Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 692
Filter
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.
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
3.
Front Immunol ; 11: 575977, 2020.
Article in English | MEDLINE | ID: mdl-33123159

ABSTRACT

Human Parainfluenza Virus-3 (HPIV3) causes severe respiratory illness in immunocompromised patients and lacks approved anti-viral therapies. A phase I study of adoptively transferred virus-specific T-cells (VSTs) targeting HPIV3 following bone marrow transplantation is underway (NCT03180216). We sought to identify immunodominant epitopes within HPIV3 Matrix protein and their cross-reactivity against related viral proteins. VSTs were generated from peripheral blood of healthy donors by ex-vivo expansion after stimulation with a 15-mer peptide library encompassing HPIV3 matrix protein. Epitope mapping was performed using IFN-γ ELIspot with combinatorial peptide pools. Flow cytometry was used to characterize products with intracellular cytokine staining. In 10 VST products tested, we discovered 12 novel immunodominant epitopes. All products recognized an epitope at the C-terminus. On IFN-γ ELISpot, individual peptides eliciting activity demonstrated mean IFN-γ spot forming units per well (SFU)/1x105 cells of 115.5 (range 24.5-247.5). VST products were polyfunctional, releasing IFN-γ and TNF-α in response to identified epitopes, which were primarily HLA Class II restricted. Peptides from Human Parainfluenza Virus-1 corresponding to the HPIV3 epitopes showed cross-reactivity for HPIV1 in 11 of 12 tested epitopes (mean cross reactivity index: 1.19). Characterization of HPIV3 epitopes may enable development of third-party VSTs to treat immune suppressed patients with HPIV infection.


Subject(s)
Adoptive Transfer , Immunodominant Epitopes , Parainfluenza Virus 1, Human/immunology , Parainfluenza Virus 3, Human/immunology , Respirovirus Infections/therapy , T-Lymphocytes/transplantation , Viral Matrix Proteins/immunology , Cells, Cultured , Clinical Trials, Phase I as Topic , Cross Reactions , Enzyme-Linked Immunospot Assay , Epitope Mapping , Host-Pathogen Interactions , Humans , Interferon-gamma/metabolism , Interferon-gamma Release Tests , Parainfluenza Virus 1, Human/pathogenicity , Parainfluenza Virus 3, Human/pathogenicity , Respirovirus Infections/immunology , Respirovirus Infections/metabolism , Respirovirus Infections/virology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
4.
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
5.
Sci Rep ; 8(1): 12533, 2018 08 22.
Article in English | MEDLINE | ID: mdl-30135435

ABSTRACT

The bat paramyxovirus B16-40 was first isolated in Korea in this study. Using the isolated virus, we could obtain not only genomic information, but also several biological characteristics of the virus. In the phylogenetic analysis, the virus was found to belong to the recently proposed genus Shaanvirus. Through sequence analyses and in vitro testing, the isolated virus was also found to have haemagglutinin-neuraminidase (HN) protein as one of the structural proteins. When mouse antiserum was generated against the isolated virus and tested, it was cross-reactive to human parainfluenza virus 1 in an indirect immunofluorescence assay but could not cross-neutralize human parainfluenza virus 1. In addition, the bat paramyxovirus B16-40 was not infectious in the mouse model. Collectively, this study provided basic information on further classification of the bat paramyxovirus B16-40 and related viruses in the proposed genus Shaanvirus.


Subject(s)
Chiroptera/virology , Paramyxoviridae/genetics , Paramyxoviridae/immunology , Phylogeny , Animals , Cross Reactions , Female , Mice, Inbred C57BL , Neuraminidase/genetics , Neutralization Tests , Parainfluenza Virus 1, Human/immunology , Paramyxoviridae/isolation & purification , Paramyxoviridae/pathogenicity , Paramyxoviridae Infections/veterinary , Republic of Korea
6.
PLoS One ; 12(7): e0181425, 2017.
Article in English | MEDLINE | ID: mdl-28742120

ABSTRACT

Asthma exacerbation can be a life-threatening condition, and is most often triggered by common respiratory viruses. Poor asthma control and worsening of respiratory function is associated with increased airway inflammation, including eosinophilia. Prevention of asthma exacerbation relies on treatment with corticosteroids, which preferentially inhibit allergic inflammation like eosinophils. Human studies demonstrate that inactivated virus can trigger eosinophil activation in vitro through antigen presentation and memory CD4+ lymphocytes. We hypothesized that animals with immunologic memory to a respiratory virus would also develop airway hyperresponsiveness in response to a UV-inactivated form of the virus if they have pre-existing allergic airway inflammation. Guinea pigs were ovalbumin-sensitized, infected with live parainfluenza virus (PIV), aerosol-challenged with ovalbumin, and then re-inoculated 60 days later with live or UV-inactivated PIV. Some animals were either treated with dexamethasone prior to the second viral exposure. Lymphocytes were isolated from parabronchial lymph nodes to confirm immunologic memory to the virus. Airway reactivity was measured and inflammation was assessed using bronchoalveolar lavage and lung histology. The induction of viral immunologic memory was confirmed in infected animals. Allergen sensitized and challenged animals developed airway hyperreactivity with eosinophilic airway inflammation when re-exposed to UV-inactivated PIV, while non-sensitized animals did not. Airway hyperreactivity in the sensitized animals was inhibited by pre-treatment with dexamethasone. We suggest that the response of allergic inflammation to virus antigen is a significant factor causing asthma exacerbation. We propose that this is one mechanism explaining how corticosteroids prevent virus-induced asthma attack.


