Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 72
Filter
1.
Biometals ; 31(1): 81-89, 2018 02.
Article in English | MEDLINE | ID: mdl-29209895

ABSTRACT

Here we report on the results obtained from an antiviral screening, including herpes simplex virus, vaccinia virus, vesicular stomatitis virus, Coxsackie B4 virus or respiratory syncytial virus, parainfluenza-3 virus, reovirus-1 and Punta Toro virus, of three 2-hydroxy-3-methoxyphenyl acylhydrazone compounds in three cell lines (i.e. human embryonic lung fibroblast cells, human cervix carcinoma cells, and African Green monkey kidney cells). Interesting antiviral EC50 values are obtained against herpes simplex virus-1 and vaccinia virus. The biological activity of acylhydrazones is often attributed to their metal coordinating abilities, so potentiometric and microcalorimetric studies are here discussed to unravel the behavior of the three 2-hydroxy-3-methoxyphenyl compounds in solution. It is worth of note that the acylhydrazone with the higher affinity for Cu(II) ions shows the best antiviral activity against herpes simplex and vaccinia virus (EC50 ~ 1.5 µM, minimal cytotoxic concentration = 60 µM, selectivity index = 40).


Subject(s)
Antiviral Agents/pharmacology , Chelating Agents/pharmacology , Hydrazones/pharmacology , Simplexvirus/drug effects , Vaccinia virus/drug effects , Animals , Antiviral Agents/chemical synthesis , Antiviral Agents/metabolism , Cell Line , Cell Line, Tumor , Chelating Agents/chemical synthesis , Chelating Agents/metabolism , Chlorocebus aethiops , Copper/metabolism , Epithelial Cells/drug effects , Epithelial Cells/virology , Fibroblasts/drug effects , Fibroblasts/virology , Humans , Hydrazones/chemical synthesis , Hydrazones/metabolism , Inhibitory Concentration 50 , Magnesium/metabolism , Manganese/metabolism , Orthoreovirus, Mammalian/drug effects , Orthoreovirus, Mammalian/growth & development , Orthoreovirus, Mammalian/metabolism , Parainfluenza Virus 3, Human/drug effects , Parainfluenza Virus 3, Human/growth & development , Parainfluenza Virus 3, Human/metabolism , Phlebovirus/drug effects , Phlebovirus/growth & development , Phlebovirus/metabolism , Respiratory Syncytial Viruses/drug effects , Respiratory Syncytial Viruses/growth & development , Respiratory Syncytial Viruses/metabolism , Simplexvirus/growth & development , Simplexvirus/metabolism , Vaccinia virus/growth & development , Vaccinia virus/metabolism , Vero Cells , Vesiculovirus/drug effects , Vesiculovirus/growth & development , Vesiculovirus/metabolism
2.
Virology ; 503: 46-51, 2017 03.
Article in English | MEDLINE | ID: mdl-28113063

ABSTRACT

Interferon (IFN) exerts its antiviral effect by inducing a large family of cellular genes, named interferon (IFN)-stimulated genes (ISGs). An intriguing member of this family is indoleamine 2,3-dioxygenase (IDO), which catalyzes the first and rate-limiting step of the main branch of tryptophan (Trp) degradation, the kynurenine pathway. We recently showed that IDO strongly inhibits human parainfluenza virus type 3 (PIV3), a significant respiratory pathogen. Here, we show that 5-hydoxytryptophan (5-HTP), the first product of an alternative branch of Trp degradation and a serotonin precursor, is essential to protect virus growth against IDO in cell culture. We also show that the apparent antiviral effect of IDO on PIV3 is not due to the generation of the kynurenine pathway metabolites, but rather due to the depletion of intracellular Trp by IDO, as a result of which this rare amino acid becomes unavailable for the alternative, proviral 5-HTP pathway.


Subject(s)
5-Hydroxytryptophan/metabolism , Antiviral Agents/pharmacology , Indoleamine-Pyrrole 2,3,-Dioxygenase/pharmacology , Parainfluenza Virus 3, Human/growth & development , Tryptophan/metabolism , Virus Replication/drug effects , 5-Hydroxytryptophan/pharmacology , A549 Cells , Animals , Cell Line, Tumor , Humans , Interferons/pharmacology , Kynurenine/metabolism , Macaca mulatta , Parainfluenza Virus 3, Human/metabolism , Respirovirus Infections/drug therapy , Tryptophan/chemistry , Virus Replication/physiology
3.
J Virol ; 90(24): 11145-11156, 2016 Dec 15.
Article in English | MEDLINE | ID: mdl-27707917

ABSTRACT

A major arm of cellular innate immunity is type I interferon (IFN), represented by IFN-α and IFN-ß. Type I IFN transcriptionally induces a large number of cellular genes, collectively known as IFN-stimulated gene (ISG) proteins, which act as antivirals. The IFIT (interferon-induced proteins with tetratricopeptide repeats) family proteins constitute a major subclass of ISG proteins and are characterized by multiple tetratricopeptide repeats (TPRs). In this study, we have interrogated IFIT proteins for the ability to inhibit the growth of human parainfluenza virus type 3 (PIV3), a nonsegmented negative-strand RNA virus of the Paramyxoviridae family and a major cause of respiratory disease in children. We found that IFIT1 significantly inhibited PIV3, whereas IFIT2, IFIT3, and IFIT5 were less effective or not at all. In further screening a set of ISG proteins we discovered that several other such proteins also inhibited PIV3, including IFITM1, IDO (indoleamine 2,3-dioxygenase), PKR (protein kinase, RNA activated), and viperin (virus inhibitory protein, endoplasmic reticulum associated, interferon inducible)/Cig5. The antiviral effect of IDO, the enzyme that catalyzes the first step of tryptophan degradation, could be counteracted by tryptophan. These results advance our knowledge of diverse ISG proteins functioning as antivirals and may provide novel approaches against PIV3. IMPORTANCE: The innate immunity of the host, typified by interferon (IFN), is a major antiviral defense. IFN inhibits virus growth by inducing a large number of IFN-stimulated gene (ISG) proteins, several of which have been shown to have specific antiviral functions. Parainfluenza virus type 3 (PIV3) is major pathogen of children, and no reliable vaccine or specific antiviral against it currently exists. In this article, we report several ISG proteins that strongly inhibit PIV3 growth, the use of which may allow a better antiviral regimen targeting PIV3.


