Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 798
Filter
1.
J Virol ; 94(21)2020 10 14.
Article in English | MEDLINE | ID: mdl-32796072

ABSTRACT

Guanylate binding protein 5 (GBP5) belongs to the GTPase subfamily, which is mainly induced by interferon gamma (IFN-γ) and is involved in many important cellular processes, including inflammasome activation and innate immunity against a wide variety of microbial pathogens. However, it is unknown whether GBP5 inhibits respiratory syncytial virus (RSV) infection. In this study, we identified GBP5 as an effector of the anti-RSV activity of IFN-γ and found that in children, the weaker immune response, especially the weaker IFN-γ response and the decreased GBP5 expression, leads to RSV susceptibility. Furthermore, we revealed that GBP5 reduced the cell-associated levels of the RSV small hydrophobic (SH) protein, which was identified as a viroporin. In contrast, overexpression of the SH protein rescued RSV replication in the presence of GBP5. The GBP5-induced decrease in intracellular SH protein levels is because GBP5 promotes the release of the SH protein into the cell culture. Moreover, the GBP5 C583A mutants with changes at the C terminus or the GBP5 ΔC mutant lacking the C-terminal region, which impairs GBP5 localization in the Golgi, could not inhibit RSV infection, whereas the GTPase-defective GBP5 maintained RSV inhibition, suggesting that Golgi localization but not the GTPase activity of GBP5 is required for RSV inhibition. Interestingly, we found that RSV infection or RSV G protein downregulates GBP5 expression by upregulating DZIP3, an E3 ligase, which induces GBP5 degradation through the K48 ubiquitination and proteasomal pathways. Thus, this study reveals a complicated interplay between host restrictive factor GBP5 and RSV infection and provides important information for understanding the pathogenesis of RSV.IMPORTANCE RSV is a highly contagious virus that causes multiple infections in infants within their first year of life. It can also easily cause infection in elderly or immunocompromised individuals, suggesting that individual differences in immunity play an important role in RSV infection. Therefore, exploring the pathogenic mechanisms of RSV and identifying essential genes which inhibit RSV infection are necessary to develop an effective strategy to control RSV infection. Here, we report that the IFN-inducible gene GBP5 potently inhibits RSV replication by reducing the cell-associated levels of the RSV small hydrophobic (SH) protein, which is a viroporin. In contrast, the RSV G protein was shown to upregulate the expression of the DZIP3 protein, an E3 ligase that degrades GBP5 through the proteasomal pathway. Our study provides important information for the understanding of the pathogenic mechanisms of RSV and host immunity as well as the complicated interplay between the virus and host.


Subject(s)
GTP-Binding Proteins/genetics , Host-Pathogen Interactions/genetics , Interferon-gamma/genetics , Respiratory Syncytial Virus Infections/genetics , Respiratory Syncytial Virus, Human/genetics , Retroviridae Proteins, Oncogenic/genetics , Adult , Child , Epithelial Cells/immunology , Epithelial Cells/virology , Female , GTP-Binding Proteins/immunology , Gene Expression Regulation , Golgi Apparatus/immunology , Golgi Apparatus/virology , Host-Pathogen Interactions/immunology , Humans , Immunity, Innate , Inflammasomes/genetics , Inflammasomes/immunology , Interferon-gamma/immunology , Male , Mutation , Proteasome Endopeptidase Complex/immunology , Proteasome Endopeptidase Complex/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/immunology , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/pathology , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus, Human/growth & development , Respiratory Syncytial Virus, Human/immunology , Retroviridae Proteins, Oncogenic/immunology , Signal Transduction , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/immunology , Ubiquitination , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology
2.
Eur J Immunol ; 50(10): 1591-1597, 2020 10.
Article in English | MEDLINE | ID: mdl-32470143

ABSTRACT

Mice bearing CT26 tumors can be cured by administration of L19-mIL12 or F8-mTNF, two antibody fusion proteins which selectively deliver their cytokine payload to the tumor. In both settings, cancer cures crucially depended on CD8+ T cells and the AH1 peptide (derived from the gp70 protein of the murine leukemia virus) acted as the main tumor-rejection antigen, with ∼50% of CD8+ T cells in the neoplastic mass being AH1-specific after therapy. In order to characterize the clonality of the T cell response, its phenotype, and activation status, we isolated CD8+ T cells from tumors and secondary lymphoid organs and submitted them to T cell receptor (TCR) and total mRNA sequencing. We found an extremely diverse repertoire of more than 40 000 unique TCR sequences, but the ten most abundant TCRs accounted for >60% of CD8+ T-cell clones in the tumor. AH1-specific TCRs were consistently found among the most abundant sequences. AH1-specific T cells in the tumor had a tissue-resident memory phenotype. Treatment with L19-mIL12 led to overexpression of IL-12 receptor and of markers of cell activation and proliferation. These data suggest that the antitumor response driven by antibody-cytokine fusions proceeds through an oligoclonal expansion and activation of tumor-infiltrating CD8+ T cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Colon/pathology , Colonic Neoplasms/therapy , Immunotherapy/methods , Leukemia Virus, Murine/genetics , Animals , Antigens, Neoplasm/immunology , Cell Line, Tumor , Colonic Neoplasms/immunology , Disease Models, Animal , Female , Humans , Immunologic Memory , Interleukin-12/therapeutic use , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Peptides/genetics , Peptides/immunology , Receptors, Antigen, T-Cell/genetics , Retroviridae Proteins, Oncogenic/genetics , Retroviridae Proteins, Oncogenic/immunology , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology
3.
Retrovirology ; 16(1): 25, 2019 09 06.
Article in English | MEDLINE | ID: mdl-31492165

