Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 220
Filter
1.
Biomed Res Int ; 2021: 1807293, 2021.
Article in English | MEDLINE | ID: mdl-34409100

ABSTRACT

Human Parainfluenza Virus Type 3 (HPIV3) is one of the main pathogens that cause acute lower respiratory tract infections in infants and young children. However, there are currently no effective antiviral drugs and vaccines. Herein, we found that a natural compound, curcumin, inhibits HPIV3 infection and has antiviral effects on entry and replication of the virus life cycle. Immunofluorescence and western blotting experiments revealed that curcumin disrupts F-actin and inhibits viral inclusion body (IB) formation, thus inhibiting virus replication. Curcumin can also downregulate cellular PI4KB and interrupt its colocalization in viral IBs. This study verified the antiviral ability of curcumin on HPIV3 infection and preliminarily elucidated its influence on viral replication, providing a theoretical basis for antiviral drug development of HPIV3 and other parainfluenza viruses.


Subject(s)
Curcumin/pharmacology , Inclusion Bodies, Viral/metabolism , Parainfluenza Virus 3, Human/physiology , Respirovirus Infections/metabolism , 1-Phosphatidylinositol 4-Kinase/genetics , 1-Phosphatidylinositol 4-Kinase/metabolism , A549 Cells , Actins/metabolism , Animals , Dogs , Down-Regulation , Drug Tapering , HeLa Cells , Humans , Inclusion Bodies, Viral/drug effects , Inclusion Bodies, Viral/genetics , Madin Darby Canine Kidney Cells , Parainfluenza Virus 3, Human/drug effects , Respirovirus Infections/drug therapy , Respirovirus Infections/genetics , Virus Internalization/drug effects , Virus Replication/drug effects
2.
Am J Respir Cell Mol Biol ; 63(6): 758-766, 2020 12.
Article in English | MEDLINE | ID: mdl-32853024

ABSTRACT

Viral pneumonias remain global health threats, as exemplified in the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic, requiring novel treatment strategies both early and late in the disease process. We have reported that mice treated before or soon after infection with a combination of inhaled Toll-like receptor (TLR) 2/6 and 9 agonists (Pam2-ODN) are broadly protected against microbial pathogens including respiratory viruses, but the mechanisms remain incompletely understood. The objective of this study was to validate strategies for immune modulation in a preclinical model of viral pneumonia and determine their mechanisms. Mice were challenged with the Sendai paramyxovirus in the presence or absence of Pam2-ODN treatment. Virus burden and host immune responses were assessed to elucidate Pam2-ODN mechanisms of action and to identify additional opportunities for therapeutic intervention. Enhanced survival of Sendai virus pneumonia with Pam2-ODN treatment was associated with reductions in lung virus burden and with virus inactivation before internalization. We noted that mortality in sham-treated mice corresponded with CD8+ T-cell lung inflammation on days 11-12 after virus challenge, after the viral burden had declined. Pam2-ODN blocked this injurious inflammation by minimizing virus burden. As an alternative intervention, depleting CD8+ T cells 8 days after viral challenge also decreased mortality. Stimulation of local innate immunity within the lungs by TLR agonists early in disease or suppression of adaptive immunity by systemic CD8+ T-cell depletion late in disease improves outcomes of viral pneumonia in mice. These data reveal opportunities for targeted immunomodulation to protect susceptible human subjects.


Subject(s)
Immunity, Innate/immunology , Lipopeptides/pharmacology , Pneumonia, Viral/drug therapy , Pneumonia/prevention & control , Respirovirus Infections/drug therapy , Sendai virus/drug effects , Viral Load/drug effects , Animals , Epithelial Cells/drug effects , Epithelial Cells/immunology , Epithelial Cells/virology , Female , Immunity, Innate/drug effects , Lung/drug effects , Lung/immunology , Lung/virology , Mice , Mice, Inbred C57BL , Pneumonia/immunology , Pneumonia/pathology , Pneumonia, Viral/immunology , Pneumonia, Viral/virology , Respirovirus Infections/immunology , Respirovirus Infections/virology , Sendai virus/immunology
3.
J Virol ; 92(2)2018 01 15.
Article in English | MEDLINE | ID: mdl-29093083