Subject(s)
Carrier Proteins/immunology , Host-Pathogen Interactions , Immunity, Innate , Interferon-alpha/immunology , Interferon-beta/immunology , Parainfluenza Virus 3, Human/immunology , A549 Cells , Adaptor Proteins, Signal Transducing , Animals , Antigens, Differentiation/genetics , Antigens, Differentiation/immunology , Carrier Proteins/genetics , Cell Line, Tumor , Epithelial Cells/drug effects , Epithelial Cells/immunology , Epithelial Cells/virology , Gene Expression Regulation , HEK293 Cells , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology , Interferon-alpha/genetics , Interferon-beta/genetics , Oxidoreductases Acting on CH-CH Group Donors , Parainfluenza Virus 3, Human/growth & development , Protein Isoforms/genetics , Protein Isoforms/immunology , Proteins/genetics , Proteins/immunology , RNA-Binding Proteins , Signal Transduction , Tryptophan/pharmacology , eIF-2 Kinase/genetics , eIF-2 Kinase/immunology
4.
PLoS One ; 10(12): e0144648, 2015.
Article in English | MEDLINE | ID: mdl-26659560

ABSTRACT

Fatty acid synthase (FASN) catalyzes the de novo synthesis of palmitate, a fatty acid utilized for synthesis of more complex fatty acids, plasma membrane structure, and post-translational palmitoylation of host and viral proteins. We have developed a potent inhibitor of FASN (TVB-3166) that reduces the production of respiratory syncytial virus (RSV) progeny in vitro from infected human lung epithelial cells (A549) and in vivo from mice challenged intranasally with RSV. Addition of TVB-3166 to the culture medium of RSV-infected A549 cells reduces viral spread without inducing cytopathic effects. The antiviral effect of the FASN inhibitor is a direct consequence of reducing de novo palmitate synthesis; similar doses are required for both antiviral activity and inhibition of palmitate production, and the addition of exogenous palmitate to TVB-3166-treated cells restores RSV production. TVB-3166 has minimal effect on RSV entry but significantly reduces viral RNA replication, protein levels, viral particle formation and infectivity of released viral particles. TVB-3166 substantially impacts viral replication, reducing production of infectious progeny 250-fold. In vivo, oral administration of TVB-3166 to RSV-A (Long)-infected BALB/c mice on normal chow, starting either on the day of infection or one day post-infection, reduces RSV lung titers 21-fold and 9-fold respectively. Further, TVB-3166 also inhibits the production of RSV B, human parainfluenza 3 (PIV3), and human rhinovirus 16 (HRV16) progeny from A549, HEp2 and HeLa cells respectively. Thus, inhibition of FASN and palmitate synthesis by TVB-3166 significantly reduces RSV progeny both in vitro and in vivo and has broad-spectrum activity against other respiratory viruses. FASN inhibition may alter the composition of regions of the host cell membrane where RSV assembly or replication occurs, or change the membrane composition of RSV progeny particles, decreasing their infectivity.


Subject(s)
Antiviral Agents/pharmacology , Enzyme Inhibitors/pharmacology , Fatty Acid Synthase, Type I/antagonists & inhibitors , Protein Processing, Post-Translational , Respiratory Syncytial Virus Infections/drug therapy , Respiratory Syncytial Viruses/drug effects , Virus Replication/drug effects , Administration, Oral , Animals , Antiviral Agents/chemical synthesis , Enzyme Inhibitors/chemical synthesis , Fatty Acid Synthase, Type I/genetics , Fatty Acid Synthase, Type I/metabolism , Gene Expression , HeLa Cells , Hep G2 Cells , Host-Pathogen Interactions , Humans , Lipoylation/drug effects , Mice , Mice, Inbred BALB C , Palmitic Acid/antagonists & inhibitors , Palmitic Acid/metabolism , Parainfluenza Virus 3, Human/drug effects , Parainfluenza Virus 3, Human/growth & development , Parainfluenza Virus 3, Human/metabolism , Respiratory Mucosa/drug effects , Respiratory Mucosa/enzymology , Respiratory Mucosa/virology , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Viruses/growth & development , Respiratory Syncytial Viruses/metabolism , Rhinovirus/drug effects , Rhinovirus/growth & development , Rhinovirus/metabolism , Viral Proteins/antagonists & inhibitors , Viral Proteins/genetics , Viral Proteins/metabolism , Virion/drug effects , Virion/growth & development , Virion/metabolism
5.
Antiviral Res ; 116: 34-44, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25637710