ABSTRACT

Of the members of the primate T cell lymphotropic virus (PTLV) family, only the human T-cell leukemia virus type-1 (HTLV-1) causes disease in humans-as the etiological agent of adult T-cell leukemia/lymphoma (ATLL), HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP), and other auto-inflammatory disorders. Despite having significant genomic organizational and structural similarities, the closely related human T-cell lymphotropic virus type-2 (HTLV-2) is considered apathogenic and has been linked with benign lymphoproliferation and mild neurological symptoms in certain infected patients. The silencing of proviral gene expression and maintenance of latency are central for the establishment of persistent infections in vivo. The conserved pX sequences of HTLV-1 and HTLV-2 encode several ancillary factors which have been shown to negatively regulate proviral gene expression, while simultaneously activating host cellular proliferative and pro-survival pathways. In particular, the ORF-II proteins, HTLV-1 p30II and HTLV-2 p28II, suppress Tax-dependent transactivation from the viral promoter-whereas p30II also inhibits PU.1-mediated inflammatory-signaling, differentially augments the expression of p53-regulated metabolic/pro-survival genes, and induces lymphoproliferation which could promote mitotic proviral replication. The ubiquitinated form of the HTLV-1 p13II protein localizes to nuclear speckles and interferes with recruitment of the p300 coactivator by the viral transactivator Tax. Further, the antisense-encoded HTLV-1 HBZ and HTLV-2 APH-2 proteins and mRNAs negatively regulate Tax-dependent proviral gene expression and activate inflammatory signaling associated with enhanced T-cell lymphoproliferation. This review will summarize our current understanding of the pX latency-maintenance factors of HTLV-1 and HTLV-2 and discuss how these products may contribute to the differences in pathogenicity between the human PTLVs.


Subject(s)
Human T-lymphotropic virus 1/genetics , Human T-lymphotropic virus 2/genetics , Transcription Factors/genetics , Viral Regulatory and Accessory Proteins/genetics , Virus Latency , Gene Expression Regulation, Viral , HTLV-I Infections/complications , HTLV-II Infections/virology , Human T-lymphotropic virus 1/pathogenicity , Human T-lymphotropic virus 2/pathogenicity , Humans , Primate T-lymphotropic virus 1/genetics , Primate T-lymphotropic virus 1/pathogenicity , Retroviridae Proteins, Oncogenic/genetics , Retroviridae Proteins, Oncogenic/metabolism
4.
J Virol ; 92(20)2018 10 15.
Article in English | MEDLINE | ID: mdl-30068647

ABSTRACT

J paramyxovirus (JPV) was first isolated from moribund mice with hemorrhagic lung lesions in Australia in 1972. It is a paramyxovirus classified under the newly proposed genus Jeilongvirus JPV has a genome of 18,954 nucleotides, consisting of eight genes in the order 3'-N-P/V/C-M-F-SH-TM-G-L-5'. JPV causes little cytopathic effect (CPE) in tissue culture cells but severe disease in mice. The small hydrophobic (SH) protein is an integral membrane protein encoded by many paramyxoviruses, such as mumps virus (MuV) and respiratory syncytial virus (RSV). However, the function of SH has not been defined in a suitable animal model. In this work, the functions of SH of JPV, MuV, and RSV have been examined by generating recombinant JPV lacking the SH protein (rJPV-ΔSH) or replacing SH of JPV with MuV SH (rJPV-MuVSH) or RSV SH (rJPV-RSVSH). rJPV-ΔSH, rJPV-MuVSH, and rJPV-RSVSH were viable and had no growth defect in tissue culture cells. However, more tumor necrosis factor alpha (TNF-α) was produced during rJPV-ΔSH infection, confirming the role of SH in inhibiting TNF-α production. rJPV-ΔSH induced more apoptosis in tissue culture cells than rJPV, rJPV-MuVSH, and rJPV-RSVSH, suggesting that SH plays a role in blocking apoptosis. Furthermore, rJPV-ΔSH was attenuated in mice compared to rJPV, rJPV-MuVSH, and rJPV-RSVSH, indicating that the SH protein plays an essential role in virulence. The results indicate that the functions of MuV SH and RSV SH are similar to that of JPV SH even though they have no sequence homology.IMPORTANCE Paramyxoviruses are associated with many devastating diseases in animals and humans. J paramyxovirus (JPV) was isolated from moribund mice in Australia in 1972. Newly isolated viruses, such as Beilong virus (BeiPV) and Tailam virus (TlmPV), have genome structures similar to that of JPV. A new paramyxovirus genus, Jeilongvirus, which contains JPV, BeiPV, and TlmPV, has been proposed. Small hydrophobic (SH) protein is present in many paramyxoviruses. Our present study investigates the role of SH protein of JPV in pathogenesis in its natural host. Understanding the pathogenic mechanism of Jeilongvirus is important to control and prevent potential diseases that may emerge from this group of viruses.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Paramyxoviridae Infections/pathology , Paramyxoviridae/growth & development , Retroviridae Proteins, Oncogenic/metabolism , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Virulence Factors/metabolism , Animals , Cell Line , Chlorocebus aethiops , Disease Models, Animal , Gene Deletion , Genetic Complementation Test , Humans , Mice , Microbial Viability , Mumps virus/genetics , Mumps virus/physiology , Paramyxoviridae Infections/virology , Respiratory Syncytial Viruses/genetics , Respiratory Syncytial Viruses/physiology , Retroviridae Proteins, Oncogenic/genetics , Virulence , Virulence Factors/genetics
5.
Virol J ; 15(1): 80, 2018 05 02.
Article in English | MEDLINE | ID: mdl-29716616

