Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 162
Filtrer
1.
J Virol ; 98(6): e0164123, 2024 Jun 13.
Article de Anglais | MEDLINE | ID: mdl-38690874

RÉSUMÉ

Numerous viruses have been found to exploit glycoconjugates expressed on human cells as their initial attachment factor for viral entry and infection. The virus-cell glycointeractome, when characterized, may serve as a template for antiviral drug design. Heparan sulfate proteoglycans extensively decorate the human cell surface and were previously described as a primary receptor for human metapneumovirus (HMPV). After respiratory syncytial virus, HMPV is the second most prevalent respiratory pathogen causing respiratory tract infection in young children. To date, there is neither vaccine nor drug available to prevent or treat HMPV infection. Using a multidisciplinary approach, we report for the first time the glycointeractome of the HMPV fusion (F) protein, a viral surface glycoprotein that is essential for target-cell recognition, attachment, and entry. Our glycan microarray and surface plasmon resonance results suggest that Galß1-3/4GlcNAc moieties that may be sialylated or fucosylated are readily recognized by HMPV F. The bound motifs are highly similar to the N-linked and O-linked glycans primarily expressed on the human lung epithelium. We demonstrate that the identified glycans have the potential to compete with the cellular receptors used for HMPV entry and consequently block HMPV infection. We found that lacto-N-neotetraose demonstrated the strongest HMPV binding inhibition in a cell infection assay. Our current findings offer an encouraging and novel avenue for the design of anti-HMPV drug candidates using oligosaccharide templates.IMPORTANCEAll cells are decorated with a dense coat of sugars that makes a sugar code. Many respiratory viruses exploit this sugar code by binding to these sugars to cause infection. Human metapneumovirus is a leading cause for acute respiratory tract infections. Despite its medical importance, there is no vaccine or antiviral drug available to prevent or treat human metapneumovirus infection. This study investigates how human metapneumovirus binds to sugars in order to more efficiently infect the human host. We found that human metapneumovirus binds to a diverse range of sugars and demonstrated that these sugars can ultimately block viral infection. Understanding how viruses can take advantage of the sugar code on our cells could identify new intervention and treatment strategies to combat viral disease.


Sujet(s)
Metapneumovirus , Infections à Paramyxoviridae , Polyosides , Metapneumovirus/métabolisme , Metapneumovirus/physiologie , Humains , Polyosides/métabolisme , Infections à Paramyxoviridae/virologie , Infections à Paramyxoviridae/métabolisme , Protéines de fusion virale/métabolisme , Pénétration virale , Attachement viral , Liaison aux protéines , Récepteurs viraux/métabolisme , Lignée cellulaire
2.
mSphere ; 9(2): e0074323, 2024 Feb 28.
Article de Anglais | MEDLINE | ID: mdl-38265200

RÉSUMÉ

Human metapneumovirus (HMPV), a member of the Pneumoviridae family, causes upper and lower respiratory tract infections in humans. In vitro studies with HMPV have mostly been performed in monolayers of undifferentiated epithelial cells. In vivo studies in cynomolgus macaques and cotton rats have shown that ciliated epithelial cells are the main target of HMPV infection, but these observations cannot be studied in monolayer systems. Here, we established an organoid-derived bronchial culture model that allows physiologically relevant studies on HMPV. Inoculation with multiple prototype HMPV viruses and recent clinical virus isolates led to differences in replication among HMPV isolates. Prolific HMPV replication in this model caused damage to the ciliary layer, including cilia loss at advanced stages post-infection. These cytopathic effects correlated with those observed in previous in vivo studies with cynomolgus macaques. The assessment of the innate immune responses in three donors upon HMPV and RSV inoculation highlighted the importance of incorporating multiple donors to account for donor-dependent variation. In conclusion, these data indicate that the organoid-derived bronchial cell culture model resembles in vivo findings and is therefore a suitable and robust model for future HMPV studies. IMPORTANCE: Human metapneumovirus (HMPV) is one of the leading causative agents of respiratory disease in humans, with no treatment or vaccine available yet. The use of primary epithelial cultures that recapitulate the tissue morphology and biochemistry of the human airways could aid in defining more relevant targets to prevent HMPV infection. For this purpose, this study established the first primary organoid-derived bronchial culture model suitable for a broad range of HMPV isolates. These bronchial cultures were assessed for HMPV replication, cellular tropism, cytopathology, and innate immune responses, where the observations were linked to previous in vivo studies with HMPV. This study exposed an important gap in the HMPV field since extensively cell-passaged prototype HMPV B viruses did not replicate in the bronchial cultures, underpinning the need to use recently isolated viruses with a controlled passage history. These results were reproducible in three different donors, supporting this model to be suitable to study HMPV infection.