Subject(s)
Asthma/virology , Parainfluenza Virus 1, Human/immunology , Respiratory Hypersensitivity/virology , Respirovirus Infections/complications , Animals , Anti-Inflammatory Agents/therapeutic use , Asthma/drug therapy , Asthma/immunology , Dexamethasone/therapeutic use , Disease Models, Animal , Female , Guinea Pigs , Humans , Immunologic Memory/drug effects , Inflammation/drug therapy , Inflammation/immunology , Inflammation/virology , Lymphocytes/immunology , Lymphocytes/virology , Respiratory Hypersensitivity/drug therapy , Respiratory Hypersensitivity/immunology , Respirovirus Infections/drug therapy , Respirovirus Infections/immunology
7.
J Virol ; 89(20): 10319-32, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26223633

ABSTRACT

UNLABELLED: Live attenuated recombinant human parainfluenza virus type 1 (rHPIV1) was investigated as a vector to express the respiratory syncytial virus (RSV) fusion (F) glycoprotein, to provide a bivalent vaccine against RSV and HPIV1. The RSV F gene was engineered to include HPIV1 transcription signals and inserted individually into three gene locations in each of the two attenuated rHPIV1 backbones. Each backbone contained a single previously described attenuating mutation that was stabilized against deattenuation, specifically, a non-temperature-sensitive deletion mutation involving 6 nucleotides in the overlapping P/C open reading frames (ORFs) (C(Δ170)) or a temperature-sensitive missense mutation in the L ORF (L(Y942A)). The insertion sites in the genome were pre-N (F1), N-P (F2), or P-M (F3) and were identical for both backbones. In vitro, the presence of the F insert reduced the rate of virus replication, but the final titers were the same as the final titer of wild-type (wt) HPIV1. High levels of RSV F expression in cultured cells were observed with rHPIV1-C(Δ170)-F1, -F2, and -F3 and rHPIV1-L(Y942A)-F1. In hamsters, the rHPIV1-C(Δ170)-F1, -F2, and -F3 vectors were moderately restricted in the nasal turbinates, highly restricted in lungs, and genetically stable in vivo. Among the C(Δ170) vectors, the F1 virus was the most immunogenic and protective against wt RSV challenge. The rHPIV1-L(Y942A) vectors were highly restricted in vivo and were not detectably immunogenic or protective, indicative of overattenuation. The C(Δ170)-F1 construct appears to be suitably attenuated and immunogenic for further development as a bivalent intranasal pediatric vaccine. IMPORTANCE: There are no vaccines for the pediatric respiratory pathogens RSV and HPIV. We are developing live attenuated RSV and HPIV vaccines for use in virus-naive infants. Live attenuated RSV strains in particular are difficult to develop due to their poor growth and physical instability, but these obstacles could be avoided by the use of a vaccine vector. We describe the development and preclinical evaluation of live attenuated rHPIV1 vectors expressing the RSV F protein. Two different attenuated rHPIV1 backbones were each engineered to express RSV F from three different gene positions. The rHPIV1-C(Δ170)-F1 vector, bearing an attenuating deletion mutation (C(Δ170)) in the P/C gene and expressing RSV F from the pre-N position, was attenuated, stable, and immunogenic against the RSV F protein and HPIV1 in the hamster model and provided substantial protection against RSV challenge. This study provides a candidate rHPIV1-RSV-F vaccine virus suitable for continued development as a bivalent vaccine against two major childhood pathogens.