ABSTRACT

Heparan sulfate (HS) is a ubiquitous glycosaminoglycan that serves as a cellular attachment site for a number of significant human pathogens, including respiratory syncytial virus (RSV), human parainfluenza virus 3 (hPIV3), and herpes simplex virus (HSV). Decoy receptors can target pathogens by binding to the receptor pocket on viral attachment proteins, acting as 'molecular sinks' and preventing the pathogen from binding to susceptible host cells. Decoy receptors functionalized with HS could bind to pathogens and prevent infection, so we generated decoy liposomes displaying HS-octasaccharide (HS-octa). These decoy liposomes significantly inhibited RSV, hPIV3, and HSV infectivity in vitro to a greater degree than the original HS-octa building block. The degree of inhibition correlated with the density of HS-octa displayed on the liposome surface. Decoy liposomes with HS-octa inhibited infection of viruses to a greater extent than either full-length heparin or HS-octa alone. Decoy liposomes were effective when added prior to infection or following the initial infection of cells in vitro. By targeting the well-conserved receptor-binding sites of HS-binding viruses, decoy liposomes functionalized with HS-octa are a promising therapeutic antiviral agent and illustrate the utility of the liposome delivery platform.


Subject(s)
Antiviral Agents/pharmacology , Heparitin Sulfate/pharmacology , Liposomes , Parainfluenza Virus 3, Human/drug effects , Respiratory Syncytial Viruses/drug effects , Simplexvirus/drug effects , Virus Replication/drug effects , Animals , Antiviral Agents/administration & dosage , Antiviral Agents/chemistry , Heparitin Sulfate/administration & dosage , Parainfluenza Virus 3, Human/growth & development , Respiratory Syncytial Viruses/growth & development , Simplexvirus/growth & development , Vero Cells
6.
mBio ; 4(5): e00803-13, 2013 Oct 22.
Article in English | MEDLINE | ID: mdl-24149514

ABSTRACT

UNLABELLED: Paramyxoviruses, enveloped RNA viruses that include human parainfluenza virus type 3 (HPIV3), cause the majority of childhood viral pneumonia. HPIV3 infection starts when the viral receptor-binding protein engages sialic acid receptors in the lung and the viral envelope fuses with the target cell membrane. Fusion/entry requires interaction between two viral surface glycoproteins: tetrameric hemagglutinin-neuraminidase (HN) and fusion protein (F). In this report, we define structural correlates of the HN features that permit infection in vivo. We have shown that viruses with an HN-F that promotes growth in cultured immortalized cells are impaired in differentiated human airway epithelial cell cultures (HAE) and in vivo and evolve in HAE into viable viruses with less fusogenic HN-F. In this report, we identify specific structural features of the HN dimer interface that modulate HN-F interaction and fusion triggering and directly impact infection. Crystal structures of HN, which promotes viral growth in vivo, show a diminished interface in the HN dimer compared to the reference strain's HN, consistent with biochemical and biological data indicating decreased dimerization and decreased interaction with F protein. The crystallographic data suggest a structural explanation for the HN's altered ability to activate F and reveal properties that are critical for infection in vivo. IMPORTANCE: Human parainfluenza viruses cause the majority of childhood cases of croup, bronchiolitis, and pneumonia worldwide. Enveloped viruses must fuse their membranes with the target cell membranes in order to initiate infection. Parainfluenza fusion proceeds via a multistep reaction orchestrated by the two glycoproteins that make up its fusion machine. In vivo, viruses adapt for survival by evolving to acquire a set of fusion machinery features that provide key clues about requirements for infection in human beings. Infection of the lung by parainfluenzavirus is determined by specific interactions between the receptor binding molecule (hemagglutinin-neuraminidase [HN]) and the fusion protein (F). Here we identify specific structural features of the HN dimer interface that modulate HN-F interaction and fusion and directly impact infection. The crystallographic and biochemical data point to a structural explanation for the HN's altered ability to activate F for fusion and reveal properties that are critical for infection by this important lung virus in vivo.


Subject(s)
HN Protein/metabolism , Parainfluenza Virus 3, Human/growth & development , Parainfluenza Virus 3, Human/metabolism , Respirovirus Infections/virology , Viral Fusion Proteins/metabolism , Viral Proteins/metabolism , Animals , Crystallography, X-Ray , Dimerization , Female , HN Protein/chemistry , HN Protein/genetics , Humans , Parainfluenza Virus 3, Human/enzymology , Parainfluenza Virus 3, Human/genetics , Protein Binding , Rats , Sigmodontinae , Viral Fusion Proteins/chemistry , Viral Fusion Proteins/genetics , Viral Proteins/chemistry , Viral Proteins/genetics
7.
Voen Med Zh ; 334(4): 27-31, 2013 Apr.
Article in Russian | MEDLINE | ID: mdl-24000610

ABSTRACT

Research is performed for investigation of cultivation peculiarities of virus parainfluenza-3 on different cell cultures and optimization of methods of virus infection of these cultures with PI-3 virus in the process of development of vaccines. Influence of a seed lot on the formation rate of a cellular monolayer of continuous cell cultures PT-80, LEK, KSTand T-1 is defined, it is established that the seed lot of 250,000 cells in 1 ml provides optimum concentration for formation of a full monolayer within the time limits of 48-72 hours. The most sensitive cell cultures (PT-80) for cultivation of parainfluenza-3 virus are selected; an optimal CCID of virus PI-3(0,1 TCD/ml), whereby we can observe the maximal accumulation of a virus within 2-4 day after infecting cell cultures, is defined.


