Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS Pathog ; 17(9): e1009908, 2021 09.
Article in English | MEDLINE | ID: mdl-34529742

ABSTRACT

Human parainfluenza virus type 1 (hPIV1) and 3 (hPIV3) cause seasonal epidemics, but little is known about their interaction with human airway cells. In this study, we determined cytopathology, replication, and progeny virion release from human airway cells during long-term infection in vitro. Both viruses readily established persistent infection without causing significant cytopathic effects. However, assembly and release of hPIV1 rapidly declined in sharp contrast to hPIV3 due to impaired viral ribonucleocapsid (vRNP) trafficking and virus assembly. Transcriptomic analysis revealed that both viruses induced similar levels of type I and III IFNs. However, hPIV1 induced specific ISGs stronger than hPIV3, such as MX2, which bound to hPIV1 vRNPs in infected cells. In addition, hPIV1 but not hPIV3 suppressed genes involved in lipid biogenesis and hPIV1 infection resulted in ubiquitination and degradation of 3-hydroxy-3-methylglutaryl-coenzyme A reductase, a rate limiting enzyme in cholesterol biosynthesis. Consequently, formation of cholesterol-rich lipid rafts was impaired in hPIV1 infected cells. These results indicate that hPIV1 is capable of regulating cholesterol biogenesis, which likely together with ISGs contributes to establishment of a quiescent infection.


Subject(s)
Cholesterol/biosynthesis , Respiratory Mucosa/virology , Respirovirus Infections/metabolism , Respirovirus Infections/virology , A549 Cells , Humans , Interferons/immunology , Parainfluenza Virus 1, Human/immunology , Parainfluenza Virus 1, Human/metabolism , Parainfluenza Virus 3, Human/immunology , Parainfluenza Virus 3, Human/metabolism , Respirovirus Infections/immunology
2.
Virology ; 510: 234-241, 2017 10.
Article in English | MEDLINE | ID: mdl-28750327

ABSTRACT

Cholesterol-rich lipid raft microdomains in the plasma membrane are considered to play a major role in the enveloped virus lifecycle. However, the functional role of cholesterol in assembly, infectivity and stability of respiratory RNA viruses is not fully understood. We previously reported that depletion of cellular cholesterol by cholesterol-reducing agents decreased production of human parainfluenza virus type 1 (hPIV1) particles by inhibiting virus assembly. In this study, we analyzed the role of cholesterol on influenza A virus (IAV) and respiratory syncytial virus (RSV) production. Unlike hPIV1, treatment of human airway cells with the agents did not decrease virus particle production. However, the released virions were less homogeneous in density and unstable. Addition of exogenous cholesterol to the released virions restored virus stability and infectivity. Collectively, these data indicate a critical role of cholesterol in maintaining IAV and RSV membrane structure that is essential for sustaining viral stability and infectivity.


Subject(s)
Cholesterol/metabolism , Influenza A virus/physiology , Respiratory Syncytial Viruses/physiology , Virus Assembly , Humans , Microbial Viability , Virion/metabolism , Virion/ultrastructure
3.
Methods Mol Biol ; 1602: 103-110, 2017.
Article in English | MEDLINE | ID: mdl-28508216

ABSTRACT

Sendai virus (SeV) is a non-segment negative-sense RNA virus that naturally infects and causes pneumonia in mice. As a prototypic member of the family Paramyxoviridae, SeV has been characterized well, and these studies revealed numerous traits of paramyxovirus biology. The reverse genetics system to rescue SeV was first established in 1995. The virus was rescued from a cloned cDNA that contains full genome sequence flanked by T7 promoter and hepatitis delta virus ribozyme. To rescue SeV, it is necessary to infect cells with a recombinant vaccinia virus vTF7.3 that expresses T7 RNA polymerase, and transfect with the SeV full genome cDNAs together with supporting plasmids encoding NP, P, and L genes under the T7 promoter. Synthesized viral RNA by T7 RNA polymerase will be encapsidated with NP and associated with a polymerase complex composed of P and L. The polymerase complex transcribes and replicates the genome, and produces progeny virions. Rescued SeV needs to be plaque purified to exclude vTF7.3 from viral stock. Reverse genetics system of SeV is relatively efficient compared to other paramyxoviruses, but alternative approaches to rescue poorly growing mutant viruses are also available.


Subject(s)
DNA, Complementary , Genome, Viral , RNA, Viral , Sendai virus/genetics , Animals , Cell Line , Gene Expression Regulation, Viral , Genetic Vectors/genetics , Humans , Plasmids/genetics , Recombination, Genetic , Transfection , Vaccinia virus/genetics , Virus Replication
4.
Virology ; 501: 127-135, 2017 01 15.
Article in English | MEDLINE | ID: mdl-27915128

ABSTRACT

Many enveloped RNA viruses utilize lipid rafts for the assembly of progeny virions, but the role of cholesterol, a major component of rafts, on paramyxovirus budding and virion formation is controversial. In this study, we analyzed the effects of FDA-approved cholesterol-reducing agents, gemfibrozil and lovastatin, on raft formation and assembly of human parainfluenza virus type 1 (hPIV1) and Sendai virus (SeV). Treatment of the human airway epithelial A549 cells with the agents, especially when combined, significantly decreased production of infectious hPIV1 and SeV. Mechanistic analysis indicated that depletion of cellular cholesterol reduced cell surface accumulation of envelope glycoproteins and association of viral matrix and nucleocapsids with raft membrane, which resulted in impaired virus budding and release from the cells. These results indicate that cellular cholesterol is required for assembly and formation of type 1 parainfluenza viruses and suggest that cholesterol could be an attractive target for antiviral agents against hPIV1.