Sujet(s)
Metapneumovirus , Infections à Paramyxoviridae , Humains , Animaux , Metapneumovirus/physiologie , Cytologie , Réplication virale , Infections à Paramyxoviridae/anatomopathologie , Épithélium , Macaca , Tropisme
3.
mSphere ; 9(1): e0057023, 2024 Jan 30.
Article de Anglais | MEDLINE | ID: mdl-38197640

RÉSUMÉ

Human metapneumovirus (HMPV) is a negative-sense single-stranded RNA virus in the Pneumoviridae family and a leading cause of acute upper and lower respiratory infections, particularly in children, immunocompromised patients, and the elderly. Although nearly every person is infected with HMPV during early childhood, re-infections occur often, highlighting difficulty in building long-term immunity. Inflammatory responses, including PD-1-mediated impairment of virus-specific CD8+ T cells (TCD8), contribute to HMPV disease severity. HMPV strains are divided into four lineages: A1, A2, B1, and B2. However, little is known about immune responses to different viral subtypes. Here, we characterize responses to four HMPV clinical isolates-TN/94-344 (A1), TN/94-49 (A2), C2-202 (B1), and TN/96-35 (B2)-in vivo in C57BL/6 (B6) mice. TN/94-49 was avirulent, while TN/94-344, C2-202, and TN/96-35 showed varying degrees of weight loss and clinical disease. Differences in disease did not correlate to virus burden in upper or lower tracts. TN/94-49 HMPV exhibited highest nose titers and delayed lung clearance. Cytokine profiles differed between HMPV isolates, with TN/96-35 inducing the broadest lung inflammatory cytokines. TN/96-35 also showed lower HMPV burden and less weight loss than other virulent isolates, suggesting a more efficient antiviral response. Interestingly, disease correlated with higher expression of T-cell chemoattractant CXCL9. All isolates elicited PD-1 upregulation and decreased IFNγ and CD107a expression in virus-specific TCD8, with little difference between HMPV subtypes. This work uncovers previously uncharacterized variations in immune responses to clinical HMPV isolates of different lineages.IMPORTANCEThis study extensively explored differences in T-cell-mediated immunity between human metapneumovirus (HMPV) clinical isolates. Much existing HMPV research has been done with strains passaged extensively in cell lines, likely acquiring mutations advantageous to in vitro replication. Clinical isolates are collected directly from human patients and have undergone <10 passages, serving as more physiologically relevant models of HMPV infection. Additionally, existing animal studies of HMPV disease mainly focus on lung pathogenesis, while HMPV infects both upper and lower airways of humans. This work highlights distinct differences in HMPV burden in upper and lower tracts between clinical isolates. Lastly, this study uniquely explores differences in host immunity between all four HMPV genetic lineages. The predominant HMPV subtype in circulation varies seasonally; thus, understanding host responses to all subgroups is critical for developing effective HMPV vaccines.


Sujet(s)
Metapneumovirus , Enfant d'âge préscolaire , Enfant , Humains , Souris , Animaux , Sujet âgé , Metapneumovirus/physiologie , Lymphocytes T CD8+ , Récepteur-1 de mort cellulaire programmée , Souris de lignée C57BL , Poumon/anatomopathologie , Perte de poids
4.
J Virol ; 97(12): e0105223, 2023 Dec 21.
Article de Anglais | MEDLINE | ID: mdl-38032197

RÉSUMÉ

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.


Sujet(s)
Metapneumovirus , Infections à Paramyxoviridae , Sujet âgé , Humains , Anticorps neutralisants , Anticorps antiviraux , Épitopes , Metapneumovirus/physiologie , Infections à Paramyxoviridae/immunologie , Protéines de fusion virale , Vaccins antiviraux/immunologie
5.
Immunohorizons ; 7(6): 398-411, 2023 06 01.
Article de Anglais | MEDLINE | ID: mdl-37261717

RÉSUMÉ

Human metapneumovirus (HMPV) is a leading cause of respiratory infection in adults >65 y. Nearly all children worldwide are seropositive for HMPV by age 5 y, but reinfections occur throughout life, and there is no licensed vaccine. Recurrent HMPV infection is mild and self-resolving in immunocompetent individuals. However, elderly individuals develop severe respiratory disease on HMPV reinfection that leads to a high risk for morbidity and mortality. In this study, we developed a mouse model to mirror HMPV reinfection in elderly humans. C57BL/6J mice were infected with HMPV at 6-7 wk old, aged in-house, and rechallenged with high-dose virus at 70 wk. Aged rechallenged mice had profound weight loss similar to primary infected mice, increased lung histopathology, and accumulated cytotoxic CD8+CD44+CD62L-CD69+CD103+ memory cells despite having undetectable lung virus titer. When aged mice 14 mo postinfection (p.i.) or young mice 5 wk p.i. were restimulated with HMPV cognate Ag to mimic epitope vaccination, aged mice had an impaired CD8+ memory response. Convalescent serum transfer from young naive or 5 wk p.i. mice into aged mice on day of infection did not protect. Aged mice vaccinated with UV-inactivated HMPV also exhibited diminished protection and poor CD8+ memory response compared with young mice. These results suggest aged individuals with HMPV reinfection have a dysregulated CD8+ memory T cell response that fails to protect and exacerbates disease. Moreover, aged mice exhibited a poor memory response to either epitope peptide or UV-inactivated vaccination, suggesting that aged CD8+ T cell dysfunction presents a barrier to effective vaccination strategies.