Subject(s)
Antibodies, Viral/biosynthesis , Respiratory Syncytial Virus Infections/prevention & control , Respirovirus Infections/prevention & control , Vaccination , Viral Fusion Proteins/immunology , Viral Vaccines/immunology , Animals , Base Sequence , Cricetulus , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Humans , Molecular Sequence Data , Parainfluenza Virus 1, Human/genetics , Parainfluenza Virus 1, Human/immunology , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus, Human/genetics , Respiratory Syncytial Virus, Human/immunology , Respirovirus Infections/immunology , Respirovirus Infections/virology , Sequence Deletion , Vaccines, Attenuated , Vaccines, Synthetic , Viral Fusion Proteins/genetics , Viral Vaccines/administration & dosage , Viral Vaccines/genetics , Virus Replication
8.
Clin Vaccine Immunol ; 22(3): 298-303, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25552633

ABSTRACT

Human parainfluenza virus type 1 (hPIV-1) is the most common cause of laryngotracheobronchitis (croup), resulting in tens of thousands of hospitalizations each year in the United States alone. No licensed vaccine is yet available. We have developed murine PIV-1 (Sendai virus [SeV]) as a live Jennerian vaccine for hPIV-1. Here, we describe vaccine testing in healthy 3- to 6-year-old hPIV-1-seropositive children in a dose escalation study. One dose of the vaccine (5 × 10(5), 5 × 10(6), or 5 × 10(7) 50% egg infectious doses) was delivered by the intranasal route to each study participant. The vaccine was well tolerated by all the study participants. There was no sign of vaccine virus replication in the airway in any participant. Most children exhibited an increase in antibody binding and neutralizing responses toward hPIV-1 within 4 weeks from the time of vaccination. In several children, antibody responses remained above incoming levels for at least 6 months after vaccination. Data suggest that SeV may provide a benefit to 3- to 6-year-old children, even when vaccine recipients have preexisting cross-reactive antibodies due to previous exposures to hPIV-1. Results encourage the testing of SeV administration in young seronegative children to protect against the serious respiratory tract diseases caused by hPIV-1 infections.


Subject(s)
Antibodies, Viral/blood , Parainfluenza Virus 1, Human/immunology , Respirovirus Infections/prevention & control , Sendai virus/immunology , Vaccines, Live, Unattenuated/administration & dosage , Vaccines, Live, Unattenuated/immunology , Viral Vaccines/administration & dosage , Administration, Intranasal , Animals , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Child , Child, Preschool , Cross Reactions , Female , Humans , Infant , Male , Mice , Sendai virus/growth & development , United States , Vaccines, DNA/administration & dosage , Vaccines, DNA/immunology , Viral Vaccines/adverse effects , Viral Vaccines/immunology
9.
BMC Pulm Med ; 14: 144, 2014 Sep 02.
Article in English | MEDLINE | ID: mdl-25178477

ABSTRACT

BACKGROUND: Viral pathogens were more commonly reported than previously estimated in community-acquired pneumonia (CAP) patients. However, the real role of virus was still controversial. METHODS: Consecutive adult patients with CAP between April and December, 2009 were prospectively enrolled. A four-fold or greater increase of IgG-titres against respiratory viruses in pair sera was tested by means of hemagglutination inhibition assay or indirect immunofluorescence. Swab samples were tested by cell culture and/or nucleic amplification tests. Viral etiology was considered definitive if at least one of the above tests was positive. RESULTS: Viral etiology was established in fifty-two (34.9%) of 149 CAP patients, twenty-two (81.5%) of 27 influenza like illness patients, and none of 75 volunteer controls. Forty-seven CAP patients were infected by a single virus (24 influenza A virus, 5 influenza B, 10 parainfluenza virus type 3 [PIV-3], 2 PIV-1, 2 adenovirus, 2 human rhinovirus and 2 coronavirus OC43), five cases by two or three viruses co-infection. Fever ≥ 39 °C (66.7%), fatigue (64.6%), and purulent sputum (52.1%) was the most common symptoms in viral pneumonia patients. On multivariate analysis, myalgia was included in the model for pneumonia associated with influenza infection. In the CURB-65 model only influenza infection was found independently associated with severe disease (CURB-65 score ≥ 3) out of variables, including age(years), sex, current smoking status, sick contact with febrile patients, numbers of comorbidity, presence of influenza infection, presence of PIV infection, with P = 0.021, OR 7.86 (95% CI 1.37-45.04). CONCLUSION: Respiratory virus was not a bystander, but pathogenic in pneumonia and was a common cause of CAP.


Subject(s)
Adenoviridae Infections/virology , Antibodies, Viral/blood , Immunoglobulin G/blood , Pneumonia, Viral/virology , RNA Virus Infections/virology , Adenoviridae/immunology , Adenoviridae Infections/blood , Adult , Aged , Coinfection/virology , Community-Acquired Infections/blood , Community-Acquired Infections/virology , Coronavirus/immunology , Coronavirus Infections/blood , Coronavirus Infections/virology , Female , Healthy Volunteers , Humans , Influenza A virus/immunology , Influenza B virus/immunology , Influenza, Human/blood , Influenza, Human/virology , Male , Middle Aged , Parainfluenza Virus 1, Human/immunology , Parainfluenza Virus 3, Human/immunology , Picornaviridae Infections/blood , Picornaviridae Infections/virology , Pneumonia, Viral/blood , Prospective Studies , RNA Virus Infections/blood , Respirovirus Infections/blood , Respirovirus Infections/virology , Rhinovirus/immunology
10.
Jpn J Infect Dis ; 67(5): 392-6, 2014.
Article in English | MEDLINE | ID: mdl-25241693