ABSTRACT

There are no approved vaccines or virus-specific treatments for human parainfluenza viruses (HPIVs), which have recently been reclassified into the species Human respirovirus 1, Human respirovirus 3, Human rubulavirus 2, and Human rubulavirus 4 These viruses cause morbidity and mortality in immunocompromised patients, including those undergoing hematopoietic cell transplant (HCT). No small-animal models for noninvasive imaging of respiratory virus infection in the HCT host exist, despite the utility that such a system would offer to monitor prolonged infection, its clearance, and treatment options. We used a luciferase-expressing reporter virus to noninvasively image in mice the infection of murine respirovirus (strain Sendai virus [SeV]), the murine counterpart of HPIV1. Independent of disease severity, the clearance of infection began approximately 21 days after HCT, largely due to the recovery of CD8+ T cells. Immunotherapy with granulocyte colony-stimulating factor (G-CSF) and adoptive transfer of natural killer (NK) cells provided a limited therapeutic benefit. Treatment with a fusion (F) protein-specific monoclonal antibody arrested the spread of lung infection and reduced the disease severity even when treatment was delayed to up to 10 days postinfection but had little observable effect on upper respiratory tract infection. Adoptive transfer of virus-specific T cells at 10 days postinfection accelerated the clearance by 5 days, reduced the extent of infection throughout the respiratory tract, and reduced the disease severity. Overall, the results support investigation of the clinical treatment of respiratory virus infection in the HCT host with monoclonal antibodies and adoptive T-cell transfer; the imaging system should be extendable to other respiratory viruses, such as respiratory syncytial virus and influenza virus.IMPORTANCE Parainfluenza viruses are a major cause of disease and death due to respiratory virus infection in the immunocompromised host, including those undergoing bone marrow transplantation. There are currently no effective treatment measures. We noninvasively imaged mice that were undergoing a bone marrow transplant and infected with Sendai virus, a murine parainfluenza virus (respirovirus). For the first time, we show the therapeutic windows of adoptive T-cell therapy and treatment with a monoclonal antibody to the fusion (F) protein in clearing Sendai virus from the respiratory tract and reducing disease severity. Mice tolerated these treatments without any detectable toxicity. These findings pave the way for studies assessing the safety of T-cell therapy against parainfluenza virus in humans. Adoptive T-cell therapy against other blood-borne viruses in humans has been shown to be safe and effective. Our model of noninvasive imaging in mice that had undergone a bone marrow transplant may be well suited to track other respiratory virus infections and develop novel preventive and therapeutic strategies.


Subject(s)
Hematopoietic Stem Cell Transplantation/adverse effects , Immunotherapy/adverse effects , Respirovirus Infections/etiology , Respirovirus Infections/virology , Sendai virus/physiology , Adoptive Transfer , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Antibodies, Viral/immunology , Antibodies, Viral/pharmacology , Female , Granulocyte Colony-Stimulating Factor/pharmacology , Immunotherapy/methods , Luminescent Measurements/methods , Lymphocyte Count , Mice , Neutrophils/drug effects , Neutrophils/immunology , Respirovirus Infections/diagnosis , Respirovirus Infections/drug therapy , Severity of Illness Index , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Viral Load
4.
PLoS One ; 12(7): e0181425, 2017.
Article in English | MEDLINE | ID: mdl-28742120

ABSTRACT

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


Subject(s)
Asthma/virology , Parainfluenza Virus 1, Human/immunology , Respiratory Hypersensitivity/virology , Respirovirus Infections/complications , Animals , Anti-Inflammatory Agents/therapeutic use , Asthma/drug therapy , Asthma/immunology , Dexamethasone/therapeutic use , Disease Models, Animal , Female , Guinea Pigs , Humans , Immunologic Memory/drug effects , Inflammation/drug therapy , Inflammation/immunology , Inflammation/virology , Lymphocytes/immunology , Lymphocytes/virology , Respiratory Hypersensitivity/drug therapy , Respiratory Hypersensitivity/immunology , Respirovirus Infections/drug therapy , Respirovirus Infections/immunology
5.
Immun Inflamm Dis ; 5(2): 98-108, 2017 06.
Article in English | MEDLINE | ID: mdl-28474501

ABSTRACT

INTRODUCTION: Asthma is major health burden throughout the world, and there are no therapies that have been shown to be able to prevent the development of disease. A severe respiratory paramyxoviral infection early in life has been demonstrated to greatly increase the risk of developing asthma. We have a mouse model of a severe respiratory paramyxoviral infection (Sendai virus, SeV) that mimics human disease, and requires early expression of the cytokine CCL28 to drive the development of post-viral airway disease. The known receptors for CCL28 are CCR3 and CCR10. However, it is not known if blockade of these receptors will prevent the development of post-viral airway disease. The objective of this study was to determine if treatment with a protein epitope mimetic antagonist of CCR10, POL7085, will provide sufficient protection against the development of post-viral airway disease. METHODS: C57BL6 mice were inoculated with SeV or UV inactivated SeV. From day 3-19 post inoculation (PI), mice were subcutaneously administered daily POL7085 or saline, or every other day anti-CCL28 mAb. On days 8, 10, and 12 PI bronchoalveolar cytokines, serum IgE, and lung cellular constituents were measured. At day 21 PI airway hyper-reactivity to methacholine and mucous cell metaplasia was measured. RESULTS: Treatment with either anti-CCL28 or POL7085 significantly reduced development of airway hyper-reactivity and mucous cell metaplasia following SeV infection. The prevention of post-viral airway disease was associated with early reductions in innate immune cells, but did not appear to be due to a reduction in IL-13 or IgE. CONCLUSIONS: Blockade of CCL28 or CCR10 during an acute severe respiratory paramyxoviral infection is sufficient to prevent the development of post-viral airway disease. However, the mechanism of action is unclear and requires further exploration.


