Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 341
Filter
1.
Methods Mol Biol ; 2556: 169-178, 2022.
Article in English | MEDLINE | ID: mdl-36175634

ABSTRACT

Human parainfluenza virus type 1 (hPIV1) and type 3 (hPIV3) are respiratory pathogen viruses that bind to terminal sialic acids of glycoconjugates on the cell surface hemagglutinin-neuraminidase glycoprotein. Sialic acid residues are linked to the galactose residue primarily by α2,3 or α2,6 linkages on the terminal of glycoprotein or glycolipids. One of the major determinants of pathogenicity or tissue tropism is virus binding or infection specificity for each sialyl linkage. Sialic linkage-modified human blood cells or mammalian cells that mainly have α2,3- or α2,6-linked sialic acid residues on the surface can be prepared by treatment with linkage-specific sialidases or sialyltransferases. These linkage-modified cells can be used in hemagglutination assays to estimate virus particles' binding specificity, hemadsorption assays to estimate virus glycoproteins' binding specificity, and virus infectivity assays. These methods contribute to identifying the specificity of sialic acid lineage recognition of the hPIV or other sialic acid-binding viruses.


Subject(s)
Parainfluenza Virus 1, Human , Paramyxoviridae Infections , Animals , Galactose , Glycolipids , HN Protein , Humans , Mammals , Membrane Glycoproteins , N-Acetylneuraminic Acid , Parainfluenza Virus 1, Human/genetics , Receptors, Virus , Sialyltransferases
2.
Influenza Other Respir Viruses ; 16(6): 1122-1132, 2022 11.
Article in English | MEDLINE | ID: mdl-35672928

ABSTRACT

BACKGROUND: Human Parainfluenza viruses (HPIV) comprise of four members of the genetically distinct genera of Respirovirus (HPIV1&3) and Orthorubulavirus (HPIV2&4), causing significant upper and lower respiratory tract infections worldwide, particularly in children. However, despite frequent molecular diagnosis, they are frequently considered collectively or with HPIV4 overlooked entirely. We therefore investigated clinical and viral epidemiological distinctions of the relatively less prevalent Orthorubulaviruses HPIV2&4 at a regional UK hospital across four autumn/winter epidemic seasons. METHODS: A retrospective audit of clinical features of all HPIV2 or HPIV4 RT-PCR-positive patients, diagnosed between 1st September 2013 and 12th April 2017 was undertaken, alongside sequencing of viral genome fragments in a representative subset of samples. RESULTS: Infection was observed across all age groups, but predominantly in children under nine and adults over 40, with almost twice as many HPIV4 as HPIV2 cases. Fever, abnormal haematology, elevated C-reactive protein and hospital admission were more frequently seen in HPIV2 than HPIV4 infection. Each of the four seasonal peaks of either HPIV2, HPIV4 or both, closely matched that of RSV, occurring in November and December and preceding that of Influenza A. A subset of viruses were partially sequenced, indicating co-circulation of multiple subtypes of both HPIV2&4, but with little variation between each epidemic season or from limited global reference sequences. CONCLUSIONS: Despite being closest known genetic relatives, our data indicates a potential difference in associated disease between HPIV2 and HPIV4, with more hospitalisation seen in HPIV2 mono-infected individuals, but a greater overall number of HPIV4 cases.


Subject(s)
Paramyxoviridae Infections , Respiratory Tract Infections , Adult , C-Reactive Protein , Child , Genomics , Humans , Molecular Epidemiology , Parainfluenza Virus 1, Human/genetics , Parainfluenza Virus 2, Human/genetics , Parainfluenza Virus 3, Human/genetics , Paramyxoviridae Infections/diagnosis , Paramyxoviridae Infections/epidemiology , Respiratory Tract Infections/epidemiology , Retrospective Studies , United Kingdom/epidemiology
3.
Zhonghua Yu Fang Yi Xue Za Zhi ; 56(2): 203-211, 2022 Feb 06.
Article in Chinese | MEDLINE | ID: mdl-35078303

ABSTRACT

Human parainfluenza viruses (HPIVs) is one of the main causes of acute respiratory tract infections in children. HPIVs have been grouped into four serotypes (HPIV1~HPIV4) according to serological and genetic variation. Different serotypes of HPIVs have diverse clinical disease spectrum, epidemic characteristics and disease burden. Based on the nucleotide variation in structural protein genes, HPIVs can be further divided into distinct genotypes and subtypes with diverse temporal and spatial distribution features. The standard molecular typing methods are helpful to clarify the gene evolution and transmission patterns of HPIVs in the process of population transmission. However, the development of molecular epidemiology of HPIVs has been hindered by the lack of a standardized molecular typing method worldwide. Therefore, this study reviewed the viral characteristics, genome structure, existing genotyping methods and evolution of HPIVs, and screened the reference strains for molecular typing, so as to improve the understanding of gene characteristics and molecular typing of HPIVs, and provide an important scientific basis for the monitoring and research of molecular epidemiology of HPIVs in China.


