Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 83
Filter
1.
J Virol ; 98(4): e0005124, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38466095

ABSTRACT

Avian metapneumovirus subgroup C (aMPV/C), an important pathogen causing acute respiratory infection in chickens and turkeys, contributes to substantial economic losses in the poultry industry worldwide. aMPV/C has been reported to induce autophagy, which is beneficial to virus replication. Sequestosome 1 (SQSTM1/P62), a selective autophagic receptor, plays a crucial role in viral replication by clearing ubiquitinated proteins. However, the relationship between SQSTM1-mediated selective autophagy and aMPV/C replication is unclear. In this study, we found that the expression of SQSTM1 negatively regulates aMPV/C replication by reducing viral protein expression and viral titers. Further studies revealed that the interaction between SQSTM1 and aMPV/C M2-2 protein is mediated via the Phox and Bem1 (PB1) domain of the former, which recognizes a ubiquitinated lysine at position 67 of the M2-2 protein, and finally degrades M2-2 via SQSTM1-mediated selective autophagy. Collectively, our results reveal that SQSTM1 degrades M2-2 via a process of selective autophagy to suppress aMPV/C replication, thereby providing novel insights for the prevention and control of aMPV/C infection.IMPORTANCEThe selective autophagy plays an important role in virus replication. As an emerging pathogen of avian respiratory virus, clarification of the effect of SQSTM1, a selective autophagic receptor, on aMPV/C replication in host cells enables us to better understand the viral pathogenesis. Previous study showed that aMPV/C infection reduced the SQSTM1 expression accompanied by virus proliferation, but the specific regulatory mechanism between them was still unclear. In this study, we demonstrated for the first time that SQSTM1 recognizes the 67th amino acid of M2-2 protein by the interaction between them, followed by M2-2 degradation via the SQSTM1-mediated selective autophagy, and finally inhibits aMPV/C replication. This information supplies the mechanism by which SQSTM1 negatively regulates viral replication, and provides new insights for preventing and controlling aMPV/C infection.


Subject(s)
Autophagy , Birds , Metapneumovirus , Proteolysis , Sequestosome-1 Protein , Viral Proteins , Virus Replication , Animals , Humans , HEK293 Cells , Metapneumovirus/classification , Metapneumovirus/growth & development , Paramyxoviridae Infections/metabolism , Paramyxoviridae Infections/veterinary , Paramyxoviridae Infections/virology , Protein Binding , Sequestosome-1 Protein/chemistry , Sequestosome-1 Protein/metabolism , Vero Cells , Viral Proteins/chemistry , Viral Proteins/metabolism , Birds/virology
2.
Front Immunol ; 13: 970750, 2022.
Article in English | MEDLINE | ID: mdl-36045682

ABSTRACT

Retinoic acid-inducible gene I (RIG-I) is a receptor that senses viral RNA and interacts with mitochondrial antiviral signaling (MAVS) protein, leading to the production of type I interferons and inflammatory cytokines to establish an antiviral state. This signaling axis is initiated by the K63-linked RIG-I ubiquitination, mediated by E3 ubiquitin ligases such as TRIM25. However, many viruses, including several members of the family Paramyxoviridae and human respiratory syncytial virus (HRSV), a member of the family Pneumoviridae, escape the immune system by targeting RIG-I/TRIM25 signaling. In this study, we screened human metapneumovirus (HMPV) open reading frames (ORFs) for their ability to block RIG-I signaling reconstituted in HEK293T cells by transfection with TRIM25 and RIG-I CARD (an N-terminal CARD domain that is constitutively active in RIG-I signaling). HMPV M2-2 was the most potent inhibitor of RIG-I/TRIM25-mediated interferon (IFN)-ß activation. M2-2 silencing induced the activation of transcription factors (IRF and NF-kB) downstream of RIG-I signaling in A549 cells. Moreover, M2-2 inhibited RIG-I ubiquitination and CARD-dependent interactions with MAVS. Immunoprecipitation revealed that M2-2 forms a stable complex with RIG-I CARD/TRIM25 via direct interaction with the SPRY domain of TRIM25. Similarly, HRSV NS1 also formed a stable complex with RIG-I CARD/TRIM25 and inhibited RIG-I ubiquitination. Notably, the inhibitory actions of HMPV M2-2 and HRSV NS1 are similar to those of V proteins of several members of the Paramyxoviridae family. In this study, we have identified a novel mechanism of immune escape by HMPV, similar to that of Pneumoviridae and Paramyxoviridae family members.