ABSTRACT

BACKGROUND: The human T-lymphotropic virus type 1 (HTLV-1) affects 2-5 million people worldwide, and is associated with a number of degenerative and infectious diseases. The Envelope glycoproteins (gp) are highly conserved among the different HTLV-1 isolates, although nucleotide substitutions in the region that codifies these proteins may influence both the infectivity and the replication of the virus. The gp46 gene has functional domains which have been associated with the inhibition of the formation of the syncytium, cell-cell transmission, and the production of antibodies. The present study investigated the genetic stability of the gp46 gene of HTLV-1 in an endemic region of Brazilian Amazonia. METHODS: Index case (IC - a sample of a given family group) carriers of HTLV-1 were investigated in the metropolitan region of Belém (Pará, Brazil) between January 2010 (registered retrospectively) and December 2015. The sequences that codify the gp46 were amplified by PCR, purified and sequenced (MF084788-MF084825). The gene was characterized using bioinformatics and Bayesian Inference. RESULTS: The 40 patients analyzed had a mean age of 45.2 years and 70% presented some type of symptom, with a predominance of pain and sensitivity, dysautonomia, and motor disorders. All patients presented the aA (Transcontinental Cosmopolitan) genotype, with an extremely low mutation rate, which is characteristic of the codifying region (aA - 1.83 × 10-4 mutations per site per year). The gp46 gene had a nucleotide diversity of between 0.00% and 2.0%. Amino acid mutations were present in 66.6% of the samples of individuals with signs/symptoms or diseases associated with HTLV-1 (p = 0.0091). Of the three most frequent mutations, the previously undescribed N93D mutant was invariably associated with symptomatic cases. CONCLUSIONS: The aA HTLV-1 subtype is predominant in the metropolitan region of Belém and presented a high degree of genetic stability in the codifying region. The rare N93D amino acid mutation may be associated with the clinical manifestations of this viral infection. IMPORTANCE: Little is known of the phylogeny of HTLV-1 in the endemic region of Brazilian Amazonia, and few complete gene sequences are available for the gp46 glycoprotein from the local population. The nucleotide sequences of the viral gp46 gene recorded in the present study confirmed the genetic stability of the region, and pointed to a homogeneous viral group, with local geographic characteristics. Further research will be necessary to more fully understand the molecular diversity of this protein, given the potential of this codifying region as a model for an effective HTLV-1 vaccine. The identification of a rare mutation (N93D), present only in symptomatic patients, should also be investigated further as a potential clinical marker. TRIAL REGISTRATION: ISRCTN 12345678, registered 28 September 2014.


Subject(s)
Endemic Diseases , Gene Products, env/genetics , HTLV-I Infections/epidemiology , Human T-lymphotropic virus 1/genetics , Mutation , Pain/epidemiology , Primary Dysautonomias/epidemiology , Retroviridae Proteins, Oncogenic/genetics , Adult , Amino Acid Substitution , Base Sequence , Bayes Theorem , Brazil/epidemiology , Computational Biology , Female , Gene Expression , Genotype , HTLV-I Infections/diagnosis , HTLV-I Infections/physiopathology , HTLV-I Infections/virology , Heterozygote , Human T-lymphotropic virus 1/isolation & purification , Human T-lymphotropic virus 1/pathogenicity , Humans , Male , Middle Aged , Pain/diagnosis , Pain/physiopathology , Pain/virology , Primary Dysautonomias/diagnosis , Primary Dysautonomias/physiopathology , Primary Dysautonomias/virology , Protein Domains , Retrospective Studies , Sequence Analysis, DNA
6.
Biochim Biophys Acta Proteins Proteom ; 1866(4): 541-548, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29458191

ABSTRACT

Entry of human T-cell lymphotropic virus type 1 (HTLV-1) into host cells is mainly mediated by interactions between the viral envelope glycoprotein surface unit (SU) and three host receptors: glucose transporter type 1, heparin/heparan sulfate proteoglycan, and neuropilin-1 (Nrp1). Here, we analyzed the interaction between HTLV-1 SU and Nrp1 using nuclear magnetic resonance and isothermal titration calorimetry. We found that two SU peptides, residues 85-94 and residues 304-312, bound directly to the Nrp1 b1 domain with affinities of 7.4 and 17.7 µM, respectively. The binding modes of both peptides were almost identical to those observed for Tuftsin and vascular endothelial growth factor A binding to the Nrp1 b1 domain. These results suggest that the C-terminal region of HTLV-1 SU contains a novel site for direct binding of virus to the Nrp1 b1 domain. Our biophysical characterization of the SU peptides may help in developing inhibitors of HTLV-1 entry.


Subject(s)
Gene Products, env/chemistry , Human T-lymphotropic virus 1/chemistry , Neuropilin-1/chemistry , Retroviridae Proteins, Oncogenic/chemistry , Binding Sites , Gene Products, env/genetics , Gene Products, env/metabolism , Human T-lymphotropic virus 1/genetics , Human T-lymphotropic virus 1/metabolism , Humans , Neuropilin-1/genetics , Neuropilin-1/metabolism , Protein Binding , Retroviridae Proteins, Oncogenic/genetics , Retroviridae Proteins, Oncogenic/metabolism
7.
Article in English | MEDLINE | ID: mdl-28861397

ABSTRACT

The Human Respiratory Syncytial Virus (hRSV) is a major cause of acute lower respiratory tract infections (ARTIs) and high rates of hospitalizations in children and in the elderly worldwide. Symptoms of hRSV infection include bronchiolitis and pneumonia. The lung pathology observed during hRSV infection is due in part to an exacerbated host immune response, characterized by immune cell infiltration to the lungs. HRSV is an enveloped virus, a member of the Pneumoviridae family, with a non-segmented genome and negative polarity-single RNA that contains 10 genes encoding for 11 proteins. These include the Fusion protein (F), the Glycoprotein (G), and the Small Hydrophobic (SH) protein, which are located on the virus surface. In addition, the Nucleoprotein (N), Phosphoprotein (P) large polymerase protein (L) part of the RNA-dependent RNA polymerase complex, the M2-1 protein as a transcription elongation factor, the M2-2 protein as a regulator of viral transcription and (M) protein all of which locate inside the virion. Apart from the structural proteins, the hRSV genome encodes for the non-structural 1 and 2 proteins (NS1 and NS2). HRSV has developed different strategies to evade the host immunity by means of the function of some of these proteins that work as virulence factors to improve the infection in the lung tissue. Also, hRSV NS-1 and NS-2 proteins have been shown to inhibit the activation of the type I interferon response. Furthermore, the hRSV nucleoprotein has been shown to inhibit the immunological synapsis between the dendritic cells and T cells during infection, resulting in an inefficient T cell activation. Here, we discuss the hRSV virulence factors and the host immunological features raised during infection with this virus.


