Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters










Publication year range
1.
Sci Rep ; 14(1): 13397, 2024 06 11.
Article in English | MEDLINE | ID: mdl-38862576

ABSTRACT

Severe fever with thrombocytopenia syndrome virus (SFTSV) is an emerging tick-borne virus with a mortality rate of up to 30%. First identified in China in 2009, it was later reported in other Asian countries, including Thailand in 2020. SFTSV has been detected in several tick species, including Rhipicephalus sanguineus, known for infesting dogs. We conducted a seroprevalence study of SFTSV in Bangkok and Nong Khai, Thailand, by analyzing 1162 human samples collected between 2019 and 2023. The testing method relied on IgG detection using ELISA and confirmed though a virus seroneutralization test. The results indicated that out of the participants, 12 (1.1%) tested positive for anti-SFTSV IgG antibodies; however, none exhibited positive results in the seroneutralization assay. Additionally, molecular detection of SFTSV, Crimean-Congo hemorrhagic fever (CCHF), Coxiella spp., Bartonella spp., and Rickettsia spp. was performed on 433 Rh. sanguineus ticks collected from 49 dogs in 2023 in Chachoengsao Province, Thailand. No evidence of these pathogens was found in ticks. These findings highlight the importance of exploring viral cross-reactivity. Furthermore, it is important to conduct additional studies to isolate SFTSV from animals and ticks in order to identify the potential transmission routes contributing to human and animal infections in Thailand.


Subject(s)
Phlebovirus , Rhipicephalus sanguineus , Severe Fever with Thrombocytopenia Syndrome , Animals , Thailand/epidemiology , Seroepidemiologic Studies , Rhipicephalus sanguineus/virology , Humans , Phlebovirus/genetics , Phlebovirus/immunology , Phlebovirus/isolation & purification , Middle Aged , Female , Male , Adult , Severe Fever with Thrombocytopenia Syndrome/epidemiology , Severe Fever with Thrombocytopenia Syndrome/virology , Severe Fever with Thrombocytopenia Syndrome/veterinary , Severe Fever with Thrombocytopenia Syndrome/diagnosis , Dogs , Aged , Adolescent , Antibodies, Viral/blood , Young Adult , Child , Child, Preschool , Aged, 80 and over , Infant , Immunoglobulin G/blood
2.
Biomed Pharmacother ; 177: 116988, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38897157

ABSTRACT

Therapeutic monoclonal antibodies have been successful in protecting vulnerable populations against SARS-CoV-2. However, their effectiveness has been hampered by the emergence of new variants. To adapt the therapeutic landscape, health authorities have based their recommendations mostly on in vitro neutralization tests. However, these do not provide a reliable understanding of the changes in the dose-effect relationship and how they may translate into clinical efficacy. Taking the example of EvusheldTM (AZD7442), we aimed to investigate how in vivo data can provide critical quantitative results and project clinical effectiveness. We used the Golden Syrian hamster model to estimate 90 % effective concentrations (EC90) of AZD7442 in vivo against SARS-CoV-2 Omicron BA.1, BA.2 and BA.5 variants. While our in vivo results confirmed the partial loss of AZD7442 activity for BA.1 and BA.2, they showed a much greater loss of efficacy against BA.5 than that obtained in vitro. We analyzed in vivo EC90s in perspective with antibody levels measured in a cohort of immunocompromised patients who received 300 mg of AZD7442. We found that a substantial proportion of patients had serum levels of anti-SARS-CoV-2 spike protein IgG above the estimated in vivo EC90 for BA.1 and BA.2 (21 % and 92 % after 1 month, respectively), but not for BA.5. These findings suggest that AZD7442 is likely to retain clinical efficacy against BA.2 and BA.1, but not against BA.5. Overall, the present study illustrates the importance of complementing in vitro investigations by preclinical studies in animal models to help predict the efficacy of monoclonal antibodies in humans.