Subject(s)
Asthma/drug therapy , Chemokines, CC/antagonists & inhibitors , Receptors, CCR10/antagonists & inhibitors , Respirovirus Infections/drug therapy , Sendai virus/immunology , Animals , Asthma/etiology , Asthma/immunology , Asthma/virology , Chemokines, CC/immunology , Humans , Mice , Receptors, CCR10/immunology , Respirovirus Infections/complications , Respirovirus Infections/immunology
6.
J Enzyme Inhib Med Chem ; 32(1): 375-402, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28276287

ABSTRACT

Respiratory syncytial virus (RSV) has been identified as a main cause of hospitalisation in infants and children. To date, the current therapeutic arsenal is limited to ribavirin and palivizumab with variable efficacy. In this work, starting from a number of in-house series of previously described anti-RSV agents based on the benzimidazole scaffold, with the aim at gaining a better understanding of the related chemical features involved in potency and safety profiles, we applied a computational study including two focussed comparative molecular fields analysis (CoMFA) and comparative molecular similarity indices analysis (CoMSIA). The results allowed us to derive useful suggestions for the design of derivatives and also to set up statistical models predicting the potency and selectivity index (SI = CC50/EC50) of any new analogue prior to synthesis. Accordingly, here, we discuss preliminary results obtained through the applied exhaustive QSAR analyses, leading to design and synthesise more effective anti-RSV agents.


Subject(s)
Antiviral Agents/pharmacology , Benzimidazoles/pharmacology , Respiratory Syncytial Viruses/drug effects , Respirovirus Infections/drug therapy , Animals , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Benzimidazoles/chemical synthesis , Benzimidazoles/chemistry , Cell Line , Chlorocebus aethiops , Dose-Response Relationship, Drug , Humans , Microbial Sensitivity Tests , Models, Molecular , Molecular Structure , Quantitative Structure-Activity Relationship , Vero Cells
7.
Virology ; 503: 46-51, 2017 03.
Article in English | MEDLINE | ID: mdl-28113063

ABSTRACT

Interferon (IFN) exerts its antiviral effect by inducing a large family of cellular genes, named interferon (IFN)-stimulated genes (ISGs). An intriguing member of this family is indoleamine 2,3-dioxygenase (IDO), which catalyzes the first and rate-limiting step of the main branch of tryptophan (Trp) degradation, the kynurenine pathway. We recently showed that IDO strongly inhibits human parainfluenza virus type 3 (PIV3), a significant respiratory pathogen. Here, we show that 5-hydoxytryptophan (5-HTP), the first product of an alternative branch of Trp degradation and a serotonin precursor, is essential to protect virus growth against IDO in cell culture. We also show that the apparent antiviral effect of IDO on PIV3 is not due to the generation of the kynurenine pathway metabolites, but rather due to the depletion of intracellular Trp by IDO, as a result of which this rare amino acid becomes unavailable for the alternative, proviral 5-HTP pathway.


Subject(s)
5-Hydroxytryptophan/metabolism , Antiviral Agents/pharmacology , Indoleamine-Pyrrole 2,3,-Dioxygenase/pharmacology , Parainfluenza Virus 3, Human/growth & development , Tryptophan/metabolism , Virus Replication/drug effects , 5-Hydroxytryptophan/pharmacology , A549 Cells , Animals , Cell Line, Tumor , Humans , Interferons/pharmacology , Kynurenine/metabolism , Macaca mulatta , Parainfluenza Virus 3, Human/metabolism , Respirovirus Infections/drug therapy , Tryptophan/chemistry , Virus Replication/physiology
8.
PLoS Pathog ; 12(9): e1005875, 2016 09.
Article in English | MEDLINE | ID: mdl-27589232

ABSTRACT

In immunocompromised patients, parainfluenza virus (PIV) infections have an increased potential to spread to the lower respiratory tract (LRT), resulting in increased morbidity and mortality. Understanding the immunologic defects that facilitate viral spread to the LRT will help in developing better management protocols. In this study, we immunosuppressed mice with dexamethasone and/or cyclophosphamide then monitored the spread of viral infection into the LRT by using a noninvasive bioluminescence imaging system and a reporter Sendai virus (murine PIV type 1). Our results show that immunosuppression led to delayed viral clearance and increased viral loads in the lungs. After cessation of cyclophosphamide treatment, viral clearance occurred before the generation of Sendai-specific antibody responses and coincided with rebounds in neutrophils, T lymphocytes, and natural killer (NK) cells. Neutrophil suppression using anti-Ly6G antibody had no effect on infection clearance, NK-cell suppression using anti-NK antibody delayed clearance, and T-cell suppression using anti-CD3 antibody resulted in no clearance (chronic infection). Therapeutic use of hematopoietic growth factors G-CSF and GM-CSF had no effect on clearance of infection. In contrast, treatment with Sendai virus-specific polysera or a monoclonal antibody limited viral spread into the lungs and accelerated clearance. Overall, noninvasive bioluminescence was shown to be a useful tool to study respiratory viral progression, revealing roles for NK and T cells, but not neutrophils, in Sendai virus clearance after treatment with dexamethasone and cyclophosphamide. Virus-specific antibodies appear to have therapeutic potential.