ABSTRACT

An epidemic of parainfluenza virus type 1 (PIV1) infection occurred in a hospital ward housing patients with severe motor and intellectual disabilities. Twenty-three infected patients exhibited persistent high fever for 4-16 days and decreased lymphocyte counts. One-half of the symptomatic patients had increased blood monocyte counts and the other half progressed to bronchitis or pneumonia. We also compared levels of 27 cytokines in the sera of 21 patients during the acute and normal phases of infection. Cytokine levels were measured with a bead immunoassay performed using the Luminex Multiplex System. Serum levels of interleukin (IL)-1Ra, C-C-motif chemokine (CCL) 2, and C-X-C-motif chemokine (CXCL) 10 significantly increased during the acute phase. In contrast, the serum level of CXCL8 decreased slightly. These results suggest the involvement of monocytes/macrophages and respiratory epithelial cells in the initial stage of PIV1 infection. A previous report using nasal wash samples also found a significant increase in levels of CXCL10 during the acute phase. Hence, CXCL10 may be a useful marker of a cytokine storm produced upon viral infection. However, alterations in levels of IL-1Ra, CCL2, and other cytokines differed between the 2 studies, suggesting that the cytokine profile produced systemically at viral infection is different from that produced at mucosal sites. Further analysis is required to clarify the mechanisms underlying cytokine production during PIV1 infections.


Subject(s)
Cytokines/blood , Parainfluenza Virus 1, Human/immunology , Respirovirus Infections/pathology , Respirovirus Infections/virology , Adolescent , Adult , Aged , Female , Humans , Leukocyte Count , Lymphocytes/immunology , Male , Middle Aged , Molecular Sequence Data , Monocytes/immunology , RNA, Viral/genetics , Sequence Analysis, DNA , Serum/chemistry , Young Adult
11.
Virology ; 433(2): 320-8, 2012 Nov 25.
Article in English | MEDLINE | ID: mdl-22959894

ABSTRACT

Human parainfluenza viruses (PIVs) cause acute respiratory illness in children, the elderly, and immunocompromised patients. PIV3 is a common cause of bronchiolitis and pneumonia, whereas PIV1 and 2 are frequent causes of upper respiratory tract illness and croup. To assess how PIV1, 2, and 3 differ with regard to replication and induction of type I interferons, interleukin-6, and relevant chemokines, we infected primary human airway epithelium (HAE) cultures from the same tissue donors and examined replication kinetics and cytokine secretion. PIV1 replicated to high titer yet did not induce cytokine secretion until late in infection, while PIV2 replicated less efficiently but induced an early cytokine peak. PIV3 replicated to high titer but induced a slower rise in cytokine secretion. The T cell chemoattractants CXCL10 and CXCL11 were the most abundant chemokines induced. Differences in replication and cytokine secretion might explain some of the differences in PIV serotype-specific pathogenesis and epidemiology.


Subject(s)
Bronchi/immunology , Bronchi/virology , Parainfluenza Virus 1, Human/physiology , Parainfluenza Virus 2, Human/physiology , Parainfluenza Virus 3, Human/physiology , Trachea/immunology , Trachea/virology , Cells, Cultured , Chemokines/biosynthesis , Cytokines/biosynthesis , Cytokines/genetics , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Interleukin-6/biosynthesis , Interleukin-6/genetics , Kinetics , Parainfluenza Virus 1, Human/classification , Parainfluenza Virus 1, Human/immunology , Parainfluenza Virus 1, Human/pathogenicity , Parainfluenza Virus 2, Human/classification , Parainfluenza Virus 2, Human/immunology , Parainfluenza Virus 2, Human/pathogenicity , Parainfluenza Virus 3, Human/classification , Parainfluenza Virus 3, Human/immunology , Parainfluenza Virus 3, Human/pathogenicity , Polymorphism, Single Nucleotide , Respiratory Mucosa/immunology , Respiratory Mucosa/virology , Serotyping , Species Specificity , Virus Replication
12.
Hum Vaccin ; 7(6): 639-45, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21508675

