Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 695
Filter
1.
Front Immunol ; 15: 1364622, 2024.
Article in English | MEDLINE | ID: mdl-38933273

ABSTRACT

Introduction: Community-acquired pneumonia (CAP) is a global health concern, with 25% of cases attributed to Streptococcus pneumoniae (Spn). Viral infections like influenza A virus (IAV), respiratory syncytial virus (RSV), and human metapneumovirus (hMPV) increase the risk of Spn, leading to severe complications due to compromised host immunity. Methods: We evaluated the efficacy of an anti-PhtD monoclonal antibody (mAb) cocktail therapy (PhtD3 + 7) in improving survival rates in three viral/bacterial coinfection models: IAV/Spn, hMPV/Spn, and RSV/Spn. Results: The PhtD3 + 7 mAb cocktail outperformed antiviral mAbs, resulting in prolonged survival. In the IAV/Spn model, it reduced bacterial titers in blood and lungs by 2-4 logs. In the hMPV/Spn model, PhtD3 + 7 provided greater protection than the hMPV-neutralizing mAb MPV467, significantly reducing bacterial titers. In the RSV/Spn model, PhtD3 + 7 offered slightly better protection than the antiviral mAb D25, uniquely decreasing bacterial titers in blood and lungs. Discussion: Given the threat of antibiotic resistance, our findings highlight the potential of anti-PhtD mAb therapy as an effective option for treating viral and secondary pneumococcal coinfections.


Subject(s)
Antibodies, Monoclonal , Coinfection , Streptococcus pneumoniae , Superinfection , Animals , Humans , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/immunology , Streptococcus pneumoniae/immunology , Mice , Superinfection/immunology , Superinfection/microbiology , Coinfection/immunology , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/drug therapy , Metapneumovirus/immunology , Influenza A virus/immunology , Disease Models, Animal , Pneumococcal Infections/immunology , Pneumococcal Infections/prevention & control , Pneumococcal Infections/drug therapy , Female , Mice, Inbred BALB C , Paramyxoviridae Infections/immunology , Paramyxoviridae Infections/drug therapy , Antibodies, Viral/immunology
2.
Nat Commun ; 15(1): 4629, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38821950

ABSTRACT

The Paramyxoviridae family encompasses medically significant RNA viruses, including human respiroviruses 1 and 3 (RV1, RV3), and zoonotic pathogens like Nipah virus (NiV). RV3, previously known as parainfluenza type 3, for which no vaccines or antivirals have been approved, causes respiratory tract infections in vulnerable populations. The RV3 fusion (F) protein is inherently metastable and will likely require prefusion (preF) stabilization for vaccine effectiveness. Here we used structure-based design to stabilize regions involved in structural transformation to generate a preF protein vaccine antigen with high expression and stability, and which, by stabilizing the coiled-coil stem region, does not require a heterologous trimerization domain. The preF candidate induces strong neutralizing antibody responses in both female naïve and pre-exposed mice and provides protection in a cotton rat challenge model (female). Despite the evolutionary distance of paramyxovirus F proteins, their structural transformation and local regions of instability are conserved, which allows successful transfer of stabilizing substitutions to the distant preF proteins of RV1 and NiV. This work presents a successful vaccine antigen design for RV3 and provides a toolbox for future paramyxovirus vaccine design and pandemic preparedness.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , Sigmodontinae , Viral Fusion Proteins , Viral Vaccines , Animals , Female , Viral Fusion Proteins/immunology , Viral Fusion Proteins/genetics , Viral Fusion Proteins/chemistry , Mice , Viral Vaccines/immunology , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Humans , Mice, Inbred BALB C , Paramyxoviridae Infections/prevention & control , Paramyxoviridae Infections/immunology , Paramyxoviridae Infections/virology , Parainfluenza Virus 3, Human/immunology , Parainfluenza Virus 3, Human/genetics
3.
Pediatrics ; 153(6)2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38738290

ABSTRACT

OBJECTIVES: Human metapneumovirus (hMPV) and parainfluenza virus type 3 (PIV3) are common respiratory illnesses in children. The safety and immunogenicity of an investigational mRNA-based vaccine, mRNA-1653, encoding membrane-anchored fusion proteins of hMPV and PIV3, was evaluated in hMPV/PIV3-seropositive children. METHODS: In this phase 1b randomized, observer-blind, placebo-controlled, dose-ranging study, hMPV/PIV3-seropositive children were enrolled sequentially into 2 dose levels of mRNA-1653 administered 2 months apart; children aged 12 to 36 months were randomized (1:1) to receive 10-µg of mRNA-1653 or placebo and children aged 12 to 59 months were randomized (3:1) to receive 30-µg of mRNA-1653 or placebo. RESULTS: Overall, 27 participants aged 18 to 55 months were randomized; 15 participants received 10-µg of mRNA-1653 (n = 8) or placebo (n = 7), whereas 12 participants received 30-µg of mRNA-1653 (n = 9) or placebo (n = 3). mRNA-1653 was well-tolerated at both dose levels. The only reported solicited local adverse reaction was tenderness at injection site; solicited systemic adverse reactions included grade 1 or 2 chills, irritability, loss of appetite, and sleepiness. A single 10-µg or 30-µg mRNA-1653 injection increased hMPV and PIV3 neutralizing antibody titers (geometric mean fold-rise ratio over baseline: hMPV-A = 2.9-6.1; hMPV-B = 6.2-13.2; PIV3 = 2.8-3.0) and preF and postF binding antibody concentrations (geometric mean fold-rise ratio: hMPV preF = 5.3-6.1; postF = 4.6-6.5 and PIV3 preF = 13.9-14.2; postF = 11.0-12.1); a second injection did not further increase antibody levels in these seropositive children. Binding antibody responses were generally preF biased. CONCLUSIONS: mRNA-1653 was well-tolerated and boosted hMPV and PIV3 antibody levels in seropositive children aged 12 to 59 months, supporting the continued development of mRNA-1653 or its components for the prevention of hMPV and PIV3.


