Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
J Virol ; 98(6): e0164123, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38690874

ABSTRACT

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


Subject(s)
Metapneumovirus , Paramyxoviridae Infections , Polysaccharides , Metapneumovirus/metabolism , Metapneumovirus/physiology , Humans , Polysaccharides/metabolism , Paramyxoviridae Infections/virology , Paramyxoviridae Infections/metabolism , Viral Fusion Proteins/metabolism , Virus Internalization , Virus Attachment , Protein Binding , Receptors, Virus/metabolism , Cell Line
2.
Antimicrob Agents Chemother ; 66(10): e0100822, 2022 10 18.
Article in English | MEDLINE | ID: mdl-36094205

ABSTRACT

Human metapneumovirus (HMPV) is recognized as an important cause of pneumonia in infants, in the elderly, and in immunocompromised individuals worldwide. The absence of an antiviral treatment or vaccine strategy against HMPV infection creates a high burden on the global health care system. Drug repurposing has become increasingly attractive for the treatment of emerging and endemic diseases as it requires less research and development costs than traditional drug discovery. In this study, we developed an in vitro medium-throughput screening assay that allows for the identification of novel anti-HMPV drugs candidates. Out of ~2,400 compounds, we identified 11 candidates with a dose-dependent inhibitory activity against HMPV infection. Additionally, we further described the mode of action of five anti-HMPV candidates with low in vitro cytotoxicity. Two entry inhibitors, Evans Blue and aurintricarboxylic acid, and three post-entry inhibitors, mycophenolic acid, mycophenolate mofetil, and 2,3,4-trihydroxybenzaldehyde, were identified. Among them, the mycophenolic acid series displayed the highest levels of inhibition, due to the blockade of intracellular guanosine synthesis. Importantly, MPA has significant potential for drug repurposing as inhibitory levels are achieved below the approved human oral dose. Our drug-repurposing strategy proved to be useful for the rapid discovery of novel hit candidates to treat HMPV infection and provide promising novel templates for drug design.


Subject(s)
Metapneumovirus , Paramyxoviridae Infections , Infant , Humans , Aged , Drug Repositioning , Mycophenolic Acid , Evans Blue/therapeutic use , Aurintricarboxylic Acid/therapeutic use , Paramyxoviridae Infections/drug therapy , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Guanosine/therapeutic use
3.
Antiviral Res ; 207: 105405, 2022 11.
Article in English | MEDLINE | ID: mdl-36084851

ABSTRACT

Human metapneumoviruses have emerged in the past decades as an important global pathogen that causes severe upper and lower respiratory tract infections. Children under the age of 2, the elderly and immunocompromised individuals are more susceptible to HMPV infection than the general population due to their suboptimal immune system. Despite the recent discovery of HMPV as a novel important respiratory virus, reports have rapidly described its epidemiology, biology, and pathogenesis. However, progress is still to be made in the development of vaccines and drugs against HMPV infection as none are currently available. Herein, we discuss the importance of HMPV and review the reported strategies for anti-HMPV drug candidates. We also present the fusion protein as a promising antiviral drug target due to its multiple roles in the HMPV lifecycle. This key viral protein has previously been targeted by a range of inhibitors, which will be discussed as they represent opportunities for future drug design.


Subject(s)
Metapneumovirus , Paramyxoviridae Infections , Respiratory Tract Infections , Aged , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Child , Humans , Paramyxoviridae Infections/drug therapy , Respiratory Tract Infections/prevention & control , Viral Proteins/genetics
4.
Micromachines (Basel) ; 12(10)2021 Sep 24.
Article in English | MEDLINE | ID: mdl-34683202

ABSTRACT

This paper reports the design, development, and testing of a novel, yet simple and low-cost portable device for the rapid detection of SARS-CoV-2. The device performs loop mediated isothermal amplification (LAMP) and provides visually distinguishable images of the fluorescence emitted from the samples. The device utilises an aluminium block embedded with a cartridge heater for isothermal heating of the sample and a single-board computer and camera for fluorescence detection. The device demonstrates promising results within 20 min using clinically relevant starting concentrations of the synthetic template. Time-to-signal data for this device are considerably lower compared to standard quantitative Polymerase Chain Reaction(qPCR) machine (~10-20 min vs. >38 min) for 1 × 102 starting template copy number. The device in its fully optimized and characterized state can potentially be used as simple to operate, rapid, sensitive, and inexpensive platform for population screening as well as point-of-need severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2) detection and patient management.