Subject(s)
Paramyxoviridae Infections , Respiratory Tract Infections , Child , Humans , Molecular Typing , Parainfluenza Virus 1, Human/genetics , Parainfluenza Virus 2, Human/genetics , Parainfluenza Virus 3, Human/genetics , Paramyxoviridae Infections/epidemiology , Respiratory Tract Infections/epidemiology
4.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-935239

ABSTRACT

Human parainfluenza viruses (HPIVs) is one of the main causes of acute respiratory tract infections in children. HPIVs have been grouped into four serotypes (HPIV1~HPIV4) according to serological and genetic variation. Different serotypes of HPIVs have diverse clinical disease spectrum, epidemic characteristics and disease burden. Based on the nucleotide variation in structural protein genes, HPIVs can be further divided into distinct genotypes and subtypes with diverse temporal and spatial distribution features. The standard molecular typing methods are helpful to clarify the gene evolution and transmission patterns of HPIVs in the process of population transmission. However, the development of molecular epidemiology of HPIVs has been hindered by the lack of a standardized molecular typing method worldwide. Therefore, this study reviewed the viral characteristics, genome structure, existing genotyping methods and evolution of HPIVs, and screened the reference strains for molecular typing, so as to improve the understanding of gene characteristics and molecular typing of HPIVs, and provide an important scientific basis for the monitoring and research of molecular epidemiology of HPIVs in China.


Subject(s)
Child , Humans , Molecular Typing , Parainfluenza Virus 1, Human/genetics , Parainfluenza Virus 2, Human/genetics , Parainfluenza Virus 3, Human/genetics , Paramyxoviridae Infections/epidemiology , Respiratory Tract Infections/epidemiology
5.
Viruses ; 13(6)2021 05 29.
Article in English | MEDLINE | ID: mdl-34072332

ABSTRACT

Human respiratory syncytial virus (HRSV), human metapneumovirus (HMPV), and human parainfluenza viruses (HPIVs) are leading causes of respiratory disease in young children, the elderly, and individuals of all ages with immunosuppression. Vaccination strategies against these pneumoviruses and paramyxoviruses are vast in number, yet no licensed vaccines are available. Here, we review development of Sendai virus (SeV), a versatile pediatric vaccine that can (a) serve as a Jennerian vaccine against HPIV1, (b) serve as a recombinant vaccine against HRSV, HPIV2, HPIV3, and HMPV, (c) accommodate foreign genes for viral glycoproteins in multiple intergenic positions, (d) induce durable, mucosal, B-cell, and T-cell immune responses without enhanced immunopathology, (e) protect cotton rats, African green monkeys, and chimpanzees from infection, and (f) be formulated into a vaccine cocktail. Clinical phase I safety trials of SeV have been completed in adults and 3-6-year-old children. Clinical testing of SeVRSV, an HRSV fusion (F) glycoprotein gene recombinant, has also been completed in adults. Positive results from these studies, and collaborative efforts with the National Institutes of Health and the Serum Institute of India assist advanced development of SeV-based vaccines. Prospects are now good for vaccine successes in infants and consequent protection against serious viral disease.


Subject(s)
Genetic Vectors/genetics , Respiratory Tract Infections/prevention & control , Respiratory Tract Infections/virology , Sendai virus/genetics , Viral Envelope Proteins/genetics , Viral Vaccines/genetics , Viruses/genetics , Animals , Antibodies, Viral/blood , Clinical Trials as Topic , Mice , Parainfluenza Virus 1, Human/genetics , Parainfluenza Virus 1, Human/immunology , Respiratory Syncytial Virus, Human/genetics , Respiratory Syncytial Virus, Human/immunology , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Viral Vaccines/immunology , Viruses/classification , Viruses/immunology
6.
Hum Vaccin Immunother ; 17(2): 554-559, 2021 02 01.
Article in English | MEDLINE | ID: mdl-32750273

ABSTRACT

SeVRSV is a replication-competent Sendai virus (SeV)-based vaccine carrying the respiratory syncytial virus (RSV) fusion protein (F) gene. Unmanipulated, non-recombinant SeV is a murine parainfluenza virus type 1 (PIV-1) and serves as a Jennerian vaccine for human PIV-1 (hPIV-1). SeV protects African green monkeys (AGM) from infection after hPIV-1 challenge. The recombinant SeVRSV additionally targets RSV and protects AGM from lower respiratory infections after RSV challenge. The present study is the first to report on the safety, viral genome detection, and immunogenicity following SeVRSV vaccination of healthy adults. Seventeen and four healthy adults received intranasal SeVRSV and PBS, respectively, followed by six months of safety monitoring. Virus genome (in nasal wash) and vaccine-specific antibodies (in sera) were monitored for two and four weeks, respectively, post-vaccination. The vaccine was well-tolerated with only mild to moderate reactions that were also present in the placebo group. No severe reactions occurred. As expected, due to preexisting immunity toward hPIV-1 and RSV in adults, vaccine genome detection was transient. There were minimal antibody responses to SeV and negligible responses to RSV F. Results encourage further studies of SeVRSV with progression toward a clinical trial in seronegative children. Abbreviations: AE-adverse event; SAE-serious adverse event; SeV-Sendai virus; RSV-respiratory syncytial virus; PIV-1-parainfluenza virus-type 1; hPIV-1-human parainfluenza virus-type 1; F-RSV fusion protein; SeVRSV-recombinant SeV carrying the RSV F gene; Ab-antibody; MSW-medically significant wheezing; NOCMC-new onset chronic medical condition, mITT-modified Intent to Treat; ALRI-acute lower respiratory tract infection.


