identified on the genomic RNA during all stages of virus assembly. Our results identify a stem-loop structure that promotes specific binding of the SFV Cp to site #1 RNA. This structure is also recognized by the Cps of the related alphaviruses chikungunya virus and Ross River virus.


Subject(s)
Alphavirus/genetics , Capsid Proteins/genetics , Capsid Proteins/metabolism , Genome, Viral/physiology , Inverted Repeat Sequences/genetics , RNA, Viral/metabolism , Alphavirus/metabolism , Binding Sites , Capsid/metabolism , Cell Line , Chikungunya virus/genetics , Chikungunya virus/metabolism , Genome, Viral/genetics , Inverted Repeat Sequences/physiology , Protein Binding , RNA, Viral/genetics , RNA-Binding Motifs , Ross River virus/genetics , Ross River virus/metabolism , Semliki forest virus/genetics , Semliki forest virus/metabolism , Virus Assembly
4.
J Virol ; 86(19): 10675-85, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22837203

ABSTRACT

Arthralgia-associated alphaviruses, including chikungunya virus (CHIKV) and Ross River virus (RRV), pose significant public health threats because of their ability to cause explosive outbreaks of debilitating arthralgia and myalgia in human populations. Although the host inflammatory response is known to contribute to the pathogenesis of alphavirus-induced arthritis and myositis, the role that Toll-like receptors (TLRs), which are major regulators of host antiviral and inflammatory responses, play in the pathogenesis of alphavirus-induced arthritis and myositis has not been extensively studied. Using a mouse model of RRV-induced myositis/arthritis, we found that myeloid differentiation primary response gene 88 (Myd88)-dependent TLR7 signaling is involved in protection from severe RRV-associated disease. Infections of Myd88- and TLR7-deficient mouse strains with RRV revealed that both Myd88 and TLR7 significantly contributed to protection from RRV-induced mortality, and both mouse strains exhibited more severe tissue damage than wild-type (WT) mice following RRV infection. While viral loads were unchanged in either Myd88 or TLR7 knockout mice compared to WT mice at early times postinfection, both Myd88 and TLR7 knockout mice exhibited higher viral loads than WT mice at late times postinfection. Furthermore, while high levels of RRV-specific antibody were produced in TLR7-deficient mice, this antibody had very little neutralizing activity and had lower affinity than WT antibody. Additionally, TLR7- and Myd88-deficient mice showed defects in germinal center activity, suggesting that TLR7-dependent signaling is critical for the development of protective antibody responses against RRV.


Subject(s)
Alphavirus Infections/metabolism , Membrane Glycoproteins/metabolism , Myeloid Differentiation Factor 88/metabolism , Ross River virus/metabolism , Toll-Like Receptor 7/metabolism , Animals , Antibodies, Neutralizing/chemistry , Biological Assay/methods , Enzyme-Linked Immunosorbent Assay/methods , Flow Cytometry/methods , Inflammation , Mice , Mice, Inbred C57BL , Mice, Transgenic , Vero Cells
5.
PLoS One ; 6(10): e25837, 2011.
Article in English | MEDLINE | ID: mdl-22016778

ABSTRACT

Toll-like Receptor 3 (TLR3) detects double-stranded (ds) RNAs to activate innate immune responses. While poly(I:C) is an excellent agonist for TLR3 in several cell lines and in human peripheral blood mononuclear cells, viral dsRNAs tend to be poor agonists, leading to the hypothesis that additional factor(s) are likely required to allow TLR3 to respond to viral dsRNAs. TLR3 signaling was examined in a lung epithelial cell line by quantifying cytokine production and in human embryonic kidney cells by quantifying luciferase reporter levels. Recombinant 1b hepatitis C virus polymerase was found to enhance TLR3 signaling in the lung epithelial BEAS-2B cells when added to the media along with either poly(I:C) or viral dsRNAs. The polymerase from the genotype 2a JFH-1 HCV was a poor enhancer of TLR3 signaling until it was mutated to favor a conformation that could bind better to a partially duplexed RNA. The 1b polymerase also co-localizes with TLR3 in endosomes. RNA-binding capsid proteins (CPs) from two positive-strand RNA viruses and the hepadenavirus hepatitis B virus (HBV) were also potent enhancers of TLR3 signaling by poly(I:C) or viral dsRNAs. A truncated version of the HBV CP that lacked an arginine-rich RNA-binding domain was unable to enhance TLR3 signaling. These results demonstrate that several viral RNA-binding proteins can enhance the dsRNA-dependent innate immune response initiated by TLR3.


