Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Viruses ; 16(7)2024 Jul 06.
Article in English | MEDLINE | ID: mdl-39066249

ABSTRACT

Human metapneumovirus (HMPV) is an important cause of acute respiratory tract infection and causes significant morbidity and mortality. There is no specific antiviral drug to treat HMPV or vaccine to prevent HMPV. This study determined if probenecid, a host-targeting antiviral drug, had prophylactic (pre-virus) or therapeutic (post-virus) efficacy to inhibit HMPV replication in LLC-MK2 cells in vitro and in the lungs of BALB/c mice. This study showed that ≥0.5 µM probenecid significantly inhibited HMPV replication in vitro, and 2-200 mg/kg probenecid prophylaxis or treatment reduced HMPV replication in BALB/c mice.


Subject(s)
Antiviral Agents , Metapneumovirus , Mice, Inbred BALB C , Paramyxoviridae Infections , Probenecid , Virus Replication , Animals , Metapneumovirus/drug effects , Metapneumovirus/physiology , Virus Replication/drug effects , Mice , Probenecid/pharmacology , Paramyxoviridae Infections/drug therapy , Paramyxoviridae Infections/virology , Antiviral Agents/pharmacology , Cell Line , Lung/virology , Humans , Respiratory Tract Infections/virology , Respiratory Tract Infections/drug therapy , Female
2.
Viruses ; 13(12)2021 12 06.
Article in English | MEDLINE | ID: mdl-34960719

ABSTRACT

Pneumoviruses include pathogenic human and animal viruses, the most known and studied being the human respiratory syncytial virus (hRSV) and the metapneumovirus (hMPV), which are the major cause of severe acute respiratory tract illness in young children worldwide, and main pathogens infecting elderly and immune-compromised people. The transcription and replication of these viruses take place in specific cytoplasmic inclusions called inclusion bodies (IBs). These activities depend on viral polymerase L, associated with its cofactor phosphoprotein P, for the recognition of the viral RNA genome encapsidated by the nucleoprotein N, forming the nucleocapsid (NC). The polymerase activities rely on diverse transient protein-protein interactions orchestrated by P playing the hub role. Among these interactions, P interacts with the NC to recruit L to the genome. The P protein also plays the role of chaperone to maintain the neosynthesized N monomeric and RNA-free (called N0) before specific encapsidation of the viral genome and antigenome. This review aims at giving an overview of recent structural information obtained for hRSV and hMPV P, N, and more specifically for P-NC and N0-P complexes that pave the way for the rational design of new antivirals against those viruses.


Subject(s)
Antiviral Agents , Drug Design , Metapneumovirus/metabolism , Nucleocapsid Proteins/metabolism , Phosphoproteins/metabolism , Respiratory Syncytial Virus, Human/metabolism , Viral Proteins/metabolism , Animals , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Humans , Metapneumovirus/drug effects , Metapneumovirus/genetics , Models, Molecular , Nucleocapsid Proteins/chemistry , Paramyxoviridae Infections/drug therapy , Paramyxoviridae Infections/virology , Phosphoproteins/chemistry , Protein Binding , Protein Conformation , RNA, Viral/chemistry , RNA, Viral/metabolism , Respiratory Syncytial Virus Infections/drug therapy , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus, Human/drug effects , Respiratory Syncytial Virus, Human/genetics , Transcription, Genetic , Viral Proteins/chemistry , Virus Replication
3.
Viruses ; 13(1)2021 Jan 19.
Article in English | MEDLINE | ID: mdl-33478119

ABSTRACT

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.


Subject(s)
Cell Culture Techniques , Coinfection , Host-Pathogen Interactions , Metapneumovirus/physiology , Microbial Interactions , Respiratory Syncytial Virus, Human/physiology , Virus Replication , Cell Line , Humans , Interferon Type I/pharmacology , Interferons/pharmacology , Metapneumovirus/drug effects , Paramyxoviridae Infections/virology , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus, Human/drug effects , Spheroids, Cellular , Interferon Lambda
4.
Virology ; 543: 43-53, 2020 04.
Article in English | MEDLINE | ID: mdl-32056846

ABSTRACT

Viruses possessing class I fusion proteins require proteolytic activation by host cell proteases to mediate fusion with the host cell membrane. The mammalian SPINT2 gene encodes a protease inhibitor that targets trypsin-like serine proteases. Here we show the protease inhibitor, SPINT2, restricts cleavage-activation efficiently for a range of influenza viruses and for human metapneumovirus (HMPV). SPINT2 treatment resulted in the cleavage and fusion inhibition of full-length influenza A/CA/04/09 (H1N1) HA, A/Aichi/68 (H3N2) HA, A/Shanghai/2/2013 (H7N9) HA and HMPV F when activated by trypsin, recombinant matriptase or KLK5. We also demonstrate that SPINT2 was able to reduce viral growth of influenza A/CA/04/09 H1N1 and A/X31 H3N2 in cell culture by inhibiting matriptase or TMPRSS2. Moreover, inhibition efficacy did not differ whether SPINT2 was added at the time of infection or 24 h post-infection. Our data suggest that the SPINT2 inhibitor has a strong potential to serve as a novel broad-spectrum antiviral.