Subject(s)
Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus Vaccines , Respiratory Syncytial Virus, Human , Adult , Animals , Antibodies, Viral , Chlorocebus aethiops , Humans , Immunogenicity, Vaccine , Parainfluenza Virus 1, Human/genetics , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Vaccines/adverse effects , Respiratory Syncytial Virus Vaccines/genetics , Respiratory Syncytial Virus, Human/genetics , Sendai virus/genetics , Viral Fusion Proteins/genetics
7.
PLoS One ; 15(12): e0243735, 2020.
Article in English | MEDLINE | ID: mdl-33315873

ABSTRACT

INTRODUCTION: Wheezing is a major problem in children, and respiratory viruses are often believed to be the causative agent. While molecular detection tools enable identification of respiratory viruses in wheezing children, it remains unclear if and how these viruses are associated with wheezing. The objective of this systematic review is to clarify the prevalence of different respiratory viruses in children with wheezing. METHODS: We performed an electronic in Pubmed and Global Index Medicus on 01 July 2019 and manual search. We performed search of studies that have detected common respiratory viruses in children ≤18 years with wheezing. We included only studies using polymerase chain reaction (PCR) assays. Study data were extracted and the quality of articles assessed. We conducted sensitivity, subgroup, publication bias, and heterogeneity analyses using a random effects model. RESULTS: The systematic review included 33 studies. Rhinovirus, with a prevalence of 35.6% (95% CI 24.6-47.3, I2 98.4%), and respiratory syncytial virus, at 31.0% (95% CI 19.9-43.3, I2 96.4%), were the most common viruses detected. The prevalence of other respiratory viruses was as follows: human bocavirus 8.1% (95% CI 5.3-11.3, I2 84.6%), human adenovirus 7.7% (95% CI 2.6-15.0, I2 91.0%), influenza virus6.5% (95% CI 2.2-12.6, I2 92.4%), human metapneumovirus5.8% (95% CI 3.4-8.8, I2 89.0%), enterovirus 4.3% (95% CI 0.1-12.9, I2 96.2%), human parainfluenza virus 3.8% (95% CI 1.5-6.9, I2 79.1%), and human coronavirus 2.2% (95% CI 0.6-4.4, I2 79.4%). CONCLUSIONS: Our results suggest that rhinovirus and respiratory syncytial virus may contribute to the etiology of wheezing in children. While the clinical implications of molecular detection of respiratory viruses remains an interesting question, this study helps to illuminate the potential of role respiratory viruses in pediatric wheezing. REVIEW REGISTRATION: PROSPERO, CRD42018115128.


Subject(s)
Respiratory Sounds/etiology , Respiratory Sounds/genetics , Respiratory Tract Infections/diagnosis , Bocavirus/genetics , Bocavirus/isolation & purification , Bocavirus/pathogenicity , Child , Child, Preschool , Coronavirus/isolation & purification , Coronavirus/pathogenicity , Humans , Orthomyxoviridae/genetics , Orthomyxoviridae/isolation & purification , Orthomyxoviridae/pathogenicity , Parainfluenza Virus 1, Human/genetics , Parainfluenza Virus 1, Human/isolation & purification , Parainfluenza Virus 1, Human/pathogenicity , Polymerase Chain Reaction , Respiratory Sounds/physiopathology , Respiratory System/pathology , Respiratory System/virology , Respiratory Tract Infections/genetics , Respiratory Tract Infections/virology
8.
Intern Med ; 59(1): 101-105, 2020 Jan 01.
Article in English | MEDLINE | ID: mdl-31511487

ABSTRACT

Two patients, a 76-year-old woman and 66-year-old woman, presented to our hospital with symptoms of lower respiratory tract infection. Both patients showed chest imaging findings of bilateral ground-glass opacities and consolidations. We initially suspected these patients of having influenza-associated pneumonia and cryptogenic organizing pneumonia, respectively, and performed bronchoalveolar lavage, but only human parainfluenza virus-1 infection was detected by multiplex polymerase chain reaction testing. These findings suggest that pneumonia due to human parainfluenza virus-1 should be included in the differential diagnosis of such cases.


Subject(s)
Bronchoalveolar Lavage Fluid/chemistry , Cryptogenic Organizing Pneumonia/diagnosis , Influenza, Human/diagnosis , Parainfluenza Virus 1, Human/genetics , Pneumonia, Viral/diagnostic imaging , RNA, Viral/analysis , Respirovirus Infections/diagnostic imaging , Aged , Bronchoalveolar Lavage Fluid/virology , Diagnosis, Differential , Female , Humans , Lung/diagnostic imaging , Lung/pathology , Multiplex Polymerase Chain Reaction , Pneumonia, Viral/pathology , Pneumonia, Viral/virology , Polymerase Chain Reaction , Respirovirus Infections/pathology , Respirovirus Infections/virology , Tomography, X-Ray Computed
9.
PLoS One ; 14(7): e0220057, 2019.
Article in English | MEDLINE | ID: mdl-31318956