Subject(s)
Interferon Type I , Metapneumovirus , Paramyxoviridae Infections/metabolism , Tripartite Motif Proteins/metabolism , Antiviral Agents , DEAD Box Protein 58/metabolism , HEK293 Cells , Humans , Immunity, Innate , Interferon Type I/metabolism , Interferon-beta/metabolism , Paramyxoviridae , Paramyxoviridae Infections/virology , Receptors, Immunologic/metabolism , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
3.
J Clin Invest ; 131(23)2021 12 01.
Article in English | MEDLINE | ID: mdl-34609969

ABSTRACT

The capacity of respiratory viruses to undergo evolution within the respiratory tract raises the possibility of evolution under the selective pressure of the host environment or drug treatment. Long-term infections in immunocompromised hosts are potential drivers of viral evolution and development of infectious variants. We showed that intrahost evolution in chronic human parainfluenza virus 3 (HPIV3) infection in immunocompromised individuals elicited mutations that favored viral entry and persistence, suggesting that similar processes may operate across enveloped respiratory viruses. We profiled longitudinal HPIV3 infections from 2 immunocompromised individuals that persisted for 278 and 98 days. Mutations accrued in the HPIV3 attachment protein hemagglutinin-neuraminidase (HN), including the first in vivo mutation in HN's receptor binding site responsible for activating the viral fusion process. Fixation of this mutation was associated with exposure to a drug that cleaves host-cell sialic acid moieties. Longitudinal adaptation of HN was associated with features that promote viral entry and persistence in cells, including greater avidity for sialic acid and more active fusion activity in vitro, but not with antibody escape. Long-term infection thus led to mutations promoting viral persistence, suggesting that host-directed therapeutics may support the evolution of viruses that alter their biophysical characteristics to persist in the face of these agents in vivo.


Subject(s)
Immunocompromised Host , Lung Diseases/virology , Lung/virology , Parainfluenza Virus 3, Human/metabolism , Paramyxoviridae Infections/virology , Adult , Binding Sites , DNA Mutational Analysis , Female , Gene Frequency , Graft vs Host Disease/drug therapy , HEK293 Cells , Humans , Leukemia, Myeloid, Acute , Mutation , Mycophenolic Acid/administration & dosage , N-Acetylneuraminic Acid/chemistry , Parainfluenza Virus 3, Human/genetics , Paramyxoviridae Infections/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/complications , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/virology , Receptors, Virus/metabolism , Sirolimus/administration & dosage , Viral Fusion Proteins/genetics , Viral Fusion Proteins/metabolism , Virus Internalization , Young Adult
4.
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
5.
J Immunother Cancer ; 9(6)2021 06.
Article in English | MEDLINE | ID: mdl-34172515

ABSTRACT

BACKGROUND: There is intense interest in developing novel oncolytic viruses, which can be used in cancer therapies along with immune cells such as natural killer (NK) cells. We have previously developed a particle-based method for in vitro expansion of highly cytotoxic human NK cells (PM21-NK cells). Here, we have tested the hypothesis that oncolytic parainfluenza virus 5 (P/V virus) can combine with PM21-NK cells for targeted killing of lung cancer cells. METHODS: PM21-NK cells were assayed for killing of P/V virus-infected A549, H1299 and Calu-1 lung cancer cells in two-dimensional (2D) and three-dimensional (3D) cultures using flow cytometry, luminescence and kinetic imaging-based methods. Blocking antibodies were used to evaluate NK cell activating receptors involved in PM21-NK cell killing of infected target cells. Media transfer experiments tested soluble factors that increase PM21-NK cell killing of both P/V virus-infected and uninfected tumor cells. RESULTS: In 2D cultures, PM21-NK cells efficiently killed P/V virus-infected cancer cells compared with non-infected cells, through involvement of the viral glycoprotein and NK cell receptors NKp30, NKp46 and NKG2D. In 3D spheroid cultures, P/V virus infection was restricted to the outer layer of the spheroid. However, PM21-NK cells were able to more efficiently kill both the outer layer of infected cells in the spheroid and progressing further to kill the uninfected interior cells. Media transfer experiments demonstrated that P/V virus infection produced both type I and type III interferons, which decreased cell growth, which contributed to a reduction in the overall number of uninfected tumor cells in conjunction with PM21-NK cells. Across five cancer cell lines, the contribution of P/V virus infection on PM21-NK cell killing of target cells correlated with interferon induction. CONCLUSION: Our data support the potential of combining oncolytic parainfluenza virus with PM21-NK cell adoptive therapy against lung cancer.