Subject(s)
Influenza A virus/drug effects , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/pharmacology , Metapneumovirus/drug effects , Serine Proteinase Inhibitors/pharmacology , Viral Fusion Proteins/metabolism , Animals , Cell Line , Cell Survival/drug effects , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Host-Pathogen Interactions , Humans , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H1N1 Subtype/growth & development , Influenza A Virus, H1N1 Subtype/metabolism , Influenza A Virus, H1N1 Subtype/physiology , Influenza A Virus, H3N2 Subtype/drug effects , Influenza A Virus, H3N2 Subtype/growth & development , Influenza A Virus, H3N2 Subtype/metabolism , Influenza A Virus, H3N2 Subtype/physiology , Influenza A Virus, H7N9 Subtype/drug effects , Influenza A Virus, H7N9 Subtype/growth & development , Influenza A Virus, H7N9 Subtype/metabolism , Influenza A Virus, H7N9 Subtype/physiology , Influenza A virus/growth & development , Influenza A virus/metabolism , Influenza A virus/physiology , Membrane Glycoproteins/genetics , Metapneumovirus/growth & development , Metapneumovirus/metabolism , Metapneumovirus/physiology , Peptide Hydrolases/metabolism , Protease Inhibitors/pharmacology , Recombinant Proteins/pharmacology , Serine Endopeptidases/metabolism , Serine Proteinase Inhibitors/metabolism , Trypsin Inhibitors/metabolism , Trypsin Inhibitors/pharmacology
5.
J Microbiol Biotechnol ; 29(12): 2006-2013, 2019 Dec 28.
Article in English | MEDLINE | ID: mdl-31581384

ABSTRACT

The isolation of respiratory viruses, especially from clinical specimens, often shows poor efficiency with classical cell culture methods. The lack of suitable methods to generate virus particles inhibits the development of diagnostic assays, treatments, and vaccines. We compared three inoculation methods, classical cell culture, the addition of a JAK2 inhibitor AZD1480, and centrifugation-enhanced inoculation (CEI), to replicate human respiratory syncytial virus (HRSV) and human metapneumovirus (HMPV). In addition, a combined method using AZD1480 treatment and CEI was used on throat swabs to verify that this method could increase virus isolation efficiency from human clinical specimens. Both CEI and AZD1480 treatment increased HRSV and HMPV genome replication. Also, the combined method using CEI and AZD1480 treatment enhanced virus proliferation synergistically. The combined method is particularly suited for the isolation of interferon-sensitive or slowly growing viruses from human clinical specimens.


Subject(s)
Centrifugation/methods , Pneumovirus/isolation & purification , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Virus Cultivation/methods , Humans , Metapneumovirus/drug effects , Metapneumovirus/genetics , Metapneumovirus/growth & development , Metapneumovirus/isolation & purification , Pneumovirus/drug effects , Pneumovirus/growth & development , Respiratory Syncytial Virus, Human/drug effects , Respiratory Syncytial Virus, Human/genetics , Respiratory Syncytial Virus, Human/growth & development , Respiratory Syncytial Virus, Human/isolation & purification , Specimen Handling , Virus Replication
6.
PLoS One ; 14(3): e0214646, 2019.
Article in English | MEDLINE | ID: mdl-30921418

ABSTRACT

Human metapneumovirus (hMPV) is a widely distributed pathogen responsible for acute upper and lower respiratory infections of varying severity. Previously, we reported that N-sulfonated derivatives of poly(allylamine hydrochloride) (NSPAHs) efficiently inhibit replication of the influenza virus in vitro and ex vivo. Here, we show a dose dependent inhibition of hMPV infection by NSPAHs in LLC-MK2 cells. The results showed strong antiviral properties of NSPAHs. While the activity of NSPAHs is comparable to those of carrageenans, they show better physicochemical properties and may be delivered at high concentrations. The functional assays showed that tested polymers block hMPV release from infected cells and, consequently, constrain virus spread. Moreover, further studies on viruses utilizing different egress mechanisms suggest that observed antiviral effect depend on selective inhibition of viruses budding from the cell surface.