ABSTRACT

Human Parainfluenza viruses (HPIV) type 1 and 3 are important causes of respiratory tract infections in young children globally. HPIV infections do not confer complete protective immunity so reinfections occur throughout life. Since no effective vaccine is available for the two virus subtypes, comprehensive understanding of HPIV-1 and HPIV-3 genetic and epidemic features is important for diagnosis, prevention, and treatment of HPIV-1 and HPIV-3 infections. Relatively few whole genome sequences are available for both HPIV-1 and HPIV-3 viruses, so our study sought to provide whole genome sequences from multiple countries to further the understanding of the global diversity of HPIV at a whole-genome level. We collected HPIV-1 and HPIV-3 samples and isolates from Argentina, Australia, France, Mexico, South Africa, Switzerland, and USA from the years 2003-2011 and sequenced the genomes of 40 HPIV-1 and 75 HPIV-3 viruses with Sanger and next-generation sequencing with the Ion Torrent, Illumina, and 454 platforms. Phylogenetic analysis showed that the HPIV-1 genome is evolving at an estimated rate of 4.97 × 10-4 mutations/site/year (95% highest posterior density 4.55 × 10-4 to 5.38 × 10-4) and the HPIV-3 genome is evolving at a similar rate (3.59 × 10-4 mutations/site/year, 95% highest posterior density 3.26 × 10-4 to 3.94 × 10-4). There were multiple genetically distinct lineages of both HPIV-1 and 3 circulating on a global scale. Further surveillance and whole-genome sequencing are greatly needed to better understand the spatial dynamics of these important respiratory viruses in humans.


Subject(s)
Genome, Viral , Genomics , Parainfluenza Virus 1, Human/genetics , Parainfluenza Virus 3, Human/genetics , Evolution, Molecular , Genomics/methods , High-Throughput Nucleotide Sequencing , Humans , Phylogeny , Recombination, Genetic , Selection, Genetic , Sequence Analysis, DNA
10.
Viruses ; 11(5)2019 05 05.
Article in English | MEDLINE | ID: mdl-31060278

ABSTRACT

Human parainfluenza virus (hPIV) infections are a major cause of respiratory tract illnesses in children, with currently no available vaccine or drug treatment. The surface glycoprotein haemagglutinin-neuraminidase (HN) of hPIV has a central role in the viral life cycle, including neuraminic acid-recognising receptor binding activity (early stage) and receptor-destroying activity (late stage), which makes it an ideal target for antiviral drug disovery. In this study, we showed that targeting the catalytic mechanism of hPIV-1 HN by a 2α,3ß-difluoro derivative of the known hPIV-1 inhibitor, BCX 2798, produced more potent inhibition of the neuraminidase function which is reflected by a stronger inhibition of viral replication. The difluorosialic acid-based inhibitor efficiently blocked the neuraminidase activity of HN for a prolonged period of time relative to its unsaturated neuraminic acid (Neu2en) analogue, BCX 2798 and produced a more efficient inhibition of the HN neuraminidase activity as well as in vitro viral replication. This prolonged inhibition of the hPIV-1 HN protein suggests covalent binding of the inhibitor to a key catalytic amino acid, making this compound a new lead for a novel class of more potent hPIV-1 mechanism-based inhibitors.


Subject(s)
Enzyme Inhibitors/chemistry , HN Protein/chemistry , Parainfluenza Virus 1, Human/enzymology , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Azides/chemistry , Azides/pharmacology , Biocatalysis , Enzyme Inhibitors/pharmacology , HN Protein/genetics , HN Protein/metabolism , Hexuronic Acids/chemistry , Hexuronic Acids/pharmacology , Humans , Parainfluenza Virus 1, Human/drug effects , Parainfluenza Virus 1, Human/genetics , Respirovirus Infections/virology , Virus Replication/drug effects
11.
Antiviral Res ; 167: 89-97, 2019 07.
Article in English | MEDLINE | ID: mdl-30951732

ABSTRACT

Human parainfluenza viruses cause acute respiratory tract infections and disease predominantly in young children and immunocompromised individuals. Currently, there are no vaccines to prevent hPIV infections, nor licensed anti-hPIV drugs. There is therefore a need for specific antiviral therapies to decrease the morbidity and mortality associated with hPIV diseases. Haemagglutinin-neuraminidase (HN) is one of two hPIV surface proteins with critical roles in host receptor recognition, binding and cleavage; it has been explored as a key drug development target for the past few decades with variable success. Recent advancements in computational modelling and the availability of the X-ray crystal structure of hPIV3 HN have improved our understanding of the structural and mechanistic features of HN. This review explores structural features of the HN protein that are being exploited for structure-guided inhibitor design. We describe past and present hPIV HN inhibition strategies based on sialic acid scaffolds, together with other novel approaches that decrease hPIV infectivity. Although many HN inhibitors have been developed and evaluated as anti-hPIV agents, currently only a host-directed therapy (DAS181) has succeeded in phase II clinical drug trials. Hence, the review concludes with future considerations for targeting the specific function(s) of hPIV HN and suggestions for antiviral drug design.