Subject(s)
Parainfluenza Virus 3, Human , Humans , Female , Male , Child, Preschool , Infant , Parainfluenza Virus 3, Human/immunology , Parainfluenza Virus 3, Human/genetics , Metapneumovirus/immunology , Metapneumovirus/genetics , Single-Blind Method , Paramyxoviridae Infections/prevention & control , Paramyxoviridae Infections/immunology , Antibodies, Viral/blood , Parainfluenza Vaccines/immunology , Parainfluenza Vaccines/administration & dosage , Parainfluenza Vaccines/genetics , Immunogenicity, Vaccine , RNA, Messenger
4.
mBio ; 15(5): e0055024, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38530032

ABSTRACT

Human metapneumovirus (HMPV) is a primary cause of acute respiratory infection, yet there are no approved vaccines or antiviral therapies for HMPV. Early host responses to HMPV are poorly characterized, and further understanding could identify important antiviral pathways. Type III interferon (IFN-λ) displays potent antiviral activity against respiratory viruses and is being investigated for therapeutic use. However, its role in HMPV infection remains largely unknown. Here, we show that IFN-λ is highly upregulated during HMPV infection in vitro in human and mouse airway epithelial cells and in vivo in mice. We found through several immunological and molecular assays that type II alveolar cells are the primary producers of IFN-λ. Using mouse models, we show that IFN-λ limits lung HMPV replication and restricts virus spread from upper to lower airways but does not contribute to clinical disease. Moreover, we show that IFN-λ signaling is predominantly mediated by CD45- non-immune cells. Mice lacking IFN-λ signaling showed diminished loss of ciliated epithelial cells and decreased recruitment of lung macrophages in early HMPV infection along with higher inflammatory cytokine and interferon-stimulated gene expression, suggesting that IFN-λ may maintain immunomodulatory responses. Administration of IFN-λ for prophylaxis or post-infection treatment in mice reduced viral load without inflammation-driven weight loss or clinical disease. These data offer clinical promise for IFN-λ in HMPV treatment. IMPORTANCE: Human metapneumovirus (HMPV) is a common respiratory pathogen and often contributes to severe disease, particularly in children, immunocompromised people, and the elderly. There are currently no licensed HMPV antiviral treatments or vaccines. Here, we report novel roles of host factor IFN-λ in HMPV disease that highlight therapeutic potential. We show that IFN-λ promotes lung antiviral responses by restricting lung HMPV replication and spread from upper to lower airways but does so without inducing lung immunopathology. Our data uncover recruitment of lung macrophages, regulation of ciliated epithelial cells, and modulation of inflammatory cytokines and interferon-stimulated genes as likely contributors. Moreover, we found these roles to be distinct and non-redundant, as they are not observed with knockout of, or treatment with, type I IFN. These data elucidate unique antiviral functions of IFN-λ and suggest IFN-λ augmentation as a promising therapeutic for treating HMPV disease and promoting effective vaccine responses.


Subject(s)
Interferon Lambda , Lung , Metapneumovirus , Paramyxoviridae Infections , Virus Replication , Animals , Humans , Mice , Antiviral Agents/pharmacology , Disease Models, Animal , Epithelial Cells/virology , Epithelial Cells/immunology , Interferon Lambda/immunology , Interferon Lambda/pharmacology , Interferons/immunology , Interferons/pharmacology , Lung/immunology , Lung/virology , Metapneumovirus/immunology , Metapneumovirus/genetics , Mice, Inbred C57BL , Paramyxoviridae Infections/immunology , Paramyxoviridae Infections/virology , Virus Replication/drug effects
5.
Mucosal Immunol ; 17(3): 476-490, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38176655