ABSTRACT

A Sendai virus (SeV) vector is being developed for delivery of an HIV immunogen. SeV is not known to cause disease in humans. Because it is genetically and antigenically related to human parainfluenza virus type 1 (hPIV-1), it is important to determine whether pre-existing hPIV-1 antibodies will affect immune responses elicited by a SeV vector-based vaccine. To quantify SeV neutralizing antibodies (NAb) in human serum, a sensitive virus neutralization assay was developed using a SeV vector encoding green fluorescent protein. Samples from 255 HIV-uninfected subjects from Africa, Europe, United States, and Japan, as well as from 12 confirmed hPIV-1-infected patients, were analyzed. SeV NAb titers did not vary significantly after serum was treated with receptor-destroying enzyme, indicating that non-specific hemagglutination inhibitors did not affect the assay sensitivity. A significant correlation was observed between hPIV-1 ELISA and SeV NAb titers. SeV NAb were detected in 92.5% subjects with a median titer of 60.6 and values ranging from 5.9- 11,324. The majority had titers < 1000 with 71.7% < 100 (< 5 considered negative). There was no significant difference in titer or prevalence by gender, age range or geographic origin. However, African males had a lower titer than non-Africans of either gender (p=0.007). Overall, the prevalence of SeV NAb is high and likely due to neutralization by cross-reactive hPIV-1 antibodies. Clinical trials will be needed to assess the influence of pre-existing SeV NAb on HIV-specific immune responses elicited by a SeV vaccine vector expressing HIV.


Subject(s)
AIDS Vaccines/immunology , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Sendai virus/immunology , Adolescent , Adult , Africa , Cross Reactions , Europe , Female , Genetic Vectors , Humans , Japan , Male , Middle Aged , Parainfluenza Virus 1, Human/immunology , Sendai virus/genetics , United States
13.
Vaccine ; 28(41): 6749-56, 2010 Sep 24.
Article in English | MEDLINE | ID: mdl-20682364

ABSTRACT

Sendai virus (SeV), a natural mouse pathogen, shows considerable promise as a candidate vaccine for human parainfluenza virus-type 1 (hPIV-1), and also as a vaccine vector for other serious pathogens of infants including respiratory syncytial virus (RSV). In an effort to define correlates of immunity, we examined the virus-specific serum antibody of cotton rats inoculated intranasally (I.N.) with SeV. Virus-specific antibody forming cells (AFCs) were also measured in the bone marrow, because these are considered responsible for durable serum antibody levels in other viral systems. Results showed that a single SeV inoculation was sufficient to induce virus-specific serum antibodies and bone marrow-resident AFCs that persisted for as many as 8 months post-vaccination. Given that the predominant SeV-specific serum antibody isotype was IgG, an isotype that traffics poorly to the upper respiratory tract (URT), we asked if local nasal and lung-associated antibodies and AFCs were also present. Studies showed that: (i) SeV-specific antibodies appeared in the URT and lower respiratory tract (LRT) within 7 days after immunization, (ii) corresponding AFCs were present in the diffuse-NALT (d-NALT) and lung, (iii) AFCs in the d-NALT and lung peaked at approximately 6 weeks and persisted for the lifetime of the animal, reaching a level exceeding that of the bone marrow by an order of magnitude, (iv) IgA was the dominant isotype among AFCs in the d-NALT and lung at 4-weeks post-vaccination and thereafter, and (v) antibody and AFC responses associated with the prevention of lung infection when animals were challenged with hPIV-1 just 1 week after vaccination.


Subject(s)
Antibody-Producing Cells/immunology , Parainfluenza Virus 1, Human/immunology , Respirovirus Infections/prevention & control , Sendai virus/immunology , Animals , Antibodies, Viral/biosynthesis , Antibodies, Viral/blood , Antibody Specificity , Bone Marrow Cells/immunology , Immunoglobulin A/biosynthesis , Immunoglobulin G/biosynthesis , Immunoglobulin G/blood , Lung/immunology , Nose/immunology , Respirovirus Infections/immunology , Sigmodontinae
14.
Pediatr Infect Dis J ; 29(4): e26-31, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20182399

ABSTRACT

OBJECTIVE: To describe the clinical manifestations of parainfluenza virus (PIV) infection and to characterize biochemical markers of PIV disease severity. PATIENTS AND METHODS: We reviewed the medical records of 165 children who had a nasal wash culture positive for PIV at our institution between 1998 and 2008. Nasal wash samples were assayed for 26 inflammatory mediators using Luminex bead proteomics. RESULTS: A total of 153 patients, ages 2 weeks to 12 years, with single virus infection were included in our final analysis. Fifty-two patients were infected with PIV1, 19 with PIV2, 74 with PIV3, and 8 with PIV4. Lower respiratory tract infection (LRTI) was diagnosed in 67 (44%) patients, 21 (14%) had laryngotracheobronchitis, and 49 (32%) had an upper respiratory infection other than laryngotracheobronchitis. LRTI was diagnosed in 54% of patients infected with PIV3, 35% of those infected with PIV1, 26% of those with PIV2, and 50% of those with PIV4. Compared with uninfected control patients, PIV-infected patients had higher nasal wash concentrations of interleukin-6, CX-chemokine ligand 8 (CXCL8 or interleukin-8), CCL3 (macrophage inflammatory protein-1alpha), CCL4 (macrophage inflammatory protein-1beta), CXCL9 (monokine induced by interferon gamma), and CCL5 (regulated upon activation, normal T cell expressed and secreted (RANTES). Patients with LRTI, moderate or severe illness, and PIV 1 or 3 (respirovirus) infection had higher nasal wash concentrations of CXCL8 when compared with patients with upper respiratory infection, mild illness, or PIV 2 and 4 (rubulavirus) infection (P < 0.05). CONCLUSIONS: PIV infection causes a spectrum of illnesses associated with the expression and release of several proinflammatory mediators. Of note, elevated concentrations of CXCL8 in nasal wash samples are associated with more severe forms of PIV disease.