Subject(s)
Cyclophosphamide/therapeutic use , Dexamethasone/therapeutic use , Paramyxoviridae Infections/immunology , Respirovirus Infections/pathology , Sendai virus/physiology , Animals , Disease Models, Animal , Filgrastim , Granulocyte Colony-Stimulating Factor , Granulocyte-Macrophage Colony-Stimulating Factor , Humans , Immunocompromised Host , Killer Cells, Natural/immunology , Killer Cells, Natural/virology , Lung/immunology , Lung/virology , Mice , Mice, Inbred BALB C , Neutrophils/pathology , Neutrophils/virology , Paramyxoviridae Infections/virology , Polyethylene Glycols , Recombinant Proteins , Respirovirus Infections/drug therapy , Respirovirus Infections/immunology , Respirovirus Infections/virology , T-Lymphocytes/immunology , T-Lymphocytes/virology
9.
Blood ; 127(22): 2682-92, 2016 06 02.
Article in English | MEDLINE | ID: mdl-26968533

ABSTRACT

The widespread use of multiplex molecular diagnostics has led to a significant increase in the detection of respiratory viruses in patients undergoing cytotoxic chemotherapy and hematopoietic cell transplantation (HCT). Respiratory viruses initially infect the upper respiratory tract and then progress to lower respiratory tract disease in a subset of patients. Lower respiratory tract disease can manifest itself as airflow obstruction or viral pneumonia, which can be fatal. Infection in HCT candidates may require delay of transplantation. The risk of progression differs between viruses and immunosuppressive regimens. Risk factors for progression and severity scores have been described, which may allow targeting treatment to high-risk patients. Ribavirin is the only antiviral treatment option for noninfluenza respiratory viruses; however, high-quality data demonstrating its efficacy and relative advantages of the aerosolized versus oral form are lacking. There are significant unmet needs, including data defining the virologic characteristics and clinical significance of human rhinoviruses, human coronaviruses, human metapneumovirus, and human bocavirus, as well as the need for new treatment and preventative options.


Subject(s)
Hematopoietic Stem Cell Transplantation , Pneumonia, Viral/drug therapy , Respirovirus Infections/drug therapy , Respirovirus , Ribavirin/therapeutic use , Allografts , Humans , Pneumonia, Viral/etiology , Respirovirus Infections/etiology , Risk Factors
10.
J Pediatric Infect Dis Soc ; 4(2): 114-8, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26185620

ABSTRACT

BACKGROUND: Parainfluenza virus (PIV), a common pediatric pathogen, is associated with significant morbidity in immunocompromised (IC) hosts. DAS181, a novel sialidase fusion protein inhibitor, seems to be effective against PIV in vitro and in vivo; its use in IC children has not been evaluated. METHODS: Patients were diagnosed with PIV infection using a quantitative reverse transcription-polymerase chain reaction. DAS181 was obtained under emergency investigational new drug applications and was administered via aerosol chamber or nebulizer. Patients were assessed daily for their clinical condition and adverse outcomes. RESULTS: Four pediatric hematopoietic cell transplantation (HCT) patients with PIV detected in respiratory specimens were identified and treated with DAS 181. Patients 1 and 2 were diagnosed with PIV lower respiratory tract infection (LRTI) by bronchoalveolar lavage at 9 months and 2 days after allogeneic transplantation, respectively. Patient 3 was on chemotherapy prior to planned autologous HCT at time of PIV diagnosis from a nasal swab. Patient 4 was diagnosed with PIV via nasal wash 2 days after HCT. Patients 1-3 had clinical symptoms and chest imaging consistent with LRTI. Inhaled DAS181 was administered for 5-10 days. All 4 patients tolerated therapy well. Clinical improvement in oxygen requirement and respiratory rate was observed in all patients who required oxygen at therapy initiation. Viral load decreased in all patients within 1 week of therapy and became undetectable by day 3 of therapy in patient 3. CONCLUSION: DAS181 was used to treat 4 severely IC pediatric patients with PIV disease. The drug was well tolerated. Improvement in both viral loads and symptoms after initiation of therapy was observed in all cases. This report supports prospective, randomized studies in IC patients with PIV infection.


Subject(s)
Immunocompromised Host/physiology , Parainfluenza Virus 2, Human/drug effects , Parainfluenza Virus 3, Human/drug effects , Recombinant Fusion Proteins/pharmacology , Recombinant Fusion Proteins/therapeutic use , Respiratory Tract Infections/drug therapy , Respirovirus Infections/drug therapy , Rubulavirus Infections/drug therapy , Alanine Transaminase/blood , Alanine Transaminase/drug effects , Aspartate Aminotransferases/blood , Aspartate Aminotransferases/drug effects , Child , Child, Preschool , Humans , Infant , Leukemia, Myeloid, Acute/complications , Male , Neuroblastoma/complications , Parainfluenza Virus 2, Human/physiology , Parainfluenza Virus 3, Human/physiology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/complications , Prospective Studies , Random Allocation , Recombinant Fusion Proteins/administration & dosage , Respiratory Tract Infections/diagnosis , Severe Combined Immunodeficiency/complications , Transplantation/adverse effects , Viral Load/drug effects , Viral Load/physiology
11.
PLoS One ; 10(4): e0123635, 2015.
Article in English | MEDLINE | ID: mdl-25849666