Sujet(s)
Metapneumovirus , Sujet âgé , Animaux , Humains , Souris , Épitopes , Metapneumovirus/physiologie , Souris de lignée C57BL , Acuité des besoins du patient , Réinfection
6.
J Virol ; 97(5): e0003023, 2023 05 31.
Article de Anglais | MEDLINE | ID: mdl-37092993

RÉSUMÉ

Human metapneumovirus (HMPV) is a negative-strand RNA virus that frequently causes respiratory tract infections in infants, the elderly, and the immunocompromised. A hallmark of HMPV infection is the formation of membraneless, liquid-like replication and transcription centers in the cytosol termed inclusion bodies (IBs). The HMPV phosphoprotein (P) and nucleoprotein (N) are the minimal viral proteins necessary to form IB-like structures, and both proteins are required for the viral polymerase to synthesize RNA during infection. HMPV P is a homotetramer with regions of intrinsic disorder and has several known and predicted phosphorylation sites of unknown function. In this study, we found that the P C-terminal intrinsically disordered domain (CTD) must be present to facilitate IB formation with HMPV N, while either the N-terminal intrinsically disordered domain or the central oligomerization domain was dispensable. Alanine substitution at a single tyrosine residue within the CTD abrogated IB formation and reduced coimmunoprecipitation with HMPV N. Mutations to C-terminal phosphorylation sites revealed a potential role for phosphorylation in regulating RNA synthesis and P binding partners within IBs. Phosphorylation mutations which reduced RNA synthesis in a reporter assay produced comparable results in a recombinant viral rescue system, measured as an inability to produce infectious viral particles with genomes containing these single P mutations. This work highlights the critical role HMPV P plays in facilitating a key step of the viral life cycle and reveals the potential role for phosphorylation in regulating the function of this significant viral protein. IMPORTANCE Human metapneumovirus (HMPV) infects global populations, with severe respiratory tract infections occurring in infants, the elderly, and the immunocompromised. There are currently no FDA-approved therapeutics available to prevent or treat HMPV infection. Therefore, understanding how HMPV replicates is vital for the identification of novel targets for therapeutic development. During HMPV infection, viral RNA synthesis proteins localize to membraneless structures called inclusion bodies (IBs), which are sites of genome replication and transcription. The HMPV phosphoprotein (P) is necessary for IBs to form and for the virus to synthesize RNA, but it is not known how this protein contributes to IB formation or if it is capable of regulating viral replication. We show that the C-terminal domain of P is the location of a molecular interaction driving IB formation and contains potential phosphorylation sites where amino acid charge regulates the function of the viral polymerase complex.


Sujet(s)
Metapneumovirus , Infections à Paramyxoviridae , Sujet âgé , Humains , Lignée cellulaire , Metapneumovirus/physiologie , Nucleotidyltransferases , Infections à Paramyxoviridae/virologie , Phosphoprotéines/génétique , Phosphoprotéines/métabolisme , Infections de l'appareil respiratoire , ARN , Protéines virales/génétique , Protéines virales/métabolisme , Compartiments de réplication virale/métabolisme , Réplication virale , Corps d'inclusion viraux/métabolisme
7.
Cell Rep ; 40(12): 111399, 2022 09 20.
Article de Anglais | MEDLINE | ID: mdl-36130517

RÉSUMÉ

Human metapneumovirus (hMPV) is a major cause of acute respiratory infections in infants and older adults, for which no vaccines or therapeutics are available. The viral fusion (F) glycoprotein is required for entry and is the primary target of neutralizing antibodies; however, little is known about the humoral immune response generated from natural infection. Here, using prefusion-stabilized F proteins to interrogate memory B cells from two older adults, we obtain over 700 paired non-IgM antibody sequences representing 563 clonotypes, indicative of a highly polyclonal response. Characterization of 136 monoclonal antibodies reveals broad recognition of the protein surface, with potently neutralizing antibodies targeting each antigenic site. Cryo-EM studies further reveal two non-canonical sites and the molecular basis for recognition of the apex of hMPV F by two prefusion-specific neutralizing antibodies. Collectively, these results provide insight into the humoral response to hMPV infection in older adults and will help guide vaccine development.