5.
Clin Transl Immunology ; 10(9): e1337, 2021.
Article in English | MEDLINE | ID: mdl-34527244

ABSTRACT

OBJECTIVES: The upper respiratory tract is the major entry site for Streptococcus pyogenes and influenza virus. Vaccine strategies that activate mucosal immunity could significantly reduce morbidity and mortality because of these pathogens. The severity of influenza is significantly greater if a streptococcal infection occurs during the viraemic period and generally viral infections complicated by a subsequent bacterial infection are known as super-infections. We describe an innovative vaccine strategy against influenza virus:S. pyogenes super-infection. Moreover, we provide the first description of a liposomal multi-pathogen-based platform that enables the incorporation of both viral and bacterial antigens into a vaccine and constitutes a transformative development. METHODS: Specifically, we have explored a vaccination strategy with biocompatible liposomes that express conserved streptococcal and influenza A virus B-cell epitopes on their surface and contain encapsulated diphtheria toxoid as a source of T-cell help. The vaccine is adjuvanted by inclusion of the synthetic analogue of monophosphoryl lipid A, 3D-PHAD. RESULTS: We observe that this vaccine construct induces an Immunoglobulin A (IgA) response in both mice and ferrets. Vaccination reduces viral load in ferrets from influenza challenge and protects mice from both pathogens. Notably, vaccination significantly reduces both mortality and morbidity associated with a super-infection. CONCLUSION: The vaccine design is modular and could be adapted to include B-cell epitopes from other mucosal pathogens where an IgA response is required for protection.

6.
Chem Commun (Camb) ; 57(38): 4666-4669, 2021 May 11.
Article in English | MEDLINE | ID: mdl-33977992

ABSTRACT

We determine that the substitution-inert polynuclear platinum complex (PPC) TriplatinNC is an antiviral agent and protects cells from enterovirus 71 and human metapneumovirus infection. This protection occurs through the formation of adducts with cell-surface glycosaminoglycans. Our detailed mechanistic investigation demonstrates that TriplatinNC blocks viral entry by shielding cells from virus attack, opening new directions for metalloshielding antiviral drug development.


Subject(s)
Antiviral Agents/pharmacology , Organoplatinum Compounds/pharmacology , Paramyxoviridae Infections/drug therapy , Antiviral Agents/chemistry , Cell Line , Dose-Response Relationship, Drug , Humans , Molecular Structure , Organoplatinum Compounds/chemistry
7.
Ann Hematol ; 100(7): 1723-1732, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33942128

ABSTRACT

Data on clinical use of ponatinib are limited. This prospective registry aimed to evaluate outcomes of ponatinib treatment in routine practice over 3 years (2016-2019) in Belgium (NCT03678454). Patients with chronic myeloid leukemia (CML) or Philadelphia chromosome-positive (Ph+) acute lymphoblastic leukemia (ALL) were treated with ponatinib per current label. Fifty patients (33 CML and 17 Ph+ ALL) were enrolled. Fifty-five percent of CML and 29% of Ph+ ALL patients had received ≥3 prior tyrosine kinase inhibitors (TKIs). Reasons for starting ponatinib were intolerance (40%), relapse or refractoriness (28%) to previous TKIs, progression (16%), or T315I mutation (16%). Median follow-up was 15 months for CML and 4.5 months for Ph+ ALL patients. Best response was a major molecular response in 58% of CML and 41% of Ph+ ALL patients. Of 20 patients who started ponatinib due to intolerance to previous TKIs, 9 (64%) CML and 4 (67%) Ph+ ALL achieved a major molecular response. Three-year estimates of overall survival were 85.3% and 85.6%, respectively, in CML and Ph+ ALL patients; estimated progression-free survival was 81.6% and 48.9%. Adverse reactions were reported in 34 patients (68%); rash (26%) and dry skin (10%) were most common. Reported cardiovascular adverse reactions included vascular stenosis (3), arterial hypertension (2), chest pain (1), palpitations (1), and vascular occlusion (1). This Belgian registry confirms results from the PACE clinical trial and supports routine ponatinib use in CML and Ph+ ALL patients who are resistant or intolerant to previous TKIs or with the T315I mutation.