ABSTRACT

Viral double-stranded RNA (dsRNA) is recognised by pathogen recognition receptors such as Toll-Like Receptor 3 (TLR3) and retinoic acid inducible gene-I (RIG-I), and results in cytokine and interferon production. Fas, a well characterised death receptor, has recently been shown to play a role in the inflammatory response. In this study we investigated the role of Fas in the anti-viral immune response. Stimulation of Fas on macrophages did not induce significant cytokine production. However, activation of Fas modified the response of macrophages to the viral dsRNA analogue poly I:C. In particular, poly I:C-induced IP-10 production was significantly enhanced. A similar augmentation of IP-10 by Fas was observed following stimulation with both poly A:U and Sendai virus. Fas activation suppressed poly I:C-induced phosphorylation of the MAP kinases p38 and JNK, while overexpression of the Fas adaptor protein, Fas-associated protein with death domain (FADD), activated AP-1 and inhibited poly I:C-induced IP-10 production. Consistent with an inhibitory role for AP-1 in IP-10 production, mutation of the AP-1 binding site on the IP-10 promoter resulted in augmented poly I:C-induced IP-10. These results demonstrate that engagement of the Fas receptor plays a role in modifying the innate immune response to viral RNA.


Subject(s)
Antiviral Agents/pharmacology , Cytokines/metabolism , Interleukin-10/metabolism , Macrophages/metabolism , Poly I-C/pharmacology , Signal Transduction/drug effects , Apoptosis , Blotting, Western , Cell Proliferation , Cells, Cultured , Cytokines/genetics , Fas-Associated Death Domain Protein/genetics , Fas-Associated Death Domain Protein/metabolism , Humans , Interleukin-10/genetics , Macrophages/cytology , Macrophages/drug effects , Macrophages/virology , NF-kappa B/genetics , NF-kappa B/metabolism , Phosphorylation/drug effects , RNA, Messenger/genetics , RNA, Viral/genetics , Real-Time Polymerase Chain Reaction , Respirovirus Infections/drug therapy , Respirovirus Infections/metabolism , Respirovirus Infections/virology , Reverse Transcriptase Polymerase Chain Reaction , Sendai virus/genetics , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism
12.
Antivir Chem Chemother ; 23(4): 161-71, 2014 Jan 29.
Article in English | MEDLINE | ID: mdl-23538746

ABSTRACT

BACKGROUND: Ribavirin is a broad-spectrum antiviral agent that derives some of its activity from inhibition of cellular inosine monophosphate dehydrogenase (IMPDH), resulting in lower guanosine triphosphate (GTP) levels. Here we report the biological activities of three ribavirin analogues. METHODS: Antiviral activities of test compounds were performed by in vitro cytopathic effect inhibition assays against influenza A (H1N1, H3N2 and H5N1), influenza B, measles, parainfluenza type 3 (PIV-3) and respiratory syncytial viruses. Compounds were modelled into the ribavirin 5'-monophosphate binding site of the crystallographic structure of the human type II IMPDH (hIMPDH2) ternary complex. Effects of compounds on intracellular GTP levels were performed by strong anion exchange HPLC analysis. RESULTS: Of the three compounds evaluated, the 5-ethynyl nucleoside (ETCAR) exhibited virus-inhibitory activities (at 1.2-20 µM, depending upon the virus) against most of the viruses, except for weak activity against PIV-3 (62 µM). Antiviral activity of ETCAR was similar to ribavirin; however, cytotoxicity of ETCAR was greater than ribavirin. Replacing the 5-ethynyl group with a 5-propynyl or bromo substituent (BrCAR) considerably reduced antiviral activity. Computational studies of ternary complexes of hIMPDH2 enzyme with 5'-monophosphates of the compounds helped rationalize the observed differences in biological activity. All compounds suppressed GTP levels in cells; additionally, BrCAR suppressed adenosine triphosphate and elevated uridine triphosphate levels. CONCLUSIONS: Three compounds related to ribavirin inhibited IMPDH and had weak to moderate antiviral activity. Cytotoxicity adversely affected the antiviral selectivity of ETCAR. As with ribavirin, reduction in intracellular GTP may play a role in virus inhibition.


Subject(s)
Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Nucleosides/chemistry , Nucleosides/pharmacology , Ribavirin/analogs & derivatives , Ribavirin/pharmacology , Animals , Cell Line , Humans , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H3N2 Subtype/drug effects , Influenza A Virus, H5N1 Subtype/drug effects , Influenza B virus/drug effects , Influenza, Human/drug therapy , Measles/drug therapy , Measles virus/drug effects , Models, Molecular , Orthomyxoviridae Infections/drug therapy , Parainfluenza Virus 3, Human/drug effects , Respiratory Syncytial Virus Infections/drug therapy , Respiratory Syncytial Viruses/drug effects , Respirovirus Infections/drug therapy , Triazoles/chemistry , Triazoles/pharmacology
13.
Antiviral Res ; 100(2): 562-6, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24076357