Sujet(s)
Metapneumovirus , Sujet âgé , Anticorps monoclonaux , Anticorps neutralisants , Anticorps antiviraux , Humains , Metapneumovirus/physiologie , Protéines de fusion virale
8.
J Virol ; 96(2): e0090921, 2022 01 26.
Article de Anglais | MEDLINE | ID: mdl-34730389

RÉSUMÉ

Human metapneumovirus (HMPV) causes severe respiratory diseases in young children. The HMPV RNA genome is encapsidated by the viral nucleoprotein (N), forming an RNA-N complex (NNuc), which serves as the template for genome replication and mRNA transcription by the RNA-dependent RNA polymerase (RdRp). The RdRp is formed by the association of the large polymerase subunit (L), which has RNA polymerase, capping, and methyltransferase activities, and the tetrameric phosphoprotein (P). P plays a central role in the RdRp complex by binding to NNuc and L, allowing the attachment of the L polymerase to the NNuc template. During infection these proteins concentrate in cytoplasmic inclusion bodies (IBs) where viral RNA synthesis occurs. By analogy to the closely related pneumovirus respiratory syncytial virus (RSV), it is likely that the formation of IBs depends on the interaction between HMPV P and NNuc, which has not been demonstrated yet. Here, we finely characterized the binding P-NNuc interaction domains by using recombinant proteins, combined with a functional assay for the polymerase complex activity, and the study of the recruitment of these proteins to IBs by immunofluorescence. We show that the last 6 C-terminal residues of HMPV P are necessary and sufficient for binding to NNuc and that P binds to the N-terminal domain of N (NNTD), and we identified conserved N residues critical for the interaction. Our results allowed us to propose a structural model for the HMPV P-NNuc interaction. IMPORTANCE Human metapneumovirus (HMPV) is a leading cause of severe respiratory infections in children but also affects human populations of all ages worldwide. Currently, no vaccine or efficient antiviral treatments are available for this pneumovirus. A better understanding of the molecular mechanisms involved in viral replication could help the design or discovery of specific antiviral compounds. In this work, we have investigated the interaction between two major viral proteins involved in HMPV RNA synthesis, the N and P proteins. We finely characterized their domains of interaction and identified a pocket on the surface of the N protein, a potential target of choice for the design of compounds interfering with N-P complexes and inhibiting viral replication.


Sujet(s)
Metapneumovirus/composition chimique , Protéines nucléocapside/composition chimique , Phosphoprotéines/composition chimique , Animaux , Sites de fixation , Lignée cellulaire , Cricetinae , Corps d'inclusion/métabolisme , Metapneumovirus/physiologie , Modèles moléculaires , Mutation , Protéines nucléocapside/génétique , Protéines nucléocapside/métabolisme , Phosphoprotéines/génétique , Phosphoprotéines/métabolisme , Liaison aux protéines , Motifs et domaines d'intéraction protéique , ARN viral/métabolisme , RNA replicase/métabolisme , Réplication virale
9.
PLoS One ; 16(12): e0259443, 2021.
Article de Anglais | MEDLINE | ID: mdl-34919553

RÉSUMÉ

The present study was done to identify the viral diversity, seasonality and burden associated with childhood acute respiratory tract infection (ARTI) in Sri Lanka. Nasopharyngeal aspirates (NPA) of hospitalized children (1 month-5 years) with ARTI were collected in 2 centers (wet and dry zones) from March 2013 to August 2014. Respiratory viral antigen detection by immunofluorescence assay (IFA) was used to identify the infecting viruses. IFA negative 100 NPA samples were tested for human metapeumovirus (hMPV), human bocavirus and corona viruses by polymerase chain reaction. Of the 443 and 418 NPAs, 37.2% and 39.4% were positive for any of the 8 different respiratory viruses tested from two centers studied. Viral co-infection was detected with respiratory syncytial virus (RSV) in both centers. Peak viral detection was noted in the wet zone from May-July 2013 and 2014 and in the dry zone from December-January 2014 suggesting a local seasonality for viral ARTI. RSV showed a clear seasonality with a direct correlation of monthly RSV infections with rainy days in the wet zone and an inverse correlation with temperature in both centers. The case fatality rate was 2.7% for RSV associated ARTI. The overall disability adjusted life years was 335.9 and for RSV associated ARTI it was 241.8. RSV was the commonly detected respiratory virus with an annual seasonality and distribution in rainy seasons in the dry and wet zones of Sri Lanka. Identifying the virus and seasonality will contribute to employ preventive measures and reduce the empirical use of antibiotics in resource limited settings.


