Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
1.
J Virol ; 98(3): e0112923, 2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38305155

ABSTRACT

The global circulation of clade 2.3.4.4b H5Ny highly pathogenic avian influenza viruses (HPAIVs) in poultry and wild birds, increasing mammal infections, continues to pose a public health threat and may even form a pandemic. An efficacious vaccine against H5Ny HPAIVs is crucial for emergency use and pandemic preparedness. In this study, we developed a parainfluenza virus 5 (PIV5)-based vaccine candidate expressing hemagglutinin (HA) protein of clade 2.3.4.4b H5 HPAIV, termed rPIV5-H5, and evaluated its safety and efficacy in mice and ferrets. Our results demonstrated that intranasal immunization with a single dose of rPIV5-H5 could stimulate H5-specific antibody responses, moreover, a prime-boost regimen using rPIV5-H5 stimulated robust humoral, cellular, and mucosal immune responses in mice. Challenge study showed that rPIV5-H5 prime-boost regimen provided sterile immunity against lethal clade 2.3.4.4b H5N1 virus infection in mice and ferrets. Notably, rPIV5-H5 prime-boost regimen provided protection in mice against challenge with lethal doses of heterologous clades 2.2, 2.3.2, and 2.3.4 H5N1, and clade 2.3.4.4h H5N6 viruses. These results revealed that rPIV5-H5 can elicit protective immunity against a diverse clade of highly pathogenic H5Ny virus infection in mammals, highlighting the potential of rPIV5-H5 as a pan-H5 influenza vaccine candidate for emergency use.IMPORTANCEClade 2.3.4.4b H5Ny highly pathogenic avian influenza viruses (HPAIVs) have been widely circulating in wild birds and domestic poultry all over the world, leading to infections in mammals, including humans. Here, we developed a recombinant PIV5-vectored vaccine candidate expressing the HA protein of clade 2.3.4.4b H5 virus. Intranasal immunization with rPIV5-H5 in mice induced airway mucosal IgA responses, high levels of antibodies, and robust T-cell responses. Importantly, rPIV5-H5 conferred complete protection in mice and ferrets against clade 2.3.4.4b H5N1 virus challenge, the protective immunity was extended against heterologous H5Ny viruses. Taken together, our data demonstrate that rPIV5-H5 is a promising vaccine candidate against diverse H5Ny influenza viruses in mammals.


Subject(s)
Influenza A Virus, H5N1 Subtype , Influenza A Virus, H5N6 Subtype , Influenza Vaccines , Orthomyxoviridae Infections , Parainfluenza Virus 5 , Animals , Humans , Mice , Ferrets/immunology , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Immunity, Cellular , Immunity, Humoral , Immunity, Mucosal , Influenza A Virus, H5N1 Subtype/chemistry , Influenza A Virus, H5N1 Subtype/classification , Influenza A Virus, H5N1 Subtype/genetics , Influenza A Virus, H5N1 Subtype/immunology , Influenza A Virus, H5N6 Subtype/chemistry , Influenza A Virus, H5N6 Subtype/classification , Influenza A Virus, H5N6 Subtype/genetics , Influenza A Virus, H5N6 Subtype/immunology , Influenza in Birds/immunology , Influenza in Birds/prevention & control , Influenza in Birds/transmission , Influenza in Birds/virology , Influenza Vaccines/administration & dosage , Influenza Vaccines/adverse effects , Influenza Vaccines/genetics , Influenza Vaccines/immunology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/transmission , Orthomyxoviridae Infections/virology , Pandemic Preparedness/methods , Parainfluenza Virus 5/genetics , Parainfluenza Virus 5/immunology , Parainfluenza Virus 5/metabolism , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/adverse effects , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Administration, Intranasal , Poultry/virology , Immunoglobulin A/immunology , T-Lymphocytes/immunology
2.
J Gen Virol ; 104(11)2023 11.
Article in English | MEDLINE | ID: mdl-37962188

ABSTRACT

Parainfluenza virus type 5 (PIV5) can either have a persistent or a lytic phenotype in cultured cells. We have previously shown that the phenotype is determined by the phosphorylation status of the phosphoprotein (P). Single amino acid substitutions at critical residues, including a serine-to-phenylalanine substitution at position 157 on P, result in a switch between persistent and lytic phenotypes. Here, using PIV5 vectors expressing either mCherry or GFP with persistent or lytic phenotypes, we show that in co-infections the persistent phenotype is dominant. Thus, in contrast to the cell death observed with cells infected solely with the lytic variant, in co-infected cells persistence is immediately established and both lytic and persistent genotypes persist. Furthermore, 10-20 % of virus released from dually infected cells contains both genotypes, indicating that PIV5 particles can package more than one genome. Co-infected cells continue to maintain both genotypes/phenotypes during cell passage, as do individual colonies of cells derived from a culture of persistently infected cells. A refinement of our model on how the dynamics of virus selection may occur in vivo is presented.