ABSTRACT

DAS181 is a novel drug in development for the treatment of influenza as well as human parainfluenza viruses (hPIVs). Previous studies demonstrated that DAS181 inhibited laboratory strains of hPIV, but no tests were conducted with primary clinical isolates of hPIV. To fill this gap, we studied six primary isolates including hPIV-2 and hPIV-3. First tests showed that the amplification of all viruses in vitro was reproducibly inhibited with DAS181 drug concentrations ranging between 0.1 and 1nM. An hPIV-3 primary clinical isolate was then tested in a cotton rat model for sensitivity to 0.3-1mg/kg drug treatments. Results showed that virus amplification in the lower respiratory tract was significantly and reproducibly inhibited by drug. Together, experiments demonstrated that DAS181 inhibited primary clinical isolates of hPIV in vitro and in vivo at doses similar to those previously described for inhibition of laboratory hPIV and influenza virus isolates.


Subject(s)
Antiviral Agents/pharmacology , Parainfluenza Virus 2, Human/drug effects , Parainfluenza Virus 3, Human/drug effects , Recombinant Fusion Proteins/pharmacology , Respirovirus Infections/drug therapy , Animals , Antiviral Agents/therapeutic use , Cells, Cultured , Disease Models, Animal , Humans , Microbial Sensitivity Tests , Parainfluenza Virus 2, Human/isolation & purification , Parainfluenza Virus 3, Human/isolation & purification , Recombinant Fusion Proteins/therapeutic use , Respiratory System/virology , Respirovirus Infections/virology , Rubulavirus Infections/virology , Sigmodontinae , Treatment Outcome , Viral Load
14.
Bone Marrow Transplant ; 48(12): 1558-61, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23912665

ABSTRACT

The prognosis for patients with respiratory syncytial virus (RSV) or parainfluenza virus type 3 (PIV3) respiratory tract infection post allogeneic haematopoietic progenitor cell transplant (HPCT) is historically poor. The use of oral ribavirin (RBV) has not been widely studied in this patient population. We examined the outcomes of 15 consecutive patients (RSV, n=13 and PIV3, n=2) treated with oral RBV post HPCT. Oral RBV was commenced at a starting dose of 10 mg/kg/day, increasing to a maximum dose of 60 mg/kg/day depending on response and tolerance. At diagnosis, seven patients had upper respiratory tract infection (URTI) and eight had lower respiratory tract infection (LRTI). The starting RBV dose of 10 mg/kg/day did not prevent the progression of URTI to LRTI in any patient. However, with dose escalation, six of the seven patients responded to RBV therapy and survived their infective episode. Of the eight patients presenting with LRTI, six patients survived their infection, again after dose escalation of RBV. There was no dose-limiting toxicity seen in any patient. Our results indicate that oral RBV has clinical efficacy in the treatment of RSV/PIV3 infection post HPCT. However, a starting dose of 10 mg/kg/day appears ineffective; we recommend a starting dose of 20 mg/kg/day in this patient group.


Subject(s)
Hematopoietic Stem Cell Transplantation/methods , Parainfluenza Virus 3, Human/isolation & purification , Respiratory Syncytial Virus Infections/drug therapy , Respiratory Syncytial Viruses/isolation & purification , Respirovirus Infections/drug therapy , Ribavirin/administration & dosage , Administration, Oral , Adult , Female , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Male , Middle Aged , Prognosis , Retrospective Studies , Transplantation Conditioning/adverse effects , Transplantation Conditioning/methods , Transplantation, Autologous , Treatment Outcome , Young Adult
15.
Am J Respir Cell Mol Biol ; 48(6): 790-6, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23449736

ABSTRACT

Respiratory virus infections cause airway hyperreactivity (AHR). Preventative strategies for virus-induced AHR remain limited. Toll-like receptors (TLRs) have been suggested as a therapeutic target because of their central role in triggering antiviral immune responses. Previous studies showed that concurrent treatment with TLR2/6 and TLR9 agonists reduced lethality and the microbial burden in murine models of bacterial and viral pneumonia. This study investigated the effects of TLR2/6 and TLR9 agonist pretreatment on parainfluenza virus pneumonia and virus-induced AHR in guinea pigs in vivo. Synthetic TLR2/6 lipopeptide agonist Pam2CSK4 and Class C oligodeoxynucleotide TLR9 agonist ODN2395, administered in combination 24 hours before virus infection, significantly reduced viral replication in the lung. Despite a fivefold reduction in viral titers, concurrent TLR2/6 and TLR9 agonist pretreatment did not prevent virus-induced AHR or virus-induced inhibitory M2 muscarinic receptor dysfunction. Interestingly, the TLR agonists independently caused non-M2-dependent AHR. These data confirm the therapeutic antiviral potential of TLR agonists, while suggesting that virus inhibition may be insufficient to prevent virus-induced airway pathophysiology. Furthermore, TLR agonists independently cause AHR, albeit through a distinctly different mechanism from that of parainfluenza virus.