Subject(s)
Parainfluenza Vaccines , Parainfluenza Virus 3, Human/growth & development , Respirovirus Infections/metabolism , Animals , Cattle , Cell Line , Humans , Parainfluenza Virus 3, Human/immunology , Respirovirus Infections/immunology , Respirovirus Infections/prevention & control
8.
mBio ; 3(3)2012.
Article in English | MEDLINE | ID: mdl-22669629

ABSTRACT

UNLABELLED: Paramyxoviruses, a family of RNA enveloped viruses that includes human parainfluenza virus type 3 (HPIV3), cause the majority of childhood croup, bronchiolitis, and pneumonia worldwide. Infection starts with host cell receptor binding and fusion of the viral envelope with the cell membrane at the cell surface. The fusion process requires interaction of the two viral surface glycoproteins, the hemagglutinin-neuraminidase (HN) and the fusion protein (F). We have previously shown that viruses with an HN/F pair that is highly fusogenic in monolayers of immortalized cells due to mutations in HN's secondary sialic acid binding site are growth impaired in differentiated human airway epithelium (HAE) cultures and in vivo. Here we have shown that adaptation of HPIV3 to growth in the lung is determined by specific features of HN and F that are different from those required for growth in cultured immortalized cells. An HPIV3 virus bearing a mutated HN (H552Q), which is fit and fusogenic in immortalized cells but unfit for growth in the lung, evolved into a less-fusogenic but viable virus in differentiated human airway epithelium. Stepwise evolution led to a progressive decrease in efficiency of fusion activation by the HN/F pair, with a mutation in F first decreasing the activation of F by HN and a mutation in HN's secondary sialic acid binding site decreasing fusion activation further and producing a stable virus. Adaptation of HPIV3 to successful growth in HAE is determined by specific features of HN and F that lead to a less easily activated fusion mechanism. IMPORTANCE: Human parainfluenza viruses (HPIVs) are paramyxoviruses that cause the majority of childhood cases of croup, bronchiolitis, and pneumonia worldwide, but there are currently no vaccines or antivirals available for treatment. Enveloped viruses must fuse their membrane with the target cell membrane in order to initiate infection. Parainfluenza virus fusion proceeds via a multistep reaction orchestrated by the two glycoproteins that make up its fusion machine. The receptor-binding hemagglutinin-neuraminidase (HN), upon receptor engagement, activates the fusion protein (F) to penetrate the target cell and mediate viral entry. In this study, we show that the precise balance of fusion activation properties of these two glycoproteins during entry is key for infection. In clinically relevant tissues, viruses evolve to acquire a set of fusion features that provide key clues about requirements for infection in human beings.


Subject(s)
Adaptation, Biological , Epithelial Cells/virology , HN Protein/genetics , Parainfluenza Virus 3, Human/physiology , Viral Fusion Proteins/genetics , Virus Internalization , Animals , Cell Line , Chlorocebus aethiops , Evolution, Molecular , Humans , Models, Molecular , Mutant Proteins/genetics , Mutation, Missense , Parainfluenza Virus 3, Human/genetics , Parainfluenza Virus 3, Human/growth & development , Parainfluenza Virus 3, Human/pathogenicity , Protein Conformation
9.
Scand J Immunol ; 66(6): 645-53, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17944814

ABSTRACT

No licensed vaccines are available to protect against parainfluenza virus type 3 (PIV3), a significant health risk for infants. In search of a safe vaccine, we used an alphavirus-based chimeric vector, consisting of Sindbis virus (SIN) structural proteins and Venezuelan equine encephalitis virus (VEE) replicon RNA, expressing the PIV3 hemagglutinin-neuraminidase (HN) glycoprotein (VEE/SIN-HN). We compared different routes of intramuscular (i.m.), intranasal (i.n.), or combined i.n. and i.m. immunizations with VEE/SIN-HN in hamsters. Six months after the final immunization, all hamsters were protected against live PIV3 i.n. challenge in nasal turbinates and lungs. This protection appeared to correlate with antibodies in serum, nasal turbinates and lungs. This is the first report demonstrating mucosal protection against PIV3 for an extended time following immunizations with an RNA replicon delivery system.


Subject(s)
Alphavirus/immunology , Mucous Membrane/immunology , Parainfluenza Vaccines/immunology , Parainfluenza Virus 3, Human/immunology , RNA, Viral/immunology , Replicon/immunology , Administration, Intranasal , Alphavirus/genetics , Animals , Antibodies, Viral/blood , Antibodies, Viral/immunology , Cricetinae , Disease Models, Animal , Encephalitis Virus, Venezuelan Equine/immunology , Humans , Immunization , Injections, Intramuscular , Parainfluenza Virus 3, Human/growth & development , RNA, Viral/genetics , Replicon/genetics , Sindbis Virus/immunology , Vaccines, Attenuated/immunology , Vaccines, Attenuated/therapeutic use , Vaccines, Synthetic/immunology
10.
Vaccine ; 25(3): 481-9, 2007 Jan 05.
Article in English | MEDLINE | ID: mdl-17052811

ABSTRACT

Parainfluenza virus type 3 (PIV3) infections continue to be a significant health risk for infants, young children, and immunocompromised adults. We describe a gene-based vaccine strategy against PIV3 using replication-defective alphavirus vectors. These RNA replicon vectors, delivered as virus-like particles and expressing the PIV3 hemagglutinin-neuraminidase glycoprotein, were shown to be highly immunogenic in mice and hamsters, inducing PIV3-specific neutralizing antibody responses. Importantly, the replicon particle-based vaccine administered intramuscularly or intranasally protected against mucosal PIV3 challenge in hamsters, preventing virus replication in both nasal turbinates and lungs. These data suggest that the alphavirus replicon platform can be useful for a PIV3 vaccine and possibly other respiratory viruses.