Subject(s)
Coinfection , Parainfluenza Virus 5 , Paramyxovirinae , Respirovirus Infections , Humans , Parainfluenza Virus 5/genetics , Phenotype
3.
J Med Virol ; 95(3): e28622, 2023 03.
Article in English | MEDLINE | ID: mdl-36846910

ABSTRACT

Parainfluenza virus 5 (PIV5) is a negative-sense, single-stranded RNA virus that can infect humans and many species of animals. Infection in these reservoir hosts is generally asymptomatic and has few safety concerns. Emerging evidence has shown that PIV5 is a promising vector for developing vaccines against human infectious diseases caused by coronaviruses, influenza, respiratory syncytial virus, rabies, HIV, or bacteria. In this review, we summarize recent progress and highlight the advantages and strategies of PIV5 as a vaccine vector to improve future vaccine design and application for clinical trials.


Subject(s)
Influenza Vaccines , Influenza, Human , Parainfluenza Virus 5 , Rabies Vaccines , Respiratory Syncytial Virus, Human , Animals , Humans , Parainfluenza Virus 5/genetics , Respiratory Syncytial Virus, Human/genetics , Parainfluenza Virus 3, Human
4.
Transbound Emerg Dis ; 69(6): 3300-3316, 2022 Nov.
Article in English | MEDLINE | ID: mdl-35964328

ABSTRACT

The number of parainfluenza virus 5 (PIV5) infection cases has increased worldwide over the past six decades; however, factors underlying this increase remain unclear. PIV5 has been emerging or re-emerging in humans and animal species. To date, no information is yet available regarding PIV5 infection in arthropod ticks. Here, we successfully isolated tick-derived PIV5 from the Ixodes persulcatus species designated as HLJ/Tick/2019 in Heilongjiang, China. Phylogenetic analysis revealed that the tick-derived PIV5 is closely related to subclade 2.2.6, which has become the dominant subtype prevalent in dogs, pigs and wildlife across China. Further experiments to understand the importance of this virus as an infectious vector revealed that a ferret animal model experimentally infected with Tick/HLJ/2019 via the oronasal and ocular inoculation routes developed moderate respiratory distress with pneumonia and neurologic tissue damage from inflammation for the first time. Further surveillance of PIV5 in vectors of viral transmission is necessary to enhance our knowledge of its ecology in reservoirs and facilitate the control of re-emerging diseases.


Subject(s)
Ixodes , Parainfluenza Virus 5 , Animals , Dogs , Humans , Ferrets , Ixodes/virology , Parainfluenza Virus 5/classification , Parainfluenza Virus 5/genetics , Parainfluenza Virus 5/isolation & purification , Phylogeny , Rubulavirus Infections/epidemiology , Rubulavirus Infections/pathology , Rubulavirus Infections/virology , Swine
5.
J Virol ; 96(8): e0025722, 2022 04 27.
Article in English | MEDLINE | ID: mdl-35373581

ABSTRACT

Aggressive B-cell lymphoma is one of the most common types of blood malignancy. Robust delivery of genes of interest into target cells, long-term gene expression, and minimal risk of secondary effects are highly desirable for translational medicine including gene therapy and studies on gene function. However, efficient gene delivery into viral or nonviral B-lymphoma cells remains a challenge. Here, we report a strategy for inducing foreign gene expression in B-lymphoma cells by using a vector based on the novel parainfluenza virus PIV5-L (a strain isolated from B cells) that enabled us to study and control the function of a gene product within B-lymphoma cells. Using enhanced green fluorescent protein (eGFP) as a reporter, we successfully rescued PIV5-L and established a one-step system to generate PIV5-L virus-like particles (L-VLPs) with efficient delivery into a broad spectrum of susceptible B-lymphoma cell lines, including Epstein-Barr virus (EBV)- or Kaposi's sarcoma-associated herpesvirus (KSHV)-transformed B-lymphoblastoid cells. Similar to lentiviral vector, the L-VLP highly expressed exogenous genes and remained stable for long periods without obvious negative effects on cell viability. Taken together, these data demonstrate that the PIV5-L-based system provides a potential new strategy for the delivery of desirable genes and the treatment of cancer. IMPORTANCE B-cell lymphoma is a common aggressive neoplastic disorder of lymphocytes. Delivery of genes of interest into B cells, particularly virus-mediated B-lymphoma cells, is still a challenge. In this study, we report that a system (L-VLP) based on the parainfluenza virus PIV5-L strain isolated from B cells had highly expressed exogenous genes and remained stable without obvious cell toxicity, which provides a potential new strategy for gene delivery and treatment of B-cell cancer.