Subject(s)
Adaptive Immunity , Host-Pathogen Interactions/immunology , Immunity, Innate , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus, Human/immunology , Viral Proteins/immunology , Virulence Factors/immunology , Aged , Child , Dendritic Cells/immunology , Genome, Viral , Glycoproteins/genetics , Humans , Immune Evasion , Immunological Synapses/immunology , Interferon Type I/metabolism , Interferons/immunology , Lung/pathology , Lymphocyte Activation , Nucleoproteins/genetics , Phosphoproteins/genetics , RNA-Dependent RNA Polymerase/genetics , Respiratory Syncytial Virus Infections/pathology , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus, Human/genetics , Respiratory Syncytial Virus, Human/pathogenicity , Respiratory Syncytial Virus, Human/physiology , Respiratory Tract Infections/immunology , Respiratory Tract Infections/virology , Retroviridae Proteins, Oncogenic/genetics , T-Lymphocytes/immunology , Viral Fusion Proteins/genetics , Viral Proteins/genetics , Viral Proteins/metabolism , Viral Proteins/physiology , Viral Structural Proteins/genetics , Viral Structural Proteins/metabolism , Virulence Factors/genetics , Virulence Factors/physiology
8.
J Gen Virol ; 98(7): 1587-1599, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28714847

ABSTRACT

Bovine respiratory syncytial virus (BRSV) is an important cause of respiratory disease in young cattle and is closely related to human RSV (HRSV), which causes severe respiratory disease in infants and the elderly. The RSV genome encodes a small hydrophobic (SH) protein with viroporin activity. Previous studies have shown that recombinant BRSV lacking the SH gene (rBRSVΔSH) is attenuated in the lungs, but not in the upper respiratory tract, of calves and mucosal vaccination with rBRSVΔSH induced long-lasting protective immunity. Attenuation of rBRSVΔSH may be due to the ability of this virus to induce an early innate response as rBRSVΔSH induces higher levels of pro-inflammatory cytokines than wild-type (wt) rBRSV. In this study, we investigated the effects of the BRSV SH protein on NF-κB p65 phosphorylation, a master step in the regulation of pro-inflammatory cytokines. Expression of SH resulted in the inhibition of NF-κB p65 phosphorylation in response to BRSV infection and extracellular lipopolysaccharide, and a reduction in the production of pro-inflammatory cytokines. In contrast, rBRSVΔSH does not inhibit NF-κB p65 phosphorylation in bovine antigen-presenting cells, including monocytes, macrophages and dendritic cells, resulting in increased expression of pro-inflammatory cytokines and increased activation of T cells compared to cells infected with wt BRSV. These findings highlight an important role for the BRSV SH protein in immune modulation.


Subject(s)
Cytokines/metabolism , Dendritic Cells/immunology , Macrophages/immunology , Monocytes/immunology , Respiratory Syncytial Virus, Bovine/metabolism , Retroviridae Proteins, Oncogenic/immunology , Transcription Factor RelA/metabolism , Animals , Cattle , Cattle Diseases/immunology , Cattle Diseases/virology , Cell Line , Dendritic Cells/metabolism , Dendritic Cells/virology , Humans , Lipopolysaccharides/metabolism , Lymphocyte Activation/immunology , Macrophages/metabolism , Macrophages/virology , Mice , Monocytes/metabolism , Monocytes/virology , NF-KappaB Inhibitor alpha/metabolism , Phosphorylation , RAW 264.7 Cells , Respiratory Syncytial Virus, Bovine/genetics , Respiratory Syncytial Virus, Bovine/immunology , Retroviridae Proteins, Oncogenic/genetics , Retroviridae Proteins, Oncogenic/metabolism , T-Lymphocytes/immunology
9.
AIDS ; 30(16): 2427-2438, 2016 10 23.
Article in English | MEDLINE | ID: mdl-27428745

ABSTRACT

Vaccination with SIVmac239Δnef provides robust protection against subsequent challenge with wild-type simian immunodeficiency virus (SIV), but safety issues have precluded designing an HIV-1 vaccine based on a live-attenuated virus concept. Safe immunogens and adjuvants that could reproduce identified immune correlates of SIVmac239Δnef protection therefore offer an alternative path for development of an HIV vaccine. Here we describe SIV envelope trimeric gp41 (gp41t) immunogens based on a protective correlate of antibodies to gp41t concentrated on the path of virus entry by the neonatal Fc receptor (FcRn) in cervical vaginal epithelium. We developed a gp41t immunogen-monophosphoryl lipid A adjuvant liposomal nanoparticle for intramuscular (i.m.) immunization and a gp41t-Fc immunogen for intranasal immunization for pilot studies in mice, rabbits, and rhesus macaques. Repeated immunizations to mimic persistent antigen exposure in infection elicited gp41t antibodies in rhesus macaques that were detectable in FcRn+ cervical vaginal epithelium, thus recapitulating one key feature of SIVmac239Δnef vaccinated and protected animals. Although this strategy did not reproduce the system of local production of antibody in SIVmac239Δnef-vaccinated animals, passive immunization experiments supported the concept that sufficiently high levels of antibody can be concentrated by the FcRn at mucosal frontlines, thus setting the stage for assessing protection against vaginal challenge by gp41t immunization.


Subject(s)
Antibodies, Viral/immunology , Gene Products, env/immunology , Retroviridae Proteins, Oncogenic/immunology , SAIDS Vaccines/immunology , Simian Immunodeficiency Virus/immunology , Viral Fusion Proteins/immunology , Adjuvants, Immunologic/administration & dosage , Administration, Intranasal , Animals , Epithelium/immunology , Gene Products, env/genetics , Histocompatibility Antigens Class I/immunology , Immunity, Mucosal , Injections, Intramuscular , Lipid A/administration & dosage , Macaca mulatta , Mice, Inbred BALB C , Rabbits , Receptors, Fc/immunology , Retroviridae Proteins, Oncogenic/genetics , SAIDS Vaccines/administration & dosage , SAIDS Vaccines/genetics , Simian Immunodeficiency Virus/genetics , Viral Fusion Proteins/genetics
10.
Microb Pathog ; 97: 38-44, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27235335

ABSTRACT

To prevent the spread of HTLV-I (Human T-lymphotropic virus type 1), a safe and effective vaccine is required. To increase immune responses against the peptide antigens can be potentiated with polymer-based nanoparticles, like chitosan (CHT) and trimethylchitosan (TMC), as delivery system/adjuvant. CHT and TMC nanoparticles loaded with recombinant proteins (env23 & env13) of gp46 were prepared by direct coating of antigens with positively charged polymers. The size of CHT and TMC nanoparticles (NPs) loaded with each antigen was about 400 nm. The physical stability of NPs was followed for 4 weeks. Both formulations showed to be stable for about 15 days. The immunogenicity of NPs loaded with antigens was studied after nasal and subcutaneous immunization in mice. Three immunizations (7.5 µg antigen) were performed with 2 weeks intervals. Two weeks after the last booster dose, sera IgG subtypes were measured. After subcutaneous administration, for both nanoparticulate antigens, serum IgG1 and IgGtotal levels were higher than antigen solution (P < 0.001). After nasal administration, for env23, IgG2a levels and IgG2a/IgG1 ratio was significantly higher than groups with subcutaneous administration (P < 0.001). Both nanoparticles showed good immunoadjuvant potential. Env23 antigen was a better candidate for vaccination against HTLV-I, as it induced higher cellular immune responses, compared with env13.