Subject(s)
Bronchial Hyperreactivity/virology , Toll-Like Receptor 2/agonists , Toll-Like Receptor 6/agonists , Toll-Like Receptor 9/agonists , Virus Replication/drug effects , Acetylcholine/pharmacology , Animals , Antiviral Agents/pharmacology , Bronchial Hyperreactivity/immunology , Bronchial Hyperreactivity/physiopathology , Bronchoconstriction , Drug Synergism , Female , Guinea Pigs , Leukocyte Count , Lipopeptides/pharmacology , Lung/drug effects , Lung/immunology , Lung/virology , Oligodeoxyribonucleotides/pharmacology , Receptor, Muscarinic M3/drug effects , Receptor, Muscarinic M3/immunology , Respirovirus Infections/drug therapy , Respirovirus Infections/immunology , Respirovirus Infections/physiopathology , Respirovirus Infections/virology , Sendai virus/immunology , Sendai virus/physiology , Specific Pathogen-Free Organisms , Toll-Like Receptor 2/immunology , Toll-Like Receptor 6/immunology , Toll-Like Receptor 9/immunology
16.
Chem Pharm Bull (Tokyo) ; 61(1): 69-74, 2013.
Article in English | MEDLINE | ID: mdl-23302587

ABSTRACT

Novel sialidase inhibitors 11 having an α-acylaminoamido group at the C-4 position of Neu5Ac2en 1 against human parainfluenza virus type 1 (hPIV-1) were synthesized using one-pot isocyanide-based four-component condensation, and their inhibitory activities against hPIV-1 sialidase were studied. Compound 11b showed inhibitory activity (IC(50)=5.1 mM) against hPIV-1 sialidase. The degree of inhibition of 11b was much weaker than that of 1 (IC(50)=0.3 mM).


Subject(s)
Antiviral Agents/chemistry , Antiviral Agents/pharmacology , N-Acetylneuraminic Acid/analogs & derivatives , Neuraminidase/antagonists & inhibitors , Parainfluenza Virus 1, Human/enzymology , Respirovirus Infections/drug therapy , Antiviral Agents/chemical synthesis , Humans , N-Acetylneuraminic Acid/chemical synthesis , N-Acetylneuraminic Acid/chemistry , N-Acetylneuraminic Acid/pharmacology , Parainfluenza Virus 1, Human/drug effects , Respirovirus Infections/virology , Structure-Activity Relationship
17.
Transpl Infect Dis ; 15(1): E28-32, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23279859

ABSTRACT

Parainfluenza virus (PIV) may cause life-threatening pneumonia in lung transplant patients and there are no proven effective therapies. We report the use of inhaled DAS181, a novel sialidase fusion protein, to treat severe PIV type 3 pneumonia in a lung transplant patient. Treatment was well tolerated and associated with improvement in oxygenation and symptoms, along with rapid clearance of PIV. DAS181 should be systematically evaluated for treatment of PIV infection in transplant recipients.


Subject(s)
Antiviral Agents/therapeutic use , Lung Transplantation/adverse effects , Parainfluenza Virus 3, Human/isolation & purification , Pneumonia, Viral/drug therapy , Recombinant Fusion Proteins/therapeutic use , Respirovirus Infections/drug therapy , Female , Humans , Middle Aged , Pneumonia, Viral/etiology , Respirovirus Infections/etiology , Severity of Illness Index , Treatment Outcome
18.
J Gen Virol ; 94(Pt 1): 59-68, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23052390

ABSTRACT

Interferon (IFN) induces an antiviral state in cells that results in alterations of the patterns and levels of parainfluenza virus type 5 (PIV5) transcripts and proteins. This study reports that IFN-stimulated gene 56/IFN-induced protein with tetratricopeptide repeats 1 (ISG56/IFIT1) is primarily responsible for these effects of IFN. It was shown that treating cells with IFN after infection resulted in an increase in virus transcription but an overall decrease in virus protein synthesis. As there was no obvious decrease in the overall levels of cellular protein synthesis in infected cells treated with IFN, these results suggested that ISG56/IFIT1 selectively inhibits the translation of viral mRNAs. This conclusion was supported by in vitro translation studies. Previous work has shown that ISG56/IFIT1 can restrict the replication of viruses lacking a 2'-O-methyltransferase activity, an enzyme that methylates the 2'-hydroxyl group of ribose sugars in the 5'-cap structures of mRNA. However, the data in the current study strongly suggested that PIV5 mRNAs are methylated at the 2'-hydroxyl group and thus that ISG56/IFIT1 selectively inhibits the translation of PIV5 mRNA by some as yet unrecognized mechanism. It was also shown that ISG56/IFIT1 is primarily responsible for the IFN-induced inhibition of PIV5.