Subject(s)
Gene Transfer Techniques , Lymphoma, B-Cell , Parainfluenza Virus 5 , Cell Line, Tumor , Genetic Therapy , Herpesvirus 4, Human/genetics , Herpesvirus 8, Human/genetics , Humans , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/therapy , Parainfluenza Virus 5/genetics
6.
Transbound Emerg Dis ; 69(5): e1510-e1525, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35179303

ABSTRACT

Parainfluenza virus 5 (PIV5), a member of paramyxoviruses, causes respiratory and neurological infection in several animal species. Whereas information on PIV5 infection in digestive system is very scarce. Here, we successfully isolated one PIV5 strain from diarrheic piglets. After four times plaque purification and ultracentrifugation, the paramyxovirus-like particles were observed by electron microscopy. The genome-wide phylogenetic analysis showed that the isolated strain was closely related to the PIV5 strain from a lesser panda and pigs in China. Therefore, we characterized this isolated PIV5 and found that this virus could haemagglutinate red blood cells from both guinea pigs and chickens. Further, we observed that this PIV5 could infect cell lines from various host species including pig, human, monkey, bovine, dog, cat, rabbit, hamster and mouse, which was confirmed with the immunofluorescent assay. To evaluate the distribution of PIV5 in the field, we developed an indirect ELISA (iELISA) for the first time to detect the specific antibodies based on recombinant nucleocapsid protein. A total of 530 porcine serum samples were tested and the PIV5-positive rate was 75.7%. To our knowledge, this is the first report describing the full characterization of PIV5 strain isolated from a diarrheic piglet. The ability of this PIV5 strain to infect a wide range of mammalian cell types indicates that PIV5 can transmit across different species, providing a remarkable insight into potential zoonosis. The virus strain and iELISA developed in this study can be used to investigate the pathogenesis, epidemiology, and zoonotic potential of PIV5.


Subject(s)
Parainfluenza Virus 5 , Animals , Cattle , Cell Line , Chickens , Dogs , Guinea Pigs , Humans , Mammals , Mice , Nucleocapsid Proteins , Parainfluenza Virus 5/genetics , Phylogeny , Rabbits , Swine
7.
Front Immunol ; 12: 623996, 2021.
Article in English | MEDLINE | ID: mdl-33717130

ABSTRACT

The search for a preventive vaccine against HIV infection remains an ongoing challenge, indicating the need for novel approaches. Parainfluenza virus 5 (PIV5) is a paramyxovirus replicating in the upper airways that is not associated with any animal or human pathology. In animal models, PIV5-vectored vaccines have shown protection against influenza, RSV, and other human pathogens. Here, we generated PIV5 vaccines expressing HIV envelope (Env) and SIV Gag and administered them intranasally to macaques, followed by boosting with virus-like particles (VLPs) containing trimeric HIV Env. Moreover, we compared the immune responses generated by PIV5-SHIV prime/VLPs boost regimen in naïve vs a control group in which pre-existing immunity to the PIV5 vector was established. We demonstrate for the first time that intranasal administration of PIV5-based HIV vaccines is safe, well-tolerated and immunogenic, and that boosting with adjuvanted trimeric Env VLPs enhances humoral and cellular immune responses. The PIV5 prime/VLPs boost regimen induced robust and durable systemic and mucosal Env-specific antibody titers with functional activities including ADCC and neutralization. This regimen also induced highly polyfunctional antigen-specific T cell responses. Importantly, we show that diminished responses due to PIV5 pre-existing immunity can be overcome in part with VLP protein boosts. Overall, these results establish that PIV5-based HIV vaccine candidates are promising and warrant further investigation including moving on to primate challenge studies.


Subject(s)
AIDS Vaccines/administration & dosage , Gene Products, gag/administration & dosage , HIV-1/immunology , Immunogenicity, Vaccine , Parainfluenza Virus 5/immunology , Simian Immunodeficiency Virus/immunology , Virion/immunology , env Gene Products, Human Immunodeficiency Virus/administration & dosage , AIDS Vaccines/genetics , AIDS Vaccines/immunology , Administration, Intranasal , Animals , Antibodies, Viral/blood , Cattle , Cell Line , Gene Products, gag/genetics , Gene Products, gag/immunology , HIV-1/genetics , Host-Pathogen Interactions , Immunity, Cellular , Immunity, Humoral , Immunity, Mucosal , Macaca mulatta , Male , Nasal Mucosa/immunology , Nasal Mucosa/virology , Parainfluenza Virus 5/genetics , Simian Immunodeficiency Virus/genetics , T-Lymphocytes/immunology , T-Lymphocytes/virology , Vaccination , Vaccines, DNA/administration & dosage , Vaccines, DNA/immunology , Virion/genetics , env Gene Products, Human Immunodeficiency Virus/genetics , env Gene Products, Human Immunodeficiency Virus/immunology
8.
Sci Rep ; 11(1): 3866, 2021 02 16.
Article in English | MEDLINE | ID: mdl-33594165