Subject(s)
Enzyme Inhibitors/pharmacology , HN Protein , N-Acetylneuraminic Acid/analogs & derivatives , Neuraminidase/antagonists & inhibitors , Paramyxoviridae Infections/drug therapy , Antiviral Agents/chemical synthesis , Antiviral Agents/pharmacology , Child , Child, Preschool , Drug Delivery Systems/methods , Drug Design , Drug Resistance, Viral/drug effects , Enzyme Inhibitors/chemical synthesis , Genome, Viral , HN Protein/chemistry , HN Protein/genetics , HN Protein/metabolism , Humans , Immunocompromised Host , N-Acetylneuraminic Acid/chemical synthesis , N-Acetylneuraminic Acid/pharmacology , Parainfluenza Virus 1, Human/drug effects , Parainfluenza Virus 1, Human/genetics , Parainfluenza Virus 3, Human/drug effects , Parainfluenza Virus 3, Human/genetics , Paramyxoviridae Infections/pathology , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Internalization/drug effects
12.
Proc Natl Acad Sci U S A ; 115(48): 12265-12270, 2018 11 27.
Article in English | MEDLINE | ID: mdl-30420505

ABSTRACT

Parainfluenza virus types 1-4 (PIV1-4) are highly infectious human pathogens, of which PIV3 is most commonly responsible for severe respiratory illness in newborns, elderly, and immunocompromised individuals. To obtain a vaccine effective against all four PIV types, we engineered mutations in each of the four PIV fusion (F) glycoproteins to stabilize their metastable prefusion states, as such stabilization had previously enabled the elicitation of high-titer neutralizing antibodies against the related respiratory syncytial virus. A cryoelectron microscopy structure of an engineered PIV3 F prefusion-stabilized trimer, bound to the prefusion-specific antibody PIA174, revealed atomic-level details for how introduced mutations improved stability as well as how a single PIA174 antibody recognized the trimeric apex of prefusion PIV3 F. Nine combinations of six newly identified disulfides and two cavity-filling mutations stabilized the prefusion PIV3 F immunogens and induced 200- to 500-fold higher neutralizing titers in mice than were elicited by PIV3 F in the postfusion conformation. For PIV1, PIV2, and PIV4, we also obtained stabilized prefusion Fs, for which prefusion versus postfusion titers were 2- to 20-fold higher. Elicited murine responses were PIV type-specific, with little cross-neutralization of other PIVs. In nonhuman primates (NHPs), quadrivalent immunization with prefusion-stabilized Fs from PIV1-4 consistently induced potent neutralizing responses against all four PIVs. For PIV3, the average elicited NHP titer from the quadrivalent immunization was more than fivefold higher than any titer observed in a cohort of over 100 human adults, highlighting the ability of a prefusion-stabilized immunogen to elicit especially potent neutralization.


Subject(s)
Parainfluenza Virus 1, Human/immunology , Parainfluenza Virus 2, Human/immunology , Parainfluenza Virus 3, Human/immunology , Parainfluenza Virus 4, Human/immunology , Respirovirus Infections/immunology , Viral Fusion Proteins/chemistry , Viral Vaccines/chemistry , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Cryoelectron Microscopy , Female , Humans , Macaca mulatta , Male , Mice , Parainfluenza Virus 1, Human/chemistry , Parainfluenza Virus 1, Human/genetics , Parainfluenza Virus 2, Human/chemistry , Parainfluenza Virus 2, Human/genetics , Parainfluenza Virus 3, Human/chemistry , Parainfluenza Virus 3, Human/genetics , Parainfluenza Virus 4, Human/chemistry , Parainfluenza Virus 4, Human/genetics , Respiratory Syncytial Virus Infections , Respirovirus Infections/prevention & control , Respirovirus Infections/virology , Viral Fusion Proteins/administration & dosage , Viral Fusion Proteins/genetics , Viral Fusion Proteins/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/genetics , Viral Vaccines/immunology
13.
Sci Rep ; 8(1): 6833, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29717150

ABSTRACT

HPIVs are serologically and genetically grouped into four species that account for up to 10% of all hospitalizations due to acute respiratory infection in children under the age of five. Genetic and epidemiological data for the four HPIVs derived from two pediatric cohorts in Viet Nam are presented. Respiratory samples were screened for HPIV1-4 by real-time PCR. Demographic and clinical data of patients infected with different HPIV were compared. We used a hemi-nested PCR approach to generate viral genome sequences from HPIV-positive samples and conducted a comprehensive phylogenetic analysis. In total, 170 samples tested positive for HPIV. HPIV3 was most commonly detected in our cohort and 80 co-detections of HPIV with other respiratory viruses were found. Phylogenetic analyses suggest local endemic circulation as well as punctuated introductions of new HPIV lineages. Viral gene flow analysis revealed that Viet Nam is a net importer of viral genetic diversity. Epidemiological analyses imply similar disease severity for all HPIV species. HPIV sequences from Viet Nam formed local clusters and were interspersed with sequences from diverse geographic regions. Combined, this new knowledge will help to investigate global HPIV circulation patterns in more detail and ultimately define more suitable vaccine strains.