Subject(s)
Antigens, Viral/immunology , Chitosan/administration & dosage , Gene Products, env/immunology , Human T-lymphotropic virus 1/immunology , Nanoparticles/administration & dosage , Retroviridae Proteins, Oncogenic/immunology , Viral Vaccines/immunology , Adjuvants, Immunologic/administration & dosage , Administration, Intranasal , Animals , Antibodies, Viral/blood , Antigens, Viral/genetics , Drug Carriers/administration & dosage , Drug Stability , Gene Products, env/genetics , Human T-lymphotropic virus 1/genetics , Immunoglobulin G/blood , Injections, Subcutaneous , Male , Mice, Inbred BALB C , Retroviridae Proteins, Oncogenic/genetics , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/genetics
11.
Viruses ; 8(2)2016 Feb 03.
Article in English | MEDLINE | ID: mdl-26848684

ABSTRACT

Although the number of human T-cell leukemia virus type-I (HTLV-I)-infected individuals in the world has been estimated at over 10 million, no prophylaxis vaccines against HTLV-I infection are available. In this study, we took a new approach for establishing the basis of protective vaccines against HTLV-I. We show here the potential of a passively administered HTLV-I neutralizing monoclonal antibody of rat origin (LAT-27) that recognizes epitopes consisting of the HTLV-I gp46 amino acids 191-196. LAT-27 completely blocked HTLV-I infection in vitro at a minimum concentration of 5 µg/mL. Neonatal rats born to mother rats pre-infused with LAT-27 were shown to have acquired a large quantity of LAT-27, and these newborns showed complete resistance against intraperitoneal infection with HTLV-I. On the other hand, when humanized immunodeficient mice were pre-infused intravenously with humanized LAT-27 (hu-LAT-27), all the mice completely resisted HTLV-I infection. These results indicate that hu-LAT-27 may have a potential for passive immunization against both horizontal and mother-to-child vertical infection with HTLV-I.


Subject(s)
Antibodies, Monoclonal/immunology , Disease Transmission, Infectious/prevention & control , Gene Products, env/immunology , HTLV-I Infections/prevention & control , HTLV-I Infections/transmission , Human T-lymphotropic virus 1/physiology , Infectious Disease Transmission, Vertical/prevention & control , Retroviridae Proteins, Oncogenic/immunology , Adult , Animals , Antibodies, Monoclonal/administration & dosage , Female , Gene Products, env/genetics , HTLV-I Infections/immunology , HTLV-I Infections/virology , Human T-lymphotropic virus 1/genetics , Human T-lymphotropic virus 1/immunology , Humans , Immunization, Passive , Infant , Male , Mice , Rats , Rats, Inbred F344 , Rats, Sprague-Dawley , Retroviridae Proteins, Oncogenic/genetics
12.
J Virol ; 89(17): 8974-81, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26085154

ABSTRACT

UNLABELLED: The small hydrophobic (SH) gene of respiratory syncytial virus (RSV), a major cause of infant hospitalization, encodes a viroporin of unknown function. SH gene knockout virus (RSV ΔSH) is partially attenuated in vivo, but not in vitro, suggesting that the SH protein may have an immunomodulatory role. RSV ΔSH has been tested as a live attenuated vaccine in humans and cattle, and here we demonstrate that it protected against viral rechallenge in mice. We compared the immune response to infection with RSV wild type and RSV ΔSH in vivo using BALB/c mice and in vitro using epithelial cells, neutrophils, and macrophages. Strikingly, the interleukin-1ß (IL-1ß) response to RSV ΔSH infection was greater than to wild-type RSV, in spite of a decreased viral load, and when IL-1ß was blocked in vivo, the viral load returned to wild-type levels. A significantly greater IL-1ß response to RSV ΔSH was also detected in vitro, with higher-magnitude responses in neutrophils and macrophages than in epithelial cells. Depleting macrophages (with clodronate liposome) and neutrophils (with anti-Ly6G/1A8) demonstrated the contribution of these cells to the IL-1ß response in vivo, the first demonstration of neutrophilic IL-1ß production in response to viral lung infection. In this study, we describe an increased IL-1ß response to RSV ΔSH, which may explain the attenuation in vivo and supports targeting the SH gene in live attenuated vaccines. IMPORTANCE: There is a pressing need for a vaccine for respiratory syncytial virus (RSV). A number of live attenuated RSV vaccine strains have been developed in which the small hydrophobic (SH) gene has been deleted, even though the function of the SH protein is unknown. The structure of the SH protein has recently been solved, showing it is a pore-forming protein (viroporin). Here, we demonstrate that the IL-1ß response to RSV ΔSH is greater in spite of a lower viral load, which contributes to the attenuation in vivo. This potentially suggests a novel method by which viruses can evade the host response. As all Pneumovirinae and some Paramyxovirinae carry similar SH genes, this new understanding may also enable the development of live attenuated vaccines for both RSV and other members of the Paramyxoviridae.