ABSTRACT

Respiratory viral infections, including human metapneumovirus (HMPV), remain a leading cause of morbidity and mortality in neonates and infants. However, the mechanisms behind the increased sensitivity to those respiratory viral infections in neonates are poorly understood. Neonates, unlike adults, have several anti-inflammatory mechanisms in the lung, including elevated baseline expression of programmed death ligand 1 (PD-L1), a ligand for the inhibitory receptor programmed cell death protein 1 (PD-1). We thus hypothesized that neonates would rely on PD-1:PD-L1 signaling to restrain antiviral CD8 responses. To test this, we developed a neonatal primary HMPV infection model using wild-type C57BL/6 (B6) and Pdcd1-/- (lacking PD-1) mice. HMPV-infected neonatal mice had increased PD-L1/PD-L2 co-expression on innate immune cells but a similar number of antigen-specific CD8+ T cells and upregulation of PD-1 to that of adult B6 mice. Neonatal CD8+ T cells had reduced interferon-gamma (IFN-γ), granzyme B, and interleukin-2 production compared with B6 adults. Pdcd1-/- neonatal CD8+ T cells had markedly increased production of IFN-γ and granzyme B compared with B6 neonates. Pdcd1-/- neonates had increased acute pathology with HMPV or influenza. Pdcd1-/- neonates infected with HMPV had long-term changes in pulmonary physiology with evidence of immunopathology and a persistent CD8+ T-cell response with increased granzyme B production. Using single-cell ribonucleic acid sequencing from a child lacking PD-1 signaling, a similar activated CD8+ T-cell signature with increased granzyme B expression was observed. These data indicate that PD-1 signaling critically limits CD8+ T-cell effector functions and prevents immunopathology in response to neonatal respiratory viral infections.


Subject(s)
Animals, Newborn , CD8-Positive T-Lymphocytes , Metapneumovirus , Mice, Knockout , Paramyxoviridae Infections , Programmed Cell Death 1 Receptor , Signal Transduction , Animals , CD8-Positive T-Lymphocytes/immunology , Programmed Cell Death 1 Receptor/metabolism , Programmed Cell Death 1 Receptor/genetics , Mice , Humans , Metapneumovirus/immunology , Paramyxoviridae Infections/immunology , B7-H1 Antigen/metabolism , B7-H1 Antigen/genetics , Mice, Inbred C57BL , Granzymes/metabolism , Granzymes/genetics , Disease Models, Animal , Respiratory Tract Infections/immunology , Respiratory Tract Infections/virology
6.
Nature ; 626(7998): 392-400, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38086420

ABSTRACT

An ideal vaccine both attenuates virus growth and disease in infected individuals and reduces the spread of infections in the population, thereby generating herd immunity. Although this strategy has proved successful by generating humoral immunity to measles, yellow fever and polio, many respiratory viruses evolve to evade pre-existing antibodies1. One approach for improving the breadth of antiviral immunity against escape variants is through the generation of memory T cells in the respiratory tract, which are positioned to respond rapidly to respiratory virus infections2-6. However, it is unknown whether memory T cells alone can effectively surveil the respiratory tract to the extent that they eliminate or greatly reduce viral transmission following exposure of an individual to infection. Here we use a mouse model of natural parainfluenza virus transmission to quantify the extent to which memory CD8+ T cells resident in the respiratory tract can provide herd immunity by reducing both the susceptibility of acquiring infection and the extent of transmission, even in the absence of virus-specific antibodies. We demonstrate that protection by resident memory CD8+ T cells requires the antiviral cytokine interferon-γ (IFNγ) and leads to altered transcriptional programming of epithelial cells within the respiratory tract. These results suggest that tissue-resident CD8+ T cells in the respiratory tract can have important roles in protecting the host against viral disease and limiting viral spread throughout the population.


Subject(s)
CD8-Positive T-Lymphocytes , Immunologic Memory , Memory T Cells , Paramyxoviridae Infections , Respiratory System , Animals , Mice , CD8-Positive T-Lymphocytes/immunology , Disease Models, Animal , Epithelial Cells/immunology , Epithelial Cells/metabolism , Immunity, Herd/immunology , Immunologic Memory/immunology , Interferon-gamma/immunology , Memory T Cells/immunology , Paramyxoviridae/immunology , Paramyxoviridae/physiology , Paramyxoviridae Infections/immunology , Paramyxoviridae Infections/prevention & control , Paramyxoviridae Infections/transmission , Paramyxoviridae Infections/virology , Respiratory System/cytology , Respiratory System/immunology , Respiratory System/virology , Transcription, Genetic , Humans
7.
J Virol ; 97(12): e0105223, 2023 Dec 21.
Article in English | MEDLINE | ID: mdl-38032197

ABSTRACT

IMPORTANCE: Human metapneumovirus (hMPV) is a common pathogen causing lower respiratory tract infections worldwide and can develop severe symptoms in high-risk populations such as infants, the elderly, and immunocompromised patients. There are no approved hMPV vaccines or neutralizing antibodies available for therapeutic or prophylactic use. The trimeric hMPV fusion F protein is the major target of neutralizing antibodies in human sera. Understanding the immune recognition of antibodies to hMPV-F antigen will provide critical insights into developing efficacious hMPV monoclonal antibodies and vaccines.