Subject(s)
Anticholesteremic Agents/pharmacology , Cholesterol/metabolism , Parainfluenza Virus 1, Human/drug effects , Paramyxoviridae Infections/virology , Virus Assembly/drug effects , Cell Membrane/metabolism , Cell Membrane/virology , Humans , Parainfluenza Virus 1, Human/genetics , Parainfluenza Virus 1, Human/physiology , Paramyxoviridae Infections/drug therapy , Paramyxoviridae Infections/metabolism , Protein Transport , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Release/drug effects
5.
Virology ; 487: 11-8, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26484934

ABSTRACT

Paramyxoviruses replicate in the cytoplasm of infected cells and newly synthesized viral nucleocapsids (vRNPs) are transported to the plasma membrane to be incorporated into progeny virions. In this study, we analyzed the impact of the Rab11-mediated recycling pathway in Sendai virus (SeV) and human parainfluenza virus type 1 (hPIV1) vRNP transport. We found that suppression of Rab11 expression caused vRNP aggregation in the cytoplasm and reduced progeny virion formation. Overexpression of constitutively active Rab11Q70L, but not dominant negative Rab11S25N co-localized with vRNP, showing that vRNP specifically recognizes the GTP-bound active form of Rab11. Moreover, Rab11Q70L co-localized with the dominant negative tails of all three subtypes of myosins, Va, Vb, and Vc, while SeV and hPIV1 vRNPs co-localized with only myosin Vb and Vc. These results highlight the critical role of Rab11 in vRNP trafficking, and suggest a specificity in the recycling endosomes parainfluenza viruses utilize for virus assembly.


Subject(s)
Myosin Type V/metabolism , Parainfluenza Virus 1, Human/metabolism , Sendai virus/metabolism , Virus Assembly/genetics , rab GTP-Binding Proteins/genetics , Animals , Cell Line , Endosomes/metabolism , HeLa Cells , Humans , Macaca mulatta , Nucleocapsid/metabolism , Parainfluenza Virus 1, Human/genetics , Paramyxoviridae Infections , Protein Transport/genetics , Protein Transport/physiology , RNA Interference , RNA, Small Interfering , Sendai virus/genetics , rab GTP-Binding Proteins/biosynthesis , rab4 GTP-Binding Proteins/metabolism
6.
J Infect Dis ; 210(7): 1052-61, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-24719474

ABSTRACT

BACKGROUND: We report the first-in-human safety and immunogenicity assessment of a prototype hexon chimeric adenovirus (Ad) serotype 5 (Ad5) vector containing the hexon hypervariable regions of Ad serotype 48 (Ad48) and expressing human immunodeficiency virus (HIV) type 1 EnvA. METHODS: Forty-eight Ad5 and Ad48 seronegative, HIV-uninfected subjects were enrolled in a randomized, double-blind, placebo-controlled, dose escalation phase 1 study. Four groups of 12 subjects received 10(9) to 10(11) viral particles (vp) of the Ad5HVR48.EnvA.01 vaccine (n = 10 per group) or placebo (n = 2 per group) at week 0 or weeks 0, 4, and 24. Safety and immunogenicity were assessed. RESULTS: Self-limited reactogenicity was observed after the initial immunization in the highest (10(11) vp) dose group. Responses in vaccinees included Ad48 neutralizing antibody (nAb) titers higher than Ad5 nAb titers, EnvA-specific enzyme-linked immunosorbent assay titers, and EnvA-specific enzyme-linked immunospot assay responses, and these responses generally persisted at week 52. At week 28 in the 10(9), 10(10), and 10(11) vp 3-dose groups, geometric mean EnvA enzyme-linked immunosorbent assay titers were 5721, 10 929, and 3420, respectively, and Ad48 nAb titers were a median of 1.7-fold higher than for Ad5. CONCLUSIONS: Ad5HVR48.ENVA.01 was safe, well tolerated, and immunogenic at all doses tested. Vector-elicited nAb responses were greater for Ad48 than Ad5, confirming that Ad-specific nAbs in humans are primarily, but not exclusively, directed against the hexon hypervariable regions. Clinical Trials Registration. NCT00695877.