Subject(s)
Hepacivirus/metabolism , Immunity, Innate , RNA, Double-Stranded/metabolism , RNA-Binding Proteins/metabolism , Signal Transduction/immunology , Toll-Like Receptor 3/metabolism , Viral Proteins/metabolism , Capsid Proteins/metabolism , Endosomes/metabolism , Genotype , HEK293 Cells , Hepacivirus/enzymology , Hepacivirus/genetics , Humans , Models, Molecular , Poly I-C/metabolism , Protein Conformation , Protein Transport , RNA, Viral/metabolism , RNA-Dependent RNA Polymerase/chemistry , RNA-Dependent RNA Polymerase/metabolism , Ross River virus/metabolism
6.
Proc Natl Acad Sci U S A ; 108(29): 12048-53, 2011 Jul 19.
Article in English | MEDLINE | ID: mdl-21730129

ABSTRACT

Arthrogenic alphaviruses, such as Ross River virus (RRV), chikungunya, Sindbis, mayaro and o'nyong-nyong viruses circulate endemically worldwide, frequently causing outbreaks of polyarthritis. The exact mechanisms of how alphaviruses induce polyarthritis remain ill defined, although macrophages are known to play a key role. Macrophage migration inhibitory factor (MIF) is an important cytokine involved in rheumatoid arthritis pathogenesis. Here, we characterize the role of MIF in alphavirus-induced arthritides using a mouse model of RRV-induced arthritis, which has many characteristics of RRV disease in humans. RRV-infected WT mice developed severe disease associated with up-regulated MIF expression in serum and tissues, which corresponded to severe inflammation and tissue damage. MIF-deficient (MIF(-/-)) mice developed mild disease accompanied by a reduction in inflammatory infiltrates and muscle destruction in the tissues, despite having viral titers similar to WT mice. In addition, reconstitution of MIF into MIF(-/-) mice exacerbated RRV disease and treatment of mice with MIF antagonist ameliorated disease in WT mice. Collectively, these findings suggest that MIF plays a critical role in determining the clinical severity of alphavirus-induced musculoskeletal disease and may provide a target for the development of antiviral pharmaceuticals. The prospect being that early treatment with MIF-blocking pharmaceuticals may curtail the debilitating arthritis associated with alphaviral infections.


Subject(s)
Arthritis/virology , Gene Expression Regulation/physiology , Intramolecular Oxidoreductases/metabolism , Macrophage Migration-Inhibitory Factors/metabolism , Myositis/virology , Ross River virus/metabolism , Analysis of Variance , Animals , Arthritis/metabolism , Arthritis/physiopathology , Chemokine CCL2/metabolism , Enzyme-Linked Immunosorbent Assay , Histological Techniques , Interferon-gamma/metabolism , Intramolecular Oxidoreductases/antagonists & inhibitors , Macrophage Migration-Inhibitory Factors/antagonists & inhibitors , Mice , Mice, Inbred C57BL , Mice, Knockout , Myositis/metabolism , Myositis/physiopathology , Reverse Transcriptase Polymerase Chain Reaction
7.
J Gene Med ; 13(3): 181-7, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21394858

ABSTRACT

BACKGROUND: Lentiviral vectors (LV) are promising vaccines because they transduce dendritic cells (DC) in vivo. To translate LV vaccines into clinical trials, bulk production will be necessary. The present study aimed to find a suitable envelope for LV vaccine production from stable packaging cells because the commonly used vesicular stomatitis virus envelope (VSV-G) is cytotoxic. METHODS: The envelope from Ross river virus (RRV) was selected. It can infect mouse and human cells, allowing testing in animals before clinical translation. We used VSV-G for comparison. Vectors produced with each envelope were titred on human 293T cells and mouse 3T3 cells. RESULTS: RRV-pseudotyped LV (RRV-LV) infected mouse myeloid DC in culture and immunized mice. An approximately 50-fold higher dose of RRV-LV than VSV-G-LV was required to generate a similar T cell response. The RRV-LV could also be used to infect human mDC and to prime a human T cell immune response. CONCLUSIONS: RRV envelope is a suitable candidate to be used for the construction of an LV producer cell line. LV vaccines with RRV envelope can be tested in mice and in human immune cell cultures. The higher dose of RRV-LV required for vaccine efficacy compared to VSV-G-LV will partly be offset by ease of production.