Subject(s)
Killer Cells, Natural/metabolism , Lung Neoplasms/virology , Oncolytic Viruses/metabolism , Paramyxoviridae Infections/metabolism , Spheroids, Cellular/metabolism , Humans , Imaging, Three-Dimensional , Interferon Type I , Interferons , Lung Neoplasms/immunology , Signal Transduction , Interferon Lambda
6.
Viruses ; 12(12)2020 12 02.
Article in English | MEDLINE | ID: mdl-33276587

ABSTRACT

The paramyxo- and pneumovirus family includes a wide range of viruses that can cause respiratory and/or systemic infections in humans and animals. The significant disease burden of these viruses is further exacerbated by the limited therapeutics that are currently available. Host cellular proteins that can antagonize or limit virus replication are therefore a promising area of research to identify candidate molecules with the potential for host-targeted therapies. Host proteins known as host cell restriction factors are constitutively expressed and/or induced in response to virus infection and include proteins from interferon-stimulated genes (ISGs). Many ISG proteins have been identified but relatively few have been characterized in detail and most studies have focused on studying their antiviral activities against particular viruses, such as influenza A viruses and human immunodeficiency virus (HIV)-1. This review summarizes current literature regarding host cell restriction factors against paramyxo- and pneumoviruses, on which there is more limited data. Alongside discussion of known restriction factors, this review also considers viral countermeasures in overcoming host restriction, the strengths and limitations in different experimental approaches in studies reported to date, and the challenges in reconciling differences between in vitro and in vivo data. Furthermore, this review provides an outlook regarding the landscape of emerging technologies and tools available to study host cell restriction factors, as well as the suitability of these proteins as targets for broad-spectrum antiviral therapeutics.


Subject(s)
Host-Pathogen Interactions , Paramyxoviridae Infections/virology , Paramyxovirinae/physiology , Pneumovirus Infections/virology , Pneumovirus/physiology , Animals , Biomarkers , Gene Expression Regulation, Viral , Host Specificity , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Immunity, Innate , Paramyxoviridae Infections/genetics , Paramyxoviridae Infections/metabolism , Pneumovirus Infections/genetics , Pneumovirus Infections/metabolism , Viral Tropism , Virus Replication
7.
Viruses ; 12(4)2020 03 28.
Article in English | MEDLINE | ID: mdl-32231136

ABSTRACT

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.


Subject(s)
Apoptosis , Host-Pathogen Interactions , Metapneumovirus/physiology , Protein Serine-Threonine Kinases/metabolism , Proteome , Animals , Chlorocebus aethiops , Chromatography, Liquid , Computational Biology/methods , Gene Silencing , Mass Spectrometry , Paramyxoviridae Infections/metabolism , Paramyxoviridae Infections/virology , Poultry Diseases/metabolism , Poultry Diseases/virology , Protein Serine-Threonine Kinases/genetics , Reactive Oxygen Species/metabolism , Reproducibility of Results , Vero Cells , Virus Release , Virus Replication
8.
J Virol ; 94(6)2020 02 28.
Article in English | MEDLINE | ID: mdl-31896588

ABSTRACT

Influenza viruses are highly infectious and are the leading cause of human respiratory diseases and may trigger severe epidemics and occasional pandemics. Although antiviral drugs against influenza viruses have been developed, there is an urgent need to design new strategies to develop influenza virus inhibitors due to the increasing resistance of viruses toward currently available drugs. In this study, we examined the antiviral activity of natural compounds against the following influenza virus strains: A/WSN/33 (H1N1), A/Udorn/72 (H3N2), and B/Lee/40. Papaverine (a nonnarcotic alkaloid that has been used for the treatment of heart disease, impotency, and psychosis) was found to be an effective inhibitor of multiple strains of influenza virus. Kinetic studies demonstrated that papaverine inhibited influenza virus infection at a late stage in the virus life cycle. An alteration in influenza virus morphology and viral ribonucleoprotein (vRNP) localization was observed as an effect of papaverine treatment. Papaverine is a well-known phosphodiesterase inhibitor and also modifies the mitogen-activated protein kinase (MAPK) pathway by downregulating the phosphorylation of MEK and extracellular signal-regulated kinase (ERK). Thus, the modulation of host cell signaling pathways by papaverine may be associated with the nuclear retention of vRNPs and the reduction of influenza virus titers. Interestingly, papaverine also inhibited paramyxoviruses parainfluenza virus 5 (PIV5), human parainfluenza virus 3 (HPIV3), and respiratory syncytial virus (RSV) infections. We propose that papaverine can be a potential candidate to be used as an antiviral agent against a broad range of influenza viruses and paramyxoviruses.IMPORTANCE Influenza viruses are important human pathogens that are the causative agents of epidemics and pandemics. Despite the availability of an annual vaccine, a large number of cases occur every year globally. Here, we report that papaverine, a vasodilator, shows inhibitory action against various strains of influenza virus as well as the paramyxoviruses PIV5, HPIV3, and RSV. A significant effect of papaverine on the influenza virus morphology was observed. Papaverine treatment of influenza-virus-infected cells resulted in the inhibition of virus at a later time in the virus life cycle through the suppression of nuclear export of vRNP and also interfered with the host cellular cAMP and MEK/ERK cascade pathways. This study explores the use of papaverine as an effective inhibitor of both influenza viruses as well as paramyxoviruses.