Subject(s)
Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Metapneumovirus/drug effects , Polyamines/chemistry , Polyamines/pharmacology , Sulfonic Acids/chemistry , Animals , Antiviral Agents/chemical synthesis , Cell Line , Humans , Metapneumovirus/physiology , Polyamines/chemical synthesis , Virion/drug effects , Virion/physiology , Virus Internalization/drug effects , Virus Replication/drug effects
7.
Antivir Chem Chemother ; 27: 2040206619830197, 2019.
Article in English | MEDLINE | ID: mdl-30759993

ABSTRACT

Human metapneumovirus, a paramyxovirus discovered in 2001, is a major cause of lower respiratory infection in adults and children worldwide. There are no licensed vaccines or drugs for human metapneumovirus. We developed a fluorescent, cell-based medium-throughput screening assay for human metapneumovirus that captures inhibitors of all stages of the viral lifecycle except budding of progeny virus particles from the cell membrane. We optimized and validated the assay and performed a successful medium-throughput screening. A number of hits were identified, several of which were confirmed to inhibit viral replication in secondary assays. This assay offers potential to discover new antivirals for human metapneumovirus and related respiratory viruses. Compounds discovered using the medium-throughput screening may also provide useful probes of viral biology.


Subject(s)
Antiviral Agents/pharmacology , Drug Discovery , Drug Evaluation, Preclinical/methods , Metapneumovirus/drug effects , Animals , Antiviral Agents/isolation & purification , Cell Line , Humans , Metapneumovirus/pathogenicity , Metapneumovirus/physiology , Microbial Sensitivity Tests , Respiratory Tract Infections/microbiology , Serial Passage , Virus Replication/drug effects
8.
J Gen Virol ; 99(10): 1367-1380, 2018 10.
Article in English | MEDLINE | ID: mdl-30102144

ABSTRACT

Thrombin has been demonstrated to be involved in several viral diseases including human metapneumovirus (hMPV) infections. We previously showed that immediate administration of thrombin inhibitor argatroban post-infection protected mice against hMPV disease. This current work aims at determining whether warfarin and heparin, two other anticoagulants inhibiting thrombin formation and activities, may also be used for treatment against hMPV in vivo. We found that immediate injections of argatroban, warfarin or heparin after virus challenge protected mice against hMPV infection, as evidenced by decreased or no mortality, less weight loss, reduced viral load and attenuated inflammation. However, delayed treatments starting 1 day post-infection with argatroban or warfarin almost did not impact the survival whereas delayed treatment with heparin induced an increased mortality during infection. Moreover, these treatments also did not reduce weight loss, viral replication and inflammation. In agreement with these results, thrombin generation was decreased upon immediate anticoagulant treatments but was unaltered upon delayed treatments. Thus, thrombin generation occurs at the onset of hMPV infection and thrombin inhibition may be only useful for the treatment of this disease when initiated in the early stage. In this case, heparin is not recommended because of its reduced efficacy on mortality in infected mice whereas argatroban and warfarin appear as safe and effective drugs for the treatment of hMPV disease. The antiviral and anti-inflammatory effects of argatroban occur via thrombin-dependent pathways whereas the mechanisms by which warfarin exerts its beneficial effects against hMPV infection were not elucidated and need to be further studied.


Subject(s)
Anticoagulants/administration & dosage , Heparin/administration & dosage , Paramyxoviridae Infections/drug therapy , Warfarin/administration & dosage , Animals , Arginine/analogs & derivatives , Disease Models, Animal , Metapneumovirus/drug effects , Metapneumovirus/isolation & purification , Mice , Paramyxoviridae Infections/pathology , Paramyxoviridae Infections/virology , Pipecolic Acids/administration & dosage , Sulfonamides , Survival Analysis , Treatment Outcome , Viral Load , Virus Replication/drug effects
9.
EMBO Mol Med ; 10(2): 175-187, 2018 02.
Article in English | MEDLINE | ID: mdl-29217660

ABSTRACT

Human respiratory syncytial virus (hRSV) and human metapneumovirus (hMPV), two members of the Pneumoviridae family, account for the majority of severe lower respiratory tract infections worldwide in very young children. They are also a frequent cause of morbidity and mortality in the elderly and immunocompromised adults. High levels of neutralizing antibodies, mostly directed against the viral fusion (F) glycoprotein, correlate with protection against either hRSV or hMPV However, no cross-neutralization is observed in polyclonal antibody responses raised after virus infection or immunization with purified F proteins. Based on crystal structures of hRSV F and hMPV F, we designed chimeric F proteins in which certain residues of well-characterized antigenic sites were swapped between the two antigens. The antigenic changes were monitored by ELISA with virus-specific monoclonal antibodies. Inoculation of mice with these chimeras induced polyclonal cross-neutralizing antibody responses, and mice were protected against challenge with the virus used for grafting of the heterologous antigenic site. These results provide a proof of principle for chimeric fusion proteins as single immunogens that can induce cross-neutralizing antibody and protective responses against more than one human pneumovirus.