Subject(s)
Molecular Epidemiology/methods , Parainfluenza Virus 1, Human/genetics , Parainfluenza Virus 2, Human/genetics , Parainfluenza Virus 3, Human/genetics , Parainfluenza Virus 4, Human/genetics , Respiratory Tract Infections/epidemiology , Respirovirus Infections/epidemiology , Acute Disease , Adolescent , Chi-Square Distribution , Child , Child, Preschool , Female , Follow-Up Studies , Genetic Variation , Genome, Viral , Humans , Infant , Infant, Newborn , Male , Phylogeny , Real-Time Polymerase Chain Reaction , Severity of Illness Index , Statistics, Nonparametric , Vietnam/epidemiology , Whole Genome Sequencing
14.
Food Environ Virol ; 10(2): 133-140, 2018 06.
Article in English | MEDLINE | ID: mdl-29196954

ABSTRACT

The aim of this study was to evaluate the potential role of office fomites in respiratory (human parainfluenza virus 1-HPIV1, human parainfluenza virus 3-HPIV3) and enteric (norovirus GI-NoV GI, norovirus GII-NoV GII) viruses transmission by assessing the occurrence of these viruses on surfaces in office buildings. Between 2016 and 2017, a total of 130 surfaces from open-space and non-open-space rooms in office buildings located in one city were evaluated for HPIV1, HPIV3, NoV GI, and NoV GII viral RNA presence. Detection of viruses was performed by RT-qPCR method. Study revealed 27 positive samples, among them 59.3% were HPIV3-positive, 25.9% HPIV1-positive, and 14.8% NoV GII-positive. All tested surfaces were NoV GI-negative. Statistical analysis of obtained data showed that the surfaces of office equipment including computer keyboards and mice, telephones, and desktops were significantly more contaminated with respiratory viruses than the surfaces of building equipment elements such as door handles, light switches, or ventilation tracts (χ 2 p = 0.006; Fisher's Exact p = 0.004). All examined surfaces were significantly more contaminated with HPIVs than NoVs (χ 2 p = 0.002; Fisher's Exact p = 0.003). Office fomites in open-space rooms were more often contaminated with HPIVs than with NoVs (χ 2 p = 0.016; Fisher's Exact p = 0.013). The highest average concentration of HPIVs RNA copies was observed on telephones (1.66 × 102 copies/100 cm2), while NoVs on the light switches (1.40 × 102 copies/100 cm2). However, the Kruskal-Wallis test did not show statistically significant differences in concentration levels of viral RNA copies on surfaces between the all tested samples. This study unequivocally showed that individuals in office environment may have contact with both respiratory and enteric viral particles present on frequently touched surfaces.


Subject(s)
Caliciviridae Infections/virology , Fomites/virology , Norovirus/isolation & purification , Parainfluenza Virus 1, Human/isolation & purification , Parainfluenza Virus 3, Human/isolation & purification , Respirovirus Infections/virology , Caliciviridae Infections/transmission , Genome, Viral/genetics , Humans , Norovirus/genetics , Parainfluenza Virus 1, Human/genetics , Parainfluenza Virus 3, Human/genetics , Prevalence , RNA, Viral/genetics , RNA, Viral/isolation & purification , Respirovirus Infections/transmission , Reverse Transcriptase Polymerase Chain Reaction
15.
Indian J Med Res ; 145(2): 189-193, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28639594

ABSTRACT

BACKGROUND & OBJECTIVES: Wheezing is a common problem in children under five with acute respiratory infections (ARIs). Viruses are known to be responsible for a considerable proportion of ARIs in children. This study was undertaken to know the viral aetiology of wheezing among the children less than five years of age, admitted to a tertiary care hospital in eastern India. METHODS: Seventy five children, under the age of five years admitted with wheezing, were included in the study. Throat and nasal swabs were collected, and real-time multiplex polymerase chain reaction (PCR) assay was used to screen for influenza 1 and 2, respiratory syncytial virus (RSV), parainfluenza virus (PIV) 1, 2, 3 and 4, rhinovirus, human meta-pneumovirus, bocavirus (HBoV), Coronavirus, adenovirus, Enterovirus and Parechovirus. RESULTS: The total viral detection rate was 28.57 per cent. Viral RNA markers were detected from children diagnosed to be having pneumonia (3 cases), bronchiolitis (9 cases), episodic wheeze (2 cases) and multitrigger wheeze (6 cases). RSV was the most common virus (35%) followed by PIV1, 2 and 3 (20%), HBoV (10%) and rhinovirus (5%). However, mixed infection was observed in 30 per cent of cases. INTERPRETATION & CONCLUSIONS: The study reported the presence of respiratory viral agents in 28.57 per cent of children with wheezing; RSV and PIV were most common, accounting to 55 per cent of the total cases. Mixed infection was reported in 30 per cent of cases. Seasonal variation in the occurrence of these viruses was also noted. Further studies need to be done with a large sample and longer follow up period to verify these findings.