3.
Emerg Microbes Infect ; 13(1): 2307510, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38240255

ABSTRACT

Serological studies of COVID-19 convalescent patients have identified polyclonal lineage-specific and cross-reactive antibodies (Abs), with varying effector functions against virus variants. Individual specificities of anti-SARS-CoV-2 Abs and their impact on infectivity by other variants have been little investigated to date. Here, we dissected at a monoclonal level neutralizing and enhancing Abs elicited by early variants and how they affect infectivity of emerging variants. B cells from 13 convalescent patients originally infected by D614G or Alpha variants were immortalized to isolate 445 naturally-produced anti-SARS-CoV-2 Abs. Monoclonal antibodies (mAbs) were tested for their abilities to impact the cytopathic effect of D614G, Delta, and Omicron (BA.1) variants. Ninety-eight exhibited robust neutralization against at least one of the three variant types, while 309 showed minimal or no impact on infectivity. Thirty-eight mAbs enhanced infectivity of SARS-CoV-2. Infection with D614G/Alpha variants generated variant-specific (65 neutralizing Abs, 35 enhancing Abs) and cross-reactive (18 neutralizing Abs, 3 enhancing Abs) mAbs. Interestingly, among the neutralizing mAbs with cross-reactivity restricted to two of the three variants tested, none demonstrated specific neutralization of the Delta and Omicron variants. In contrast, cross-reactive mAbs enhancing infectivity (n = 3) were found exclusively specific to Delta and Omicron variants. Notably, two mAbs that amplified in vitro the cytopathic effect of the Delta variant also exhibited neutralization against Omicron. These findings shed light on functional diversity of cross-reactive Abs generated during SARS-CoV-2 infection and illustrate how the balance between neutralizing and enhancing Abs facilitate variant emergence.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , Antibodies, Blocking , Antibodies, Neutralizing , Antibodies, Monoclonal , Antibodies, Viral , Spike Glycoprotein, Coronavirus
4.
PLoS Pathog ; 18(9): e1010799, 2022 09.
Article in English | MEDLINE | ID: mdl-36067253

ABSTRACT

The binding of the SARS-CoV-2 spike to angiotensin-converting enzyme 2 (ACE2) promotes virus entry into the cell. Targeting this interaction represents a promising strategy to generate antivirals. By screening a phage-display library of biosynthetic protein sequences build on a rigid alpha-helicoidal HEAT-like scaffold (named αReps), we selected candidates recognizing the spike receptor binding domain (RBD). Two of them (F9 and C2) bind the RBD with affinities in the nM range, displaying neutralisation activity in vitro and recognizing distinct sites, F9 overlapping the ACE2 binding motif. The F9-C2 fusion protein and a trivalent αRep form (C2-foldon) display 0.1 nM affinities and EC50 of 8-18 nM for neutralization of SARS-CoV-2. In hamsters, F9-C2 instillation in the nasal cavity before or during infections effectively reduced the replication of a SARS-CoV-2 strain harbouring the D614G mutation in the nasal epithelium. Furthermore, F9-C2 and/or C2-foldon effectively neutralized SARS-CoV-2 variants (including delta and omicron variants) with EC50 values ranging from 13 to 32 nM. With their high stability and their high potency against SARS-CoV-2 variants, αReps provide a promising tool for SARS-CoV-2 therapeutics to target the nasal cavity and mitigate virus dissemination in the proximal environment.


Subject(s)
Angiotensin-Converting Enzyme 2 , COVID-19 Drug Treatment , Recombinant Fusion Proteins , Spike Glycoprotein, Coronavirus , Angiotensin-Converting Enzyme 2/chemistry , Angiotensin-Converting Enzyme 2/metabolism , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Humans , Peptidyl-Dipeptidase A/metabolism , Protein Binding , Recombinant Fusion Proteins/pharmacology , Recombinant Fusion Proteins/therapeutic use , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , SARS-CoV-2/genetics , Spike Glycoprotein, Coronavirus/metabolism
5.
Sci Rep ; 12(1): 12609, 2022 07 23.
Article in English | MEDLINE | ID: mdl-35871089