Subject(s)
Metapneumovirus , Paramyxoviridae Infections , Aged , Humans , Antibodies, Neutralizing , Antibodies, Viral , Epitopes , Metapneumovirus/physiology , Paramyxoviridae Infections/immunology , Viral Fusion Proteins , Viral Vaccines/immunology
8.
Front Immunol ; 14: 1173605, 2023.
Article in English | MEDLINE | ID: mdl-37435074

ABSTRACT

Human metapneumovirus (HMPV) is a pneumovirus that may cause severe respiratory disease in humans. HMPV infection has been found to increase susceptibility to bacterial superinfections leading to increased morbidity and mortality. The molecular mechanisms underlying HMPV-mediated increase in bacterial susceptibility are poorly understood and largely understudied. Type I interferons (IFNs), while critical for antiviral defenses, may often have detrimental effects by skewing the host immune response and cytokine output of immune cells. It is currently unknown if HMPV skews the inflammatory response in human macrophages triggered by bacterial stimuli. Here we report that HMPV pre-infection impacts production of specific cytokines. HMPV strongly suppresses IL-1ß transcription in response to LPS or heat-killed Pseudomonas aeruginosa and Streptococcus pneumonia, while enhancing mRNA levels of IL-6, TNF-α and IFN-ß. We demonstrate that in human macrophages the HMPV-mediated suppression of IL-1ß transcription requires TANK-binding kinase 1 (TBK1) and signaling via the IFN-ß-IFNAR axis. Interestingly, our results show that HMPV pre-infection did not impair the LPS-stimulated activation of NF-κB and HIF-1α, transcription factors that stimulate IL-1ß mRNA synthesis in human cells. Furthermore, we determined that sequential HMPV-LPS treatment resulted in accumulation of the repressive epigenetic mark H3K27me3 at the IL1B promoter. Thus, for the first time we present data revealing the molecular mechanisms by which HMPV shapes the cytokine output of human macrophages exposed to bacterial pathogens/LPS, which appears to be dependent on epigenetic reprogramming at the IL1B promoter leading to reduced synthesis of IL-1ß. These results may improve current understanding of the role of type I IFNs in respiratory disease mediated not only by HMPV, but also by other respiratory viruses that are associated with superinfections.


Subject(s)
Bacterial Infections , Interferon-beta , Interleukin-1beta , Paramyxoviridae Infections , Superinfection , Humans , Cytokines , Metapneumovirus , Transcription, Genetic , Bacterial Infections/immunology , Paramyxoviridae Infections/immunology
9.
Viruses ; 15(2)2023 02 09.
Article in English | MEDLINE | ID: mdl-36851691

ABSTRACT

Human metapneumovirus (hMPV) is an important cause of respiratory disease in immunocompromised individuals, yet hMPV infection has not been modeled before in immunocompromised animals. In this work, cotton rats S. hispidus immunosuppressed by cyclophosphamide were infected with hMPV, and viral replication and pulmonary inflammation in these animals were compared to those in normal hMPV-infected S. hispidus. The efficacy of prophylactic and therapeutic administration of the anti-hMPV antibody MPV467 was also evaluated. Immunosuppressed animals had higher pulmonary and nasal titers of hMPV on day 5 post-infection compared to normal animals, and large amounts of hMPV were still present in the respiratory tract of immunosuppressed animals on days 7 and 9 post-infection, indicating prolonged viral replication. Immunosuppression was accompanied by reduced pulmonary histopathology in hMPV-infected cotton rats compared to normal animals; however, a delayed increase in pathology and pulmonary chemokine expression was seen in immunosuppressed cotton rats. Prophylactic and therapeutic MPV467 treatments protected both upper and lower respiratory tracts against hMPV infection. The lung pathology and pulmonary expression of IP-10 and MIP-1α mRNA were reduced by therapeutic MPV467 administration. These results indicate that immunosuppressed cotton rats represent a useful model for studying hMPV pathogenesis and for evaluating therapeutics that could alleviate hMPV-induced disease in immunocompromised subjects.


Subject(s)
Immunocompromised Host , Metapneumovirus , Paramyxoviridae Infections , Sigmodontinae , Animals , Humans , Chemokine CCL3 , Immunocompromised Host/immunology , Immunosuppression Therapy , Paramyxoviridae Infections/immunology , Paramyxoviridae Infections/virology , Sigmodontinae/immunology , Sigmodontinae/virology , Disease Models, Animal
10.
Viruses ; 14(1)2022 01 12.
Article in English | MEDLINE | ID: mdl-35062341

ABSTRACT

Particles of many paramyxoviruses include small amounts of proteins with a molecular weight of about 20 kDa. These proteins, termed "C", are basic, have low amino acid homology and some secondary structure conservation. C proteins are encoded in alternative reading frames of the phosphoprotein gene. Some viruses express nested sets of C proteins that exert their functions in different locations: In the nucleus, they interfere with cellular transcription factors that elicit innate immune responses; in the cytoplasm, they associate with viral ribonucleocapsids and control polymerase processivity and orderly replication, thereby minimizing the activation of innate immunity. In addition, certain C proteins can directly bind to, and interfere with the function of, several cytoplasmic proteins required for interferon induction, interferon signaling and inflammation. Some C proteins are also required for efficient virus particle assembly and budding. C-deficient viruses can be grown in certain transformed cell lines but are not pathogenic in natural hosts. C proteins affect the same host functions as other phosphoprotein gene-encoded proteins named V but use different strategies for this purpose. Multiple independent systems to counteract host defenses may ensure efficient immune evasion and facilitate virus adaptation to new hosts and tissue environments.