Subject(s)
Coinfection/virology , Influenza, Human/virology , Respiratory Sounds/physiopathology , Respiratory Tract Infections/virology , Child , Child, Preschool , Coinfection/genetics , Female , Humans , India , Infant , Infant, Newborn , Influenza, Human/epidemiology , Influenza, Human/genetics , Male , Parainfluenza Virus 1, Human/genetics , Parainfluenza Virus 1, Human/isolation & purification , Parainfluenza Virus 1, Human/pathogenicity , Respiratory Sounds/etiology , Respiratory Syncytial Virus, Human/genetics , Respiratory Syncytial Virus, Human/isolation & purification , Respiratory Syncytial Virus, Human/pathogenicity , Respiratory Tract Infections/epidemiology , Respiratory Tract Infections/physiopathology , Rhinovirus/genetics , Rhinovirus/isolation & purification , Rhinovirus/pathogenicity
16.
J Med Microbiol ; 66(2): 160-168, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28266286

ABSTRACT

PURPOSE: To genetically explore the fusion protein gene (F) in human parainfluenza virus type 1 (HPIV1) and type 3 (HPIV3) strains, we analysed them in patients with acute respiratory infections in Eastern Japan from 2011 to 2015. METHODOLOGY: We constructed phylogenetic trees based on the HPIV and HPIV3 F gene using the maximum likelihood method and conducted P-distance and selective pressure analyses. We also predicted the linear epitopes of the protein in the prototype strains. Furthermore, we mapped the amino acid substitutions of the proteins. RESULTS: Nineteen strains of HPIV1 and 53 strains of HPIV3 were detected among the clinical acute respiratory infection cases. The phylogenetic trees indicated that the HPIV1 and HPIV3 strains were classified into clusters II and III and cluster C, respectively. The P-distance values of the HPIV1 and HPIV3 F genes were <0.03. Two positive selection sites were inferred in the HPIV1 (aa 8 and aa 10), and one positive selection site was inferred in the HPIV3 (aa 108), but over 10 negative selection sites were inferred. Four epitopes were predicted for the HPIV1 prototype strains, while five epitopes were predicted for the HPIV3 prototype strain. A positive selection site (aa 108) or the HPIV3 F protein was involved in the predicted epitope. Additionally, we found that an amino acid substitution (R73K) in the LC76627 HPIV3 strain presumably may affect the resistance to neutralization by antibodies. CONCLUSION: The F gene of HPIV1 and HPIV3 was relatively well conserved in the eastern part of Japan during the investigation period.


Subject(s)
Parainfluenza Virus 1, Human/genetics , Parainfluenza Virus 3, Human/genetics , Respiratory Tract Infections/epidemiology , Respirovirus Infections/epidemiology , Viral Fusion Proteins/genetics , Acute Disease , Adolescent , Adult , Aged , Aged, 80 and over , Amino Acid Substitution , Child , Child, Preschool , Epitopes/genetics , Epitopes/metabolism , Female , Humans , Infant , Japan/epidemiology , Likelihood Functions , Male , Middle Aged , Parainfluenza Virus 1, Human/isolation & purification , Parainfluenza Virus 3, Human/isolation & purification , Phylogeny , RNA, Viral/genetics , RNA, Viral/isolation & purification , Respiratory Tract Infections/virology , Respirovirus Infections/virology , Sequence Analysis, RNA , Viral Fusion Proteins/metabolism , Young Adult
17.
J Virol ; 91(10)2017 05 15.
Article in English | MEDLINE | ID: mdl-28250127

ABSTRACT

The recent 2014-2016 Ebola virus (EBOV) outbreak prompted increased efforts to develop vaccines against EBOV disease. We describe the development and preclinical evaluation of an attenuated recombinant human parainfluenza virus type 1 (rHPIV1) expressing the membrane-anchored form of EBOV glycoprotein GP, as an intranasal (i.n.) EBOV vaccine. GP was codon optimized and expressed either as a full-length protein or as an engineered chimeric form in which its transmembrane and cytoplasmic tail (TMCT) domains were replaced with those of the HPIV1 F protein in an effort to enhance packaging into the vector particle and immunogenicity. GP was inserted either preceding the N gene (pre-N) or between the N and P genes (N-P) of rHPIV1 bearing a stabilized attenuating mutation in the P/C gene (CΔ170). The constructs grew to high titers and efficiently and stably expressed GP. Viruses were attenuated, replicating at low titers over several days, in the respiratory tract of African green monkeys (AGMs). Two doses of candidates expressing GP from the pre-N position elicited higher GP neutralizing serum antibody titers than the N-P viruses, and unmodified GP induced higher levels than its TMCT counterpart. Unmodified EBOV GP was packaged into the HPIV1 particle, and the TMCT modification did not increase packaging or immunogenicity but rather reduced the stability of GP expression during in vivo replication. In conclusion, we identified an attenuated and immunogenic i.n. vaccine candidate expressing GP from the pre-N position. It is expected to be well tolerated in humans and is available for clinical evaluation.IMPORTANCE EBOV hemorrhagic fever is one of the most lethal viral infections and lacks a licensed vaccine. Contact of fluids from infected individuals, including droplets or aerosols, with mucosal surfaces is an important route of EBOV spread during a natural outbreak, and aerosols also might be exploited for intentional virus spread. Therefore, vaccines that protect against mucosal as well as systemic inoculation are needed. We evaluated a version of human parainfluenza virus type 1 (HPIV1) bearing a stabilized attenuating mutation in the P/C gene (CΔ170) as an intranasal vaccine vector to express the EBOV glycoprotein GP. We evaluated expression from two different genome positions (pre-N and N-P) and investigated the use of vector packaging signals. African green monkeys immunized with two doses of the vector expressing GP from the pre-N position developed high titers of GP neutralizing serum antibodies. The attenuated vaccine candidate is expected to be safe and immunogenic and is available for clinical development.