Subject(s)
Antiviral Agents/pharmacology , Drug Repositioning , Orthomyxoviridae Infections , Orthomyxoviridae/metabolism , Papaverine/pharmacology , Paramyxoviridae Infections , Paramyxovirinae/metabolism , Animals , Dogs , Drug Evaluation, Preclinical , HEK293 Cells , Humans , MAP Kinase Signaling System/drug effects , Madin Darby Canine Kidney Cells , Orthomyxoviridae Infections/drug therapy , Orthomyxoviridae Infections/metabolism , Orthomyxoviridae Infections/pathology , Paramyxoviridae Infections/metabolism , Paramyxoviridae Infections/pathology
9.
Mediators Inflamm ; 2019: 4964239, 2019.
Article in English | MEDLINE | ID: mdl-31686982

ABSTRACT

Human metapneumovirus (HMPV) may cause severe respiratory disease. The early innate immune response to viruses like HMPV is characterized by induction of antiviral interferons (IFNs) and proinflammatory immune mediators that are essential in shaping adaptive immune responses. Although innate immune responses to HMPV have been comprehensively studied in mice and murine immune cells, there is less information on these responses in human cells, comparing different cell types infected with the same HMPV strain. The aim of this study was to characterize the HMPV-induced mRNA expression of critical innate immune mediators in human primary cells relevant for airway disease. In particular, we determined type I versus type III IFN expression in human epithelial cells and monocyte-derived macrophages (MDMs) and dendritic cells (MDDCs). In epithelial cells, HMPV induced only low levels of IFN-ß mRNA, while a robust mRNA expression of IFN-λs was found in epithelial cells, MDMs, and MDDCs. In addition, we determined induction of the interferon regulatory factors (IRFs) IRF1, IRF3, and IRF7 and critical inflammatory cytokines (IL-6, IP-10, and IL-1ß). Interestingly, IRF1 mRNA was predominantly induced in MDMs and MDDCs. Overall, our results suggest that for HMPV infection of MDDCs, MDMs, NECs, and A549 cells (the cell types examined), cell type is a strong determinator of the ability of HMPV to induce different innate immune mediators. HMPV induces the transcription of IFN-ß and IRF1 to higher extents in MDMs and MDDCs than in A549s and NECs, whereas the induction of type III IFN-λ and IRF7 is considerable in MDMs, MDDCs, and A549 epithelial cells.


Subject(s)
Immunity, Innate/physiology , Metapneumovirus/pathogenicity , Paramyxoviridae Infections/immunology , A549 Cells , Cells, Cultured , Chemokine CXCL10/metabolism , Fluorescent Antibody Technique , Humans , Immunity, Innate/genetics , Interleukin-6/metabolism , Macrophages/metabolism , Metapneumovirus/immunology , Microscopy, Confocal , Paramyxoviridae Infections/metabolism , RNA, Messenger/metabolism , Signal Transduction
10.
Proc Natl Acad Sci U S A ; 116(43): 21514-21520, 2019 10 22.
Article in English | MEDLINE | ID: mdl-31591233