Subject(s)
Interleukin-1beta/immunology , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Viruses/genetics , Respiratory Syncytial Viruses/immunology , Retroviridae Proteins, Oncogenic/genetics , Animals , Cell Line , Epithelial Cells/immunology , Epithelial Cells/virology , Female , Gene Deletion , Gene Knockout Techniques , Humans , Interleukin-1beta/biosynthesis , Macrophages/immunology , Macrophages/virology , Mice , Mice, Inbred BALB C , Neutrophils/immunology , Neutrophils/virology , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus Vaccines/immunology , Respiratory Syncytial Viruses/growth & development , Vaccination , Vaccines, Attenuated/immunology , Viral Load/immunology
13.
Viruses ; 7(6): 2858-83, 2015 Jun 04.
Article in English | MEDLINE | ID: mdl-26053927

ABSTRACT

Viroporins are members of a rapidly growing family of channel-forming small polypeptides found in viruses. The present review will be focused on recent structural and protein-protein interaction information involving two viroporins found in enveloped viruses that target the respiratory tract; (i) the envelope protein in coronaviruses and (ii) the small hydrophobic protein in paramyxoviruses. Deletion of these two viroporins leads to viral attenuation in vivo, whereas data from cell culture shows involvement in the regulation of stress and inflammation. The channel activity and structure of some representative members of these viroporins have been recently characterized in some detail. In addition, searches for protein-protein interactions using yeast-two hybrid techniques have shed light on possible functional roles for their exposed cytoplasmic domains. A deeper analysis of these interactions should not only provide a more complete overview of the multiple functions of these viroporins, but also suggest novel strategies that target protein-protein interactions as much needed antivirals. These should complement current efforts to block viroporin channel activity.


Subject(s)
Coronavirus/physiology , Paramyxovirinae/physiology , Porins/metabolism , Retroviridae Proteins, Oncogenic/metabolism , Viral Envelope Proteins/metabolism , Animals , Antiviral Agents/isolation & purification , Antiviral Agents/pharmacology , Gene Deletion , Humans , Protein Binding , Protein Conformation , Protein Interaction Mapping , Retroviridae Proteins, Oncogenic/antagonists & inhibitors , Retroviridae Proteins, Oncogenic/genetics , Two-Hybrid System Techniques , Viral Envelope Proteins/antagonists & inhibitors , Viral Envelope Proteins/genetics
14.
J Vet Diagn Invest ; 26(4): 496-506, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24842287

ABSTRACT

A multiplex amplification refractory mutation system reverse transcription polymerase chain reaction (ARMS RT-PCR) was developed for the differential diagnosis of Feline leukemia virus (FeLV) vaccine and wild-type strains based on a point mutation between the vaccine strain (S) and the wild-type strain (T) located in the p27 gene. This system was further upgraded to obtain a real-time ARMS RT-PCR (ARMS qRT-PCR) with a high-resolution melt analysis (HRMA) platform. The genotyping of various strains of FeLV was determined by comparing the HRMA curves with the defined wild-type FeLV (strain TW1), and the results were expressed as a percentage confidence. The detection limits of ARMS RT-PCR and ARMS qRT-PCR combined with HRMA were 100 and 1 copies of transcribed FeLV RNA per 0.5 ml of sample, respectively. No false-positive results were obtained with 6 unrelated pathogens and 1 feline cell line. Twelve FeLV Taiwan strains were correctly identified using ARMS qRT-PCR combined with HRMA. The genotypes of the strains matched the defined FeLV wild-type strain genotype with at least 91.17% confidence. A higher degree of sequence polymorphism was found throughout the p27 gene compared with the long terminal repeat region. In conclusion, the current study describes the phylogenetic relationship of the FeLV Taiwan strains and demonstrates that the developed ARMS RT-PCR assay is able to be used to detect the replication of a vaccine strain that has not been properly inactivated, thus acting as a safety check for the quality of FeLV vaccines.


Subject(s)
Cat Diseases/virology , Leukemia Virus, Feline/classification , Multiplex Polymerase Chain Reaction/veterinary , Reverse Transcriptase Polymerase Chain Reaction/veterinary , Animals , Cats , Leukemia Virus, Feline/genetics , Multiplex Polymerase Chain Reaction/methods , Point Mutation , Retroviridae Proteins, Oncogenic/genetics , Reverse Transcriptase Polymerase Chain Reaction/methods , Sequence Analysis, RNA , Terminal Repeat Sequences/genetics , Viral Proteins/genetics , Viral Vaccines/genetics
15.
J Gen Virol ; 95(Pt 6): 1244-1254, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24700100

ABSTRACT

Bovine respiratory syncytial virus (BRSV) causes inflammation and obstruction of the small airways, leading to severe respiratory disease in young calves. The virus is closely related to human (H)RSV, a major cause of bronchiolitis and pneumonia in young children. The ability to manipulate the genome of RSV has provided opportunities for the development of stable, live attenuated RSV vaccines. The role of the SH protein in the pathogenesis of BRSV was evaluated in vitro and in vivo using a recombinant (r)BRSV in which the SH gene had been deleted. Infection of bovine epithelial cells and monocytes with rBRSVΔSH, in vitro, resulted in an increase in apoptosis, and higher levels of TNF-α and IL-1ß compared with cells infected with parental, wild-type (WT) rBRSV. Although replication of rBRSVΔSH and WT rBRSV, in vitro, were similar, the replication of rBRSVΔSH was moderately reduced in the lower, but not the upper, respiratory tract of experimentally infected calves. Despite the greater ability of rBRSVΔSH to induce pro-inflammatory cytokines, in vitro, the pulmonary inflammatory response in rBRSVΔSH-infected calves was significantly reduced compared with that in calves inoculated with WT rBRSV, 6 days previously. Virus lacking SH appeared to be as immunogenic and effective in inducing resistance to virulent virus challenge, 6 months later, as the parental rBRSV. These findings suggest that rBRSVΔSH may be an ideal live attenuated virus vaccine candidate, combining safety with a high level of immunogenicity.