Sujet(s)
Infections à coronavirus/épidémiologie , Infections à Paramyxoviridae/épidémiologie , Infections à Parvoviridae/épidémiologie , Infections à virus respiratoire syncytial/épidémiologie , Infections de l'appareil respiratoire/épidémiologie , Charge virale , Enfant hospitalisé , Enfant d'âge préscolaire , Co-infection , Coronavirus/pathogénicité , Coronavirus/physiologie , Infections à coronavirus/mortalité , Infections à coronavirus/virologie , Espérance de vie corrigée de l'incapacité/tendances , Femelle , Bocavirus humain/pathogénicité , Bocavirus humain/physiologie , Humains , Incidence , Nourrisson , Mâle , Metapneumovirus/pathogénicité , Metapneumovirus/physiologie , Infections à Paramyxoviridae/mortalité , Infections à Paramyxoviridae/virologie , Infections à Parvoviridae/mortalité , Infections à Parvoviridae/virologie , Infections à virus respiratoire syncytial/mortalité , Infections à virus respiratoire syncytial/virologie , Virus respiratoire syncytial humain/pathogénicité , Virus respiratoire syncytial humain/physiologie , Infections de l'appareil respiratoire/mortalité , Infections de l'appareil respiratoire/virologie , Saisons , Sri Lanka/épidémiologie , Analyse de survie
10.
Infect Genet Evol ; 96: 105096, 2021 12.
Article de Anglais | MEDLINE | ID: mdl-34601094

RÉSUMÉ

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.


Sujet(s)
Glycoprotéines/métabolisme , Immunité innée , Infections à Paramyxoviridae/immunologie , Protéines virales/métabolisme , Cellules A549 , Humains , Metapneumovirus/physiologie , Infections à Paramyxoviridae/virologie
11.
Viruses ; 13(6)2021 06 21.
Article de Anglais | MEDLINE | ID: mdl-34205489

RÉSUMÉ

The recently discovered exchange protein directly activated by cAMP (EPAC), compared with protein kinase A (PKA), is a fairly new family of cAMP effectors. Soon after the discovery, EPAC has shown its significance in many diseases including its emerging role in infectious diseases. In a recent study, we demonstrated that EPAC, but not PKA, is a promising therapeutic target to regulate respiratory syncytial virus (RSV) replication and its associated inflammation. In mammals, there are two isoforms of EPAC-EPAC1 and EPAC2. Unlike other viruses, including Middle East respiratory syndrome coronavirus (MERS-CoV) and Ebola virus, which use EPAC1 to regulate viral replication, RSV uses EPAC2 to control its replication and associated cytokine/chemokine responses. To determine whether EPAC2 protein has a broad impact on other respiratory viral infections, we used an EPAC2-specific inhibitor, MAY0132, to examine the functions of EPAC2 in human metapneumovirus (HMPV) and adenovirus (AdV) infections. HMPV is a negative-sense single-stranded RNA virus belonging to the family Pneumoviridae, which also includes RSV, while AdV is a double-stranded DNA virus. Treatment with an EPAC1-specific inhibitor was also included to investigate the impact of EPAC1 on these two viruses. We found that the replication of HMPV, AdV, and RSV and the viral-induced immune mediators are significantly impaired by MAY0132, while an EPAC1-specific inhibitor, CE3F4, does not impact or slightly impacts, demonstrating that EPAC2 could serve as a novel common therapeutic target to control these viruses, all of which do not have effective treatment and prevention strategies.


Sujet(s)
Adenoviridae/physiologie , Facteurs d'échange de nucléotides guanyliques/génétique , Facteurs d'échange de nucléotides guanyliques/métabolisme , Metapneumovirus/physiologie , Virus respiratoire syncytial humain/physiologie , Réplication virale , Cellules A549 , Lignée cellulaire , Chimiokines/immunologie , Cyclic AMP-Dependent Protein Kinases/antagonistes et inhibiteurs , Cyclic AMP-Dependent Protein Kinases/métabolisme , Cellules épithéliales/effets des médicaments et des substances chimiques , Cellules épithéliales/virologie , Facteurs d'échange de nucléotides guanyliques/antagonistes et inhibiteurs , Cellules HEK293 , Humains , Quinoléines/pharmacologie
13.
Viruses ; 13(1)2021 Jan 19.
Article de Anglais | MEDLINE | ID: mdl-33478119

RÉSUMÉ

Respiratory viral infections constitute a global public health concern. Among prevalent respiratory viruses, two pneumoviruses can be life-threatening in high-risk populations. In young children, they constitute the first cause of hospitalization due to severe lower respiratory tract diseases. A better understanding of their pathogenesis is still needed as there are no approved efficient anti-viral nor vaccine against pneumoviruses. We studied Respiratory Syncytial virus (RSV) and human Metapneumovirus (HMPV) in single and dual infections in three-dimensional cultures, a highly relevant model to study viral respiratory infections of the airway epithelium. Our investigation showed that HMPV is less pathogenic than RSV in this model. Compared to RSV, HMPV replicated less efficiently, induced a lower immune response, did not block cilia beating, and was more sensitive to IFNs. In dual infections, RSV-infected epithelia were less permissive to HMPV. By neutralizing IFNs in co-infection assays, we partially prevented HMPV inhibition by RSV and significantly increased the number of co-infected cells in the tissue. This suggests that interference in dual infection would be at least partly mediated by the host immune response. In summary, this work provides new insight regarding virus-host and virus-virus interactions of pneumoviruses in the airway epithelium. This could be helpful for the proper handling of at-risk patients.