ABSTRACT

The bat-borne paramyxovirus, Sosuga virus (SosV), is one of many paramyxoviruses recently identified and classified within the newly established genus Pararubulavirus, family Paramyxoviridae The envelope surface of SosV presents a receptor-binding protein (RBP), SosV-RBP, which facilitates host-cell attachment and entry. Unlike closely related hemagglutinin neuraminidase RBPs from other genera of the Paramyxoviridae, SosV-RBP and other pararubulavirus RBPs lack many of the stringently conserved residues required for sialic acid recognition and hydrolysis. We determined the crystal structure of the globular head region of SosV-RBP, revealing that while the glycoprotein presents a classical paramyxoviral six-bladed ß-propeller fold and structurally classifies in close proximity to paramyxoviral RBPs with hemagglutinin-neuraminidase (HN) functionality, it presents a receptor-binding face incongruent with sialic acid recognition. Hemadsorption and neuraminidase activity analysis confirms the limited capacity of SosV-RBP to interact with sialic acid in vitro and indicates that SosV-RBP undergoes a nonclassical route of host-cell entry. The close overall structural conservation of SosV-RBP with other classical HN RBPs supports a model by which pararubulaviruses only recently diverged from sialic acid binding functionality.


Subject(s)
HN Protein/chemistry , Paramyxoviridae Infections/virology , Paramyxoviridae/physiology , Viral Proteins/chemistry , Virus Internalization , HN Protein/genetics , HN Protein/metabolism , Humans , N-Acetylneuraminic Acid/metabolism , Paramyxoviridae/chemistry , Paramyxoviridae/genetics , Paramyxoviridae Infections/metabolism , Receptors, Virus/genetics , Receptors, Virus/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Attachment
11.
Adv Med Sci ; 64(2): 338-343, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31022559

ABSTRACT

PURPOSE: Respiratory viral infection and nonsteroidal anti-inflammatory drugs (NSAIDs) may affect arachidonic acid (AA) metabolism in the airway epithelium, however their joint effect has not been studied. We hypothesized, that alternations of AA metabolism in human airway epithelial cells (ECs) - induced by Parainfluenza virus type 3 (PIV3) - may be modified by concomitant treatment with NSAIDs. MATERIALS AND METHODS: Nasal (RPMI 2650) and bronchial (BEAS-2B) epithelial cells were cultured into confluence and then infected with PIV3. Prostaglandin E2 (PGE2) and 15-hydroxyeicosatetraenoic acid (15-HETE) levels in cell supernatants were measured by ELISA and expression of cyclooxygenase-1 (COX-1), cyclooxygenase-2 (COX-2), 5-lipoxygenase (5-LO) and 15-lipoxygenase (15-LO) mRNA in cells was evaluated after reverse transcription with real-time polymerase chain reactions. RESULTS: PGE2 generation was decreased by PIV3 infection in the upper airway epithelial cells, and increased in the lower airway epithelial cells. Both naproxen and celecoxib induced significant reduction in PGE2 release in both infected and non-infected upper and lower airway epithelial cells. However, in PIV3-infected epithelial cells celecoxib inhibited PGE2 release and COX-2 expression to significantly higher degree as compared to non-infected cells. 15-HETE generation or COX-1, 5-LO and 15-LO expression were not affected by the virus infection or by NSAIDs. CONCLUSION: Virus infection in airway epithelial cells enhances inhibitory effect of NSAIDs on prostaglandin E2 generation.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cyclooxygenase 2/metabolism , Dinoprostone/metabolism , Paramyxoviridae Infections/metabolism , Celecoxib/pharmacology , Cell Line , Epithelial Cells , Humans , Real-Time Polymerase Chain Reaction
12.
Virology ; 531: 183-191, 2019 05.
Article in English | MEDLINE | ID: mdl-30927711

ABSTRACT

Human metapneumovirus (hMPV) is an important cause of acute lower respiratory tract infections in infants, elderly and immunocompromised individuals. Ingenuity pathway analysis of microarrays data showed that 20% of genes affected by hMPV infection of airway epithelial cells (AECs) were related to metabolism. We found that levels of the glycolytic pathway enzymes hexokinase 2, pyruvate kinase M2, and lactate dehydrogenase A were significantly upregulated in normal human AECs upon hMPV infection, as well as levels of enzymes belonging to the hexosamine biosynthetic and glycosylation pathways. On the other hand, expression of the majority of the enzymes belonging to the tricarboxylic acid cycle was significantly diminished. Inhibition of hexokinase 2 and of the glycosylating enzyme O-linked N-acetylglucosamine transferase led to a significant reduction in hMPV titer, indicating that metabolic changes induced by hMPV infection play a major role during the virus life cycle, and could be explored as potential antiviral targets.