Subject(s)
AIDS Vaccines/adverse effects , AIDS Vaccines/immunology , Adenoviruses, Human/genetics , Capsid Proteins/genetics , Gene Expression , HIV-1/genetics , env Gene Products, Human Immunodeficiency Virus/genetics , AIDS Vaccines/administration & dosage , AIDS Vaccines/genetics , Adolescent , Adult , Antibodies, Neutralizing/blood , Double-Blind Method , Drug Carriers , Drug-Related Side Effects and Adverse Reactions/epidemiology , Drug-Related Side Effects and Adverse Reactions/pathology , Enzyme-Linked Immunosorbent Assay , Enzyme-Linked Immunospot Assay , Female , Genetic Vectors , HIV Antibodies/blood , Humans , Leukocytes, Mononuclear/immunology , Male , Middle Aged , Placebos/administration & dosage , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/adverse effects , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Young Adult
7.
J Infect Dis ; 207(12): 1888-97, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23482644

ABSTRACT

BACKGROUND: Modified vaccinia Ankara (MVA-BN, IMVAMUNE) is emerging as a primary immunogen and as a delivery system to treat or prevent a wide range of diseases. Defining the safety and immunogenicity of MVA-BN in key populations is therefore important. METHODS: We performed a dose-escalation study of MVA-BN administered subcutaneously in 2 doses, one on day 0 and another on day 28. Twenty-four hematopoietic stem cell transplant recipients were enrolled sequentially into the study, and vaccine or placebo was administered under a randomized, double-blind allocation. Ten subjects received vaccine containing 10(7) median tissue culture infective doses (TCID50) of MVA-BN, 10 subjects received vaccine containing 10(8) TCID50 of MVA-BN, and 4 subjects received placebo. RESULTS: MVA-BN was generally well tolerated at both doses. No vaccine-related serious adverse events were identified. Transient local reactogenicity was more frequently seen at the higher dose. Neutralizing antibodies (NAb) to Vaccinia virus (VACV) were elicited by both doses of MVA-BN and were greater for the higher dose. Median peak anti-VACV NAb titers were 1:49 in the lower-dose group and 1:118 in the higher-dose group. T-cell immune responses to VACV were detected by an interferon γ enzyme-linked immunosorbent spot assay and were higher in the higher-dose group. CONCLUSIONS: MVA-BN is safe, well tolerated, and immunogenic in HSCT recipients. These data support the use of 10(8) TCID50 of MVA-BN in this population. CLINICAL TRIALS REGISTRATION: NCT00565929.


Subject(s)
Hematopoietic Stem Cell Transplantation/adverse effects , Smallpox Vaccine/administration & dosage , Vaccination , Vaccinia virus/immunology , Vaccinia/prevention & control , Adult , Antibodies, Neutralizing/biosynthesis , Antibodies, Viral/biosynthesis , Antigens, Viral/immunology , Cell Line , Dose-Response Relationship, Immunologic , Double-Blind Method , Female , Humans , Immunity, Cellular , Immunocompromised Host , Male , Middle Aged , Recombinant Proteins , Smallpox Vaccine/adverse effects , Smallpox Vaccine/immunology , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/adverse effects , Vaccines, Attenuated/immunology , Vaccinia/immunology , Vaccinia/virology , Vaccinia virus/genetics , Young Adult
8.
Nat Med ; 18(11): 1688-92, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23086475

ABSTRACT

Neutralizing antibodies are likely to play a crucial part in a preventative HIV-1 vaccine. Although efforts to elicit broadly cross-neutralizing (BCN) antibodies by vaccination have been unsuccessful, a minority of individuals naturally develop these antibodies after many years of infection. How such antibodies arise, and the role of viral evolution in shaping these responses, is unknown. Here we show, in two HIV-1-infected individuals who developed BCN antibodies targeting the glycan at Asn332 on the gp120 envelope, that this glycan was absent on the initial infecting virus. However, this BCN epitope evolved within 6 months, through immune escape from earlier strain-specific antibodies that resulted in a shift of a glycan to position 332. Both viruses that lacked the glycan at amino acid 332 were resistant to the Asn332-dependent BCN monoclonal antibody PGT128 (ref. 8), whereas escaped variants that acquired this glycan were sensitive. Analysis of large sequence and neutralization data sets showed the 332 glycan to be significantly under-represented in transmitted subtype C viruses compared to chronic viruses, with the absence of this glycan corresponding with resistance to PGT128. These findings highlight the dynamic interplay between early antibodies and viral escape in driving the evolution of conserved BCN antibody epitopes.


Subject(s)
Antibodies, Neutralizing , Epitopes , HIV-1 , HIV , Polysaccharides , AIDS Vaccines/immunology , AIDS Vaccines/therapeutic use , Amino Acid Sequence , Antibodies, Neutralizing/genetics , Antibodies, Neutralizing/immunology , Antibody Specificity , Epitopes/genetics , Epitopes/immunology , Evolution, Molecular , Female , Genome, Viral , HIV/genetics , HIV/immunology , HIV/pathogenicity , HIV Envelope Protein gp120/immunology , HIV Infections/immunology , HIV Infections/prevention & control , HIV Infections/virology , HIV Seropositivity/genetics , HIV Seropositivity/immunology , HIV-1/genetics , HIV-1/immunology , HIV-1/pathogenicity , Humans , Molecular Sequence Data , Mutagenesis, Site-Directed , Neutralization Tests , Polysaccharides/genetics , Polysaccharides/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...