Subject(s)
Carrier Proteins/biosynthesis , Interferon-alpha/pharmacology , Respirovirus Infections/virology , Respirovirus/drug effects , Respirovirus/genetics , Viral Proteins/biosynthesis , Adaptor Proteins, Signal Transducing , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line , Cell Line, Tumor , Chlorocebus aethiops , DNA Replication , Gene Knockdown Techniques , Humans , Interferon alpha-2 , Protein Biosynthesis , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Viral/genetics , RNA-Binding Proteins , Recombinant Proteins/pharmacology , Respirovirus/metabolism , Respirovirus Infections/drug therapy , Respirovirus Infections/metabolism , Transcription, Genetic , Vero Cells , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Replication/drug effects , Virus Replication/genetics
19.
Adv Exp Med Biol ; 756: 291-301, 2013.
Article in English | MEDLINE | ID: mdl-22836647

ABSTRACT

Clinicians often do not consider the presence of more than one viral etiologic agent in respiratory infection, and in many cases they order diagnostics for influenza viruses or recently even only for A(H1N1)2009 virus. However, in a substantial number of patients with a respiratory tract disease, co-infection with various viral pathogens has been confirmed. Although the association between the occurrence of co-infection and substantially higher severity of disease is still unclear, a rapid and proper diagnostics of wide spectrum of viral respiratory pathogens reveals an accurate picture of the disease and is essential for appropriate therapeutic management and control of infection. In the present study we reported five cases of multiple respiratory infection in hospitalized immunosuppressed patients: two double infections with influenza virus (IV) type A/respiratory syncytial virus (RSV) type A and IV type A/coronavirus (CoV) OC43, one infection with four viruses - IV type A/RSV type A and B/CoV OC43, and two cases of mixed infections caused by five viral agents - IV type A and B/RSV type A and B/ parainfluenza type 3 or CoV OC43. Each patient had an underlying chronic disease and received immunosuppressive treatment. Despite a low number of tested specimens, our study shows that the inclusions of multiplex PCR methods for diagnostics of respiratory tract infections and the extension of diagnostic strategies by clinicians to detect viruses other than influenza are very important and make a contribution to identifying the true rate of co-infections and their correlation with the clinical symptoms and severity of disease.


Subject(s)
Coinfection , Immunocompromised Host , Respiratory Tract Infections , Adult , Aged , Antiviral Agents/therapeutic use , Child, Preschool , Coronavirus , Coronavirus Infections/complications , Coronavirus Infections/diagnosis , Coronavirus Infections/drug therapy , Female , Humans , Influenza, Human/complications , Influenza, Human/diagnosis , Influenza, Human/drug therapy , Alphainfluenzavirus , Male , Middle Aged , Oseltamivir/therapeutic use , Parainfluenza Virus 3, Human , Respiratory Syncytial Virus Infections/complications , Respiratory Syncytial Virus Infections/diagnosis , Respiratory Syncytial Virus Infections/drug therapy , Respiratory Syncytial Viruses/genetics , Respiratory Tract Infections/diagnosis , Respiratory Tract Infections/drug therapy , Respiratory Tract Infections/virology , Respirovirus Infections/complications , Respirovirus Infections/diagnosis , Respirovirus Infections/drug therapy
20.
Antivir Chem Chemother ; 23(3): 113-28, 2012 Dec 07.
Article in English | MEDLINE | ID: mdl-23234699

ABSTRACT

BACKGROUND: Amantadine constitutes an interesting, diamond crystal lattice-shaped, antivirally active amine with an inhibitory effect on influenza A viruses causing common 'flu' in humans. Unfortunately, amantadine forfeited most of its therapeutic potential because of resistance development in recent influenza A virus isolates. The antiviral efficacy of amantadine congeners can be chemically modified, resulting in re-constitution, improvement and/or extension of antiviral activities mediated by amino-adamantyls. METHODS: Newly synthesized compounds were evaluated towards HIV type-1 (HIV-1) replication in primary human lymphocytes. One N-phenacyl amantadine derivative was investigated for inhibiting the in vitro replication of respiratory viruses (influenza A viruses, influenza B virus, human parainfluenza virus type 3 and severe acute respiratory syndrome coronavirus). RESULTS: Two ketone-stabilized 1-adamantyl singlet nitrenes were discovered serendipitously. To our best knowledge these are the first persistently stable nitrenes to be reported. Their structure was proved by determining the X-ray single crystal structure of one hydrolytic elaboration product. This salt adduct revealed an incommensurately modulated crystal structure, which was solved by extensive computational refinement. We could show that ketone-stabilized 1-adamantyl singlet nitrenes are versatile synthons for the synthesis of antiviral drug candidates. An amantadine-folate conjugate was inhibitory on HIV-1 replication in primary human lymphocytes, and one N-phenacyl amantadine derivative was inhibitory towards low pathogenic avian influenza A virus (H5N1) replication in vitro. CONCLUSIONS: These results indicate that the aromatic-aliphatic ketone-stabilized 1-adamantyl singlet nitrenes, beyond being of fundamental interest in organic chemistry, represent versatile synthons for the synthesis of new amantadine-related potentially antiviral drugs.


Subject(s)
Amantadine/pharmacology , Antiviral Agents/pharmacology , HIV-1/drug effects , Imines/pharmacology , Influenza A virus/drug effects , Influenza B virus/drug effects , Parainfluenza Virus 3, Human/drug effects , Severe acute respiratory syndrome-related coronavirus/drug effects , Amantadine/analogs & derivatives , Antiviral Agents/chemistry , Cells, Cultured , Crystallography, X-Ray , Humans , Imines/chemistry , Influenza, Human/drug therapy , Models, Molecular , Respirovirus Infections/drug therapy , Severe Acute Respiratory Syndrome/drug therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...