Subject(s)
Inflammation Mediators/metabolism , Paramyxoviridae Infections/immunology , Paramyxoviridae Infections/physiopathology , Respiratory Tract Infections , Bronchitis/immunology , Bronchitis/physiopathology , Bronchitis/virology , Child , Child, Preschool , Humans , Infant , Infant, Newborn , Interleukin-8/metabolism , Laryngitis/immunology , Laryngitis/physiopathology , Laryngitis/virology , Nasal Lavage Fluid/immunology , Parainfluenza Virus 1, Human/immunology , Parainfluenza Virus 1, Human/pathogenicity , Parainfluenza Virus 2, Human/immunology , Parainfluenza Virus 2, Human/pathogenicity , Parainfluenza Virus 3, Human/immunology , Parainfluenza Virus 3, Human/pathogenicity , Parainfluenza Virus 4, Human/immunology , Parainfluenza Virus 4, Human/pathogenicity , Paramyxoviridae Infections/virology , Respiratory Tract Infections/immunology , Respiratory Tract Infections/physiopathology , Respiratory Tract Infections/virology , Severity of Illness Index , Tracheitis/immunology , Tracheitis/physiopathology , Tracheitis/virology
15.
Vaccine ; 28(3): 767-79, 2010 Jan 08.
Article in English | MEDLINE | ID: mdl-19857454

ABSTRACT

A novel recombinant human parainfluenza virus type 1 (rHPIV1), rHPIV1-C+P, in which the overlapping open reading frames of the C and P genes were separated in order to introduce mutations into the C gene without affecting P, was generated. Infectious rHPIV1-C+P was readily recovered and replicated as efficiently as HPIV1 wild type (wt) in vitro and in African green monkeys (AGMs). rHPIV1-C+P expressed increased levels of C protein and, surprisingly, activated the type I IFN and apoptosis responses more strongly than HPIV1 wt. rHPIV1-C+P provided a useful backbone for recovering an attenuated P/C gene mutation (Delta 84-85), which was previously unrecoverable, likely due to detrimental effects of the deletion on the P protein. rHPIV1-C(Delta 84-85)+P and an additional mutant, rHPIV1-C(Delta 169-170)+P, were found to replicate to similar titers in vitro and to activate the type I IFN and apoptosis responses to a similar degree as rHPIV1-C+P. rHPIV1-C(Delta 84-85)+P was found to be highly attenuated in AGMs, and all viruses were immunogenic and effective in protecting AGMs against challenge with HPIV1 wt. rHPIV1-C(Delta 84-85)+P will be investigated as a potential live-attenuated vaccine candidate for HPIV1.


Subject(s)
Parainfluenza Vaccines/immunology , Parainfluenza Virus 1, Human/immunology , Parainfluenza Virus 1, Human/pathogenicity , Phosphoproteins/genetics , Sequence Deletion , Viral Proteins/genetics , Animals , Antibodies, Viral/blood , Apoptosis , Base Sequence , Cell Line , Chlorocebus aethiops , Humans , Interferon Type I/biosynthesis , Molecular Sequence Data , Parainfluenza Vaccines/genetics , Parainfluenza Virus 1, Human/genetics , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Viral Load , Virulence , Virus Replication
16.
J Gen Virol ; 90(Pt 8): 1906-1915, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19423549

ABSTRACT

Human parainfluenza virus type 1 (HPIV-1) and Sendai virus (SeV) are highly homologous in structure and sequence, whilst maintaining distinct host ranges. These viruses express accessory proteins from their P/C gene that are known to have activities against innate immunity. The accessory proteins expressed from the P/C gene of these viruses are different. In addition to the nested set of C proteins, SeV expresses V protein from edited P mRNA, which is not expressed by HPIV-1. This study evaluated the host specificity and role of the P/C gene products in anti-interferon (IFN) and anti-apoptosis activity by characterizing a recombinant SeV, rSeVhP, in which the SeV P/C gene was replaced with that of HPIV-1. Unlike SeV, rSeVhP infection strongly activated IFN regulatory transcription factor (IRF)-3 and nuclear factor-kappaB, resulting in an increased level of IFN-beta induction compared with SeV in murine cells. In contrast, activation of IRF-3 was not observed in rSeVhP-infected human A549 cells. rSeVhPSV, which expressed SeV V protein from an inserted gene in rSeVhP, induced less IFN-beta than rSeVhP, suggesting that V contributes to the suppression of IFN production in murine cells. Furthermore, rSeVhP induced apoptotic cell death in murine but not in A549 cells. These data indicate the functional difference in P/C gene products from SeV and HPIV-1 in antagonizing IFN induction and apoptosis, which is likely to be one of the major factors for pathogenicity in specific hosts.