Subject(s)
Antineoplastic Agents/therapeutic use , Imidazoles/therapeutic use , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Protein Kinase Inhibitors/therapeutic use , Pyridazines/therapeutic use , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/adverse effects , Belgium , Cardiovascular Diseases/chemically induced , Drug Eruptions/etiology , Drug Substitution , Female , Follow-Up Studies , Fusion Proteins, bcr-abl/antagonists & inhibitors , Humans , Ichthyosis/chemically induced , Imidazoles/adverse effects , Kaplan-Meier Estimate , Male , Middle Aged , Progression-Free Survival , Prospective Studies , Protein Kinase Inhibitors/adverse effects , Pyridazines/adverse effects , Registries , Salvage Therapy , Treatment Outcome , Young Adult
8.
mBio ; 12(2)2021 03 30.
Article in English | MEDLINE | ID: mdl-33785634

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a recently emerged virus that causes coronavirus infectious disease 2019 (COVID-19). SARS-CoV-2 spike protein, like SARS-CoV-1, uses the angiotensin converting enzyme 2 (ACE2) as a cellular receptor to initiate infection. Compounds that interfere with the SARS-CoV-2 spike protein receptor binding domain protein (RBD)-ACE2 receptor interaction may function as entry inhibitors. Here, we used a dual strategy of molecular docking and surface plasmon resonance (SPR) screening of compound libraries to identify those that bind to human ACE2 or the SARS-CoV-2 spike protein receptor binding domain (RBD). Molecular modeling screening interrogated 57,641 compounds and focused on the region of ACE2 that is engaged by RBD of the SARS-CoV-2 spike glycoprotein and vice versa. SPR screening used immobilized human ACE2 and SARS-CoV-2 Spike protein to evaluate the binding of these proteins to a library of 3,141 compounds. These combined screens identified compounds from these libraries that bind at KD (equilibrium dissociation constant) <3 µM affinity to their respective targets, 17 for ACE2 and 6 for SARS-CoV-2 RBD. Twelve ACE2 binders and six of the RBD binders compete with the RBD-ACE2 interaction in an SPR-based competition assay. These compounds included registered drugs and dyes used in biomedical applications. A Vero-E6 cell-based SARS-CoV-2 infection assay was used to evaluate infection blockade by candidate entry inhibitors. Three compounds demonstrated dose-dependent antiviral in vitro potency-Evans blue, sodium lifitegrast, and lumacaftor. This study has identified potential drugs for repurposing as SARS-CoV-2 entry inhibitors or as chemical scaffolds for drug development.IMPORTANCE SARS-CoV-2, the causative agent of COVID-19, has caused more than 60 million cases worldwide with almost 1.5 million deaths as of November 2020. Repurposing existing drugs is the most rapid path to clinical intervention for emerging diseases. Using an in silico screen of 57,641 compounds and a biophysical screen of 3,141 compounds, we identified 22 compounds that bound to either the angiotensin converting enzyme 2 (ACE2) and/or the SARS-CoV-2 spike protein receptor binding domain (SARS-CoV-2 spike protein RBD). Nine of these drugs were identified by both screening methods. Three of the identified compounds, Evans blue, sodium lifitegrast, and lumacaftor, were found to inhibit viral replication in a Vero-E6 cell-based SARS-CoV-2 infection assay and may have utility as repurposed therapeutics. All 22 identified compounds provide scaffolds for the development of new chemical entities for the treatment of COVID-19.