Subject(s)
Antibodies, Neutralizing/immunology , Metapneumovirus , Paramyxoviridae Infections , Recombinant Fusion Proteins/immunology , Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus, Human , Viral Fusion Proteins/immunology , Animals , Humans , Immunization , Metapneumovirus/drug effects , Metapneumovirus/immunology , Mice , Models, Animal , Paramyxoviridae Infections/drug therapy , Paramyxoviridae Infections/immunology , Recombinant Fusion Proteins/pharmacology , Respiratory Syncytial Virus Infections/drug therapy , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus, Human/drug effects , Respiratory Syncytial Virus, Human/immunology , Vaccines, Synthetic , Viral Fusion Proteins/pharmacology , Viral Vaccines
10.
Br J Pharmacol ; 175(2): 388-403, 2018 01.
Article in English | MEDLINE | ID: mdl-29105740

ABSTRACT

BACKGROUND AND PURPOSE: Protease-activated receptor 1 (PAR1) has been demonstrated to be involved in the pathogenesis of viral diseases. However, its role remains controversial. The goal of our study was to investigate the contribution of PAR1 to respiratory syncytial virus (RSV) and human metapneumovirus (hMPV) infections. EXPERIMENTAL APPROACH: Pharmacological approaches were used to investigate the role of PAR1 during RSV and hMPV infection, in vitro using epithelial A549 cells and in vivo using a mouse model of virus infection. KEY RESULTS: In vitro, the PAR1 antagonist RWJ-56110 reduced the replication of RSV and hMPV in A549 cells. In agreement with these results, RWJ-56110-treated mice were protected against RSV and hMPV infections, as indicated by less weight loss and mortality. This protective effect in mice correlated with decreased lung viral replication and inflammation. In contrast, hMPV-infected mice treated with the PAR1 agonist TFLLR-NH2 showed increased mortality, as compared to infected mice, which were left untreated. Thrombin generation was shown to occur downstream of PAR1 activation in infected mice via tissue factor exposure as part of the inflammatory response, and thrombin inhibition by argatroban reduced the pathogenicity of the infection with no additive effect to that induced by PAR1 inhibition. CONCLUSION AND IMPLICATIONS: These data show that PAR1 plays a detrimental role during RSV and hMPV infections in mice via, at least, a thrombin-dependent mechanism. Thus, the use of PAR1 antagonists and thrombin inhibitors may have potential as a novel approach for the treatment of RSV and hMPV infections.


Subject(s)
Indazoles/pharmacology , Paramyxoviridae Infections/virology , Receptor, PAR-1/antagonists & inhibitors , Respiratory Syncytial Virus, Human/drug effects , Thrombin/pharmacology , Urea/analogs & derivatives , Virus Replication/drug effects , Animals , Arginine/analogs & derivatives , Cells, Cultured , Female , Humans , Metapneumovirus/drug effects , Mice , Oligopeptides/pharmacology , Paramyxoviridae Infections/mortality , Pipecolic Acids/pharmacology , Receptor, PAR-1/agonists , Sulfonamides , Urea/pharmacology , Weight Loss/drug effects
11.
Expert Rev Anti Infect Ther ; 15(6): 545-568, 2017 06.
Article in English | MEDLINE | ID: mdl-28277820

ABSTRACT

INTRODUCTION: Influenza-Like Illness is a leading cause of hospitalization in children. Disease burden due to influenza and other respiratory viral infections is reported on a population level, but clinical scores measuring individual changes in disease severity are urgently needed. Areas covered: We present a composite clinical score allowing individual patient data analyses of disease severity based on systematic literature review and WHO-criteria for uncomplicated and complicated disease. The 22-item ViVI Disease Severity Score showed a normal distribution in a pediatric cohort of 6073 children aged 0-18 years (mean age 3.13; S.D. 3.89; range: 0 to 18.79). Expert commentary: The ViVI Score was correlated with risk of antibiotic use as well as need for hospitalization and intensive care. The ViVI Score was used to track children with influenza, respiratory syncytial virus, human metapneumovirus, human rhinovirus, and adenovirus infections and is fully compliant with regulatory data standards. The ViVI Disease Severity Score mobile application allows physicians to measure disease severity at the point-of care thereby taking clinical trials to the next level.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Mobile Applications/statistics & numerical data , Respiratory Tract Infections/diagnosis , Respiratory Tract Infections/drug therapy , Adenoviridae/drug effects , Adenoviridae/growth & development , Adenoviridae/pathogenicity , Adolescent , Child , Child, Preschool , Clinical Trials as Topic , Coinfection , Female , Humans , Infant , Influenza A virus/drug effects , Influenza A virus/growth & development , Influenza A virus/pathogenicity , Influenza B virus/drug effects , Influenza B virus/growth & development , Influenza B virus/pathogenicity , Male , Metapneumovirus/drug effects , Metapneumovirus/growth & development , Metapneumovirus/pathogenicity , Respiratory Syncytial Virus, Human/drug effects , Respiratory Syncytial Virus, Human/growth & development , Respiratory Syncytial Virus, Human/pathogenicity , Respiratory Tract Infections/pathology , Respiratory Tract Infections/virology , Rhinovirus/drug effects , Rhinovirus/growth & development , Rhinovirus/pathogenicity , Severity of Illness Index
12.
Med J Malaysia ; 72(6): 372-373, 2017 12.
Article in English | MEDLINE | ID: mdl-29308778