Subject(s)
Apoptosis , Interferons/antagonists & inhibitors , Parainfluenza Virus 1, Human/immunology , Phosphoproteins/genetics , Phosphoproteins/immunology , Sendai virus/genetics , Viral Proteins/genetics , Viral Proteins/immunology , Animals , Cell Line , Humans , Mice , Parainfluenza Virus 1, Human/genetics , Recombination, Genetic , Sendai virus/immunology
17.
Vaccine ; 27(12): 1848-57, 2009 Mar 13.
Article in English | MEDLINE | ID: mdl-19200447

ABSTRACT

The human parainfluenza viruses (hPIVs) and respiratory syncytial viruses (RSVs) are the leading causes of hospitalizations due to respiratory viral disease in infants and young children, but no vaccines are yet available. Here we describe the use of recombinant Sendai viruses (rSeVs) as candidate vaccine vectors for these respiratory viruses in a cotton rat model. Two new Sendai virus (SeV)-based hPIV-2 vaccine constructs were generated by inserting the fusion (F) gene or the hemagglutinin-neuraminidase (HN) gene from hPIV-2 into the rSeV genome. The inoculation of either vaccine into cotton rats elicited neutralizing antibodies toward both homologous and heterologous hPIV-2 virus isolates. The vaccines elicited robust and durable antibodies toward hPIV-2, and cotton rats immunized with individual or mixed vaccines were fully protected against hPIV-2 infections of the lower respiratory tract. The immune responses toward a single inoculation with rSeV vaccines were long-lasting and cotton rats were protected against viral challenge for as long as 11 months after vaccination. One inoculation with a mixture of the hPIV-2-HN-expressing construct and two additional rSeVs (expressing the F protein of RSV and the HN protein of hPIV-3) resulted in protection against challenge viruses hPIV-1, hPIV-2, hPIV-3, and RSV. Results identify SeV vectors as promising vaccine candidates for four different paramyxoviruses, each responsible for serious respiratory infections in children.


Subject(s)
Parainfluenza Vaccines/therapeutic use , Parainfluenza Virus 1, Human/immunology , Parainfluenza Virus 2, Human/immunology , Parainfluenza Virus 3, Human/immunology , Paramyxoviridae Infections/immunology , Paramyxoviridae Infections/prevention & control , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus, Human/immunology , Sendai virus/genetics , Animals , Enzyme-Linked Immunosorbent Assay , Female , Genetic Vectors , Humans , Neutralization Tests , Sigmodontinae , Vaccines, Synthetic/immunology
18.
Pediatr Infect Dis J ; 27(10 Suppl): S123-5, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18820572

ABSTRACT

Parainfluenza viruses (PIV) have been generally disregarded as pathogens in spite of their importance in pediatric lower respiratory illness. Because PIVs account for 17% of hospitalized illness associated virus isolation, the development of PIV vaccine would be a major advance in preventing lower respiratory tract infection in infants and young children. We will review in detail several PIV vaccine candidates and recent newer approaches to PIV vaccine development. Intranasally administered bovine PIV3 (bPIV3) vaccine and cold-adapted PIV3 vaccine have been evaluated throughout the pediatric age spectrum. BPIV3 does not give a robust response to the heterotypic human strain although seroconversion rate to bPIV3 is 57-65%. However, bPIV3 vaccine is being used as an attenuated backbone for insertion of human PIV3 hemagglutinin-neuraminidase and fusion (F) proteins and a surface protein, F, of respiratory syncytial virus. The effectiveness of this vaccine against both PIV3 and RSV challenge has been demonstrated in African green monkeys. The cold-adapted PIV3 vaccine has been extensively evaluated and is safe and immunogenic in seronegative children with a seroconversion rate of 79%. These promising candidates deserve to enter into efficacy trials both for their ability to prevent PIV3 disease and as a model of protection against respiratory illness by mucosal vaccination.