Subject(s)
Alphavirus/genetics , Parainfluenza Vaccines/immunology , Parainfluenza Virus 3, Human/immunology , Paramyxoviridae Infections/immunology , Paramyxoviridae Infections/prevention & control , RNA, Viral/genetics , RNA, Viral/immunology , Replicon/genetics , Replicon/immunology , Administration, Intranasal , Animals , Antibodies, Viral/immunology , Cricetinae , Encephalitis Virus, Venezuelan Equine/immunology , Enzyme-Linked Immunosorbent Assay , Female , Hemagglutination Inhibition Tests , Mesocricetus , Mice , Mice, Inbred BALB C , Neutralization Tests , Parainfluenza Virus 3, Human/growth & development , Sindbis Virus/immunology , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
11.
J Clin Invest ; 115(7): 1688-98, 2005 Jul.
Article in English | MEDLINE | ID: mdl-16007245

ABSTRACT

Human parainfluenza viruses cause several serious respiratory diseases in children for which there is no effective prevention or therapy. Parainfluenza viruses initiate infection by binding to cell surface receptors and then, via coordinated action of the 2 viral surface glycoproteins, fuse directly with the cell membrane to release the viral replication machinery into the host cell's cytoplasm. During this process, the receptor-binding molecule must trigger the viral fusion protein to mediate fusion and entry of the virus into a cell. This review explores the binding and entry into cells of parainfluenza virus type 3, focusing on how the receptor-binding molecule triggers the fusion process. There are several steps during the process of binding, triggering, and fusion that are now understood at the molecular level, and each of these steps represents potential targets for interrupting infection.


Subject(s)
Parainfluenza Virus 3, Human/physiology , Parainfluenza Virus 3, Human/pathogenicity , Respirovirus Infections/therapy , Respirovirus Infections/virology , Antiviral Agents/pharmacology , Binding Sites , Child , HN Protein/chemistry , HN Protein/physiology , Humans , Influenza, Human/etiology , Influenza, Human/therapy , Influenza, Human/virology , Membrane Fusion/drug effects , Membrane Fusion/physiology , Models, Biological , Models, Molecular , Neuraminidase/antagonists & inhibitors , Parainfluenza Virus 3, Human/growth & development , Receptors, Virus/physiology , Respirovirus Infections/etiology , Viral Fusion Proteins/physiology , Viral Proteins/physiology , Virulence
12.
J Clin Virol ; 30(1): 100-5, 2004 May.
Article in English | MEDLINE | ID: mdl-15072762

ABSTRACT

Respiratory viruses cause significant morbidity and mortality. The management of these infections can be improved by a rapid diagnosis and administration of available virus-specific therapy. The goal of this study was to compare R-Mix, an engineered tissue monolayer for rapid shell vial (SV) diagnosis of viral respiratory infections, with conventional tissue culture (TC) and conventional respiratory SV (primary rhesus monkey kidney (RhMK) and Hep2 monolayers). The primary outcome measure was sensitivity for detection of influenza A and B, respiratory syncytial virus, parainfluenza 1-3, and adenovirus. The study was performed in two phases: (1) the three methods were compared using 250 nasal washes from children with lower respiratory tract infections; (2) a modified R-Mix SV harvesting schedule (SV were harvested at 24 and 120 h) was compared with TC and conventional RhMK/Hep2 SV using 311 respiratory specimens. A total of 110 viruses were identified in the first and 55 in the second phase. Diagnostic accuracies of R-Mix harvested at 24, 48, and 120 h were 98%, whereas for TC varied between 99 and 100%, and for RhMK/Hep2 SV between 98 and 99%. Sensitivities of R-Mix harvested at 24, 48, and 120 h were 26, 75, and 47%, respectively, whereas for TC varied between 60 and 94%, and for RhMK/Hep2 SV between 62 and 85%. R-Mix harvested at 48 h represent a valuable substitute for RhMK/Hep2 SV because they have comparable sensitivities and diagnostic accuracies, but R-Mix offers several technical advantages. In contrast, R-Mix harvested at 24h did not seem a very useful diagnostic tool. The utility of R-Mix harvested at 120 h, which accelerated the diagnosis of 16% of positive specimens in study phase 2, needs further investigation.


Subject(s)
Orthomyxoviridae Infections/diagnosis , Orthomyxoviridae/isolation & purification , Paramyxoviridae Infections/diagnosis , Paramyxoviridae/isolation & purification , Respiratory Tract Infections/diagnosis , Adenoviruses, Human/growth & development , Adenoviruses, Human/isolation & purification , Animals , Cell Line , Child , Humans , Influenza A virus/growth & development , Influenza A virus/isolation & purification , Influenza B virus/growth & development , Influenza B virus/isolation & purification , Nasal Lavage Fluid/virology , Orthomyxoviridae/growth & development , Orthomyxoviridae Infections/virology , Parainfluenza Virus 1, Human/growth & development , Parainfluenza Virus 1, Human/isolation & purification , Parainfluenza Virus 2, Human/growth & development , Parainfluenza Virus 2, Human/isolation & purification , Parainfluenza Virus 3, Human/growth & development , Parainfluenza Virus 3, Human/isolation & purification , Paramyxoviridae/growth & development , Paramyxoviridae Infections/virology , Predictive Value of Tests , Respiratory Syncytial Virus, Human/growth & development , Respiratory Syncytial Virus, Human/isolation & purification , Respiratory Tract Infections/virology , Sensitivity and Specificity , Time Factors , Virus Cultivation
13.
J Clin Virol ; 29(4): 320-2, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15018862