Subject(s)
Genetic Vectors/metabolism , Lentivirus/genetics , Ross River virus/metabolism , Vaccines, Synthetic/biosynthesis , Viral Envelope Proteins/metabolism , 3T3 Cells , Animals , Cell Line , Dendritic Cells/metabolism , Enzyme-Linked Immunospot Assay , Gene Transfer Techniques , Humans , Mice , Mice, Inbred C57BL , Ross River virus/genetics , T-Lymphocytes/metabolism , Transduction, Genetic , Vaccines, Synthetic/immunology , Vesicular stomatitis Indiana virus/genetics , Vesicular stomatitis Indiana virus/metabolism , Viral Envelope Proteins/genetics , Viral Envelope Proteins/toxicity
8.
J Virol ; 82(24): 12374-83, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18922878

ABSTRACT

Alphaviruses are mosquito-transmitted viruses that cause significant human disease, and understanding how these pathogens successfully transition from the mosquito vector to the vertebrate host is an important area of research. Previous studies demonstrated that mosquito and mammalian-cell-derived alphaviruses differentially induce type I interferons (alpha/beta interferon [IFN-alpha/beta]) in myeloid dendritic cells (mDCs), where the mosquito cell-derived virus is a poor inducer of IFN-alpha/beta compared to the mammalian-cell-derived virus. Furthermore, the reduced IFN-alpha/beta induction by the mosquito cell-derived virus is attributed to differential N-linked glycosylation. To further evaluate the role of viral envelope glycans in regulating the IFN-alpha/beta response, studies were performed to assess whether the mosquito cell-derived virus actively inhibits IFN-alpha/beta induction or is simply a poor inducer of IFN-alpha/beta. Coinfection studies using mammalian- and mosquito cell-derived Ross River virus (mam-RRV and mos-RRV, respectively) indicated that mos-RRV was unable to suppress IFN-alpha/beta induction by mam-RRV in mDC cultures. Additionally, a panel of mutant viruses lacking either individual or multiple N-linked glycosylation sites was used to demonstrate that N-linked glycans were essential for high-level IFN-alpha/beta induction by the mammalian-cell-derived virus. These results suggest that the failure of the mosquito cell-derived virus to induce IFN-alpha/beta is due to a lack of complex carbohydrates on the virion rather than the active suppression of the DC antiviral response.


Subject(s)
Dendritic Cells/metabolism , Interferon Type I/biosynthesis , Myeloid Cells/metabolism , Polysaccharides/metabolism , Ross River virus/metabolism , Viral Envelope Proteins/metabolism , Aedes , Animals , Cell Line , Cricetinae , Genome, Viral/genetics , Mutation/genetics , Ross River virus/genetics , Viral Envelope Proteins/genetics
9.
J Virol ; 80(18): 8848-55, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16940497

ABSTRACT

The assembly of the alphavirus nucleocapsid core has been investigated using an in vitro assembly system. The C-terminal two-thirds of capsid protein (CP), residues 81 to 264 in Sindbis virus (SINV), have been previously shown to have all the RNA-CP and CP-CP contacts required for core assembly in vitro. Helix I, which is located in the N-terminal dispensable region of the CP, has been proposed to stabilize the core by forming a coiled coil in the CP dimer formed by the interaction of residues 81 to 264. We examined the ability of heterologous alphavirus CPs to dimerize and form phenotypically mixed core-like particles (CLPs) using an in vitro assembly system. The CPs of SINV and Ross River virus (RRV) do not form phenotypically mixed CLPs, but SINV and Western equine encephalitis virus CPs do form mixed cores. In addition, CP dimers do not form between SINV and RRV in these assembly reactions. In contrast, an N-terminal truncated SINV CP (residues 81 to 264) forms phenotypically mixed CLPs when it is assembled with full-length heterologous CPs, suggesting that the region that controls the mixing is present in the N-terminal 80 residues. Furthermore, this result suggests that the dimeric interaction, which was absent between SINV and RRV CPs, can be restored by the removal of the N-terminal 80 residues of the SINV CP. We mapped the determinant that is responsible for phenotypic mixing onto helix I by using domain swapping experiments. Thus, discrimination of the CP partner in alphavirus core assembly appears to be dependent on helix I sequence compatibility. These results suggest that helix I provides one of the important interactions during nucleocapsid core formation and may play a regulatory role during the early steps of the assembly process.


Subject(s)
Alphavirus/genetics , Capsid Proteins/chemistry , Sindbis Virus/metabolism , Alphavirus/metabolism , Amino Acid Sequence , Capsid Proteins/physiology , Cross-Linking Reagents/pharmacology , Dimerization , Molecular Sequence Data , Mutation , Nucleocapsid/chemistry , Phenotype , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Ross River virus/metabolism , Sequence Homology, Amino Acid , Virus Assembly
10.
Int J Mol Med ; 15(6): 999-1003, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15870906

ABSTRACT

We have evaluated a chimeric, two-component Sindbis virus packaging system. As expected, use of this combination of two modified helper RNA species prevented formation of infection competent Sindbis viruses as analyzed by serial passaging. We observed, however, that vectors produced using this method were able to spread in BHK cell cultures and formed clusters of transgene positive cells that did not display cytopathic effects for up to 3 days post-transduction. Formation of spreading Sindbis virus vectors required only one of the helper components--the chimera with a deleted Ross River virus capsid and the Sindbis virus envelope glycoproteins. Spreading was also demonstrated in two rat glioma cell lines, 9L and BT4C, showing that this phenomenon was not limited to BHK cells. Our results warrant further characterization of split helper Sindbis virus vectors and imply their utility in gene therapy approaches where spreading of transgene expression and consequently high gene transfer rate could be beneficial.