Subject(s)
Parainfluenza Vaccines , Parainfluenza Virus 3, Human/immunology , Respirovirus Infections/prevention & control , Adult , Child , Cold Temperature , Croup/prevention & control , Humans , Infant , Mutation , Parainfluenza Vaccines/genetics , Parainfluenza Vaccines/immunology , Parainfluenza Virus 1, Human/immunology , Parainfluenza Virus 2, Human/immunology , Parainfluenza Virus 3, Bovine/genetics , Parainfluenza Virus 3, Bovine/immunology , Parainfluenza Virus 3, Human/genetics , Parainfluenza Virus 4, Human/immunology , Respirovirus Infections/immunology , Respirovirus Infections/virology , Rubulavirus Infections/immunology , Rubulavirus Infections/prevention & control , Rubulavirus Infections/virology
19.
J Allergy Clin Immunol ; 122(1): 69-77, 77.e1-2, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18472150

ABSTRACT

BACKGROUND: The most frequent trigger of asthma exacerbation is infection with common airway viruses; however, the precise mechanism regulating such severe reactions is not understood. The presence of airway eosinophil products is a unique feature detected in asthmatic airways. Using an animal model, we previously demonstrated that T cells play an important role in regulating an eosinophil-dependant pathway of virus-induced airway hyperreactivity. We hypothesize that human eosinophils respond to viruses, although only after interaction with T cells. OBJECTIVES: We sought to determine whether eosinophils can respond to airway viruses in vitro and determine the mechanism of response. METHODS: An in vitro coculture model of human eosinophils, antigen-presenting cells, and T cells was used with parainfluenza virus, respiratory syncytial virus, or rhinovirus. We measured release of eosinophil peroxidase (EPO) in concert with T-cell proliferation, cytokine release, and changes in T-cell phenotype. RESULTS: The viruses induced release of EPO when coincubated in the presence of antigen-presenting cells (dendritic cells or macrophages) and T cells. Virus-mediated release was associated with proliferation of CD3(+)CD4(+) T cells and release of cytokines. UV inactivation of the virus did not prevent virus-induced EPO release or T-cell proliferation. Proliferating CD4(+) T cells show increased expression of CD25 and CD45RO. CD8(+) T cells were not activated. CONCLUSION: Virus-induced EPO release can occur in the context of antigen presentation to CD4(+) T cells.


Subject(s)
Antigen-Presenting Cells/immunology , CD4-Positive T-Lymphocytes/immunology , Eosinophil Peroxidase/metabolism , Eosinophils/immunology , T-Lymphocyte Subsets/immunology , Viruses/immunology , Antigen Presentation , Cell Proliferation , Cells, Cultured , Coculture Techniques , Cytokines/biosynthesis , Cytokines/immunology , Humans , Parainfluenza Virus 1, Human/immunology , Respiratory Syncytial Virus, Human/immunology , Rhinovirus/immunology , T-Lymphocytes , Viruses/metabolism
20.
Virus Res ; 133(2): 178-86, 2008 May.
Article in English | MEDLINE | ID: mdl-18249456

ABSTRACT

Human parainfluenza virus types 1 and 3 (HPIV1 and HPIV3, respectively), members of the virus family Paramyxoviridae, are common causes of lower respiratory tract infections in infants, young children, the immunocompromised, the chronically ill, and the elderly. In order to synthesize recombinant HPIV1 and HPIV3 nucleocapsid proteins, the coding sequences were cloned into the yeast Saccharomyces cerevisiae expression vector pFGG3 under control of GAL7 promoter. A high level of recombinant virus nucleocapsid proteins expression (20-24 mg l(-1) of yeast culture) was obtained. Electron microscopy demonstrated the assembly of typical herring-bone structures of purified recombinant nucleocapsid proteins, characteristic for other paramyxoviruses. These structures contained host RNA, which was resistant to RNase treatment. The nucleocapsid proteins were stable in yeast and were easily purified by caesium chloride gradient ultracentrifugation. Therefore, this system proved to be simple, efficient and cost-effective, suitable for high-level production of parainfluenza virus nucleocapsids as nucleocapsid-like particles. When used as coating antigens in an indirect ELISA, the recombinant N proteins reacted with sera of patients infected with HPIV1 or 3. Serological assays to detect HPIV-specific antibodies could be designed on this basis.


Subject(s)
Nucleocapsid Proteins/metabolism , Parainfluenza Virus 1, Human/metabolism , Parainfluenza Virus 3, Human/metabolism , Recombinant Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Amino Acid Sequence , Antibodies, Viral/blood , Antibodies, Viral/immunology , Enzyme-Linked Immunosorbent Assay , Humans , Microscopy, Electron , Molecular Sequence Data , Nucleocapsid Proteins/chemistry , Nucleocapsid Proteins/genetics , Nucleocapsid Proteins/immunology , Parainfluenza Virus 1, Human/genetics , Parainfluenza Virus 1, Human/immunology , Parainfluenza Virus 3, Human/genetics , Parainfluenza Virus 3, Human/immunology , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Respirovirus Infections/diagnosis
SELECTION OF CITATIONS
SEARCH DETAIL
...