Subject(s)
Epithelial Cells/metabolism , Metapneumovirus/physiology , Paramyxoviridae Infections/metabolism , Respiratory Mucosa/metabolism , Cell Line , Epithelial Cells/virology , Glycolysis , Hexosamines/biosynthesis , Humans , Metabolic Networks and Pathways , Metapneumovirus/genetics , Oxidative Phosphorylation , Paramyxoviridae Infections/genetics , Paramyxoviridae Infections/physiopathology , Paramyxoviridae Infections/virology , Respiratory Mucosa/virology , Virus Replication
13.
PLoS Pathog ; 15(2): e1007561, 2019 02.
Article in English | MEDLINE | ID: mdl-30742688

ABSTRACT

Paramyxoviruses can establish persistent infections both in vitro and in vivo, some of which lead to chronic disease. However, little is known about the molecular events that contribute to the establishment of persistent infections by RNA viruses. Using parainfluenza virus type 5 (PIV5) as a model we show that phosphorylation of the P protein, which is a key component of the viral RNA polymerase complex, determines whether or not viral transcription and replication becomes repressed at late times after infection. If the virus becomes repressed, persistence is established, but if not, the infected cells die. We found that single amino acid changes at various positions within the P protein switched the infection phenotype from lytic to persistent. Lytic variants replicated to higher titres in mice than persistent variants and caused greater infiltration of immune cells into infected lungs but were cleared more rapidly. We propose that during the acute phases of viral infection in vivo, lytic variants of PIV5 will be selected but, as the adaptive immune response develops, variants in which viral replication can be repressed will be selected, leading to the establishment of prolonged, persistent infections. We suggest that similar selection processes may operate for other RNA viruses.


Subject(s)
Paramyxoviridae Infections/genetics , Paramyxoviridae/genetics , Phosphoproteins/genetics , Viral Proteins/genetics , A549 Cells , Amino Acid Substitution/genetics , Animals , DNA-Directed RNA Polymerases/genetics , DNA-Directed RNA Polymerases/metabolism , Female , HEK293 Cells , Humans , Mice , Mice, Inbred BALB C , Parainfluenza Virus 5/genetics , Parainfluenza Virus 5/pathogenicity , Paramyxoviridae/pathogenicity , Paramyxoviridae Infections/metabolism , Phosphoproteins/metabolism , Phosphoproteins/physiology , Phosphorylation , RNA, Viral , Viral Proteins/metabolism , Viral Proteins/physiology , Virus Replication
14.
Viruses ; 10(9)2018 09 18.
Article in English | MEDLINE | ID: mdl-30231515

ABSTRACT

Human metapneumovirus (HMPV) is one of the leading causes of respiratory diseases in infants and children worldwide. Although this pathogen infects mainly young children, elderly and immunocompromised people can be also seriously affected. To date, there is no commercial vaccine available against it. Upon HMPV infection, the host innate arm of defense produces interferons (IFNs), which are critical for limiting HMPV replication. In this review, we offer an updated landscape of the HMPV mediated-IFN response in different models as well as some of the defense tactics employed by the virus to circumvent IFN response.


Subject(s)
Host-Pathogen Interactions , Interferons/metabolism , Metapneumovirus/physiology , Paramyxoviridae Infections/metabolism , Paramyxoviridae Infections/virology , Animals , Disease Models, Animal , Gene Expression Regulation, Viral , Genome, Viral , Genomics , Humans , Paramyxoviridae Infections/epidemiology , Virus Replication
15.
Cell Immunol ; 331: 161-167, 2018 09.
Article in English | MEDLINE | ID: mdl-30077332

ABSTRACT

Human metapneumovirus (hMPV) is a common cause of respiratory infections in children. However, the precise mechanisms underlying the development of hMPV-induced pulmonary pathology remain unknown. Studies show that IL-17 plays an important role in some inflammatory diseases of the airways, including asthma and chronic obstructive pulmonary disease. Here, we generated an IL-17 KO murine model of hMPV infection and used it to characterize the role of IL-17 hMPV-induced pulmonary inflammation. The results demonstrated that the defect in IL-17 resulted in less neutrophil influx into the lungs, along with reduced ventilatory function. Meanwhile, viral infection in IL-17 KO mice increased regulatory T cells (Tregs) and reduced Th1 and Th2 cells in the lung, suggesting that lack of IL-17 skews the immune response in the lung toward an anti-inflammatory profile, as exhibited by a greater number of Treg cells and fewer Th1 and Th2 cells.