ABSTRACT

The replacement of the Omicron BA.1 variant of SARS-CoV-2 by the BA.2 and the rapid growth of the BA.5 sub lineage, which have both different sets of mutations in the spike glycoprotein, alters the spectrum of activity of therapeutic antibodies currently licensed in the European Union. Using clinical strains of the Omicron BA.2 and BA.5 variants, we compared the neutralising power of monoclonal antibodies against the Omicron BA.1, BA.2 and BA.5 variants, using an ancestral strain (lineage B.1, D614G) and a Delta variant strain as reference. Sotrovimab/Vir-7831 is less active against BA.2 than against BA.1 (fold change reduction ~ 1,4) and even less active against BA.5 (fold change reduction ~ 2.7). Within the Evusheld /AZD7442 cocktail, Cilgavimab/AZD1061 is more active against BA.2 and BA.5 than against BA.1 (fold change increase ~ 32), whilst the very low activity of Tixagevimab/AZD8895 against BA.1 is not enhanced against BA.2 nor BA.5. In total, compared to BA.1, the activity of the Evusheld/AZD7442 is significantly improved against BA.2 while BA.5 is intermediate but closer to BA.2.


Subject(s)
COVID-19 Drug Treatment , Spike Glycoprotein, Coronavirus , Antibodies, Monoclonal , Antibodies, Monoclonal, Humanized , Antibodies, Neutralizing , Antibodies, Viral , Drug Combinations , Humans , SARS-CoV-2/genetics , Spike Glycoprotein, Coronavirus/genetics
6.
Commun Biol ; 5(1): 225, 2022 03 10.
Article in English | MEDLINE | ID: mdl-35273335

ABSTRACT

Late 2020, SARS-CoV-2 Alpha variant emerged in United Kingdom and gradually replaced G614 strains initially involved in the global spread of the pandemic. In this study, we use a Syrian hamster model to compare a clinical strain of Alpha variant with an ancestral G614 strain. The Alpha variant succeed to infect animals and to induce a pathology that mimics COVID-19. However, both strains replicate to almost the same level and induced a comparable disease and immune response. A slight fitness advantage is noted for the G614 strain during competition and transmission experiments. These data do not corroborate the epidemiological situation observed during the first half of 2021 in humans nor reports that showed a more rapid replication of Alpha variant in human reconstituted bronchial epithelium. This study highlights the need to combine data from different laboratories using various animal models to decipher the biological properties of newly emerging SARS-CoV-2 variants.


Subject(s)
COVID-19 , Disease Models, Animal , Mesocricetus , SARS-CoV-2/physiology , Animals , Antibodies, Neutralizing/blood , COVID-19/blood , COVID-19/immunology , COVID-19/virology , Cytokines/genetics , Female , Gastrointestinal Tract/virology , Genome, Viral , Lung/virology , Nasal Lavage Fluid/virology , SARS-CoV-2/genetics , Virus Replication
7.
Sci Rep ; 12(1): 4683, 2022 03 18.
Article in English | MEDLINE | ID: mdl-35304531

ABSTRACT

The emergence and rapid spread of the Omicron variant of SARS-CoV-2, which has more than 30 substitutions in the spike glycoprotein, compromises the efficacy of currently available vaccines and therapeutic antibodies. Using a clinical strain of the Omicron variant, we analyzed the neutralizing power of eight currently used monoclonal antibodies compared to the ancestral B.1 BavPat1 D614G strain. We observed that six of these antibodies have lost their ability to neutralize the Omicron variant. Of the antibodies still having neutralizing activity, Sotrovimab/Vir-7831 shows the smallest reduction in activity, with a factor change of 3.1. Cilgavimab/AZD1061 alone shows a reduction in efficacy of 15.8, resulting in a significant loss of activity for the Evusheld cocktail (42.6-fold reduction) in which the other antibody, Tixagevimab, does not retain significant activity against Omicron. Our results suggest that the clinical efficacy of the initially proposed doses should be rapidly evaluated and the possible need to modify doses or propose combination therapies should be considered.