Subject(s)
Genetic Vectors , Helper Viruses/genetics , Sindbis Virus/genetics , Amino Acid Sequence , Animals , Brain Neoplasms/therapy , Capsid/chemistry , Cell Line , Cricetinae , Cricetulus , Flow Cytometry , Gene Deletion , Genetic Therapy , Glioma/therapy , Green Fluorescent Proteins/metabolism , Helper Viruses/physiology , Microscopy, Fluorescence , RNA, Viral/genetics , Rats , Recombinant Fusion Proteins/metabolism , Replicon , Ross River virus/genetics , Ross River virus/metabolism , Sindbis Virus/metabolism , Sindbis Virus/physiology , Time Factors , Transduction, Genetic , Transgenes , Viral Envelope Proteins/genetics , Virus Replication
11.
J Virol ; 79(3): 1765-71, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15650201

ABSTRACT

Alphavirus glycoproteins have broad host ranges. Human immunodeficiency virus type 1 (HIV-1) vectors pseudotyped with their glycoproteins could extend the range of tissues that can be transduced in both humans and animal models. Here, we established stable producer cell lines for HIV vectors pseudotyped with alphavirus Ross River virus (RRV) and Semliki Forest virus (SFV) glycoproteins E2E1. RRV E2E1-stable clones could routinely produce high-titer pseudotyped vectors for at least 5 months. SFV E2E1-stable clones, however, produced relatively low titers. We examined the properties of RRV E2E1-pseudotyped vectors [HIV-1(RRV)] and compared them with amphotropic murine leukemia virus Env- and vesicular stomatitis virus glycoprotein G-pseudotyped vectors. HIV-1(RRV) displayed a number of characteristics which would be advantageous in ex vivo and in vivo experiments, including resistance to inactivation by heat-labile components in fresh human sera and thermostability at 37 degrees C. Upon single-step concentration by ultracentrifugation of HIV-1(RRV), we could achieve vector stocks with titers up to 6 x 10(7) IU/ml. HIV-1(RRV) efficiently transduced cells from several different species, including murine primary dendritic cells, but failed to transduce human and murine T cells as well as human hematopoietic stem cells (HSC). These results indicate that HIV-1(RRV) could be used in a number of applications including animal model experiments and suggest that expression of RRV cellular receptors is limited or absent in certain cell types such as T cells and human HSC.


Subject(s)
Alphavirus/genetics , Genetic Vectors , Glycoproteins/metabolism , HIV-1/genetics , Viral Envelope Proteins/metabolism , Alphavirus/metabolism , Animals , Cell Line , Cells, Cultured , Dendritic Cells/virology , Glycoproteins/genetics , HIV-1/physiology , Hematopoietic Stem Cells/virology , Humans , Mice , Ross River virus/genetics , Ross River virus/metabolism , Semliki forest virus/genetics , Semliki forest virus/metabolism , T-Lymphocytes/virology , Transduction, Genetic , Viral Envelope Proteins/genetics , Virus Assembly
12.
J Virol ; 78(3): 1421-30, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14722297

ABSTRACT

Ross River virus (RRV) and Semliki Forest virus (SFV) are two alphaviruses that have a high degree of amino acid homology, as well as a very broad host range. We show here that envelope glycoproteins derived from both viruses can pseudotype human immunodeficiency virus type 1 (HIV-1)-derived lentivirus vectors. Both RRV and SFV glycoproteins considerably expand the host range of the lentivirus vector, and vectors can be efficiently concentrated by ultracentrifugation. A systematic analysis comparing the alphaviral glycoproteins to the vesicular stomatitis virus glycoprotein (VSV-G) revealed that lentivirus vectors incorporate RRV glycoproteins with an efficiency comparable to that of VSV-G. Both pseudotypes have comparable physical titers, but infectious titers with the RRV pseudotype are lower than with VSV-G. Incorporation of SFV glycoproteins into lentivirus vector is less efficient, leading to decreased physical and infectious titers. The transduction rates with VSV-G-, RRV-, and SFV-pseudotyped lentivirus vectors into adherent cell lines can be significantly increased by using a combination of Polybrene and plates coated with CH-296 recombinant fibronectin fragments. Together, our data suggest that RRV and SFV glycoproteins might be suitable as alternatives to VSV-G for pseudotyping lentivirus vectors.