Subject(s)
Interleukin-17/immunology , Metapneumovirus/immunology , Paramyxoviridae Infections/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Cytokines/immunology , Cytokines/metabolism , Humans , Interleukin-17/genetics , Interleukin-17/metabolism , Lung/immunology , Lung/metabolism , Lung/virology , Metapneumovirus/physiology , Mice, Inbred C57BL , Mice, Knockout , Paramyxoviridae Infections/metabolism , Paramyxoviridae Infections/virology , Pneumonia/immunology , Pneumonia/metabolism , Pneumonia/virology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/virology , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/virology , Th1 Cells/immunology , Th1 Cells/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism , Virus Replication/immunology
16.
BMC Res Notes ; 11(1): 432, 2018 Jul 03.
Article in English | MEDLINE | ID: mdl-29970194

ABSTRACT

OBJECTIVE: Human metapneumovirus (HMPV) and respiratory syncytial virus (RSV) are responsible for respiratory diseases, mostly in children. Despite the clinical and epidemiological similarities between these two pneumoviruses, they elicit different immune responses. This work aims to further our understanding of the differential immune response induced by these respiratory viruses by determining the changes of small non-coding RNAs (miRNAs), which regulate gene expression and are involved in numerous cellular processes including the immune system. RESULTS: In the present study, we analyzed the expression of miRNA transcripts of human dendritic cells infected with RSV or HMPV by high throughput sequencing using Illumina sequencing technology. Further validation of miRNA expression by quantitative polymerase chain reaction indicated that HMPV infection up-regulated the expression of 2 miRNAs (hsa-miR-182-5p and hsa-miR-4634), while RSV infection induced significant expression of 3 miRNAs (hsa-miR-4448, hsa-miR-30a-5p and hsa-miR-4634). The predominant miRNA induced by both viruses was hsa-miR-4634.


Subject(s)
Dendritic Cells , Metapneumovirus/metabolism , MicroRNAs/metabolism , Paramyxoviridae Infections/metabolism , Respiratory Syncytial Virus Infections/metabolism , Child , Humans , Respiratory Syncytial Virus, Human
17.
J Virol ; 92(6)2018 03 15.
Article in English | MEDLINE | ID: mdl-29321315

ABSTRACT

Paramyxovirus V proteins are known antagonists of the RIG-I-like receptor (RLR)-mediated interferon induction pathway, interacting with and inhibiting the RLR MDA5. We report interactions between the Nipah virus V protein and both RIG-I regulatory protein TRIM25 and RIG-I. We also observed interactions between these host proteins and the V proteins of measles virus, Sendai virus, and parainfluenza virus. These interactions are mediated by the conserved C-terminal domain of the V protein, which binds to the tandem caspase activation and recruitment domains (CARDs) of RIG-I (the region of TRIM25 ubiquitination) and to the SPRY domain of TRIM25, which mediates TRIM25 interaction with the RIG-I CARDs. Furthermore, we show that V interaction with TRIM25 and RIG-I prevents TRIM25-mediated ubiquitination of RIG-I and disrupts downstream RIG-I signaling to the mitochondrial antiviral signaling protein. This is a novel mechanism for innate immune inhibition by paramyxovirus V proteins, distinct from other known V protein functions such as MDA5 and STAT1 antagonism.IMPORTANCE The host RIG-I signaling pathway is a key early obstacle to paramyxovirus infection, as it results in rapid induction of an antiviral response. This study shows that paramyxovirus V proteins interact with and inhibit the activation of RIG-I, thereby interrupting the antiviral signaling pathway and facilitating virus replication.


Subject(s)
DEAD Box Protein 58/metabolism , Paramyxoviridae Infections/metabolism , Paramyxoviridae/physiology , Signal Transduction , Transcription Factors/metabolism , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Viral Proteins/metabolism , Virus Replication , A549 Cells , Animals , DEAD Box Protein 58/genetics , Dogs , HeLa Cells , Humans , Madin Darby Canine Kidney Cells , Multienzyme Complexes/genetics , Multienzyme Complexes/metabolism , Paramyxoviridae Infections/genetics , Receptors, Immunologic , Transcription Factors/genetics , Tripartite Motif Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitination/genetics , Viral Proteins/genetics
18.
Virology ; 501: 127-135, 2017 01 15.
Article in English | MEDLINE | ID: mdl-27915128

ABSTRACT

Many enveloped RNA viruses utilize lipid rafts for the assembly of progeny virions, but the role of cholesterol, a major component of rafts, on paramyxovirus budding and virion formation is controversial. In this study, we analyzed the effects of FDA-approved cholesterol-reducing agents, gemfibrozil and lovastatin, on raft formation and assembly of human parainfluenza virus type 1 (hPIV1) and Sendai virus (SeV). Treatment of the human airway epithelial A549 cells with the agents, especially when combined, significantly decreased production of infectious hPIV1 and SeV. Mechanistic analysis indicated that depletion of cellular cholesterol reduced cell surface accumulation of envelope glycoproteins and association of viral matrix and nucleocapsids with raft membrane, which resulted in impaired virus budding and release from the cells. These results indicate that cellular cholesterol is required for assembly and formation of type 1 parainfluenza viruses and suggest that cholesterol could be an attractive target for antiviral agents against hPIV1.