Sujet(s)
Techniques de culture cellulaire , Co-infection , Interactions hôte-pathogène , Metapneumovirus/physiologie , Interactions microbiennes , Virus respiratoire syncytial humain/physiologie , Réplication virale , Lignée cellulaire , Humains , Interféron de type I/pharmacologie , Interférons/pharmacologie , Metapneumovirus/effets des médicaments et des substances chimiques , Infections à Paramyxoviridae/virologie , Infections à virus respiratoire syncytial/virologie , Virus respiratoire syncytial humain/effets des médicaments et des substances chimiques , Sphéroïdes de cellules , Interféron lambda
14.
Viruses ; 13(1)2021 Jan 16.
Article de Anglais | MEDLINE | ID: mdl-33466993

RÉSUMÉ

BACKGROUND: Type-1 cryoglobulinemia (CG) is a rare disease associated with B-cell lymphoproliferative disorder. Some viral infections, such as Epstein-Barr Virus infections, are known to cause malignant lymphoproliferation, like certain B-cell lymphomas. However, their role in the pathogenesis of chronic lymphocytic leukemia (CLL) is still debatable. Here, we report a unique case of Type-1 CG associated to a CLL transformation diagnosed in the course of a human metapneumovirus (hMPV) infection. CASE PRESENTATION: A 91-year-old man was initially hospitalized for delirium. In a context of febrile rhinorrhea, the diagnosis of hMPV infection was made by molecular assay (RT-PCR) on nasopharyngeal swab. Owing to hyperlymphocytosis that developed during the course of the infection and unexplained peripheral neuropathy, a type-1 IgG Kappa CG secondary to a CLL was diagnosed. The patient was not treated for the CLL because of Binet A stage classification and his poor physical condition. CONCLUSIONS: We report the unique observation in the literature of CLL transformation and hMPV infection. We provide a mini review on the pivotal role of viruses in CLL pathophysiology.


Sujet(s)
Transformation cellulaire virale , Prédisposition aux maladies , Leucémie chronique lymphocytaire à cellules B/diagnostic , Leucémie chronique lymphocytaire à cellules B/étiologie , Metapneumovirus/physiologie , Infections à Paramyxoviridae/complications , Infections à Paramyxoviridae/virologie , Sujet âgé de 80 ans ou plus , Marqueurs biologiques , Évolution clonale , Cryoglobulinémie/diagnostic , Cryoglobulinémie/étiologie , Humains , Immunoglobuline G/sang , Chaines légères kappa des immunoglobulines/sang , Immunophénotypage , Mâle
15.
J Virol ; 94(20)2020 09 29.
Article de Anglais | MEDLINE | ID: mdl-32759319

RÉSUMÉ

Respiratory syncytial virus (RSV) and human metapneumovirus (HMPV) are two of the leading causes of respiratory infections in children and elderly and immunocompromised patients worldwide. There is no approved treatment for HMPV and only one prophylactic treatment against RSV, palivizumab, for high-risk infants. Better understanding of the viral lifecycles in a more relevant model system may help identify novel therapeutic targets. By utilizing three-dimensional (3-D) human airway tissues to examine viral infection in a physiologically relevant model system, we showed that RSV infects and spreads more efficiently than HMPV, with the latter requiring higher multiplicities of infection (MOIs) to yield similar levels of infection. Apical ciliated cells were the target for both viruses, but RSV apical release was significantly more efficient than HMPV. In RSV- or HMPV-infected cells, cytosolic inclusion bodies containing the nucleoprotein, phosphoprotein, and respective viral genomic RNA were clearly observed in human airway epithelial (HAE) culture. In HMPV-infected cells, actin-based filamentous extensions were more common (35.8%) than those found in RSV-infected cells (4.4%). Interestingly, neither RSV nor HMPV formed syncytia in HAE tissues. Palivizumab and nirsevimab effectively inhibited entry and spread of RSV in HAE tissues, with nirsevimab displaying significantly higher potency than palivizumab. In contrast, 54G10 completely inhibited HMPV entry but only modestly reduced viral spread, suggesting HMPV may use alternative mechanisms for spread. These results represent the first comparative analysis of infection by the two pneumoviruses in a physiologically relevant model, demonstrating an interesting dichotomy in the mechanisms of infection, spread, and consequent inhibition of the viral lifecycles by neutralizing monoclonal antibodies.IMPORTANCE Respiratory syncytial virus and human metapneumovirus are leading causes of respiratory illness worldwide, but limited treatment options are available. To better target these viruses, we examined key aspects of the viral life cycle in three-dimensional (3-D) human airway tissues. Both viruses establish efficient infection through the apical surface, but efficient spread and apical release were seen for respiratory syncytial virus (RSV) but not human metapneumovirus (HMPV). Both viruses form inclusion bodies, minimally composed of nucleoprotein (N), phosphoprotein (P), and viral RNA (vRNA), indicating that these structures are critical for replication in this more physiological model. HMPV formed significantly more long, filamentous actin-based extensions in human airway epithelial (HAE) tissues than RSV, suggesting HMPV may promote cell-to-cell spread via these extensions. Lastly, RSV entry and spread were fully inhibited by neutralizing antibodies palivizumab and the novel nirsevimab. In contrast, while HMPV entry was fully inhibited by 54G10, a neutralizing antibody, spread was only modestly reduced, further supporting a cell-to-cell spread mechanism.