Subject(s)
COVID-19 Drug Treatment , Viral Envelope Proteins , Antibodies, Monoclonal , Antibodies, Monoclonal, Humanized , Antibodies, Neutralizing , Antibodies, Viral/therapeutic use , Humans , Membrane Glycoproteins , Neutralization Tests , SARS-CoV-2 , Spike Glycoprotein, Coronavirus
8.
PLoS One ; 16(12): e0260958, 2021.
Article in English | MEDLINE | ID: mdl-34855904

ABSTRACT

SARS-CoV-2 variants are emerging with potential increased transmissibility highlighting the great unmet medical need for new therapies. Niclosamide is a potent anti-SARS-CoV-2 agent that has advanced in clinical development. We validate the potent antiviral efficacy of niclosamide in a SARS-CoV-2 human airway model. Furthermore, niclosamide remains its potency against the D614G, Alpha (B.1.1.7), Beta (B.1.351), and Delta (B.1.617.2) variants. Our data further support the potent anti-SARS-CoV-2 properties of niclosamide and highlights its great potential as a therapeutic agent for COVID-19.


Subject(s)
Antiviral Agents/therapeutic use , COVID-19 Drug Treatment , Niclosamide/therapeutic use , SARS-CoV-2/drug effects , Animals , Caco-2 Cells , Chlorocebus aethiops , Humans , Inhibitory Concentration 50 , Respiratory Mucosa/virology , Vero Cells
9.
iScience ; 24(11): 103329, 2021 Nov 19.
Article in English | MEDLINE | ID: mdl-34697603

ABSTRACT

Since the beginning of the COVID-19 pandemics, variants have emerged. Some of them display increased transmissibility and/or resistance to immune response. Most of the mutations involved in the functional adaptation are found in the receptor-binding motif (RBM), close to the interface with the receptor ACE2. We thus developed a fast molecular assay to detect mutations in the RBM coding sequence. After amplification, the amplicon is heat-denatured and hybridized with an amplicon of reference. The presence of a mutation can be detected using a mismatch-specific endonuclease and the cleavage pattern is analyzed by capillary electrophoresis. The method was validated on RNA of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants produced in vitro before being implemented for clinical samples. The assay showed 97.8% sensitivity and 97.8% specificity. The procedure can be set up for high-throughput identification of the presence of mutations and serve as a first-line screening to select the samples for full genome sequencing.

10.
mBio ; 12(4): e0085021, 2021 08 31.
Article in English | MEDLINE | ID: mdl-34225487

ABSTRACT

Since its emergence in 2019, circulating populations of the new coronavirus (CoV) continuously acquired genetic diversity. At the end of 2020, a variant named 20I/501Y.V1 (lineage B.1.1.7) emerged and replaced other circulating strains in several regions. This phenomenon has been poorly associated with biological evidence that this variant and the original strain exhibit different phenotypic characteristics. Here, we analyze the replication ability of this new variant in different cellular models using for comparison an ancestral D614G European strain (lineage B1). Results from comparative replication kinetics experiments in vitro and in a human reconstituted bronchial epithelium showed no difference. However, when both viruses were put in competition in human reconstituted bronchial epithelium, the 20I/501Y.V1 variant outcompeted the ancestral strain. All together, these findings demonstrate that this new variant replicates more efficiently and may contribute to a better understanding of the progressive replacement of circulating strains by the severe acute respiratory CoV-2 (SARS-CoV-2) 20I/501Y.V1 variant. IMPORTANCE The emergence of several SARS-CoV-2 variants raised numerous questions concerning the future course of the pandemic. We are currently observing a replacement of the circulating viruses by the variant from the United Kingdom known as 20I/501Y.V1, from the B.1.1.7 lineage, but there is little biological evidence that this new variant exhibits a different phenotype. In the present study, we used different cellular models to assess the replication ability of the 20I/501Y.V1 variant. Our results showed that this variant replicates more efficiently in human reconstituted bronchial epithelium, which may explain why it spreads so rapidly in human populations.