Subject(s)
Anticholesteremic Agents/pharmacology , Cholesterol/metabolism , Parainfluenza Virus 1, Human/drug effects , Paramyxoviridae Infections/virology , Virus Assembly/drug effects , Cell Membrane/metabolism , Cell Membrane/virology , Humans , Parainfluenza Virus 1, Human/genetics , Parainfluenza Virus 1, Human/physiology , Paramyxoviridae Infections/drug therapy , Paramyxoviridae Infections/metabolism , Protein Transport , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Release/drug effects
19.
Oncotarget ; 7(41): 66468-66479, 2016 Oct 11.
Article in English | MEDLINE | ID: mdl-27634893

ABSTRACT

The innate sensing system is equipped with PRRs specialized in recognizing molecular structures (PAMPs) of various pathogens. This leads to the induction of anti-viral genes and inhibition of virus growth. Human Metapneumovirus (HMPV) is a major respiratory virus that causes an upper and lower respiratory tract infection in children. In this study we show that upon HMPV infection, the innate sensing system detects the viral RNA through the RIG-I sensor leading to induction of CEACAM1 expression. We further show that CEACAM1 is induced via binding of IRF3 to the CEACAM1 promoter. We demonstrate that induction of CEACAM1 suppresses the viral loads via inhibition of the translation machinery in the infected cells in an SHP2-dependent manner. In summary, we show here that HMPV-infected cells upregulates CEACAM1 to restrict HMPV infection.


Subject(s)
Antigens, CD/immunology , Cell Adhesion Molecules/immunology , Immunity, Innate/immunology , Paramyxoviridae Infections/immunology , Animals , Antigens, CD/biosynthesis , Cell Adhesion Molecules/biosynthesis , Chlorocebus aethiops , Humans , Metapneumovirus/immunology , Paramyxoviridae Infections/metabolism , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism , Respiratory Mucosa/virology , Respiratory Tract Infections/immunology , Respiratory Tract Infections/metabolism , Respiratory Tract Infections/virology , Up-Regulation , Vero Cells
20.
PLoS Pathog ; 11(12): e1005303, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26629703

ABSTRACT

Human metapneumovirus (HMPV), a member of the Paramyxoviridae family, is a leading cause of lower respiratory illness. Although receptor binding is thought to initiate fusion at the plasma membrane for paramyxoviruses, the entry mechanism for HMPV is largely uncharacterized. Here we sought to determine whether HMPV initiates fusion at the plasma membrane or following internalization. To study the HMPV entry process in human bronchial epithelial (BEAS-2B) cells, we used fluorescence microscopy, an R18-dequenching fusion assay, and developed a quantitative, fluorescence microscopy assay to follow virus binding, internalization, membrane fusion, and visualize the cellular site of HMPV fusion. We found that HMPV particles are internalized into human bronchial epithelial cells before fusing with endosomes. Using chemical inhibitors and RNA interference, we determined that HMPV particles are internalized via clathrin-mediated endocytosis in a dynamin-dependent manner. HMPV fusion and productive infection are promoted by RGD-binding integrin engagement, internalization, actin polymerization, and dynamin. Further, HMPV fusion is pH-independent, although infection with rare strains is modestly inhibited by RNA interference or chemical inhibition of endosomal acidification. Thus, HMPV can enter via endocytosis, but the viral fusion machinery is not triggered by low pH. Together, our results indicate that HMPV is capable of entering host cells by multiple pathways, including membrane fusion from endosomal compartments.


Subject(s)
Metapneumovirus/physiology , Paramyxoviridae Infections/metabolism , Respiratory Mucosa/virology , Virus Internalization , Bronchi/virology , Cell Line , Endosomes/metabolism , Flow Cytometry , Humans , Microscopy, Confocal , RNA, Small Interfering , Transfection , Viral Fusion Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...