Subject(s)
Immunity, Innate/immunology , Paramyxoviridae Infections/immunology , Paramyxovirinae/physiology , Phosphoproteins/immunology , Viral Proteins/immunology , Virus Replication/physiology , Animals , Defective Interfering Viruses , Genome, Viral , Humans , Immune Evasion , Inflammasomes , Open Reading Frames , Paramyxovirinae/genetics , Phosphoproteins/chemistry , Phosphoproteins/genetics , Phosphoproteins/metabolism , Phylogeny , Viral Proteins/chemistry , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Assembly
11.
Jpn J Infect Dis ; 75(1): 56-62, 2022 Jan 24.
Article in English | MEDLINE | ID: mdl-34193665

ABSTRACT

Both human respiratory syncytial virus (RSV) and human metapneumovirus (hMPV) cause immune-mediated under-five acute respiratory infections (ARIs), but differences in their disease pathogenesis, if any, are not well-known. This study was undertaken to analyze the epidemiological and immunological features of RSV and hMPV infections. Nasopharyngeal aspirates from children (aged 2 months to 5 years) with ARI, presenting to our tertiary care center between December 2013 and March 2016, were subjected to real-time polymerase chain reaction for the detection of RSV and hMPV. Positive samples were analyzed for co-infection and cytokine levels. Of the 349 nasopharyngeal aspirates, RSV was detected in 40.68% (142/349), hMPV in 6.59% (23/349), and both in 1.4% (5/349). Co-infections were common, with rhinovirus being the most common co-offender. The demographic and clinical parameters of RSV- and hMPV-infected children were comparable. The MMP-9/TIMP-1 ratio was significantly higher in RSV-mediated ARI and IFN-γ in hMPV-mediated ARI. Both RSV and hMPV are common among North Indian children with ARI, and coinfections are common. Their clinical features are non-discriminatory, and molecular diagnosis should be utilized to ascertain their individual epidemiology. The differences in their immune-pathogenesis (MMP-9/TIMP-1 ratio in RSV and IFN-γ in hMPV) could serve as useful tools for developing newer drugs.


Subject(s)
Cytokines/immunology , Paramyxoviridae Infections , Respiratory Syncytial Virus Infections , Respiratory Tract Infections , Child, Preschool , Humans , Infant , Metapneumovirus , Paramyxoviridae Infections/epidemiology , Paramyxoviridae Infections/immunology , Respiratory Syncytial Virus Infections/epidemiology , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus, Human , Respiratory Tract Infections/epidemiology , Respiratory Tract Infections/immunology
12.
Sci Rep ; 11(1): 20769, 2021 10 21.
Article in English | MEDLINE | ID: mdl-34675220

ABSTRACT

Human metapneumovirus (HMPV) is a leading cause of acute lower respiratory tract illness in children and adults. Repeated infections are common and can be severe in young, elderly, and immunocompromised persons due to short-lived protective humoral immunity. In turn, few protective T cell epitopes have been identified in humans. Thus, we infected transgenic mice expressing the common human HLA MHC-I allele B*07:02 (HLA-B7) with HMPV and screened a robust library of overlapping and computationally predicted HLA-B7 binding peptides. Six HLA-B7-restricted CD8+ T cell epitopes were identified using ELISPOT screening in the F, M, and N proteins, with M195-203 (M195) eliciting the strongest responses. MHC-tetramer flow cytometric staining confirmed HLA-B7 epitope-specific CD8+ T cells migrated to lungs and spleen of HMPV-immune mice. Immunization with pooled HLA-B7-restricted peptides reduced viral titer and protected mice from virulent infection. Finally, we confirmed that CD8+ T cells from HLA-B7 positive humans also recognize the identified epitopes. These results enable identification of HMPV-specific CD8+ T cells in humans and help to inform future HMPV vaccine design.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , HLA-B7 Antigen/immunology , Metapneumovirus/immunology , Paramyxoviridae Infections/immunology , Animals , Cells, Cultured , Epitopes, T-Lymphocyte/therapeutic use , Humans , Interferon-gamma/immunology , Mice, Inbred C57BL , Mice, Transgenic , Paramyxoviridae Infections/prevention & control , Peptides/immunology , Peptides/therapeutic use , Viral Vaccines/immunology , Viral Vaccines/therapeutic use
13.
Infect Genet Evol ; 96: 105096, 2021 12.
Article in English | MEDLINE | ID: mdl-34601094

ABSTRACT

Human metapneumovirus (HMPV), an unsegmented negative-strand RNA virus, is the second most detected respiratory pathogen and one of the leading causes of respiratory illness in infants and immunodeficient individuals. HMPV infection of permissive cells in culture triggers a transient IFN response, which is efficiently suppressed later in infection. We report that two structural glycoproteins of the virus - namely G (Glycoprotein) and SH (Small Hydrophobic) - suppress the type I interferon (IFN) response in cell culture. This is manifested by inhibition of diverse steps of IFN induction and response, such as phosphorylation and nuclear translocation of IFN regulatory factor-3 and -7 (IRF3, IRF7), major transcription factors of the IFN gene. Furthermore, HMPV suppresses the cellular response to IFN by inhibiting the phosphorylation of STAT1 (Signal Transducer and Activator of Transcription 1), required for the induction of IFN-stimulated genes that act as antivirals. Site-directed mutagenesis revealed an important role of critical cysteine (Cys) residues in the Cys-rich carboxy terminal region of the SH protein in IFN suppression, whereas for G, the ectodomain plays a role. These results shed light on the mechanism of IFN suppression by HMPV, and may also offer avenues for new antiviral approaches in the future.