Sujet(s)
Anticorps neutralisants/pharmacologie , Anticorps antiviraux/pharmacologie , Metapneumovirus/physiologie , Muqueuse respiratoire , Infections à virus respiratoire syncytial , Virus respiratoires syncytiaux/physiologie , Réplication virale/effets des médicaments et des substances chimiques , Lignée cellulaire , Humains , Muqueuse respiratoire/métabolisme , Muqueuse respiratoire/virologie , Infections à virus respiratoire syncytial/métabolisme , Infections à virus respiratoire syncytial/transmission
16.
Viruses ; 12(7)2020 07 04.
Article de Anglais | MEDLINE | ID: mdl-32635475

RÉSUMÉ

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.


Sujet(s)
Metapneumovirus/physiologie , Infections à Paramyxoviridae/immunologie , Facteur de transcription STAT-2/immunologie , Animaux , Femelle , Spécificité d'hôte , Humains , Immunité innée , Interférons/génétique , Interférons/immunologie , Mâle , Metapneumovirus/génétique , Souris , Souris de lignée C57BL , Souris knockout , Infections à Paramyxoviridae/génétique , Infections à Paramyxoviridae/virologie , Facteur de transcription STAT-2/génétique , Lymphocytes auxiliaires Th2
17.
Viruses ; 12(6)2020 06 12.
Article de Anglais | MEDLINE | ID: mdl-32545470

RÉSUMÉ

The human respiratory syncytial virus (hRSV) and human Metapneumovirus (hMPV) are two of the leading etiological agents of acute lower respiratory tract infections, which constitute the main cause of mortality in infants. However, there are currently approved vaccines for neither hRSV nor hMPV. Moreover, despite the similarity between the pathology caused by both viruses, the immune response elicited by the host is different in each case. In this review, we discuss how dendritic cells, alveolar macrophages, neutrophils, eosinophils, natural killer cells, innate lymphoid cells, and the complement system regulate both pathogenesis and the resolution of hRSV and hMPV infections. The roles that these cells play during infections by either of these viruses will help us to better understand the illnesses they cause. We also discuss several controversial findings, relative to some of these innate immune components. To better understand the inflammation in the lungs, the role of the respiratory epithelium in the recruitment of innate immune cells is briefly discussed. Finally, we review the main prophylactic strategies and current vaccine candidates against both hRSV and hMPV.


Sujet(s)
Metapneumovirus/physiologie , Infections à Paramyxoviridae/immunologie , Infections à virus respiratoire syncytial/immunologie , Virus respiratoire syncytial humain/physiologie , Animaux , Humains , Immunité innée , Cellules tueuses naturelles/immunologie , Metapneumovirus/génétique , Granulocytes neutrophiles/immunologie , Infections à Paramyxoviridae/génétique , Infections à Paramyxoviridae/virologie , Infections à virus respiratoire syncytial/génétique , Infections à virus respiratoire syncytial/virologie , Virus respiratoire syncytial humain/génétique
18.
Viruses ; 12(5)2020 05 14.
Article de Anglais | MEDLINE | ID: mdl-32423043

RÉSUMÉ

Viruses are the most common cause of acute respiratory tract infections (ARTI). Human metapneumovirus (hMPV) frequently causes viral pneumonia which can become life-threatening if the virus spreads to the lungs. Even though hMPV was only isolated in 2001, this negative-stranded RNA virus has probably been circulating in the human population for many decades. Interestingly, almost all adults have serologic evidence of hMPV infection. A well-established host immune response is evoked when hMPV infection occurs. However, the virus has evolved to circumvent and even exploit the host immune response. Further, infection with hMPV induces a weak memory response, and re-infections during life are common. In this review, we provide a comprehensive overview of the different cell types involved in the immune response in order to better understand the immunopathology induced by hMPV. Such knowledge may contribute to the development of vaccines and therapeutics directed against hMPV.