Subject(s)
Genetic Vectors , HIV-1/genetics , Lentivirus/genetics , Ross River virus/genetics , Semliki forest virus/genetics , Viral Envelope Proteins/metabolism , Animals , Cell Line , Chlorocebus aethiops , HeLa Cells , Humans , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , NIH 3T3 Cells , Ross River virus/metabolism , Semliki forest virus/metabolism , Transduction, Genetic , Vero Cells , Viral Envelope Proteins/genetics
13.
J Virol ; 76(18): 9378-88, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12186920

ABSTRACT

Vectors derived from lentiviruses provide a promising gene delivery system. We examined the in vivo gene transfer efficiency and tissue or cell tropism of a feline immunodeficiency virus (FIV)-based lentiviral vector pseudotyped with the glycoproteins from Ross River Virus (RRV). RRV glycoproteins were efficiently incorporated into FIV virions, generating preparations of FIV vector, which after concentration attain titers up to 1.5 x 10(8) TU/ml. After systemic administration, RRV-pseudotyped FIV vectors (RRV/FIV) predominantly transduced the liver of recipient mice. Transduction efficiency in the liver with the RRV/FIV was ca. 20-fold higher than that achieved with the vesicular stomatitis virus G protein (VSV-G) pseudotype. Moreover, in comparison to VSV-G, the RRV glycoproteins caused less cytotoxicity, as determined from the levels of glutamic pyruvic transaminase and glutamic oxalacetic transaminase in serum. Although hepatocytes were the main liver cell type transduced, nonhepatocytes (mainly Kupffer cells) were also transduced. The percentages of the transduced nonhepatocytes were comparable between RRV and VSV-G pseudotypes and did not correlate with the production of antibody against the transgene product. After injection into brain, RRV/FIV preferentially transduced neuroglial cells (astrocytes and oligodendrocytes). In contrast to the VSV-G protein that targets predominantly neurons, <10% of the brain cells transduced with the RRV pseudotyped vector were neurons. Finally, the gene transfer efficiencies of RRV/FIV after direct application to skeletal muscle or airway were also examined and, although transgene-expressing cells were detected, their proportions were low. Our data support the utility of RRV glycoprotein-pseudotyped FIV lentiviral vectors for hepatocyte- and neuroglia-related disease applications.


Subject(s)
Capsid Proteins , Capsid/genetics , Gene Transfer Techniques , Genetic Vectors , Immunodeficiency Virus, Feline/genetics , Membrane Glycoproteins/genetics , Ross River virus/genetics , Viral Envelope Proteins/genetics , Animals , Cats , Hepatocytes/metabolism , Hepatocytes/virology , Immunodeficiency Virus, Feline/metabolism , Liver/cytology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Muscle, Skeletal/metabolism , Muscle, Skeletal/virology , Neuroglia/metabolism , Neuroglia/virology , Ross River virus/metabolism , Transduction, Genetic , Transgenes/genetics , Transgenes/physiology , Virion/genetics , Virion/metabolism , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
14.
J Virol ; 75(14): 6303-9, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11413296

ABSTRACT

Passage of Ross River virus strain NB5092 in avian cells has been previously shown to select for virus variants that have enhanced replication in these cells. Sequencing of these variants identified two independent sites that might be responsible for the phenotype. We now demonstrate, using a molecular cDNA clone of the wild-type T48 strain, that an amino acid substitution at residue 218 in the E2 glycoprotein can account for the phenotype. Substitutions that replaced the wild-type asparagine with basic residues had enhanced replication in avian cells while acidic or neutral residues had little or no observable effect. Ross River virus mutants that had increased replication in avian cells also grew better in BHK cells than the wild-type virus, whereas the remaining mutants were unaffected in growth. Replication in both BHK and avian cells of Ross River virus mutants N218K and N218R was inhibited by the presence of heparin or by the pretreatment of the cells with heparinase. Binding of the mutants, but not of the wild type, to a heparin-Sepharose column produced binding comparable to that of Sindbis virus, which has previously been shown to bind heparin. Replication of these mutants was also adversely affected when they were grown in a CHO cell line that was deficient in heparan sulfate production. These results demonstrate that amino acid 218 of the E2 glycoprotein can be modified to create an heparan sulfate binding site and this modification expands the host range of Ross River virus in cultured cells to cells of avian origin.