Subject(s)
COVID-19/transmission , Genetic Fitness/genetics , SARS-CoV-2/growth & development , SARS-CoV-2/genetics , Virus Replication/genetics , Animals , COVID-19/pathology , Caco-2 Cells , Cell Line , Chlorocebus aethiops , Humans , Respiratory Mucosa/virology , Vero Cells , Viral Load
11.
Molecules ; 26(5)2021 Mar 09.
Article in English | MEDLINE | ID: mdl-33803417

ABSTRACT

A series of hitherto unknown (1,4-disubstituted-1,2,3-triazol)-(E)-2-methyl-but-2-enyl nucleosides phosphonate prodrugs bearing 4-substituted-1,2,3-triazoles were prepared in a straight approach through an olefin acyclic cross metathesis as the key synthetic step. All novel compounds were evaluated for their antiviral activities against HBV, HIV and SARS-CoV-2. Among these molecules, only compound 15j, a hexadecyloxypropyl (HDP)/(isopropyloxycarbonyl-oxymethyl)-ester (POC) prodrug, showed activity against HBV in Huh7 cell cultures with 62% inhibition at 10 µM, without significant cytotoxicity (IC50 = 66.4 µM in HepG2 cells, IC50 = 43.1 µM in HepG2 cells) at 10 µM.


Subject(s)
Antiviral Agents/chemical synthesis , Antiviral Agents/pharmacology , Azo Compounds/chemistry , Nucleosides/chemistry , Organophosphonates/chemistry , Prodrugs/chemical synthesis , Prodrugs/pharmacology , Alkenes/chemistry , Animals , Cell Line, Tumor , Chlorocebus aethiops , HIV-1/drug effects , Hepatitis B virus/drug effects , Humans , Magnetic Resonance Spectroscopy , Methylation , SARS-CoV-2/drug effects , Structure-Activity Relationship , Triazoles/chemistry , Vero Cells
12.
Euro Surveill ; 24(50)2019 12.
Article in English | MEDLINE | ID: mdl-31847946

ABSTRACT

BackgroundNeurotropic arboviruses are increasingly recognised as causative agents of neurological disease in Europe but underdiagnosis is still suspected. Capability for accurate diagnosis is a prerequisite for adequate clinical and public health response.AimTo improve diagnostic capability in EVD-LabNet laboratories, we organised an external quality assessment (EQA) focusing on molecular detection of Toscana (TOSV), Usutu (USUV), West Nile (WNV) and tick-borne encephalitis viruses (TBEV).MethodsSixty-nine laboratories were invited. The EQA panel included two WNV RNA-positive samples (lineages 1 and 2), two TOSV RNA-positive samples (lineages A and B), one TBEV RNA-positive sample (Western subtype), one USUV RNA-positive sample and four negative samples. The EQA focused on overall capability rather than sensitivity of the used techniques. Only detection of one, clinically relevant, concentration per virus species and lineage was assessed.ResultsThe final EQA analysis included 51 laboratories from 35 countries; 44 of these laboratories were from 28 of 31 countries in the European Union/European Economic Area (EU/EEA). USUV diagnostic capability was lowest (28 laboratories in 18 countries), WNV detection capacity was highest (48 laboratories in 32 countries). Twenty-five laboratories were able to test the whole EQA panel, of which only 11 provided completely correct results. The highest scores were observed for WNV and TOSV (92%), followed by TBEV (86%) and USUV (75%).ConclusionWe observed wide variety in extraction methods and RT-PCR tests, showing a profound absence of standardisation across European laboratories. Overall, the results were not satisfactory; capacity and capability need to be improved in 40 laboratories.