ABSTRACT

We report a rare case of adult human metapneumovirus (HMPV) in a healthy 32-year-old man. There was dramatic deterioration in his condition developing pneumonia with Type-I respiratory failure and encephalitis. He needed mechanical ventilation in the intensive care setting and was treated with intravenous ribavirin. Post-extubation he remained severely physically and cognitively impaired despite rehabilitation. Treatment of HMPV pneumonia is at present, still without specific antiviral therapy. Managing HMPV-encephalitis remained supportive and challenging. More definite treatment strategies are needed.


Subject(s)
Encephalitis/drug therapy , Metapneumovirus/drug effects , Paramyxoviridae Infections/drug therapy , Adult , Encephalitis/diagnostic imaging , Encephalitis/etiology , Humans , Male , Paramyxoviridae Infections/complications , Paramyxoviridae Infections/rehabilitation , Respiratory Insufficiency/etiology , Treatment Outcome
13.
J Virol ; 90(20): 9237-50, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27489270

ABSTRACT

UNLABELLED: Human metapneumovirus (HMPV), a recently discovered paramyxovirus, infects nearly 100% of the world population and causes severe respiratory disease in infants, the elderly, and immunocompromised patients. We previously showed that HMPV binds heparan sulfate proteoglycans (HSPGs) and that HMPV binding requires only the viral fusion (F) protein. To characterize the features of this interaction critical for HMPV binding and the role of this interaction in infection in relevant models, we utilized sulfated polysaccharides, heparan sulfate mimetics, and occluding compounds. Iota-carrageenan demonstrated potent anti-HMPV activity by inhibiting binding to lung cells mediated by the F protein. Furthermore, analysis of a minilibrary of variably sulfated derivatives of Escherichia coli K5 polysaccharide mimicking the HS structure revealed that the highly O-sulfated K5 polysaccharides inhibited HMPV infection, identifying a potential feature of HS critical for HMPV binding. The peptide dendrimer SB105-A10, which binds HS, reduced binding and infection in an F-dependent manner, suggesting that occlusion of HS at the target cell surface is sufficient to prevent infection. HMPV infection was also inhibited by these compounds during apical infection of polarized airway tissues, suggesting that these interactions take place during HMPV infection in a physiologically relevant model. These results reveal key features of the interaction between HMPV and HS, supporting the hypothesis that apical HS in the airway serves as a binding factor during infection, and HS modulating compounds may serve as a platform for potential antiviral development. IMPORTANCE: Human metapneumovirus (HMPV) is a paramyxovirus that causes respiratory disease worldwide. It has been previously shown that HMPV requires binding to heparan sulfate on the surfaces of target cells for attachment and infection. In this study, we characterize the key features of this binding interaction using heparan sulfate mimetics, identify an important sulfate modification, and demonstrate that these interactions occur at the apical surface of polarized airway tissues. These findings provide insights into the initial binding step of HMPV infection that has potential for antiviral development.


Subject(s)
Antiviral Agents/pharmacology , Heparitin Sulfate/metabolism , Metapneumovirus/drug effects , Paramyxoviridae Infections/drug therapy , Respiratory System/metabolism , Respiratory System/virology , A549 Cells , Bacterial Capsules/metabolism , Cell Line , Cell Line, Tumor , Dendrimers/metabolism , Dendrimers/pharmacology , Escherichia coli/metabolism , Heparan Sulfate Proteoglycans/metabolism , Humans , Peptides/pharmacology , Viral Fusion Proteins/metabolism
14.
Cancer Lett ; 379(1): 100-6, 2016 08 28.
Article in English | MEDLINE | ID: mdl-27260872