ABSTRACT

Parainfluenza virus type 5 (PIV-5) causes respiratory infection in several animal species and humans. Canine parainfluenza virus type 5 (CPIV-5) causes respiratory disease in domestic dogs worldwide. In this study, we conducted a cross-sectional survey of CPIV-5 in dogs with respiratory symptoms from small animal hospitals in Thailand from November 2015 to December 2018. Our results showed that 32 out of 571 nasal swab samples (5.6%) were positive for CPIV-5 by RT-PCR specific to the NP gene. To characterize the viruses, three representative CPIV-5 were subjected to whole genome sequencing, and an additional ten CPIV-5 were subjected to HN, F, SH and V/P gene sequencing. Pairwise sequence comparison and phylogenetic analysis showed that Thai CPIV-5 was closely related to the CPIV-5 isolated from China and Korea. In conclusion, this study constitutes a whole genome characterization of CPIV-5 from dogs in Thailand. The surveillance of CPIV-5 should be further investigated at a larger scale to determine the dynamics, distribution and potential zoonotic transmission of CPIV-5.


Subject(s)
Genome, Viral , Parainfluenza Virus 5/genetics , Rubulavirus Infections/veterinary , Animals , Cross-Sectional Studies , Dogs , Rubulavirus Infections/virology , Thailand
9.
Vet Microbiol ; 250: 108859, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33039727

ABSTRACT

Pigs are an important reservoir for human influenza viruses, and influenza causes significant economic loss to the swine industry. As demonstrated during the 2009 H1N1 pandemic, control of swine influenza virus infection is a critical step toward blocking emergence of human influenza virus. An effective vaccine that can induce broadly protective immunity against heterologous influenza virus strains is critically needed. In our previous studies [McCormick et al., 2015; PLoS One, 10(6):e0127649], we used molecular breeding (DNA shuffling) strategies to increase the breadth of the variable and conserved epitopes expressed within a single influenza A virus chimeric hemagglutinin (HA) protein. Chimeric HAs were constructed using parental HAs from the 2009 pandemic virus and swine influenza viruses that had a history of zoonotic transmission to humans. In the current study, we used parainfluenza virus 5 (PIV-5) as a vector to express one of these chimeric HA antigens, HA-113. Recombinant PIV-5 expressing HA-113 (PIV5-113) were rescued, and immunogenicity and protective efficacy were tested in both mouse and pig models. The results showed that PIV5-113 can protect mice and pigs against challenge with viruses expressing parental HAs. The protective immunity was extended against other genetically diversified influenza H1-expressing viruses. Our work demonstrates that PIV5-based influenza vaccines are efficacious as vaccines for pigs. The PIV5 vaccine vector and chimeric HA-113 antigen are discussed in the context of the development of universal influenza vaccines and the potential contribution of PIV5-113 as a candidate universal vaccine.


Subject(s)
Antibodies, Viral/blood , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza Vaccines/immunology , Orthomyxoviridae Infections/prevention & control , Parainfluenza Virus 5/genetics , Swine Diseases/prevention & control , Animals , Antibodies, Viral/immunology , Cross Protection , Disease Models, Animal , Female , Genetic Vectors , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Immunogenicity, Vaccine , Mice , Mice, Inbred BALB C , Swine , Swine Diseases/virology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology
10.
Virulence ; 11(1): 1024-1040, 2020 12.
Article in English | MEDLINE | ID: mdl-32799724

ABSTRACT

BURKHOLDERIA MALLEI: is a highly pathogenic bacterium that causes the fatal zoonosis glanders. The organism specifies multiple membrane proteins, which represent prime targets for the development of countermeasures given their location at the host-pathogen interface. We investigated one of these proteins, Pal, and discovered that it is involved in the ability of B. mallei to resist complement-mediated killing and replicate inside host cells in vitro, is expressed in vivo and induces antibodies during the course of infection, and contributes to virulence in a mouse model of aerosol infection. A mutant in the pal gene of the B. mallei wild-type strain ATCC 23344 was found to be especially attenuated, as BALB/c mice challenged with the equivalent of 5,350 LD50 completely cleared infection. Based on these findings, we tested the hypothesis that a vaccine containing the Pal protein elicits protective immunity against aerosol challenge. To achieve this, the pal gene was cloned in the vaccine vector Parainfluenza Virus 5 (PIV5) and mice immunized with the virus were infected with a lethal dose of B. mallei. These experiments revealed that a single dose of PIV5 expressing Pal provided 80% survival over a period of 40 days post-challenge. In contrast, only 10% of mice vaccinated with a PIV5 control virus construct survived infection. Taken together, our data establish that the Peptidoglycan-associated lipoprotein Pal is a critical virulence determinant of B. mallei and effective target for developing a glanders vaccine.