Subject(s)
Heparitin Sulfate/metabolism , Ross River virus/pathogenicity , Viral Envelope Proteins/metabolism , Virus Replication , Amino Acid Substitution , Animals , Cell Membrane/metabolism , Cell Membrane/virology , Cells, Cultured , Chick Embryo , Heparin/pharmacology , Heparin Lyase/pharmacology , Point Mutation , Protein Binding/drug effects , Ross River virus/metabolism , Sepharose/analogs & derivatives , Sepharose/metabolism , Viral Envelope Proteins/genetics
15.
J Virol ; 75(6): 2653-9, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11222688

ABSTRACT

Pseudotyped retroviruses have important applications as vectors for gene transfer and gene therapy and as tools for the study of viral glycoprotein function. Recombinant Moloney murine leukemia virus (Mo-MuLV)-based retrovirus particles efficiently incorporate the glycoproteins of the alphavirus Ross River virus (RRV) and utilize them for entry into cells. Stable cell lines that produce the RRV glycoprotein-pseudotyped retroviruses for prolonged periods of time have been constructed. The pseudotyped viruses have a broadened host range, can be concentrated to high titer, and mediate stable transduction of genes into cells. The RRV glycoprotein-pseudotyped retroviruses and the cells that produce them have been employed to demonstrate that RRV glycoprotein-mediated viral entry occurs through endocytosis and that membrane fusion requires acidic pH. Alphavirus glycoprotein-pseudotyped retroviruses have significant advantages as reagents for the study of the biochemistry and prevention of alphavirus entry and as preferred vectors for stable gene transfer and gene therapy protocols.


Subject(s)
Capsid Proteins , Capsid/genetics , Membrane Glycoproteins/genetics , Moloney murine leukemia virus/metabolism , Recombinant Fusion Proteins/metabolism , Ross River virus/genetics , Viral Envelope Proteins/genetics , 3T3 Cells , Animals , Capsid/metabolism , Cell Fusion , Cell Line , Genetic Vectors , Hydrogen-Ion Concentration , Membrane Fusion , Membrane Glycoproteins/metabolism , Mice , Moloney murine leukemia virus/genetics , Moloney murine leukemia virus/physiology , Ross River virus/metabolism , Transduction, Genetic , Viral Envelope Proteins/metabolism
16.
J Virol ; 74(18): 8376-81, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10954537

ABSTRACT

Ross River virus (RRV) is an indigenous Australian arthropod-borne alphavirus responsible for epidemic polyarthritis (EPA), myalgia, and lethargy in humans. Macrophages and monocytes have been associated with human RRV disease, and previous studies have shown that RRV is capable of infecting macrophages via both a natural virus receptor and by Fc receptor-mediated antibody-dependent enhancement (ADE). Similar to other viruses, such as human immunodeficiency virus and dengue virus, ADE infection results in dramatic RRV growth increases for in vitro macrophage cultures. This study demonstrates that RRV could resist lipopolysaccharide (LPS)-induced antiviral activity in macrophage cultures when infection was via the ADE pathway. Investigation of this infection pathway found that RRV was able to suppress the transcription and translation of key antiviral genes (tumor necrosis factor and inducible nitric oxide synthase) in LPS-stimulated macrophages by disrupting the transcription into mRNA of the genes coding for the associated transcription factors IRF-1 and NF-kappaB. The transcription of non-antiviral control genes was not perturbed by RRV-ADE infection, and de novo protein synthesis also was not significantly affected in RRV-ADE infected cells. The ADE pathway of infection allowed RRV to specifically target antiviral genes in macrophages, resulting in unrestricted virus replication. As ADE has been observed for several virus families and associated with disease and adverse vaccination outcomes, these findings may have broad relevance to viral disease formation and antiviral vaccination strategies.


Subject(s)
Alphavirus Infections/metabolism , Antibody-Dependent Enhancement , Gene Expression Regulation, Viral , Macrophages/metabolism , Ross River virus/genetics , Alphavirus Infections/immunology , Animals , Cell Line , Chlorocebus aethiops , DNA-Binding Proteins/metabolism , Flow Cytometry , Humans , Interferon Regulatory Factor-1 , Lipopolysaccharides/pharmacology , Mice , NF-kappa B/metabolism , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Phosphoproteins/metabolism , Ross River virus/metabolism , Ross River virus/pathogenicity , Transcription Factors/metabolism , Tumor Necrosis Factor-alpha/metabolism , Vero Cells , Viral Plaque Assay
17.
J Virol ; 71(4): 2819-29, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9060637