Subject(s)
Encephalitis Viruses, Tick-Borne/genetics , Flavivirus/genetics , Laboratories/standards , Molecular Diagnostic Techniques/standards , Quality Assurance, Health Care/standards , Sandfly fever Naples virus/genetics , Encephalitis Viruses, Tick-Borne/isolation & purification , Encephalitis, Tick-Borne/diagnosis , Flavivirus/isolation & purification , Flavivirus Infections/diagnosis , Humans , Phlebotomus Fever/diagnosis , Quality Control , Sandfly fever Naples virus/isolation & purification , West Nile Fever/virology , West Nile virus/immunology
13.
Viruses ; 11(10)2019 10 09.
Article in English | MEDLINE | ID: mdl-31601017

ABSTRACT

Mayaro (MAYV) is an emerging arthropod-borne virus belonging to the Alphavirus genus of the Togaviridae family. Although forest-dwelling Haemagogus mosquitoes have been considered as its main vector, the virus has also been detected in circulating Aedes ssp mosquitoes. Here we assess the susceptibility of Aedes aegypti and Aedes albopictus to infection with MAYV and their innate immune response at an early stage of infection. Aedes albopictus was more susceptible to infection with MAYV than Ae. aegypti. Analysis of transcript levels of twenty immunity-related genes by real-time PCR in the midgut of both mosquitoes infected with MAYV revealed increased expression of several immune genes, including CLIP-domain serine proteases, the anti-microbial peptides defensin A, E, cecropin E, and the virus inducible gene. The regulation of certain genes appeared to be Aedes species-dependent. Infection of Ae. aegypti with MAYV resulted in increased levels of myeloid differentiation2-related lipid recognition protein (ML26A) transcripts, as compared to Ae. albopictus. Increased expression levels of thio-ester-containing protein 22 (TEP22) and Niemann-Pick type C1 (NPC1) gene transcripts were observed in infected Ae. albopictus, but not Ae. aegypti. The differences in these gene expression levels during MAYV infection could explain the variation in susceptibility observed in both mosquito species.


Subject(s)
Aedes/virology , Alphavirus Infections/transmission , Alphavirus/immunology , Immunity, Innate , Aedes/immunology , Animals , Antimicrobial Cationic Peptides/genetics , Antimicrobial Cationic Peptides/metabolism , Gene Expression/immunology , Gene Expression Profiling , Immunity, Innate/genetics , Mosquito Vectors/virology , Real-Time Polymerase Chain Reaction , Serine Proteases/genetics , Serine Proteases/metabolism
14.
Viruses ; 11(9)2019 08 29.
Article in English | MEDLINE | ID: mdl-31470617

ABSTRACT

Mayaro virus (MAYV) is an emerging arthritogenic alphavirus belonging to the Togaviridae family. Infection leads to a dengue-like illness accompanied by severe polyarthralgia. However, the molecular and cellular mechanisms of arthritis as a result of MAYV infection remain poorly understood. In the present study, we assess the susceptibility of human chondrocytes (HC), fibroblast-like synoviocytes and osteoblasts that are the major cell types involved in osteoarthritis, to infection with MAYV. We show that these cells are highly permissive to MAYV infection and that viral RNA copy number and viral titers increase over time in infected cells. Knowing that HC are the primary cells in articular cartilage and are essential for maintaining the cartilaginous matrix, gene expression studies were conducted in MAYV-infected primary HC using polymerase chain reaction (PCR) arrays. The infection of the latter cells resulted in an induction in the expression of several matrix metalloproteinases (MMP) including MMP1, MMP7, MMP8, MMP10, MMP13, MMP14 and MMP15 which could be involved in the destruction of articular cartilage. Infected HC were also found to express significantly increased levels of various IFN-stimulated genes and arthritogenic mediators such as TNF-α and IL-6. In conclusion, MAYV-infected primary HC overexpress arthritis-related genes, which may contribute to joint degradation and pathogenesis.