Subject(s)
Glycoproteins/metabolism , Immunity, Innate , Paramyxoviridae Infections/immunology , Viral Proteins/metabolism , A549 Cells , Humans , Metapneumovirus/physiology , Paramyxoviridae Infections/virology
14.
J Virol ; 95(18): e0059321, 2021 08 25.
Article in English | MEDLINE | ID: mdl-34160259

ABSTRACT

Human metapneumovirus (hMPV) is an important cause of acute viral respiratory infection. As the only target of neutralizing antibodies, the hMPV fusion (F) protein has been a major focus for vaccine development and targeting by drugs and monoclonal antibodies (MAbs). While X-ray structures of trimeric prefusion and postfusion hMPV F proteins from genotype A, and monomeric prefusion hMPV F protein from genotype B have been determined, structural data for the postfusion conformation for genotype B is lacking. We determined the crystal structure of this protein and compared the structural differences of postfusion hMPV F between hMPV A and B genotypes. We also assessed the receptor binding properties of the hMPV F protein to heparin and heparan sulfate (HS). A library of HS oligomers was used to verify the HS binding activity of hMPV F, and several compounds showed binding to predominantly prefusion hMPV F, but had limited binding to postfusion hMPV F. Furthermore, MAbs to antigenic sites III and the 66-87 intratrimeric epitope block heparin binding. In addition, we evaluated the efficacy of postfusion hMPV B2 F protein as a vaccine candidate in BALB/c mice. Mice immunized with hMPV B2 postfusion F protein showed a balanced Th1/Th2 immune response and generated neutralizing antibodies against both subgroup A2 and B2 hMPV strains, which protected the mice from hMPV challenge. Antibody competition analysis revealed the antibodies generated by immunization target two known antigenic sites (III and IV) on the hMPV F protein. Overall, this study provides new characteristics of the hMPV F protein, which may be informative for vaccine and therapy development. IMPORTANCE Human metapneumovirus (hMPV) is an important cause of viral respiratory disease. In this paper, we report the X-ray crystal structure of the hMPV fusion (F) protein in the postfusion conformation from genotype B. We also assessed binding of the hMPV F protein to heparin and heparan sulfate, a previously reported receptor for the hMPV F protein. Furthermore, we determined the immunogenicity and protective efficacy of postfusion hMPV B2 F protein, which is the first study using a homogenous conformation of the protein. Antibodies generated in response to vaccination give a balanced Th1/Th2 response and target two previously discovered neutralizing epitopes.


Subject(s)
Antibodies, Viral/immunology , Epitopes/immunology , Heparin/metabolism , Metapneumovirus/immunology , Paramyxoviridae Infections/immunology , Viral Fusion Proteins/chemistry , Viral Fusion Proteins/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Female , Heparin/analogs & derivatives , Humans , Immunization , Male , Mice , Mice, Inbred BALB C , Paramyxoviridae Infections/metabolism , Paramyxoviridae Infections/virology , Protein Binding , Protein Conformation , Proteoglycans/metabolism , Th1 Cells/immunology , Th2 Cells/immunology , Viral Fusion Proteins/metabolism
15.
Viruses ; 13(3)2021 03 22.
Article in English | MEDLINE | ID: mdl-33809875

ABSTRACT

Human metapneumovirus (hMPV) is one of the main pathogens responsible for acute respiratory infections in children up to 5 years of age, contributing substantially to health burden. The worldwide economic and social impact of this virus is significant and must be addressed. The structural components of hMPV (either proteins or genetic material) can be detected by several receptors expressed by host cells through the engagement of pattern recognition receptors. The recognition of the structural components of hMPV can promote the signaling of the immune response to clear the infection, leading to the activation of several pathways, such as those related to the interferon response. Even so, several intrinsic factors are capable of modulating the immune response or directly inhibiting the replication of hMPV. This article will discuss the current knowledge regarding the innate and adaptive immune response during hMPV infections. Accordingly, the host intrinsic components capable of modulating the immune response and the elements capable of restricting viral replication during hMPV infections will be examined.