Subject(s)
Angiotensin-Converting Enzyme 2/metabolism , Antiviral Agents/pharmacology , COVID-19 Drug Treatment , Spike Glycoprotein, Coronavirus/metabolism , Virus Attachment/drug effects , Virus Replication/drug effects , Aminopyridines/pharmacology , Animals , Benzodioxoles/pharmacology , Cell Line , Chlorocebus aethiops , Drug Evaluation, Preclinical , Drug Repositioning , Evans Blue/pharmacology , Humans , Molecular Docking Simulation , Phenylalanine/analogs & derivatives , Phenylalanine/pharmacology , Protein Binding/drug effects , SARS-CoV-2/drug effects , SARS-CoV-2/physiology , Sulfones/pharmacology , Surface Plasmon Resonance , Vero Cells
9.
Acta Clin Belg ; 76(2): 98-105, 2021 Apr.
Article in English | MEDLINE | ID: mdl-31551014

ABSTRACT

Objectives: Currently, there is no standard treatment for patients with acute myeloid leukaemia (AML) ineligible for standard induction chemotherapy (IC). This study aimed to report real-world evidence data on the efficacy and safety of decitabine in this patient group.Methods: This study was a Belgian, retrospective, non-interventional, multicentre registry of patients ≥ 65 years, with newly-diagnosed de novo or secondary AML ineligible for IC. Patients were treated according to routine clinical practice. Overall survival (OS), progression-free survival (PFS) and transfusion independence for ≥8 consecutive weeks were evaluated.Results: Forty-five patients were enrolled, including 67% (n = 30) with secondary AML. Median OS and PFS were 7.3 months (95% CI: 2.2-11.1) and 4.1 months (95% CI: 2.1-7.6) respectively. A subpopulation analysis showed that patients treated with ≥4 cycles (n = 21) had significantly better outcomes compared to patients receiving <4 cycles (median OS 17.5 vs 1.6 months; median PFS 17.5 vs. 1.4 months). Twenty-five percent and 58% of patients that were respectively RBC or platelet transfusion-dependent at baseline became transfusion independent during treatment.Conclusion: This real-world data confirms that decitabine can lead to transfusion independence and longer OS in AML patients, particularly after administering ≥4 cycles, as indicated in the summary of product characteristics.


Subject(s)
Antimetabolites, Antineoplastic , Leukemia, Myeloid, Acute , Aged , Antimetabolites, Antineoplastic/therapeutic use , Azacitidine/therapeutic use , Belgium/epidemiology , Decitabine/therapeutic use , Humans , Leukemia, Myeloid, Acute/drug therapy , Registries , Retrospective Studies , Treatment Outcome
10.
ACS Infect Dis ; 5(10): 1708-1717, 2019 10 11.
Article in English | MEDLINE | ID: mdl-31307190

ABSTRACT

Enterovirus 71 (EV71) is a major etiological agent of hand, foot, and mouth disease, for which there is no antiviral therapy. We have developed densely sulfated disaccharide heparan sulfate (HS) analogues that are potent small molecule inhibitors of EV71 infection, binding to the viral capsid and acting as decoy receptors to block early events of virus replication. The simplified structures, more potent than defined HS disaccharides and with no significant anticoagulant activity, offer promise as anti-EV71 agents.