Subject(s)
Alphavirus Infections/virology , Alphavirus/physiology , Arthritis/genetics , Chondrocytes/virology , Alphavirus/immunology , Alphavirus Infections/genetics , Alphavirus Infections/immunology , Cell Adhesion/genetics , Cell Survival , Cells, Cultured , Chondrocytes/immunology , Cytokines/genetics , Cytokines/metabolism , Extracellular Matrix/genetics , Gene Expression Profiling , Humans , Matrix Metalloproteinases/genetics , Osteoblasts/virology , RNA, Viral/metabolism , Synoviocytes/virology
15.
Antiviral Res ; 168: 109-113, 2019 08.
Article in English | MEDLINE | ID: mdl-31085207

ABSTRACT

Dengue fever is the most widespread of the human arbovirus diseases, with approximately one third of the world's population at risk of infection. Dengue viruses are members of the genus Flavivirus (family Flaviviridae) and, antigenically, they separate as four closely related serotypes (1-4) that share 60-75% amino acid homology. This genetic diversity complicates the process of antiviral drug discovery. Thus, currently no approved dengue-specific therapeutic treatments are available. With the aim of providing an efficient tool for dengue virus drug discovery, a collection of nineteen dengue viruses, representing the genotypic diversity within the four serotypes, was developed. After phylogenetic analysis of the full-length genomes, we selected relevant strains from the EVAg collection at Aix-Marseille University and completed the virus collection, using a reverse genetic system based on the infectious sub-genomic amplicons technique. Finally, we evaluated this dengue virus collection against three published dengue inhibitory compounds. NITD008, which targets the highly conserved active site of the viral NS5 polymerase enzyme, exhibited similar antiviral potencies against each of the different dengue genotypes in the panel. Compounds targeting less conserved protein subdomains, such as the capsid inhibitor ST-148, or SDM25N, a ∂ opioid receptor antagonist which indirectly targets NS4B, exhibited larger differences in potency against the various genotypes of dengue viruses. These results illustrate the importance of a phylogenetically based dengue virus reference panel for dengue antiviral research. The collection developed in this study, which includes such representative dengue viruses, has been made available to the scientific community through the European Virus Archive to evaluate novel DENV antiviral candidates.


Subject(s)
Antiviral Agents/pharmacology , Dengue Virus/drug effects , Dengue Virus/genetics , Dengue Virus/classification , Drug Evaluation, Preclinical , Genetic Variation , Genome, Viral/genetics , Genotype , Humans , Phylogeny , Serogroup , Viral Proteins/antagonists & inhibitors , Viral Proteins/genetics
17.
PLoS Negl Trop Dis ; 12(1): e0006203, 2018 01.
Article in English | MEDLINE | ID: mdl-29377886

ABSTRACT

Few data on dengue epidemiology are available for Lao PDR. Here, we provide information on the complexity of dengue epidemiology in the country, demonstrating dynamic circulation that varies over space and time, according to serotype. We recruited 1,912 consenting patients presenting with WHO dengue criteria at Mahosot Hospital, Vientiane (central Laos), between 2006 and 2010. Between 2008 and 2010, 1,413 patients with undifferentiated fever were also recruited at Luang Namtha (LNT) Provincial Hospital (northern Laos) and 555 at Salavan (SV) Provincial Hospital (southern Laos). We report significant variations in Dengue virus (DENV) circulation between the three sites. Peaks of DENV infection were observed in the rainy seasons, although 11% of confirmed cases in the provinces and 4.6% in the capital were detected during the dry and cool seasons (between December and February). Four DENV serotypes were detected among the 867 RT-PCR positive patients: 76.9% DENV-1, 9.6% DENV-2, 7.7% DENV-4 and 5.3% DENV-3. DENV-1 was the predominant serotype throughout the study except in LNT in 2008 and 2009 when it was DENV-2. Before July 2009, DENV-2 was not detected in SV and only rarely detected in Vientiane. DENV-3 and DENV-4 were commonly detected in Vientiane, before 2008 for DENV-4 and after 2009 for DENV-3. The phylogenetic analyses of DENV envelope sequences suggest concurrent multiple introductions of new strains as well as active DENV circulation throughout Laos and with neighboring countries. It is therefore of great importance to develop and strengthen a year-round nation-wide surveillance network in order to collect data that would allow anticipation of public health issues caused by the occurrence of large dengue outbreaks.