Subject(s)
Adaptive Immunity , Immunity, Innate , Metapneumovirus/immunology , Paramyxoviridae Infections/immunology , Child, Preschool , Host Microbial Interactions , Humans
16.
Vet Immunol Immunopathol ; 231: 110151, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33227621

ABSTRACT

Nucleocapsid (N) protein is the most highly expressed of all avian metapneumovirus (aMPV) viral proteins and stimulates a substantial immune response in infected animals. Codon optimized recombinant N (rec-N) protein from aMPV subtypes A, B, and C were expressed using the baculoviral expression system in Trichoplusia ni (Tni) insect cells. A mixture of purified rec-N antigens from each subtype was used as a coating antigen and was evaluated in indirect ELISA (iELISA) to assess antibody response in serum samples collected from experimentally infected chickens and turkeys with different aMPV subtypes. Also, archived field serum samples that were collected from different poultry submissions were used. Receiver operating characteristic (ROC) analysis was performed using chicken and turkey serum samples that were confirmed by indirect fluorescent antibody (IFA) test for serostatus (positive n = 270, negative n = 610). The ROC analysis showed sensitivity and specificity of 97 % at a cut-off value of 0.25. The rec-N iELISA was compared with a commercial whole virus-based APV kit. The rec-N iELISA showed comparable results in detecting antibody response in aMPV infected chicken sera but was more sensitive in detecting early antibody response in aMPV infected turkey serum samples. Our results further confirm the presence of aMPV antibodies in Canadian domestic poultry populations. The developed aMPV-rec N iELISA offers a safe and valuable alternative to whole virus-based iELISA for serodiagnosis and seroepidemiological surveillance of the disease in domestic poultry.


Subject(s)
Antibodies, Viral/blood , Enzyme-Linked Immunosorbent Assay/veterinary , Metapneumovirus/immunology , Nucleocapsid/genetics , Paramyxoviridae Infections/veterinary , Poultry Diseases/immunology , Animals , Antibodies, Viral/immunology , Chickens/immunology , Chlorocebus aethiops , Enzyme-Linked Immunosorbent Assay/methods , Nucleocapsid/immunology , Paramyxoviridae Infections/blood , Paramyxoviridae Infections/immunology , Poultry Diseases/blood , Poultry Diseases/virology , Turkeys/immunology , Vero Cells
17.
Acta Med Acad ; 49(2): 130-143, 2020 Aug.
Article in English | MEDLINE | ID: mdl-33189119

ABSTRACT

In this review, we discuss the latest developments in research pertaining to virus-induced asthma exacerbations and consider recent advances in treatment options. Asthma is a chronic disease of the airways that continues to impose a substantial clinical burden worldwide. Asthma exacerbations, characterised by an acute deterioration in respiratory symptoms and airflow obstruction, are associated with significant morbidity and mortality. These episodes are most commonly triggered by respiratory virus infections. The mechanisms underlying the pathogenesis of virus-induced exacerbations have been the focus of extensive biomedical research. Developing a robust understanding of the interplay between respiratory viruses and the host immune response will be critical for developing more efficacious, targeted therapies for exacerbations. CONCLUSION: There has been significant recent progress in our understanding of the mechanisms underlying virus-induced airway inflammation in asthma and these advances will underpin the development of future clinical therapies.


Subject(s)
Anti-Asthmatic Agents/therapeutic use , Antiviral Agents/therapeutic use , Asthma/drug therapy , Respiratory Tract Infections/drug therapy , Virus Diseases/drug therapy , Adenovirus Infections, Human/drug therapy , Adenovirus Infections, Human/immunology , Adenovirus Infections, Human/physiopathology , Administration, Inhalation , Asthma/immunology , Asthma/physiopathology , Coronavirus Infections/drug therapy , Coronavirus Infections/immunology , Coronavirus Infections/physiopathology , Disease Progression , Humans , Influenza, Human/drug therapy , Influenza, Human/immunology , Influenza, Human/physiopathology , Interferon-beta/therapeutic use , Macrolides/therapeutic use , Omalizumab/therapeutic use , Paramyxoviridae Infections/drug therapy , Paramyxoviridae Infections/immunology , Paramyxoviridae Infections/physiopathology , Picornaviridae Infections/drug therapy , Picornaviridae Infections/immunology , Picornaviridae Infections/physiopathology , Respiratory Syncytial Virus Infections/drug therapy , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/physiopathology , Respiratory Tract Infections/immunology , Respiratory Tract Infections/physiopathology , Virus Diseases/immunology , Virus Diseases/physiopathology
18.
Antiviral Res ; 184: 104903, 2020 12.
Article in English | MEDLINE | ID: mdl-32800881