ABSTRACT

Alphaviruses are a well-characterized group of positive-strand RNA viruses. The identification of cis-acting elements in their genomes and their replication strategy have made them useful as vectors for the expression of heterologous genes. In infected cells, the nonstructural proteins, required for replication and transcription of the viral genes, are translated from the genomic RNA; the structural proteins, the capsid protein that interacts with the RNA to form the nucleocapsid and the proteins embedded in the lipid envelope, are translated from a subgenomic mRNA and can be replaced by heterologous genes. Such modified genomes are self-replicating (replicons); they can be introduced into the cells by transfection and can also be packaged into extracellular particles with defective helper (DH) RNAs. The particular DH RNA determines how well it is replicated and to what extent it is packaged. One potential complication of this system has been that recombination between the replicon genome and the DH RNA may occur. The studies described here were designed to prevent recombination by expressing the capsid protein from one DH RNA and the virus membrane proteins from a second helper RNA. Recombination to yield a nonsegmented infectious virus genome would then require several independent crossover events. There is a translational enhancer located downstream of the initiating AUG in the RNA of the capsid gene that had to be conserved in the second helper to achieve high-level expression of the viral glycoproteins. For this reason, we modified the capsid protein gene in two ways: the first was to use the capsid protein gene from a different alphavirus, Ross River virus, and the second was to make deletions in that gene to maintain the translational enhancer in the RNA but to eliminate the positively charged region in the protein that should be essential for the specific and nonspecific interactions with RNA. Transfections with replicon RNA and the deleted chimeric DH RNA as the only helper resulted in the high-level production of particles that were almost completely devoid of RNA. The inclusion of a helper expressing an intact Sindbis virus capsid protein gene led to the production of high levels of packaged replicons. Recombinants were not detected even after several undiluted passages.


Subject(s)
Defective Viruses/genetics , Helper Viruses/genetics , Replicon , Sindbis Virus/genetics , Virus Assembly , Amino Acid Sequence , Animals , Capsid/genetics , Cell Line , Cricetinae , Defective Viruses/physiology , Helper Viruses/physiology , Molecular Sequence Data , RNA, Viral , Recombination, Genetic , Ross River virus/genetics , Ross River virus/metabolism , Sindbis Virus/metabolism , Sindbis Virus/physiology , Sindbis Virus/ultrastructure , Transfection , Viral Envelope Proteins/genetics , Virion/physiology
18.
J Virol ; 70(11): 7900-9, 1996 Nov.
Article in English | MEDLINE | ID: mdl-8892913

ABSTRACT

Sindbis virus and Ross River virus are alphaviruses whose nonstructural proteins share 64% identity and whose structural proteins share 48% identity. Starting from full-length cDNA clones of both viruses, we have generated two reciprocal Sindbis-Ross River chimeric viruses in which the structural and nonstructural regions have been exchanged. These chimeric viruses replicate readily in several cell lines. Both chimeras grow more poorly than do the parental viruses, with the chimera containing Sindbis virus nonstructural proteins and Ross River virus structural proteins growing considerably better in both mosquito and Vero cell lines than the reciprocal chimera does. The reduction in replicative capacity in comparison with the parental viruses appears to result at least in part from a reduction in RNA synthesis, which suggests that the structural proteins or sequence elements within the structural region interact with the nonstructural proteins or sequence elements within the nonstructural region, that these interactions are required for efficient RNA replication, and that these interactions are suboptimal in the chimeras. The chimeras are able to infect mice, but their growth is attenuated. Western equine encephalitis virus, a virus widely distributed throughout the Americas, has been previously shown to have arisen by natural recombination between two distinct alphaviruses, but other naturally occurring recombinant alphaviruses have not been found. The present results suggest that most nonstructural/structural chimeras that might arise by natural recombination will be viable but that interactions between different regions of the genome, some of which were previously known but some of which remain unknown, limit the ability of such recombinants to become established.


Subject(s)
Reassortant Viruses/pathogenicity , Ross River virus/pathogenicity , Sindbis Virus/pathogenicity , Viral Nonstructural Proteins/metabolism , Viral Structural Proteins/metabolism , Animals , Cell Line , Chlorocebus aethiops , Cricetinae , DNA, Complementary , Mice , RNA, Viral/biosynthesis , Reassortant Viruses/genetics , Reassortant Viruses/metabolism , Ross River virus/genetics , Ross River virus/metabolism , Sindbis Virus/genetics , Sindbis Virus/metabolism , Vero Cells , Viral Nonstructural Proteins/genetics , Viral Structural Proteins/genetics
19.
Virology ; 151(2): 222-32, 1986 Jun.
Article in English | MEDLINE | ID: mdl-3010551