Subject(s)
Bacterial Vaccines/immunology , Burkholderia mallei/chemistry , Burkholderia mallei/pathogenicity , Lipoproteins/immunology , Melioidosis/prevention & control , Peptidoglycan/chemistry , Aerosols , Animals , Bacterial Vaccines/administration & dosage , Burkholderia mallei/immunology , Cell Line , Female , Genetic Vectors , Immunization , Lipoproteins/administration & dosage , Macrophages/microbiology , Melioidosis/immunology , Mice , Mice, Inbred BALB C , Parainfluenza Virus 5/genetics , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology , Virulence
11.
J Virol ; 94(13)2020 06 16.
Article in English | MEDLINE | ID: mdl-32295916

ABSTRACT

During the replication of parainfluenza virus 5 (PIV5), copyback defective virus genomes (DVGs) are erroneously produced and are packaged into "infectious" virus particles. Copyback DVGs are the primary inducers of innate intracellular responses, including the interferon (IFN) response. While DVGs can interfere with the replication of nondefective (ND) virus genomes and activate the IFN-induction cascade before ND PIV5 can block the production of IFN, we demonstrate that the converse is also true, i.e., high levels of ND virus can block the ability of DVGs to activate the IFN-induction cascade. By following the replication and amplification of DVGs in A549 cells that are deficient in a variety of innate intracellular antiviral responses, we show that DVGs induce an uncharacterized IFN-independent innate response(s) that limits their replication. High-throughput sequencing was used to characterize the molecular structure of copyback DVGs. While there appears to be no sequence-specific break or rejoining points for the generation of copyback DVGs, our findings suggest there are region, size, and/or structural preferences selected for during for their amplification.IMPORTANCE Copyback defective virus genomes (DVGs) are powerful inducers of innate immune responses both in vitro and in vivo They impact the outcome of natural infections, may help drive virus-host coevolution, and promote virus persistence. Due to their potent interfering and immunostimulatory properties, DVGs may also be used therapeutically as antivirals and vaccine adjuvants. However, little is known of the host cell restrictions which limit their amplification. We show here that the generation of copyback DVGs readily occurs during parainfluenza virus 5 (PIV5) replication, but that their subsequent amplification is restricted by the induction of innate intracellular responses. Molecular characterization of PIV5 copyback DVGs suggests that while there are no genome sequence-specific breaks or rejoin points for the generation of copyback DVGs, genome region, size, and structural preferences are selected for during their evolution and amplification.


Subject(s)
Immunity, Innate/immunology , Parainfluenza Virus 5/genetics , Parainfluenza Virus 5/immunology , A549 Cells , Animals , Base Sequence/genetics , Cell Line , Chlorocebus aethiops , Cytoplasm , Defective Viruses/genetics , Genome, Viral/genetics , High-Throughput Nucleotide Sequencing/methods , Humans , Interferons/genetics , RNA, Viral/genetics , Vero Cells , Virion/genetics , Virus Diseases/genetics , Virus Replication/genetics
12.
mBio ; 11(2)2020 04 07.
Article in English | MEDLINE | ID: mdl-32265331

ABSTRACT

Middle East respiratory syndrome coronavirus (MERS-CoV) can cause severe and fatal acute respiratory disease in humans and remains endemic in the Middle East since first being identified in 2012. There are currently no approved vaccines or therapies available for MERS-CoV. In this study, we evaluated parainfluenza virus 5 (PIV5)-based vaccine expressing the MERS-CoV envelope spike protein (PIV5/MERS-S) in a human DPP4 knockin C57BL/6 congenic mouse model (hDPP4 KI). Following a single-dose intranasal immunization, PIV5-MERS-S induced neutralizing antibody and robust T cell responses in hDPP4 KI mice. A single intranasal administration of 104 PFU PIV5-MERS-S provided complete protection against a lethal challenge with mouse-adapted MERS-CoV (MERSMA6.1.2) and improved virus clearance in the lung. In comparison, single-dose intramuscular immunization with 106 PFU UV-inactivated MERSMA6.1.2 mixed with Imject alum provided protection to only 25% of immunized mice. Intriguingly, an influx of eosinophils was observed only in the lungs of mice immunized with inactivated MERS-CoV, suggestive of a hypersensitivity-type response. Overall, our study indicated that PIV5-MERS-S is a promising effective vaccine candidate against MERS-CoV infection.IMPORTANCE MERS-CoV causes lethal infection in humans, and there is no vaccine. Our work demonstrates that PIV5 is a promising vector for developing a MERS vaccine. Furthermore, success of PIV5-based MERS vaccine can be employed to develop a vaccine for emerging CoVs such as SARS-CoV-2, which causes COVID-19.