Subject(s)
Antiviral Agents/pharmacology , Enterovirus A, Human/drug effects , Heparitin Sulfate/analogs & derivatives , Heparitin Sulfate/pharmacology , Cell Line , Dose-Response Relationship, Drug , Enterovirus Infections/drug therapy , Humans , Somatomedins , Virus Attachment/drug effects , Virus Replication/drug effects
11.
Antiviral Res ; 167: 89-97, 2019 07.
Article in English | MEDLINE | ID: mdl-30951732

ABSTRACT

Human parainfluenza viruses cause acute respiratory tract infections and disease predominantly in young children and immunocompromised individuals. Currently, there are no vaccines to prevent hPIV infections, nor licensed anti-hPIV drugs. There is therefore a need for specific antiviral therapies to decrease the morbidity and mortality associated with hPIV diseases. Haemagglutinin-neuraminidase (HN) is one of two hPIV surface proteins with critical roles in host receptor recognition, binding and cleavage; it has been explored as a key drug development target for the past few decades with variable success. Recent advancements in computational modelling and the availability of the X-ray crystal structure of hPIV3 HN have improved our understanding of the structural and mechanistic features of HN. This review explores structural features of the HN protein that are being exploited for structure-guided inhibitor design. We describe past and present hPIV HN inhibition strategies based on sialic acid scaffolds, together with other novel approaches that decrease hPIV infectivity. Although many HN inhibitors have been developed and evaluated as anti-hPIV agents, currently only a host-directed therapy (DAS181) has succeeded in phase II clinical drug trials. Hence, the review concludes with future considerations for targeting the specific function(s) of hPIV HN and suggestions for antiviral drug design.


Subject(s)
Enzyme Inhibitors/pharmacology , HN Protein , N-Acetylneuraminic Acid/analogs & derivatives , Neuraminidase/antagonists & inhibitors , Paramyxoviridae Infections/drug therapy , Antiviral Agents/chemical synthesis , Antiviral Agents/pharmacology , Child , Child, Preschool , Drug Delivery Systems/methods , Drug Design , Drug Resistance, Viral/drug effects , Enzyme Inhibitors/chemical synthesis , Genome, Viral , HN Protein/chemistry , HN Protein/genetics , HN Protein/metabolism , Humans , Immunocompromised Host , N-Acetylneuraminic Acid/chemical synthesis , N-Acetylneuraminic Acid/pharmacology , Parainfluenza Virus 1, Human/drug effects , Parainfluenza Virus 1, Human/genetics , Parainfluenza Virus 3, Human/drug effects , Parainfluenza Virus 3, Human/genetics , Paramyxoviridae Infections/pathology , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Internalization/drug effects
12.
ChemMedChem ; 13(8): 785-789, 2018 04 23.
Article in English | MEDLINE | ID: mdl-29453852

ABSTRACT

Influenza virus infection continues to cause significant, often severe, respiratory illness worldwide. A validated target for the development of anti-influenza agents is the virus surface protein sialidase. In the current study, we have discovered a highly potent inhibitor of influenza virus sialidase, based on a novel sialosyl sulfonate template. The synthesised 3-guanidino sialosyl α-sulfonate, a sulfonozanamivir analogue, inhibits viral replication in vitro at the nanomolar level, comparable to that of the anti-influenza drug zanamivir. Using protein X-ray crystallography we show that the sialosyl α-sulfonate template binds within the sialidase active site in a 1 C4 chair conformation. The C1-sulfonate moiety forms crucial and strong-binding interactions with the active site's triarginyl cluster, while the 3-guanidino moiety interacts significantly with conserved active site residues. This sulfonozanamivir analogue provides a new direction in anti-influenza virus drug development.