ABSTRACT

BACKGROUND: A 5-year-old boy with acute lymphoblastic leukaemia (ALL) received a haematopoietic stem cell transplant (HSCT) from his father and was monitored for the presence of respiratory viruses. STUDY DESIGN: Nasal washes were taken during the transplant period and tested by culture and real-time PCR. RESULTS: Fifteen days prior to transplant parainfluenza virus 3 (PIV3) was isolated by culture and confirmed by immunofluoresence (IF) from a nasal wash specimen. Subsequent samples were negative by both IF and culture so the pre-transplant conditioning was started. One week after the HSCT the patient developed respiratory symptoms which progressively deteriorated. The IF and culture for PIV3 became positive again only after the symptoms deteriorated. However, retrospectively, a real-time PCR assay showed that the PIV3 was detectable throughout the conditioning phase and the amount of virus increased at the time of reappearance of respiratory symptoms post-HSCT. CONCLUSIONS: Management of PIV3 infection is important in HSCT. The use of this real-time PCR assay could improve diagnosis and management of PIV3 infection.


Subject(s)
Hematopoietic Stem Cell Transplantation , Parainfluenza Virus 3, Human/isolation & purification , Precursor Cell Lymphoblastic Leukemia-Lymphoma/complications , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Respirovirus Infections/virology , Child, Preschool , Humans , Male , Nasal Lavage Fluid/virology , Parainfluenza Virus 3, Human/genetics , Parainfluenza Virus 3, Human/growth & development , Polymerase Chain Reaction
14.
J Ethnopharmacol ; 79(3): 365-8, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11849843

ABSTRACT

The flower of Trollius chinensis Bunge is used for treating upper respiratory infections, pharyngitis, tonsillitis, and bronchitis in Chinese folk medicine. The antiviral activities of the crude extract, total flavonoids, orientin, vitexin and proglobeflowery acid isolated from the flowers of T. chinensis against parainfluenza type 3 (Para 3) virus were investigated. The results showed that the crude extract and total flavonoids exhibited a weak antiviral activity against Para 3. Orientin and vitexin demonstrated potent or moderate antiviral activity against Para 3. Proglobeflowery acid showed weak antiviral activity against Para 3.


Subject(s)
Antiviral Agents/pharmacology , Drugs, Chinese Herbal/pharmacology , Flavonoids/pharmacology , Parabens/pharmacology , Parainfluenza Virus 3, Human/drug effects , Ranunculaceae , Antiviral Agents/chemistry , Antiviral Agents/isolation & purification , Cells, Cultured , Cytopathogenic Effect, Viral/drug effects , Drug Evaluation, Preclinical/methods , Drug Evaluation, Preclinical/statistics & numerical data , Flavonoids/isolation & purification , Humans , Inhibitory Concentration 50 , Parabens/chemistry , Parabens/isolation & purification , Parainfluenza Virus 3, Human/growth & development , Plant Extracts/chemistry , Plant Extracts/isolation & purification , Plant Extracts/pharmacology , Plant Structures
15.
J Virol ; 75(21): 10498-504, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11581420

ABSTRACT

The chimeric recombinant virus rHPIV3-N(B), a version of human parainfluenza virus type 3 (HPIV3) that is attenuated due to the presence of the bovine PIV3 nucleocapsid (N) protein open reading frame (ORF) in place of the HPIV3 ORF, was modified to encode the measles virus hemagglutinin (HA) inserted as an additional, supernumerary gene between the HPIV3 P and M genes. This recombinant, designated rHPIV3-N(B)HA, replicated like its attenuated rHPIV3-N(B) parent virus in vitro and in the upper and lower respiratory tracts of rhesus monkeys, indicating that the insertion of the measles virus HA did not further attenuate rHPIV3-N(B) in vitro or in vivo. Monkeys immunized with rHPIV3-N(B)HA developed a vigorous immune response to both measles virus and HPIV3, with serum antibody titers to both measles virus (neutralizing antibody) and HPIV3 (hemagglutination inhibiting antibody) of over 1:500. An attenuated HPIV3 expressing a major protective antigen of measles virus provides a method for immunization against measles by the intranasal route, a route that has been shown with HPIV3 and respiratory syncytial virus vaccines to be relatively refractory to the neutralizing and immunosuppressive effects of maternally derived virus-specific serum antibodies. It should now be possible to induce a protective immune response against measles virus in 6-month-old infants, an age group that in developing areas of the world is not responsive to the current measles virus vaccine.


Subject(s)
Hemagglutinins, Viral/immunology , Measles Vaccine/immunology , Parainfluenza Virus 3, Human/genetics , Respirovirus/genetics , Vaccines, Synthetic/immunology , Virus Replication , Animals , Antibodies, Viral/blood , Cattle , Chimera , Humans , Immunization , Macaca mulatta , Parainfluenza Virus 3, Human/growth & development , Parainfluenza Virus 3, Human/immunology , Respiratory System/virology , Vaccines, Attenuated/immunology
16.
J Clin Virol ; 22(1): 101-10, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11418357