Subject(s)
Coronavirus Infections/prevention & control , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/immunology , Viral Vaccines/genetics , Viral Vaccines/immunology , Administration, Intranasal , Animals , Antibodies, Viral/blood , Coronavirus Infections/immunology , Coronavirus Infections/mortality , Disease Models, Animal , Immunization , Mice , Mice, Inbred C57BL , Parainfluenza Virus 5/genetics , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
13.
Arch Virol ; 165(4): 799-807, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32100137

ABSTRACT

We previously found that infection with human parainfluenza virus type 2 (hPIV-2), a member of the genus Orthorubulavirus, family Paramyxoviridae, causes filamentous actin (F-actin) formation to promote viral growth. In the present study, we investigated whether similar regulation of F-actin formation is observed in infections with other rubulaviruses, such as parainfluenza virus type 5 (PIV-5) and simian virus 41 (SV41). Infection with these viruses caused F-actin formation and RhoA activation, which promoted viral growth. These results indicate that RhoA-induced F-actin formation is important for efficient growth of these rubulaviruses. Only SV41 and hPIV-2 V and P proteins bound to Graf1, while the V and P proteins of PIV-5, mumps virus, and hPIV-4 did not bind to Graf1. In contrast, the V proteins of these rubulaviruses bound to both inactive RhoA and profilin 2. These results suggest that there are common and unique mechanisms involved in regulation of F-actin formation by members of the genus Orthorubulavirus.


Subject(s)
Actins/metabolism , Parainfluenza Virus 2, Human/metabolism , Parainfluenza Virus 5/metabolism , Rubulavirus Infections/metabolism , Rubulavirus/metabolism , Actins/chemistry , Actins/genetics , Animals , Cell Line , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , Host-Pathogen Interactions , Humans , Parainfluenza Virus 2, Human/genetics , Parainfluenza Virus 2, Human/growth & development , Parainfluenza Virus 5/genetics , Parainfluenza Virus 5/growth & development , Protein Binding , Rubulavirus/genetics , Rubulavirus/growth & development , Rubulavirus Infections/genetics , Rubulavirus Infections/virology , Viral Proteins/genetics , Viral Proteins/metabolism , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
14.
J Gen Virol ; 101(5): 467-472, 2020 05.
Article in English | MEDLINE | ID: mdl-32100701

ABSTRACT

The paramyxoviruses Hendra virus (HeV) and parainfluenza virus 5 (PIV5) require the fusion (F) protein to efficiently infect cells. For fusion to occur, F undergoes dramatic, essentially irreversible conformational changes to merge the viral and cell membranes into a continuous bilayer. Recently, a transmembrane (TM) domain leucine/isoleucine (L/I) zipper was shown to be critical in maintaining the expression, stability and pre-fusion conformation of HeV F, allowing for fine-tuned timing of membrane fusion. To analyse the effect of the TM domain L/I zipper in another paramyxovirus, we created alanine mutations to the TM domain of PIV5 F, a paramyxovirus model system. Our data show that while the PIV5 F TM L/I zipper does not significantly affect total expression and only modestly affects surface expression and pre-fusion stability, it is critical for fusogenic activity. These results suggest that the roles of TM L/I zipper motifs differ among members of the family Paramyxoviridae.


Subject(s)
Cell Membrane/genetics , Isoleucine/genetics , Leucine/genetics , Mutation/genetics , Parainfluenza Virus 5/genetics , Protein Domains/genetics , Viral Fusion Proteins/genetics , Amino Acid Sequence , Animals , Cell Line , Chlorocebus aethiops , Membrane Fusion/genetics , Paramyxovirinae/genetics , Vero Cells
16.
Int J Mol Sci ; 21(2)2020 Jan 13.
Article in English | MEDLINE | ID: mdl-31941046

ABSTRACT

The etiology of viral encephalitis in cattle often remains unresolved, posing a potential risk for animal and human health. In metagenomics studies of cattle with bovine non-suppurative encephalitis, parainfluenza virus 5 (PIV5) was identified in three brain samples. Interestingly, in two of these animals, bovine herpesvirus 6 and bovine astrovirus CH13 were additionally found. We investigated the role of PIV5 in bovine non-suppurative encephalitis and further characterized the three cases. With traditional sequencing methods, we completed the three PIV5 genomes, which were compared to one another. However, in comparison to already described PIV5 strains, unique features were revealed, like an 81 nucleotide longer open reading frame encoding the small hydrophobic (SH) protein. With in situ techniques, we demonstrated PIV5 antigen and RNA in one animal and found a broad cell tropism of PIV5 in the brain. Comparative quantitative analyses revealed a high viral load of PIV5 in the in situ positive animal and therefore, we propose that PIV5 was probably the cause of the disease. With this study, we clearly show that PIV5 is capable of naturally infecting different brain cell types in cattle in vivo and therefore it is a probable cause of encephalitis and neurological disease in cattle.