Subject(s)
Ebola Vaccines/genetics , Ebola Vaccines/immunology , Ebolavirus/chemistry , Hemorrhagic Fever, Ebola/prevention & control , Parainfluenza Virus 1, Human/genetics , Viral Envelope Proteins/genetics , Administration, Intranasal , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Chlorocebus aethiops , Ebola Vaccines/administration & dosage , Ebolavirus/genetics , Ebolavirus/immunology , Genetic Vectors , Glycoproteins/genetics , Glycoproteins/immunology , Hemorrhagic Fever, Ebola/immunology , Humans , Respiratory System/virology , Vaccination , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/chemistry , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Virus Replication
18.
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
19.
Intervirology ; 60(5): 181-189, 2017.
Article in English | MEDLINE | ID: mdl-29510403

ABSTRACT

BACKGROUND: The families Paramyxoviridae and Pneumoviridae comprise a broad spectrum of viral pathogens that affect human health. The matrix (M) protein of these viruses has a central role in their life cycle. In line with this, molecular characteristics of the M proteins from variable viruses that circulated in Croatia were investigated. METHODS: Sequences of the M proteins of human parainfluenza virus (HPIV) 1-3 within the family Paramyxoviridae, human metapneumovirus (HMPV), and human respiratory syncytial virus from the family Pneumoviridae were obtained and analyzed. RESULTS: M proteins were very diverse among HPIVs, but highly conserved within each virus. More variability was seen in nucleotide sequences of M proteins from the Pneumoviridae family. An insertion of 8 nucleotides in the 3' untranslated region in 1 HMPV M gene sequence was discovered (HR347-12). As there are no samples with such an insertion in the database, this insertion is of interest and requires further research. CONCLUSION: While we have confirmed that M proteins were conserved among individual viruses, any changes that are observed should be given attention and further researched. Of special interest is inclusion of HPIV2 M proteins in this analysis, as these proteins have not been studied to the same extent as other paramyxoviruses.


Subject(s)
Metapneumovirus/genetics , Parainfluenza Virus 1, Human/genetics , RNA, Viral/genetics , Respiratory Syncytial Viruses/genetics , Viral Matrix Proteins/genetics , Amino Acid Sequence , Animals , Chlorocebus aethiops , Gene Expression , Genetic Variation , High-Throughput Nucleotide Sequencing , Humans , Metapneumovirus/isolation & purification , Metapneumovirus/metabolism , Parainfluenza Virus 1, Human/isolation & purification , Parainfluenza Virus 1, Human/metabolism , Paramyxoviridae Infections/virology , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Viruses/isolation & purification , Respiratory Syncytial Viruses/metabolism , Respirovirus Infections/virology , Sequence Alignment , Sequence Homology, Amino Acid , Vero Cells
20.
Bratisl Lek Listy ; 118(12): 772-776, 2017.
Article in English | MEDLINE | ID: mdl-29322811

ABSTRACT

OBJECTIVES: The aim of this study was to develop an in-house multiplex reverse transcription polymerase chain reaction (mRT-PCR), which can recognize HPIV1-4 in clinical samples. BACKGROUND: Human parainfluenza virus (HPIV) is one of the major causes of viral respiratory infections and can affect people at any age, especially infants and young children. METHODS: Four sets of specific primers targeting conserved areas of hemagglutinin-neuraminidase (HN) genes of HPIV1-4, were designed and tested with type-related plasmid controls. Specificity and sensitivity of mPCR were tested. One-step mRT-PCR was set up using a viral panel containing 10 respiratory viruses, including HPIVs. One hundred nasopharyngeal samples of respiratory infection patients were tested using the set One-step mRT-PCR. RESULTS: The specificity of set mPCR for HPIV1-4 using plasmid positive controls was proved and reaction sensitivity was measured. The specificity of set mRT-PCR was confirmed and 4 and 5 out of 100 clinical samples were HPIV1 and HPIV2 positive, respectively. CONCLUSION: The developed one-step mRT-PCR in this study is an effective and specific assay for clinical diagnosis of HPIV1 to 4 (Tab. 1, Fig. 6, Ref. 28).


Subject(s)
Paramyxoviridae/genetics , Respiratory Tract Infections/diagnosis , Respirovirus Infections/diagnosis , Rubulavirus Infections/diagnosis , Child , Child, Preschool , DNA Primers , Humans , Infant , Influenza, Human , Multiplex Polymerase Chain Reaction , Parainfluenza Virus 1, Human/genetics , Parainfluenza Virus 2, Human/genetics , Parainfluenza Virus 3, Human/genetics , Parainfluenza Virus 4, Human/genetics , Paramyxoviridae Infections/diagnosis , Paramyxoviridae Infections/virology , Respiratory Tract Infections/virology , Respirovirus Infections/virology , Reverse Transcriptase Polymerase Chain Reaction , Rubulavirus Infections/virology , Sensitivity and Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...