ABSTRACT

Caprine parainfluenza virus type3 (CPIV3) is a newly identified member of Paramyxoviridae family. CPIV3 is highly prevalence in China and showed pathogenicity to goats; in addition, CPIV3 infection causes severe clinical disease under stress and/or co-infection conditions. Viperin is one of the hundreds of interferon-stimulated genes (ISGs), and possesses a wide range of antiviral activities. The aim of this study was to systemically explore the anti-CPIV3 activity of ruminants' Viperin. CPIV3 infection up-regulated Viperin transcription but not protein expression in MDBK cells. Bovine and caprine Viperin genes (bVi and gVi) were amplified and analyzed by BLAST and multiple alignment. The obtained bVi/gVi amino acid sequences showed 99.5%-100% identity with previously submitted sequences and has variants at N-terminal domain (1-70aa) between each other. The pcDNA3.1 plasmids containing bVi and gVi genes were constructed to over-express the target proteins. CPIV3 was inoculated in MDBK cells over-expressing bVi/gVi and viral load was detected by qRT-PCR, virus titration and Western blot. Both of the bVi and gVi significantly inhibited CPIV3 genome copy numbers and viral titers at 24 and 48 hpi (P < 0.01); and viral N protein expression was also decreased, comparing with those of mock transfected group. The last 50aa C-terminal region was crucial for its anti-CPIV3 activity. In addition, the over-expression of bVi/gVi did not influence CPIV3 binding, entry and release in the cells. These results indicated the anti-CPIV3 activity occurred in viral RNA/protein synthesis progress of the viral replication cycle. The Viperin also showed similar inhibitory effect on different CPIV3 strains. The potential interaction of Viperin with viral proteins (N, P, C and V) was determined by confocal laser scanning microscopy and Co-IP assay. Co-localization of Viperin with N, P or C, but not V, was observed; while only N protein direct interacted with Viperin in Co-IP test, no matter using viral protein expressing plasmids transfected or CPIV3 infected cell samples. In conclusion, the bVi and gVi Viperin effectively inhibited CPIV3 replication potentially via the interaction of Viperin with viral N protein. The present results gave more information about antiviral activity of ruminants Viperin and provided foundation for further studies of the interaction of Viperin with CPIV3 and other related viruses.


Subject(s)
Paramyxoviridae Infections/immunology , Proteins/immunology , Virus Replication , Amino Acid Sequence , Animals , Cattle , Cell Line , Gene Expression Regulation , Goat Diseases/immunology , Goat Diseases/virology , Goats , HEK293 Cells , Host Microbial Interactions , Humans , Nucleocapsid Proteins/metabolism , Paramyxoviridae/physiology , Paramyxoviridae Infections/veterinary , Reverse Transcriptase Polymerase Chain Reaction , Viral Load
19.
Viruses ; 12(7)2020 07 20.
Article in English | MEDLINE | ID: mdl-32698530

ABSTRACT

The Pneumoviridae family includes human metapneumovirus (HMPV) and human orthopneumovirus, which is also known as a respiratory syncytial virus (HRSV). These are large enveloped, negative single-strand RNA viruses. HMPV and HRSV are the human members, which commonly infect children. HMPV, which was discovered in 2001, infects most children until the age of five, which causes an influenza-like illness. The interaction of this virus with immune cells is poorly understood. In this study, we show that HMPV evades natural killer (NK) cell attack by downregulating stress-induced ligands for the activating receptor NKG2D including: Major histocompatibility complex (MHC) class I polypeptide-related sequences A and B (MICA, MICB), UL16 binding proteins ULBP2, and ULBP3, but not ULBP1. Mechanistically, we show that the viral protein G is involved in the downregulation of ULBP2 and that the viral protein M2.2 is required for MICA and MICB downregulation. These findings emphasize the importance of NK cells, in general, and NKG2D, in particular, in controlling HMPV infection, which opens new avenues for treating HMPV.


Subject(s)
Killer Cells, Natural/immunology , Metapneumovirus/immunology , NK Cell Lectin-Like Receptor Subfamily K/immunology , Antibodies, Viral/immunology , Blotting, Western , Down-Regulation , Flow Cytometry , Humans , Paramyxoviridae Infections/immunology , Real-Time Polymerase Chain Reaction , Viral Proteins/immunology
20.
Viruses ; 12(7)2020 07 04.
Article in English | MEDLINE | ID: mdl-32635475

ABSTRACT

The host tropism of viral infection is determined by a variety of factors, from cell surface receptors to innate immune signaling. Many viruses encode proteins that interfere with host innate immune recognition in order to promote infection. STAT2 is divergent between species and therefore has a role in species restriction of some viruses. To understand the role of STAT2 in human metapneumovirus (HMPV) infection of human and murine tissues, we first infected STAT2-/- mice and found that HMPV could be serially passaged in STAT2-/-, but not WT, mice. We then used in vitro methods to show that HMPV inhibits expression of both STAT1 and STAT2 in human and primate cells, but not in mouse cells. Transfection of the murine form of STAT2 into STAT2-deficient human cells conferred resistance to STAT2 inhibition. Finally, we sought to understand the in vivo role of STAT2 by infecting hSTAT2 knock-in mice with HMPV, and found that mice had increased weight loss, inhibition of type I interferon signaling, and a Th2-polarized cytokine profile compared to WT mice. These results indicate that STAT2 is a target of HMPV in human infection, while the murine version of STAT2 restricts tropism of HMPV for murine cells and tissue.


Subject(s)
Metapneumovirus/physiology , Paramyxoviridae Infections/immunology , STAT2 Transcription Factor/immunology , Animals , Female , Host Specificity , Humans , Immunity, Innate , Interferons/genetics , Interferons/immunology , Male , Metapneumovirus/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Paramyxoviridae Infections/genetics , Paramyxoviridae Infections/virology , STAT2 Transcription Factor/genetics , Th2 Cells
SELECTION OF CITATIONS
SEARCH DETAIL
...