ABSTRACT

OBJECTIVE: To evaluate shell vials of R-Mix, a combination of mink lung cells and human adenocarcinoma cells (strains Mv1Lu and A549, respectively, Diagnostic Hybrids, Athens, OH) to detect respiratory viruses from prospective clinical respiratory specimens and frozen stocks. STUDY DESIGN: We compared the performance of R-Mix to conventional culture (CC) using tubes of PMK, Hep-2, and MRC5 to detect respiratory viruses from fresh clinical specimens. For each respiratory specimen submitted to virology, two shell vials of R-Mix were inoculated and examined twice (generally after 24 and 48 h) by an indirect test with a pool of immunofluorescent antisera to influenza A and B, adenovirus, parainfluenza 1-3 and RSV (DAKO, Carpinteria, CA). If positive, testing with monoclonal antisera was done. CCs were incubated for 10 days, examined daily for cytopathological effect, hemadsorbed twice and stained if positive. Cost comparison was done. Lastly, respiratory viruses frozen from previous years were inoculated onto R-Mix. RESULTS: R-Mix was positive for all 29 frozen virus stocks. In the clinical trial, 396 prospective specimens were inoculated into R-Mix and CC. R-Mix identified 21 specimens as respiratory virus positive; CC identified 19. Turn-around time of R-Mix for positive specimens was 1.4 days; for CC it was 5.2 days. Turn-around time of R-Mix for all specimens (positive and negative) was 2.0 days; for CC it was 9.8 days. The overall cost of R-Mix was approximately 11% more than that of CC. CONCLUSION: R-Mix enabled rapid identification of all the frozen virus stocks representing the seven major respiratory viral groups. When compared to CC, R-Mix was slightly more sensitive than three cell lines (four tubes) used in CC but it was several days faster.


Subject(s)
Adenovirus Infections, Human/virology , Influenza, Human/virology , Reagent Kits, Diagnostic/economics , Respiratory Syncytial Virus Infections/virology , Respirovirus Infections/virology , Adenovirus Infections, Human/diagnosis , Animals , Cell Line , Coculture Techniques , Costs and Cost Analysis , Freezing , Humans , Influenza A virus/growth & development , Influenza A virus/isolation & purification , Influenza B virus/growth & development , Influenza B virus/isolation & purification , Influenza, Human/diagnosis , Influenza, Human/pathology , Mink , Parainfluenza Virus 1, Human/growth & development , Parainfluenza Virus 1, Human/isolation & purification , Parainfluenza Virus 2, Human/growth & development , Parainfluenza Virus 2, Human/isolation & purification , Parainfluenza Virus 3, Human/growth & development , Parainfluenza Virus 3, Human/isolation & purification , Prospective Studies , Respiratory Syncytial Virus Infections/diagnosis , Respiratory Syncytial Virus Infections/pathology , Respirovirus Infections/diagnosis , Respirovirus Infections/pathology , Sensitivity and Specificity , Time Factors , Tumor Cells, Cultured
17.
Virus Genes ; 20(2): 173-82, 2000.
Article in English | MEDLINE | ID: mdl-10872880

ABSTRACT

The Kansas/15626/84 (Ka) and Shipping Fever (SF) strains of bovine parainfluenza virus type 3 (BPIV3) replicate less efficiently than human PIV3 (HPIV3) in the upper and lower respiratory tract of rhesus monkeys, and BPIV3 Ka is also highly attenuated in humans and is in clinical trials as a candidate vaccine against HPIV3. To initiate an investigation of the genetic basis of the observed attenuation phenotype of BPIV3 in primates, the complete genomic sequences of Ka and SF genomes were determined and compared to those of BPIV3 strain 910N and two HPIV3 strains, JS and Wash/47885/57. There is a high degree of identity between the five PIV3 viruses in their 55 nucleotide (nt) leader (83.6%) and 44 nt trailer (93.2%) sequences. The five viruses display amino acid sequence identity ranging from 58.6% for the phosphoprotein to 89.7% for the matrix protein. Interestingly, the majority of amino acid residues found to be variable at a given position in a five-way protein alignment are nonetheless identical within the viruses of either host species (BPIV3 or HPIV3). These host-specific residues might be products of distinct selective pressures on BPIV3 and HPIV3 during evolution in their respective hosts. These host-specific sequences likely include ones which are responsible for the host range differences, such as the efficient growth of BPIV3 in bovines compared to its restricted growth in primates. It should now be possible using the techniques of reverse genetics to import sequences from BPIV3 into HPIV3 and identify those nt or protein sequences which attenuate HPIV3 for primates. This information should be useful in understanding virus-host interactions and in the development of vaccines to protect against HPIV3-induced disease.


Subject(s)
Genome, Viral , Macaca mulatta/virology , Respirovirus/genetics , Virus Replication , Animals , Base Sequence , Cattle , Cell Line , Consensus Sequence , Humans , Molecular Sequence Data , Open Reading Frames , Parainfluenza Virus 3, Human/genetics , Parainfluenza Virus 3, Human/growth & development , Parainfluenza Virus 3, Human/physiology , RNA, Viral/analysis , Respirovirus/growth & development , Respirovirus/physiology , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA
18.
Microsc Res Tech ; 47(2): 114-23, 1999 Oct 15.
Article in English | MEDLINE | ID: mdl-10523790

ABSTRACT

Multifunctional involvement of actin microfilaments during viral infection has been documented in many studies. The molecular mechanism underlying this important host-virus interaction, however, remains poorly understood. We have investigated the role of actin microfilaments in the life cycle of human parainfluenza virus type 3 (HPIV3), a paramyxovirus that causes severe respiratory illness in children. In vitro transcription with purified viral ribonucleoprotein (RNP) complex showed a requirement of cellular actin, in the polymeric form, for mRNA synthesis in vitro. This was further confirmed by using recombinant actin, which interacted with the viral RNP and also activated mRNA synthesis in vitro. Consistent with the role of the polymeric form of actin, the actin microfilaments of the cytoskeletal framework participate in the virus replication in vivo. Biochemical and immunological analyses revealed the association of viral RNPs with cytoskeletal framework during early stages of infection, and involvement of these RNPs in the synthesis of mRNAs and genome-length RNA. Immunofluorescent labeling and confocal microscopy showed that the viral nucleocapsids colocalize with the actin microfilaments. Treatment of cells with cytochalasin D, which depolymerizes actin microfilaments, inhibited viral RNA synthesis and RNP accumulation. These data indicate that actin microfilaments play a critical role in HPIV3 life cycle, specifically at the level of viral transcription and replication. Involvement of the cytoskeletal framework in the life cycle of several viruses containing RNA and DNA genomes is reviewed.