ABSTRACT

Over the past decade, reported incidence of human metapneumovirus (hMPV) has increased owing to the use of molecular assays for diagnosis of respiratory viral infections in cancer patients. The seasonality of these infections, differences in sampling strategies across institutions, and small sample size of published studies make it difficult to appreciate the true incidence and impact of hMPV infections. In this systematic review, we summarized the published data on hMPV infections in hematopoietic cell transplant recipients and patients with hematologic malignancy, focusing on incidence, hMPV-associated lower respiratory tract infection (LRTI), mortality, prevention, and management with ribavirin and/or intravenous immunoglobulins. Although the incidence of hMPV infections and hMPV-associated LRTI in this patient population is similar to respiratory syncytial virus or parainfluenza virus and despite lack of directed antiviral therapy, the mortality rate remains low unless patients develop LRTI. In the absence of vaccine to prevent hMPV, infection control measures are recommended to reduce its burden in cancer patients.


Subject(s)
Hematologic Neoplasms/virology , Hematopoietic Stem Cell Transplantation/adverse effects , Metapneumovirus/pathogenicity , Paramyxoviridae Infections/virology , Respiratory Tract Infections/virology , Antiviral Agents/therapeutic use , Hematologic Neoplasms/diagnosis , Hematologic Neoplasms/mortality , Hematopoietic Stem Cell Transplantation/mortality , Humans , Immunoglobulins, Intravenous/therapeutic use , Incidence , Metapneumovirus/drug effects , Paramyxoviridae Infections/diagnosis , Paramyxoviridae Infections/drug therapy , Paramyxoviridae Infections/mortality , Predictive Value of Tests , Prognosis , Respiratory Tract Infections/diagnosis , Respiratory Tract Infections/drug therapy , Respiratory Tract Infections/mortality , Risk Factors
15.
Virus Res ; 220: 129-35, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27130629

ABSTRACT

In this study, we evaluated the immune responses of avian metapneumovirus harboring chicken Fc molecule. Stable Vero cells expressing chicken Fc chimera on its surface (Vero-cFc) were established, and we confirmed that aMPV grown in Vero-cFc incorporated host derived chimera Fc into the aMPV virions. Immunization of chicken with aMPV-cFc induced higher level of antibodies and inflammatory cytokines; (Interferon (IFN)-γ and Interleukin (IL)-1ß) compared to those of aMPV. The increased levels of antibodies and inflammatory cytokines in chicken immunized with aMPV-cFc were statistically significantly (p<0.05) to that of aMPV and control. The aMPV-cFc group also generated the highest neutralizing antibody response. After challenges, chickens immunized with aMPV-cFc showed much less pathological signs in nasal turbinates and trachea so that we could confirm aMPV-cFc induced higher protection than that of aMPV. The greater ability of aMPV harboring chicken Fc to that of aMPV presented it as a possible vaccine candidate.


Subject(s)
Antibodies, Neutralizing/biosynthesis , Antibodies, Viral/biosynthesis , Chickens/immunology , Immunoglobulin Fc Fragments/genetics , Paramyxoviridae Infections/veterinary , Poultry Diseases/prevention & control , Viral Vaccines/administration & dosage , Animals , Chickens/virology , Chlorocebus aethiops , Gene Expression , Immunization , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Interleukin-1beta/biosynthesis , Interleukin-1beta/immunology , Metapneumovirus/drug effects , Metapneumovirus/growth & development , Metapneumovirus/immunology , Paramyxoviridae Infections/immunology , Paramyxoviridae Infections/prevention & control , Paramyxoviridae Infections/virology , Plasmids/chemistry , Plasmids/immunology , Poultry Diseases/immunology , Poultry Diseases/virology , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Vero Cells , Viral Proteins/genetics , Viral Proteins/immunology , Viral Vaccines/genetics , Viral Vaccines/immunology
16.
PLoS One ; 11(5): e0155484, 2016.
Article in English | MEDLINE | ID: mdl-27171557

ABSTRACT

Human metapneumovirus (hMPV) causes severe airway infection in children that may be caused by an unfavorable immune response. The nature of the innate immune response to hMPV in naturally occurring infections in children is largely undescribed, and it is unknown if inflammasome activation is implicated in disease pathogenesis. We examined nasopharynx aspirates and blood samples from hMPV-infected children without detectable co-infections. The expression of inflammatory and antiviral genes were measured in nasal airway secretions by relative mRNA quantification while blood plasma proteins were determined by a multiplex immunoassay. Several genes were significantly up-regulated at mRNA and protein level in the hMPV infected children. Most apparent was the expression of the chemokine IP-10, the pro-inflammatory cytokine IL-18 in addition to the interferon inducible gene ISG54. Interestingly, children experiencing more severe disease, as indicated by a severity index, had significantly more often up-regulation of the inflammasome-associated genes IL-1ß and NLRP3. Overall, our data point to cytokines, particularly inflammasome-associated, that might be important in hMPV mediated lung disease and the antiviral response in children with severe infection. Our study is the first to demonstrate that inflammasome components are associated with increased illness severity in hMPV-infected children.