Subject(s)
Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Arylsulfonates/chemistry , Arylsulfonates/pharmacology , Influenza A virus/drug effects , Zanamivir/analogs & derivatives , Zanamivir/pharmacology , Catalytic Domain/drug effects , Crystallography, X-Ray , Humans , Influenza A virus/chemistry , Influenza A virus/enzymology , Influenza A virus/physiology , Influenza, Human/drug therapy , Influenza, Human/virology , Molecular Docking Simulation , Neuraminidase/antagonists & inhibitors , Neuraminidase/chemistry , Neuraminidase/metabolism , Virus Replication/drug effects
13.
Org Biomol Chem ; 15(25): 5249-5253, 2017 Jun 27.
Article in English | MEDLINE | ID: mdl-28540971

ABSTRACT

A new direction for influenza virus sialidase inhibitor development was identified using a sulfonate congener of 2-deoxy-2-ß-H N-acetylneuraminic acid. Sialosyl sulfonates can be synthesised efficiently in four steps from N-acetylneuraminic acid via a microwave assisted decarboxylation. The presence of the sulfonate group significantly increases inhibition of influenza virus sialidase and viral infection when compared to the carboxylate congener, and also to the benchmark sialidase inhibitor 2,3-dehydro-2-deoxy-N-acetylneuraminic acid, Neu5Ac2en.


Subject(s)
Antiviral Agents/pharmacology , Arylsulfonates/pharmacology , Enzyme Inhibitors/pharmacology , Influenza A virus/drug effects , Neuraminidase/antagonists & inhibitors , Virus Replication/drug effects , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Arylsulfonates/chemical synthesis , Arylsulfonates/chemistry , Carbohydrate Conformation , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Neuraminidase/metabolism , Structure-Activity Relationship
14.
Sci Rep ; 6: 24138, 2016 Apr 07.
Article in English | MEDLINE | ID: mdl-27053240

ABSTRACT

Human parainfluenza type-3 virus (hPIV-3) is one of the principal aetiological agents of acute respiratory illness in infants worldwide and also shows high disease severity in the elderly and immunocompromised, but neither therapies nor vaccines are available to treat or prevent infection, respectively. Using a multidisciplinary approach we report herein that the approved drug suramin acts as a non-competitive in vitro inhibitor of the hPIV-3 haemagglutinin-neuraminidase (HN). Furthermore, the drug inhibits viral replication in mammalian epithelial cells with an IC50 of 30 µM, when applied post-adsorption. Significantly, we show in cell-based drug-combination studies using virus infection blockade assays, that suramin acts synergistically with the anti-influenza virus drug zanamivir. Our data suggests that lower concentrations of both drugs can be used to yield high levels of inhibition. Finally, using NMR spectroscopy and in silico docking simulations we confirmed that suramin binds HN simultaneously with zanamivir. This binding event occurs most likely in the vicinity of the protein primary binding site, resulting in an enhancement of the inhibitory potential of the N-acetylneuraminic acid-based inhibitor. This study offers a potentially exciting avenue for the treatment of parainfluenza infection by a combinatorial repurposing approach of well-established approved drugs.


Subject(s)
Epithelial Cells/drug effects , Parainfluenza Virus 3, Human/drug effects , Suramin/pharmacology , Zanamivir/pharmacology , Animals , Antiviral Agents/metabolism , Antiviral Agents/pharmacology , Binding Sites , Cell Line , Drug Synergism , Epithelial Cells/virology , HN Protein/metabolism , Host-Pathogen Interactions/drug effects , Kidney/cytology , Kinetics , Macaca mulatta , Magnetic Resonance Spectroscopy , Molecular Dynamics Simulation , Parainfluenza Virus 3, Human/metabolism , Parainfluenza Virus 3, Human/physiology , Protein Binding , Suramin/metabolism , Viral Proteins/antagonists & inhibitors , Viral Proteins/metabolism , Virus Replication/drug effects , Zanamivir/metabolism
15.
Exp Gerontol ; 61: 105-12, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25446500