ABSTRACT

A mutant of RRV T48 the prototype strain of Ross River virus has been isolated with a 21-nucleotide deletion in the gene coding for the envelope glycoprotein E2. Direct sequencing of the 26 S subgenomic RNA, together with HaeIII and TaqI restriction digest analysis of cDNA to RNAs from cells infected with the mutant virus (RRV dE2) and with RRV T48, were consistent with the deletion being the only major alteration in the mutant genome. The E2 protein of RRV dE2 virions had a higher electrophoretic mobility than that of RRV T48 E2 protein. Neither RRV dE2 nor RRV T48 virions contained more than trace amounts of E3, the small envelope glycoprotein found in Semliki Forest virus. RRV dE2 generated small plaques on Vero cell monolayers; plaque formation was not temperature-sensitive between 32 and 41 degrees. By comparison with RRV T48 the infectivity of RRV dE2 virions was thermolabile at 50 degrees. In BHK cells RRV dE2 grew with similar kinetics to RRV T48. Rates of synthesis of 26 S RNA and 49 S RNA were higher in cells infected with RRV dE2 than in cells infected with RRV T48. Virus-specific protein synthesis and shut-down of host protein synthesis occurred 2-3 hr earlier in RRV dE2-infected cells than in cells infected with RRV T48. Minor differences between the two viruses were observed in the profiles of virus-specific proteins generated in infected cells. In day-old mice RRV dE2 induced less severe symptoms of hind leg paralysis than did RRV T48. A small increase in LD50 and average survival time was observed in RRV dE2-infected mice by comparison with RRV T48 infected mice. Peak titers reached by RRV dE2 in the hind leg muscle, brain, and blood of day-old mice were 3-4 log units less than the titers reached during infection with RRV T48. In week-old mice the differences in virulence between the two strains were magnified: RRV dE2 induced no detectable symptoms even when injected at high doses (8 X 10(6) PFU) whereas the LD50 and average survival time for RRV T48 were unchanged from those in day-old mice. Peak RRV dE2 titers in hind leg muscle, brain, and blood, respectively, were 2, 5, and 5 log units less than the corresponding titers for RRV T48. Peak muscle titers reached by RRV dE2 were similar (approximately 10(8) PFU/g tissue) in day-old mice where lethality was high and in week-old mice where the virus was avirulent.(ABSTRACT TRUNCATED AT 400 WORDS)


Subject(s)
Alphavirus/genetics , Genes, Viral , RNA, Viral/biosynthesis , Ross River virus/genetics , Viral Envelope Proteins/genetics , Viral Proteins/biosynthesis , Animals , Brain/microbiology , Cell Line , Chlorocebus aethiops , Chromosome Deletion , Cricetinae , Female , Hot Temperature , Male , Mice , Muscles/microbiology , Mutation , Ross River virus/growth & development , Ross River virus/isolation & purification , Ross River virus/metabolism , Ross River virus/pathogenicity , Viral Plaque Assay , Virion/physiology , Virulence
20.
Arch Virol ; 63(3-4): 263-74, 1980.
Article in English | MEDLINE | ID: mdl-7356398

ABSTRACT

Polypeptide synthesis was examined in mosquito cells during the establishment of a persistent infection with two alphaviruses, Ross River virus (RRV) and Semliki Forest virus (SFV), and in vertebrate cells cytopathically-infected with the same viruses. In Aedes albopictus cell, RRV reached peak titres at 34--48 hours p.i. At 12 hours 85 per cent of cells assayed as infected by infective centre assay; by 48 hours when persistence was established, virus production was reduced and less than 5 per cent of cells assayed as infected. There was no shut-down of host polypeptide synthesis during infection. Viral polypeptide synthesis was maximal between 10 and 24 hours p.i. The major viral polypeptides labelled were nucleocapsid protein and envelope protein(s). The precursor polypeptide p95 which was prominent in infected BHK cells was not detected in mosquito cells. Similar results were obtained on SFV infection. During the establishment of persistence there was a coordinate decline in the synthesis of RRV polypeptides, reaching undetectable levels by 72 hours p.i. Subculturing persitently-infected cells led to a small increase in viral polypeptide synthesis and virus titre. In contrast, during RRV growth in BHK celos host protein synthesis was severly inhibited and by 9--11 hours p.i. virus-specific polypeptide synthesis represented more than 90 per cent of total protein synthetic activity.


Subject(s)
Arboviruses/growth & development , Ross River virus/growth & development , Semliki forest virus/growth & development , Viral Proteins/biosynthesis , Aedes , Animals , Capsid/biosynthesis , Cell Line , Cricetinae , Protein Biosynthesis , Protein Precursors/biosynthesis , Ross River virus/metabolism , Semliki forest virus/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...