Subject(s)
Antiviral Agents/therapeutic use , Cytokines/genetics , Metapneumovirus/pathogenicity , Paramyxoviridae Infections/genetics , Paramyxoviridae Infections/virology , Antiviral Agents/pharmacology , Case-Control Studies , Child , Child, Preschool , Cytokines/metabolism , Female , Gene Expression Regulation/drug effects , Genotype , Humans , Infant , Inflammasomes/genetics , Inflammasomes/metabolism , Interferons/genetics , Interferons/metabolism , Male , Metapneumovirus/drug effects , Nasopharynx/pathology , Nasopharynx/virology , Paramyxoviridae Infections/blood , Paramyxoviridae Infections/drug therapy
17.
Antimicrob Agents Chemother ; 60(8): 4620-9, 2016 08.
Article in English | MEDLINE | ID: mdl-27185803

ABSTRACT

The clinical impact of infections with respiratory viruses belonging to the family Paramyxoviridae argues for the development of antiviral therapies with broad-spectrum activity. Favipiravir (T-705) has demonstrated potent antiviral activity against multiple RNA virus families and is presently in clinical evaluation for the treatment of influenza. Here we demonstrate in vitro activity of T-705 against the paramyxoviruses human metapneumovirus (HMPV), respiratory syncytial virus, human parainfluenza virus, measles virus, Newcastle disease virus, and avian metapneumovirus. In addition, we demonstrate activity against HMPV in hamsters. T-705 treatment inhibited replication of all paramyxoviruses tested in vitro, with 90% effective concentration (EC90) values of 8 to 40 µM. Treatment of HMPV-challenged hamsters with T-705 at 200 mg/kg of body weight/day resulted in 100% protection from infection of the lungs. In all treated and challenged animals, viral RNA remained detectable in the respiratory tract. The observation that T-705 treatment had a significant effect on infectious viral titers, with a limited effect on viral genome titers, is in agreement with its proposed mode of action of viral mutagenesis. However, next-generation sequencing of viral genomes isolated from treated and challenged hamsters did not reveal (hyper)mutation. Polymerase activity assays revealed a specific effect of T-705 on the activity of the HMPV polymerase. With the reported antiviral activity of T-705 against a broad range of RNA virus families, this small molecule is a promising broad-range antiviral drug candidate for limiting the viral burden of paramyxoviruses and for evaluation for treatment of infections with (re)emerging viruses, such as the henipaviruses.


Subject(s)
Amides/pharmacology , Antiviral Agents/pharmacology , Metapneumovirus/drug effects , Paramyxoviridae Infections/drug therapy , Pyrazines/pharmacology , Animals , Antibodies, Viral/immunology , Chlorocebus aethiops , Cricetinae , HEK293 Cells , Humans , Lung/virology , Mesocricetus , Respiratory Syncytial Viruses/drug effects , Vero Cells , Virus Replication/drug effects
18.
Mol Cell Probes ; 29(6): 485-491, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26028611

ABSTRACT

To understand the molecular mechanisms of Avian metapneumovirus (aMPV) and the requirements involved in the infection and fusion, trypsin treatment was done in the different stages of virus; before infection, during entry and after virus infection followed by aMPV infection. The growth kinetics of aMPV was compared in time dependent manner. The effect of trypsin was found in the later stage of aMPV infection increasing the numbers of infected cells with the significant higher titer of infectious virions to that of trypsin treated before infection, during entry and aMPV. A serine protease inhibitor reduced aMPV replication in a significant way, whereas cysteine peptidase (E-64), aspartic protease (pepstatin A), and metalloprotease (phosphoramidon) inhibitors had no effect on aMPV replication. Inoculation of aMPV on Vero cells expressing the membrane-associated protease TMPRSS2 resulted in higher virus titers than that inoculated on normal Vero cells and is statistically significant (p < 0.05). Also, an inhibitor of clathrin/caveolae-mediated endocytosis had no effect on virus progeny, indicating that aMPV does not use the endocytic pathway for entry but undergoes direct fusion. The effect of lysosomotropic agents was not significant, suggesting that aMPV does not require low-pH environment in endosomes to fuse its envelope with the plasma membrane.