Subject(s)
Antigens, Viral , Cattle Diseases , Encephalitis, Viral , Genome, Viral , Parainfluenza Virus 5 , RNA, Viral , Rubulavirus Infections , Animals , Antigens, Viral/genetics , Antigens, Viral/metabolism , Cattle , Cattle Diseases/genetics , Cattle Diseases/metabolism , Cattle Diseases/virology , Encephalitis, Viral/genetics , Encephalitis, Viral/metabolism , Encephalitis, Viral/virology , Parainfluenza Virus 5/genetics , Parainfluenza Virus 5/metabolism , RNA, Viral/genetics , RNA, Viral/metabolism , Rubulavirus Infections/genetics , Rubulavirus Infections/metabolism
17.
Viruses ; 11(9)2019 08 23.
Article in English | MEDLINE | ID: mdl-31450796

ABSTRACT

A distinct Russian Mammalian orthorubulavirus 5 (PIV5) was detected in cell culture exhibiting cytopathic effect and hypothesized to be contaminated by a scientist with respiratory symptoms. The identification of the divergent strain indicated a lack of knowledge on the diversity of PIV5 strains and calls for surveillance of global PIV5 strains.


Subject(s)
Parainfluenza Virus 5 , Vero Cells/virology , Animals , Cell Line , Chlorocebus aethiops , Cytopathogenic Effect, Viral , Genome, Viral , Humans , Mammals/virology , Parainfluenza Virus 5/classification , Parainfluenza Virus 5/genetics , Parainfluenza Virus 5/isolation & purification , Phylogeny , Rubulavirus Infections/virology , Russia , Whole Genome Sequencing
18.
J Virol ; 93(17)2019 09 01.
Article in English | MEDLINE | ID: mdl-31217248

ABSTRACT

Enveloped viruses utilize surface glycoproteins to bind and fuse with a target cell membrane. The zoonotic Hendra virus (HeV), a member of the family Paramyxoviridae, utilizes the attachment protein (G) and the fusion protein (F) to perform these critical functions. Upon triggering, the trimeric F protein undergoes a large, irreversible conformation change to drive membrane fusion. Previously, we have shown that the transmembrane (TM) domain of the F protein, separate from the rest of the protein, is present in a monomer-trimer equilibrium. This TM-TM association contributes to the stability of the prefusion form of the protein, supporting a role for TM-TM interactions in the control of F protein conformational changes. To determine the impact of disrupting TM-TM interactions, constructs expressing the HeV F TM with limited flanking sequences were synthesized. Coexpression of these constructs with HeV F resulted in dramatic reductions in the stability of F protein expression and fusion activity. In contrast, no effects were observed when the HeV F TM constructs were coexpressed with the nonhomologous parainfluenza virus 5 (PIV5) fusion protein, indicating a requirement for specific interactions. To further examine this, a TM peptide homologous to the PIV5 F TM domain was synthesized. Addition of the peptide prior to infection inhibited infection with PIV5 but did not significantly affect infection with human metapneumovirus, a related virus. These results indicate that targeted disruption of TM-TM interactions significantly impact viral fusion protein stability and function, presenting these interactions as a novel target for antiviral development.IMPORTANCE Enveloped viruses require virus-cell membrane fusion to release the viral genome and replicate. The viral fusion protein triggers from the pre- to the postfusion conformation, an essentially irreversible change, to drive membrane fusion. We found that small proteins containing the TM and a limited flanking region homologous to the fusion protein of the zoonotic Hendra virus reduced protein expression and fusion activity. The introduction of exogenous TM peptides may displace a TM domain, disrupting native TM-TM interactions and globally destabilizing the fusion protein. Supporting this hypothesis, we showed that a sequence-specific transmembrane peptide dramatically reduced viral infection in another enveloped virus model, suggesting a broader inhibitory mechanism. Viral fusion protein TM-TM interactions are important for protein function, and disruption of these interactions dramatically reduces protein stability.