Subject(s)
Cytokines/biosynthesis , Genes, Viral , Respiratory Syncytial Virus, Bovine/genetics , Respiratory Syncytial Virus, Bovine/immunology , Animals , Apoptosis , Cattle , Cattle Diseases/immunology , Cattle Diseases/prevention & control , Cattle Diseases/virology , Gene Deletion , Humans , Immunity, Mucosal , Inflammation Mediators/metabolism , Interleukin-1beta/biosynthesis , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Infections/veterinary , Respiratory Syncytial Virus Vaccines/genetics , Respiratory Syncytial Virus Vaccines/immunology , Respiratory Syncytial Virus, Bovine/pathogenicity , Respiratory Syncytial Virus, Human/immunology , Respiratory Syncytial Virus, Human/pathogenicity , Respiratory System/virology , Retroviridae Proteins, Oncogenic/genetics , Retroviridae Proteins, Oncogenic/immunology , Tumor Necrosis Factor-alpha/biosynthesis , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Virulence/genetics , Virulence/immunology
16.
J Virol ; 88(11): 6423-33, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24672047

ABSTRACT

UNLABELLED: Human metapneumovirus (HMPV) encodes three glycoproteins: the glycoprotein, which plays a role in glycosaminoglycan binding, the fusion (F) protein, which is necessary and sufficient for both viral binding to the target cell and fusion between the cellular plasma membrane and the viral membrane, and the small hydrophobic (SH) protein, whose function is unclear. The SH protein of the closely related respiratory syncytial virus has been suggested to function as a viroporin, as it forms oligomeric structures consistent with a pore and alters membrane permeability. Our analysis indicates that both the full-length HMPV SH protein and the isolated SH protein transmembrane domain can associate into higher-order oligomers. In addition, HMPV SH expression resulted in increases in permeability to hygromycin B and alteration of subcellular localization of a fluorescent dye, indicating that SH affects membrane permeability. These results suggest that the HMPV SH protein has several characteristics consistent with a putative viroporin. Interestingly, we also report that expression of the HMPV SH protein can significantly decrease HMPV F protein-promoted membrane fusion activity, with the SH extracellular domain and transmembrane domain playing a key role in this inhibition. These results suggest that the HMPV SH protein could regulate both membrane permeability and fusion protein function during viral infection. IMPORTANCE: Human metapneumovirus (HMPV), first identified in 2001, is a causative agent of severe respiratory tract disease worldwide. The small hydrophobic (SH) protein is one of three glycoproteins encoded by all strains of HMPV, but the function of the HMPV SH protein is unknown. We have determined that the HMPV SH protein can alter the permeability of cellular membranes, suggesting that HMPV SH is a member of a class of proteins termed viroporins, which modulate membrane permeability to facilitate critical steps in a viral life cycle. We also demonstrated that HMPV SH can inhibit the membrane fusion function of the HMPV fusion protein. This work suggests that the HMPV SH protein has several functions, though the steps in the HMPV life cycle impacted by these functions remain to be clarified.


Subject(s)
Cell Membrane/metabolism , Metapneumovirus/genetics , Retroviridae Proteins, Oncogenic/metabolism , Viral Regulatory and Accessory Proteins/metabolism , Animals , COS Cells , Chlorocebus aethiops , Giant Cells/physiology , Giant Cells/virology , Humans , Hygromycin B , Microscopy, Confocal , Permeability , Plasmids/genetics , Retroviridae Proteins, Oncogenic/genetics , Ultracentrifugation , Vero Cells , Viral Regulatory and Accessory Proteins/genetics , Virus Internalization
17.
J Virol ; 88(2): 992-1001, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24198407

ABSTRACT

We previously delineated a highly conserved immunosuppressive (IS) domain within murine and primate retroviral envelope proteins that is critical for virus propagation in vivo. The envelope-mediated immunosuppression was assessed by the ability of the proteins, when expressed by allogeneic tumor cells normally rejected by engrafted mice, to allow these cells to escape, at least transiently, immune rejection. Using this approach, we identified key residues whose mutation (i) specifically abolishes immunosuppressive activity without affecting the "mechanical" function of the envelope protein and (ii) significantly enhances humoral and cellular immune responses elicited against the virus. The objective of this work was to study the immunosuppressive activity of the envelope protein (p15E) of feline leukemia virus (FeLV) and evaluate the effect of its abolition on the efficacy of a vaccine against FeLV. Here we demonstrate that the FeLV envelope protein is immunosuppressive in vivo and that this immunosuppressive activity can be "switched off" by targeted mutation of a specific amino acid. As a result of the introduction of the mutated envelope sequence into a previously well characterized canarypox virus-vectored vaccine (ALVAC-FeLV), the frequency of vaccine-induced FeLV-specific gamma interferon (IFN-γ)-producing cells was increased, whereas conversely, the frequency of vaccine-induced FeLV-specific interleukin-10 (IL-10)-producing cells was reduced. This shift in the IFN-γ/IL-10 response was associated with a higher efficacy of ALVAC-FeLV against FeLV infection. This study demonstrates that FeLV p15E is immunosuppressive in vivo, that the immunosuppressive domain of p15E can modulate the FeLV-specific immune response, and that the efficacy of FeLV vaccines can be enhanced by inhibiting the immunosuppressive activity of the IS domain through an appropriate mutation.


Subject(s)
Canarypox virus/genetics , Gene Products, env/chemistry , Gene Products, env/immunology , Immunosuppressive Agents/chemistry , Leukemia Virus, Feline/genetics , Leukemia, Feline/immunology , Mutation, Missense , Retroviridae Proteins, Oncogenic/genetics , Viral Vaccines/genetics , Animals , Canarypox virus/metabolism , Cats , Female , Gene Products, env/administration & dosage , Gene Products, env/genetics , Genetic Vectors/genetics , Genetic Vectors/metabolism , Immunosuppressive Agents/administration & dosage , Immunosuppressive Agents/immunology , Interferons/genetics , Interferons/immunology , Interleukin-10/genetics , Interleukin-10/immunology , Leukemia Virus, Feline/chemistry , Leukemia Virus, Feline/immunology , Leukemia, Feline/prevention & control , Leukemia, Feline/virology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Protein Structure, Tertiary , Retroviridae Proteins, Oncogenic/administration & dosage , Retroviridae Proteins, Oncogenic/chemistry , Retroviridae Proteins, Oncogenic/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/chemistry , Viral Vaccines/immunology
18.
J Med Virol ; 86(4): 547-57, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24009128