Subject(s)
Metapneumovirus/physiology , Trypsin/pharmacology , Virus Internalization/drug effects , Virus Replication/drug effects , Animals , Chlorocebus aethiops , Membrane Fusion/drug effects , Metapneumovirus/classification , Metapneumovirus/drug effects , Paramyxoviridae Infections/veterinary , Paramyxoviridae Infections/virology , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Vero Cells , Viral Load/drug effects
19.
Planta ; 242(1): 69-76, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25828350

ABSTRACT

MAIN CONCLUSION: The HRA2pl peptide expressed by transient transformation in N. tabacum plants is capable of inhibiting the binding of the human metapneumovirus to HEp-2 cells at the fusion stage. Human metapneumovirus (hMPV) is an agent responsible for acute respiratory infections that mainly affects children under 3 years, the elderly and immunocompromised patients. In children younger than 5 years, respiratory tract infections account for 20 % of deaths worldwide. However, there is currently no treatment or vaccine available against hMPV. The production of a safe, efficient and low cost treatment against this virus is a current challenge. Plants provide a system for recombinant protein production that is cost effective and is easier to scale up to an industrial level than other platforms; in addition, the plant tissue may be used as raw food, dried or, alternatively, proteins may be partially or fully purified and administered in aerosol or capsules as dry powder. In this study, we designed a gene expressing an antiviral peptide against hMPV based on the heptad repeat A domain of the F protein of the virus. We produced the recombinant peptide by a viral transient expression system (Magnifection(®)) in Nicotiana tabacum plants. The efficacy of this antiviral peptide was confirmed by in vitro assays in HEp-2 cell line. This is a promising result that can offer a prophylactic approach against hMPV.


Subject(s)
Antiviral Agents/chemistry , Metapneumovirus/physiology , Nicotiana/genetics , Peptides/pharmacology , Transformation, Genetic , Virus Internalization/drug effects , Amino Acid Sequence , Antiviral Agents/pharmacology , Biological Assay , Cell Death/drug effects , Cell Line, Tumor , Drug Design , Humans , Metapneumovirus/drug effects , Molecular Sequence Data , Paramyxoviridae Infections/pathology , Paramyxoviridae Infections/virology , Peptides/chemistry , Plants, Genetically Modified , Transformation, Genetic/drug effects
20.
Eur J Immunol ; 45(6): 1680-95, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25763996

ABSTRACT

Human metapneumovirus (hMPV) is a leading cause of acute respiratory tract infections in children and the elderly. The mechanism by which this virus triggers an inflammatory response still remains unknown. Here, we evaluated whether the thymic stromal lymphopoietin (TSLP) pathway contributes to lung inflammation upon hMPV infection. We found that hMPV infection promotes TSLP expression both in human airway epithelial cells and in the mouse lung. hMPV infection induced lung infiltration of OX40L(+) CD11b(+) DCs. Mice lacking the TSLP receptor deficient mice (tslpr(-/-) ) showed reduced lung inflammation and hMPV replication. These mice displayed a decreased number of neutrophils as well a reduction in levels of thymus and activation-regulated chemokine/CCL17, IL-5, IL-13, and TNF-α in the airways upon hMPV infection. Furthermore, a higher frequency of CD4(+) and CD8(+) T cells was found in tslpr(-/-) mice compared to WT mice, which could contribute to controlling viral spread. Depletion of neutrophils in WT and tslpr(-/-) mice decreased inflammation and hMPV replication. Remarkably, blockage of TSLP or OX40L with specific Abs reduced lung inflammation and viral replication following hMPV challenge in mice. Altogether, these results suggest that activation of the TSLP pathway is pivotal in the development of pulmonary pathology and pulmonary hMPV replication.


Subject(s)
Cytokines/metabolism , Metapneumovirus/physiology , Paramyxoviridae Infections/metabolism , Paramyxoviridae Infections/virology , Pneumonia, Viral/metabolism , Pneumonia, Viral/virology , Signal Transduction , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/pharmacology , Cell Line , Cytokines/genetics , Dendritic Cells/immunology , Dendritic Cells/metabolism , Disease Models, Animal , Epithelial Cells/metabolism , Epithelial Cells/virology , Gene Expression , Humans , Interleukin-33 , Interleukin-8/genetics , Interleukin-8/metabolism , Interleukins/genetics , Interleukins/metabolism , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Metapneumovirus/drug effects , Mice , Neutrophils/immunology , Neutrophils/metabolism , OX40 Ligand/antagonists & inhibitors , OX40 Ligand/genetics , OX40 Ligand/metabolism , Paramyxoviridae Infections/drug therapy , Paramyxoviridae Infections/genetics , Paramyxoviridae Infections/pathology , Pneumonia, Viral/drug therapy , Pneumonia, Viral/genetics , Pneumonia, Viral/pathology , Receptors, Cytokine/antagonists & inhibitors , Receptors, Cytokine/deficiency , Signal Transduction/drug effects , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/pathology , Virus Replication , Thymic Stromal Lymphopoietin
SELECTION OF CITATIONS
SEARCH DETAIL