Subject(s)
Dengue Virus/classification , Dengue Virus/isolation & purification , Dengue/epidemiology , Molecular Epidemiology , Serogroup , Adolescent , Adult , Child , Child, Preschool , Dengue Virus/genetics , Epidemiological Monitoring , Humans , Laos/epidemiology , Middle Aged , Phylogeography , Seasons , Topography, Medical , Young Adult
18.
J Clin Microbiol ; 55(11): 3219-3226, 2017 11.
Article in English | MEDLINE | ID: mdl-28835479

ABSTRACT

Zika virus (ZIKV) infections are a significant public health concern. A strong capability for ZIKV detection is an absolute requirement for adequate preparedness and response strategies and individual patient care. The objective of this study was to assess and improve the capability of European expert laboratories for molecular testing for ZIKV through an external quality assessment (EQA) scheme. Laboratories were provided a panel of 12 samples which included negative samples, samples containing African- or Asian-lineage ZIKV at various concentrations (103 to 109 copies/ml), and samples containing dengue virus, yellow fever virus, or chikungunya virus. The results were analyzed on the basis of the outcomes of testing for the samples and the extraction and detection method used. Samples with a ZIKV RNA status scored correctly by >50% of the laboratories were designated the core sample. A total of 85 panel outcomes were submitted by 50 laboratories in 31 countries. The results designated all samples as core samples. Thirty-three percent (28/85) of the panel outcomes identified all samples. Analysis at the laboratory level showed that only 40% of the laboratories (20/50), representing 45% of the countries, scored sufficiently; i.e., they had at least one test operational that scored all core samples correctly. There is a need for improvement of the molecular detection of ZIKV in 60% of the participating laboratories. While the specificity of the tests was more robust, the results of the EQA showed large variation in test sensitivity. Improvements should focus on both nucleic acid extraction and ZIKV detection methods.


Subject(s)
Laboratories , Laboratory Proficiency Testing , Zika Virus Infection/diagnosis , Zika Virus/isolation & purification , Europe , Humans , Sensitivity and Specificity
19.
Genome Announc ; 5(17)2017 Apr 27.
Article in English | MEDLINE | ID: mdl-28450505

ABSTRACT

We report here the complete coding sequences of two strains of dengue virus type 2, isolated in France from patients returning from Burkina Faso in November 2016. Both strains (cosmopolitan genotype) are almost identical (99.91% nucleotide identity) and closely related to a strain circulating in Burkina Faso in 1983.

20.
Sci Rep ; 6: 39384, 2016 12 19.
Article in English | MEDLINE | ID: mdl-27991555

ABSTRACT

Zika virus (ZIKV), a typical example of a re-emerging pathogen, recently caused large outbreaks in Pacific islands and the Americas, associated with congenital diseases and neurological complications. Deciphering the natural history, ecology and pathophysiology of this mosquito-borne pathogen requires effective reverse genetics tools. In the current study, using the bacterium-free 'Infectious Subgenomic Amplicons' (ISA) method, we generated and made available to the scientific community via the non-profit European Virus Archive collection, two simple and performing reverse genetics systems for ZIKV. One is based on an Asian ZIKV strain belonging to the outbreak lineage (French Polynesia 2013). The second was designed from the sequence of a low-passaged ZIKV African strain (Dakar 1984). Using the ISA procedure, we derived wild-type and a variety of specifically engineered ZIKVs in days (intra- and inter-lineage chimeras). Since they are based on low-passaged ZIKV strains, these engineered viruses provide ideal tools to study the effect of genetic changes observed in different evolutionary time-scales of ZIKV as well as pathophysiology of ZIKV infections.


Subject(s)
Zika Virus Infection/virology , Zika Virus/genetics , Africa/epidemiology , Asia/epidemiology , Biological Evolution , Cell Line , Cell Line, Tumor , Disease Outbreaks , Female , HEK293 Cells , Humans , Middle Aged , Phylogeny , Polynesia/epidemiology , Reverse Genetics/methods , Zika Virus Infection/epidemiology
SELECTION OF CITATIONS
SEARCH DETAIL
...