Subject(s)
Actin Cytoskeleton/chemistry , Actins/physiology , Parainfluenza Virus 3, Human/physiology , Transcription, Genetic , Virus Replication , Actin Cytoskeleton/physiology , Actins/pharmacology , Cells, Cultured , Cytochalasin D/pharmacology , Humans , Immunoblotting , Parainfluenza Virus 3, Human/genetics , Parainfluenza Virus 3, Human/growth & development , RNA, Messenger/biosynthesis , Recombinant Proteins/biosynthesis , Ribonucleoproteins/drug effects , Ribonucleoproteins/physiology , Transcription, Genetic/drug effects , Viral Proteins/drug effects , Viral Proteins/physiology , Virus Replication/drug effects
19.
J Infect Dis ; 179(6): 1345-51, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10228053

ABSTRACT

Two parainfluenza virus type 3 (PIV3) vaccine candidates-cp45, a live attenuated derivative of the JS wild type (wt), and a replication-defective vaccinia virus recombinant expressing the hemagglutinin-neuraminidase or fusion glycoprotein of human PIV3 (modified vaccinia virus Ankara [MVA]/PIV3 recombinants)-were evaluated in rhesus monkeys to determine whether successful immunization could be achieved in the presence of passively transferred PIV3 antibodies. The cp45 virus, administered intranasally (in) and intratracheally (it) in the presence of high levels of PIV3 antibodies, replicated efficiently in the nasopharynx and protected against challenge with wt human PIV3. The MVA recombinants, administered in, it, and intramuscularly in the absence of passive antibody, conferred protection against replication of PIV3 wt challenge virus, but this was largely abrogated when immunization occurred in the presence of passive antibodies. Because immunization for the prevention of HPIV3 disease must occur in early infancy when maternal antibodies are present, the live attenuated cp45 virus continues to be a promising vaccine candidate for this age group.


Subject(s)
Antigens, Viral/therapeutic use , Parainfluenza Virus 3, Human/immunology , Respirovirus Infections/prevention & control , Vaccination , Viral Vaccines/therapeutic use , Animals , Antibodies, Viral/blood , Antibodies, Viral/therapeutic use , Immunization, Passive , Macaca mulatta , Parainfluenza Virus 3, Human/growth & development , Respiratory System/virology , Vaccines, Synthetic/therapeutic use , Virus Replication
20.
J Virol ; 73(2): 1374-81, 1999 Feb.
Article in English | MEDLINE | ID: mdl-9882342

ABSTRACT

The live-attenuated human parainfluenza virus 3 (PIV3) cold-passage 45 (cp45) candidate vaccine was shown previously to be safe, immunogenic, and phenotypically stable in seronegative human infants. Previous findings indicated that each of the three amino acid substitutions in the L polymerase protein of cp45 independently confers the temperature-sensitive (ts) and attenuation (att) phenotypes but not the cold-adaptation (ca) phenotype (29). cp45 contains 12 additional potentially important point mutations in other proteins (N, C, M, F, and hemagglutinin-neuraminidase [HN]) or in cis-acting sequences (the leader region and the transcription gene start [GS] signal of the N gene), and their contribution to these phenotypes was undefined. To further characterize the genetic basis for the ts, ca, and att phenotypes of this promising vaccine candidate, we constructed, using a reverse genetics system, a recombinant cp45 virus that contained all 15 cp45-specific mutations mentioned above, and found that it was essentially indistinguishable from the biologically derived cp45 on the basis of plaque size, level of temperature sensitivity, cold adaptation, level of replication in the upper and lower respiratory tract of hamsters, and ability to protect hamsters from subsequent wild-type PIV3 challenge. We then constructed recombinant viruses containing the cp45 mutations in individual proteins as well as several combinations of mutations. Analysis of these recombinant viruses revealed that multiple cp45 mutations distributed throughout the genome contribute to the ts, ca, and att phenotypes. In addition to the mutations in the L gene, at least one other mutation in the 3' N region (i.e., including the leader, N GS, and N coding changes) contributes to the ts phenotype. A recombinant virus containing all the cp45 mutations except those in L was more ts than cp45, illustrating the complex nature of this phenotype. The ca phenotype of cp45 also is a complex composite phenotype, reflecting contributions of at least three separate genetic elements, namely, mutations within the 3' N region, the L protein, and the C-M-F-HN region. The att phenotype is a composite of both ts and non-ts mutations. Attenuating ts mutations are located in the L protein, and non-ts attenuating mutations are located in the C and F proteins. The presence of multiple ts and non-ts attenuating mutations in cp45 likely contributes to the high level of attenuation and phenotypic stability of this promising vaccine candidate.


Subject(s)
Mutation , Parainfluenza Virus 3, Human/genetics , Viral Vaccines/genetics , Adaptation, Biological , Animals , Cell Line , Cold Temperature , Cricetinae , Humans , Macaca mulatta , Parainfluenza Virus 3, Human/growth & development , Parainfluenza Virus 3, Human/immunology , Phenotype , Vaccines, Attenuated , Viral Plaque Assay , Viral Vaccines/immunology
SELECTION OF CITATIONS
SEARCH DETAIL