Subject(s)
Paramyxovirinae/metabolism , Peptides/pharmacology , Viral Fusion Proteins/chemistry , Viral Fusion Proteins/genetics , Animals , Binding Sites/drug effects , Chlorocebus aethiops , Hendra Virus/chemistry , Hendra Virus/genetics , Hendra Virus/metabolism , Hydrophobic and Hydrophilic Interactions/drug effects , Parainfluenza Virus 5/chemistry , Parainfluenza Virus 5/genetics , Parainfluenza Virus 5/metabolism , Paramyxovirinae/chemistry , Paramyxovirinae/genetics , Protein Conformation/drug effects , Protein Domains/drug effects , Protein Stability , Vero Cells , Viral Fusion Proteins/drug effects
19.
Viruses ; 11(5)2019 05 10.
Article in English | MEDLINE | ID: mdl-31083335

ABSTRACT

Previous results have shown that infection with the cytoplasmic-replicating parainfluenza virus 5 mutant P/V-CPI- sensitizes cells to DNA damaging agents, resulting in the enhanced killing of airway cancer cells. Here, we have tested the hypothesis that histone deacetylase (HDAC) inhibitors can also act with P/V-CPI- infection to enhance cancer cell killing. Using human small cell lung cancer and laryngeal cancer cell lines, 10 HDAC inhibitors were tested for their effect on viability of P/V-CPI- infected cells. HDAC inhibitors such as scriptaid enhanced caspase-3/7, -8 and -9 activity induced by P/V-CPI- and overall cell toxicity. Scriptaid-mediated enhanced killing was eliminated in lung cancer cells that were engineered to express a protein which sequesters double stranded RNA. Scriptaid also enhanced cancer cell killing by two other negative strand RNA viruses - the La Crosse virus and vesicular stomatitis virus. Scriptaid treatment enhanced the spread of the P/V-CPI- virus through a population of cancer cells, and suppressed interferon-beta induction through blocking phosphorylation and nuclear translocation of Interferon Regulatory Factor 3 (IRF-3). Taken together, these data support a role for combinations of a cytoplasmic-replicating RNA virus such as the P/V-CPI- mutant along with chemotherapeutic agents.


Subject(s)
Histone Deacetylase Inhibitors/pharmacology , Interferon-beta/biosynthesis , Neoplasms/metabolism , Oncolytic Viruses/physiology , Parainfluenza Virus 5/physiology , Caspases/genetics , Caspases/metabolism , Cell Line, Tumor , Humans , Hydroxylamines/pharmacology , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/metabolism , Neoplasms/genetics , Neoplasms/therapy , Neoplasms/virology , Oncolytic Viruses/genetics , Parainfluenza Virus 5/genetics , Quinolines/pharmacology
20.
PLoS One ; 14(5): e0216944, 2019.
Article in English | MEDLINE | ID: mdl-31100083

ABSTRACT

Most viruses are known to spontaneously generate defective viral genomes (DVG) due to errors during replication. These DVGs are subgenomic and contain deletions that render them unable to complete a full replication cycle in the absence of a co-infecting, non-defective helper virus. DVGs, especially of the copyback type, frequently observed with paramyxoviruses, have been recognized to be important triggers of the antiviral innate immune response. DVGs have therefore gained interest for their potential to alter the attenuation and immunogenicity of vaccines. To investigate this potential, accurate identification and quantification of DVGs is essential. Conventional methods, such as RT-PCR, are labor intensive and will only detect primer sequence-specific species. High throughput sequencing (HTS) is much better suited for this undertaking. Here, we present an HTS-based algorithm called DVG-profiler to identify and quantify all DVG sequences in an HTS data set generated from a virus preparation. DVG-profiler identifies DVG breakpoints relative to a reference genome and reports the directionality of each segment from within the same read. The specificity and sensitivity of the algorithm was assessed using both in silico data sets as well as HTS data obtained from parainfluenza virus 5, Sendai virus and mumps virus preparations. HTS data from the latter were also compared with conventional RT-PCR data and with data obtained using an alternative algorithm. The data presented here demonstrate the high specificity, sensitivity, and robustness of DVG-profiler. This algorithm was implemented within an open source cloud-based computing environment for analyzing HTS data. DVG-profiler might prove valuable not only in basic virus research but also in monitoring live attenuated vaccines for DVG content and to assure vaccine lot to lot consistency.


Subject(s)
Algorithms , Chromosome Mapping/statistics & numerical data , Defective Viruses/genetics , Genome, Viral , Mumps virus/genetics , Parainfluenza Virus 5/genetics , Sendai virus/genetics , Animals , Chromosome Mapping/methods , DNA Primers/chemical synthesis , DNA Primers/metabolism , Datasets as Topic , Defective Viruses/classification , High-Throughput Nucleotide Sequencing/statistics & numerical data , Humans , Molecular Typing , Mumps virus/classification , Parainfluenza Virus 5/classification , Real-Time Polymerase Chain Reaction , Sendai virus/classification , Sensitivity and Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...