ABSTRACT

BACKGROUND/OBJECTIVES: Geriatric patients are highly susceptible to infections. While reduced lymphocyte count has been associated with age, other studies found no change in WBC counts with age. Increased circulating white blood cell (WBC) count has been associated with cardiovascular (CV) diseases and frailty but there are discrepancies. Frailty, geriatric conditions, cardiovascular diseases and WBC count have also been associated with low grade inflammation. Association between geriatric conditions and WBC has been scarcely studied. The aim of the study is to assess the association between WBC and geriatric conditions, CV diseases, and seric IL-6 levels. DESIGN, SETTING, PARTICIPANTS, MEASUREMENTS: We recruited 100 subjects in the general population and hospitalized for chronic medical conditions (age, 23-96years). We collected information on clinical status (medical history, comorbidities, treatments and geriatric syndromes), biological parameters (hematological tests, cytomegalovirus serology) and cytokine production (basal IL-6). Using stepwise backward multivariate analyses, we defined which set of clinical and biological variables could be predictive of increased total and differential WBC counts. RESULTS: We found that low-grade inflammation is independently associated with total WBC, monocyte and neutrophil counts, but not geriatric conditions. CV diseases were the only significant associated factor for high monocyte count. CONCLUSION: In this study, we observed that differential and total WBC counts do not seem to be associated with geriatric conditions but with CV diseases, low-grade inflammation and telomere length.


Subject(s)
Aging/immunology , Cardiovascular Diseases/etiology , Interleukin-6/blood , Leukocyte Count , Telomere , Adult , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Monocytes/immunology , Neutrophils/immunology
16.
Nat Commun ; 5: 5268, 2014 Oct 20.
Article in English | MEDLINE | ID: mdl-25327774

ABSTRACT

Human parainfluenza viruses (hPIVs) cause upper and lower respiratory tract disease in children that results in a significant number of hospitalizations and impacts health systems worldwide. To date, neither antiviral drugs nor vaccines are approved for clinical use against parainfluenza virus, which reinforces the urgent need for new therapeutic discovery strategies. Here we use a multidisciplinary approach to develop potent inhibitors that target a structural feature within the hPIV type 3 haemagglutinin-neuraminidase (hPIV-3 HN). These dual-acting designer inhibitors represent the most potent designer compounds and efficiently block both hPIV cell entry and virion progeny release. We also define the binding mode of these inhibitors in the presence of whole-inactivated hPIV and recombinantly expressed hPIV-3 HN by Saturation Transfer Difference NMR spectroscopy. Collectively, our study provides an antiviral preclinical candidate and a new direction towards the discovery of potential anti-parainfluenza drugs.


Subject(s)
Antiviral Agents/chemistry , Drug Design , Hemagglutinins, Viral/chemistry , Neuraminidase/antagonists & inhibitors , Parainfluenza Virus 3, Human , Catalytic Domain , Cell Line, Tumor , Computer Simulation , Crystallography, X-Ray , Enzyme Inhibitors/chemistry , Enzyme-Linked Immunosorbent Assay , Epitope Mapping , Humans , Inhibitory Concentration 50 , Magnetic Resonance Spectroscopy , Molecular Conformation , Molecular Dynamics Simulation , Neuraminic Acids/chemistry , Recombinant Proteins/chemistry , Solvents/chemistry , Surface Properties
17.
Emerg Microbes Infect ; 3(9): e62, 2014 Sep.
Article in English | MEDLINE | ID: mdl-26038755

ABSTRACT

Enterovirus 71 (EV71) causes severe central nervous system infections, leading to cardiopulmonary complications and death in young children. There is an urgent unmet medical need for new pharmaceutical agents to control EV71 infections. Using a multidisciplinary approach, we found that the approved pediatric antiparasitic drug suramin blocked EV71 infectivity by a novel mechanism of action that involves binding of the naphtalentrisulonic acid group of suramin to the viral capsid. Moreover, we demonstrate that when suramin is used in vivo at doses equivalent to or lower than the highest dose already used in humans, it significantly decreased mortality in mice challenged with a lethal dose of EV71 and peak viral load in adult rhesus monkeys. Thus, suramin inhibits EV71 infection by neutralizing virus particles prior to cell attachment. Consequently, these findings identify suramin as a clinical candidate for further development as a therapeutic or prophylactic treatment for severe EV71 infection.

SELECTION OF CITATIONS
SEARCH DETAIL
...