Sujet(s)
Immunité cellulaire , Metapneumovirus/immunologie , Infections à Paramyxoviridae/immunologie , Infections de l'appareil respiratoire/immunologie , Humains , Échappement immunitaire , Immunité innée , Poumon/immunologie , Poumon/anatomopathologie , Poumon/virologie , Metapneumovirus/pathogénicité , Metapneumovirus/physiologie , Infections à Paramyxoviridae/anatomopathologie , Infections à Paramyxoviridae/virologie , Infections de l'appareil respiratoire/anatomopathologie , Infections de l'appareil respiratoire/virologie , Réplication virale
19.
Virulence ; 11(1): 580-593, 2020 12.
Article de Anglais | MEDLINE | ID: mdl-32463330

RÉSUMÉ

Human respiratory syncytial virus (hRSV) and human metapneumovirus (hMPV) cause acute respiratory tract infections in children worldwide. Natural killer T (NKT) cells are unconventional T lymphocytes, and their TCRs recognize glycolipids bound to the MHC-I-like molecule, CD1d. These cells modulate the inflammatory response in viral infections. Here, we evaluated the contribution of NKT cells in both hRSV and hMPV infections. A significant decrease in the number of neutrophils, eosinophils, and CD103+DCs infiltrating to the lungs, as well as an increased production of IFN-γ, were observed upon hRSV-infection in CD1d-deficient BALB/c mice, as compared to wild-type control mice. However, this effect was not observed in the CD1d-deficient BALB/c group, upon infection with hMPV. Importantly, reduced expression of CD1d in CD11b+ DCs and epithelial cells was found in hRSV -but not hMPV-infected mice. Besides, a reduction in the expression of CD1d in alveolar macrophages of lungs from hRSV- and hMPV-infected mice was found. Such reduction of CD1d expression interfered with NKT cells activation, and consequently IL-2 secretion, as characterized by in vitro experiments for both hRSV and hMPV infections. Furthermore, increased numbers of NKT cells recruited to the lungs in response to hRSV- but not hMPV-infection was detected, resulting in a reduction in the expression of IFN-γ and IL-2 by these cells. In conclusion, both hRSV and hMPV might be differently impairing NKT cells function and contributing to the immune response triggered by these viruses.


Sujet(s)
Cellules T tueuses naturelles/immunologie , Infections à Paramyxoviridae/immunologie , Infections à virus respiratoire syncytial/immunologie , Infections de l'appareil respiratoire/virologie , Réplication virale/immunologie , Animaux , Antigène CD1d/génétique , Antigène CD1d/immunologie , Humains , Poumon/immunologie , Poumon/virologie , Macrophages alvéolaires/immunologie , Macrophages alvéolaires/virologie , Mâle , Metapneumovirus/pathogénicité , Metapneumovirus/physiologie , Souris de lignée BALB C , Souris de lignée C57BL , Cellules T tueuses naturelles/anatomopathologie , Virus respiratoire syncytial humain/pathogénicité , Virus respiratoire syncytial humain/physiologie
20.
Viruses ; 12(4)2020 03 28.
Article de Anglais | MEDLINE | ID: mdl-32231136

RÉSUMÉ

Avian metapneumovirus subtype C (aMPV/C) causes an acute respiratory disease that has caused serious economic losses in the Chinese poultry industry. In the present study, we first explored the protein profile in aMPV/C-infected Vero cells using iTRAQ quantitative proteomics. A total of 921 of 7034 proteins were identified as significantly altered by aMPV/C infection. Three selected proteins were confirmed by Western blot analysis. Bioinformatics GO analysis revealed multiple signaling pathways involving cell cycle, endocytosis, and PI3K-Akt, mTOR, MAPK and p53 signaling pathways, which might participate in viral infection. In this analysis, we found that PLK2 expression was upregulated by aMPV/C infection and investigated whether it contributed to aMPV/C-mediated cellular dysfunction. Suppressing PLK2 attenuated aMPV/C-induced reactive oxygen species (ROS) production and p53-dependent apoptosis and reduced virus release. These results in a mammalian cell line suggest that high PLK2 expression correlates with aMPV/C-induced apoptosis and viral replication, providing new insight into the potential avian host cellular response to aMPV/C infection and antiviral targets.


Sujet(s)
Apoptose , Interactions hôte-pathogène , Metapneumovirus/physiologie , Protein-Serine-Threonine Kinases/métabolisme , Protéome , Animaux , Chlorocebus aethiops , Chromatographie en phase liquide , Biologie informatique/méthodes , Extinction de l'expression des gènes , Spectrométrie de masse , Infections à Paramyxoviridae/métabolisme , Infections à Paramyxoviridae/virologie , Maladies de la volaille/métabolisme , Maladies de la volaille/virologie , Protein-Serine-Threonine Kinases/génétique , Espèces réactives de l'oxygène/métabolisme , Reproductibilité des résultats , Cellules Vero , Libération de particules virales , Réplication virale
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...