ABSTRACT

The SH glycoprotein of human metapneumovirus (HMPV) is twice the size of that of human respiratory syncytial virus and possesses a large, hydrophilic luminal domain. The glycoprotein is located on the surface of the virion and of virus infected cells and, if immunogenic, might be expected to play a role in anti-viral immunity. Initial attempts to study anti-SH antibody immunogenicity were thwarted by the instability of the SH gene on passage both in human bronchial epithelial cells and in mice. Repeated passage of virus isolates in human bronchial epithelial cells in culture resulted in the appearance and eventual predominance of HMPV mutants lacking all or most of the luminal domain of SH coincidental with the loss of productive infection in mouse lungs. Where infection was established in mice with an early cell culture passage, the virus recovered from mouse lung differed markedly from the inoculum, carrying 19 coding mutations in the SH luminal domain. Immunization of mice with a mutant virus variant expressing only 14 amino acids of the luminal domain of SH induced a cross-reactive antibody response to both the F glycoprotein and the SH glycoprotein but a largely sub-group specific response to the G glycoprotein. Similar patterns of response were achieved by immunization with individual HMPV glycoproteins expressed from recombinant vaccinia viruses. Recombinant truncated SH glycoprotein induced sub-group cross-reactive antibodies capable of neutralizing wild-type virus. Recombinant F glycoprotein also induced cross-reactive neutralizing antibodies whilst recombinant G glycoprotein induced largely strain-specific, non-neutralizing antibodies.


Subject(s)
Antibodies, Viral/immunology , Glycoproteins/immunology , Metapneumovirus/immunology , Paramyxoviridae Infections/immunology , Viral Proteins/immunology , Animals , Antibodies, Neutralizing/immunology , Base Sequence , Cell Line , Genetic Variation , Glycoproteins/genetics , HeLa Cells , Humans , Immunization , Mice , Neutralization Tests , Retroviridae Proteins, Oncogenic/genetics , Retroviridae Proteins, Oncogenic/immunology , Sequence Analysis, RNA , Viral Proteins/genetics , Viral Vaccines/immunology
19.
J Virol ; 87(16): 9344-52, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23785214

ABSTRACT

Human T lymphotropic virus type 1 (HTLV-1) mainly causes adult T cell leukemia and predominantly immortalizes/transforms CD4(+) T cells in culture. HTLV-2 is aleukemic and predominantly immortalizes/transforms CD8(+) T cells in culture. We have shown previously that the viral envelope is the genetic determinant of the differential T cell tropism in culture. The surface component (SU) of the HTLV-1 envelope is responsible for binding to the cellular receptors for entry. Here, we dissect the HTLV-1 SU further to identify key domains that are involved in determining the immortalization tropism. We generated HTLV-1 envelope recombinant virus containing the HTLV-2 SU domain. HTLV-1/SU2 was capable of infecting and immortalizing freshly isolated peripheral blood mononuclear cells in culture. HTLV-1/SU2 shifted the CD4(+) T cell immortalization tropism of wild-type HTLV-1 (wtHTLV-1) to a CD8(+) T cell preference. Furthermore, a single amino acid substitution, N195D, in HTLV-1 SU (Ach.195) resulted in a shift to a CD8(+) T cell immortalization tropism preference. Longitudinal phenotyping analyses of the in vitro transformation process revealed that CD4(+) T cells emerged as the predominant population by week 5 in wtHTLV-1 cultures, while CD8(+) T cells emerged as the predominant population by weeks 4 and 7 in wtHTLV-2 and Ach.195 cultures, respectively. Our results indicate that SU domain independently influences the preferential T cell immortalization tropism irrespective of the envelope counterpart transmembrane (TM) domain. We further showed that asparagine at position 195 in HTLV-1 SU is involved in determining this CD4(+) T cell immortalization tropism. The slower emergence of the CD8(+) T cell predominance in Ach.195-infected cultures suggests that other residues/domains contribute to this tropism preference.


Subject(s)
CD4-Positive T-Lymphocytes/virology , Cell Transformation, Viral , Gene Products, env/metabolism , Human T-lymphotropic virus 1/physiology , Human T-lymphotropic virus 1/pathogenicity , Retroviridae Proteins, Oncogenic/metabolism , Viral Tropism , Virulence Factors/metabolism , Amino Acid Substitution , Cells, Cultured , DNA Mutational Analysis , Gene Products, env/genetics , Human T-lymphotropic virus 1/genetics , Humans , Mutant Proteins/genetics , Mutant Proteins/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Retroviridae Proteins, Oncogenic/genetics
20.
Avian Pathol ; 42(3): 283-9, 2013.
Article in English | MEDLINE | ID: mdl-23650927

ABSTRACT

In recent years, special attention has been paid to real-time polymerase chain reaction (PCR) for avian metapneumovirus (AMPV) diagnosis, due to its numerous advantages over classical PCR. A new multiplex quantitative real-time reverse transcription-PCR (qRT-PCR) with molecular beacon probe assay, designed to target the SH gene, was developed. The test was evaluated in terms of specificity, sensitivity and repeatability, and compared with conventional RT nested-PCR based on the G gene. All of the AMPV subtype A and B strains tested were amplified and specifically detected while no amplification occurred with other non-target bird respiratory pathogens. The detection limit of the assay was 10(-0.41) median infectious dose/ml and 10(1.15) median infectious dose/ml when the AMPV-B strain IT/Ty/B/Vr240/87 and the AMPV-A strain IT/Ty/A/259-01/03 were used, respectively, as templates. In all cases, the amplification efficiency was approximately 2 and the error values were <0.2. Standard curves, generated either using the serial dilution of an RNA suspension or RNA extracted from the serial dilution of titrated viral suspensions as templates, exhibited good linearity (R (2)>0.9375) between crossing point values and virus quantities, making the assay herein designed reliable for quantification. When the newly developed qRT-PCR was compared with a conventional RT nested-PCR, it showed greater sensitivity with RNA extracted from both positive controls and from experimentally infected birds. This assay can be effectively used for the detection, identification, differentiation and quantitation of AMPV subtype A or subtype B to assist in disease diagnosis and to carry out rapid surveillance with high levels of sensitivity and specificity.


Subject(s)
Metapneumovirus/genetics , Real-Time Polymerase Chain Reaction/veterinary , Retroviridae Proteins, Oncogenic/genetics , Reverse Transcriptase Polymerase Chain Reaction/veterinary , DNA Primers , GTP-Binding Proteins/genetics , Real-Time Polymerase Chain Reaction/methods , Reproducibility of Results , Reverse Transcriptase Polymerase Chain Reaction/methods , Sensitivity and Specificity , Species Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...