Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
1.
J Biol Chem ; 296: 100701, 2021.
Article in English | MEDLINE | ID: mdl-33895135

ABSTRACT

The acid sphingomyelinase/ceramide system has been shown to be important for cellular infection with at least some viruses, for instance, rhinovirus or severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Functional inhibition of the acid sphingomyelinase using tricyclic antidepressants prevented infection of epithelial cells, for instance with SARS-CoV-2. The structure of ambroxol, that is, trans-4-[(2,4-dibromanilin-6-yl)-methyamino]-cyclohexanol, a mucolytic drug applied by inhalation, suggests that the drug might inhibit the acid sphingomyelinase and thereby infection with SARS-CoV-2. To test this, we used vesicular stomatitis virus pseudoviral particles presenting SARS-CoV-2 spike protein on their surface (pp-VSV-SARS-CoV-2 spike), a bona fide system for mimicking SARS-CoV-2 entry into cells. Viral uptake and formation of ceramide localization were determined by fluorescence microscopy, activity of the acid sphingomyelinase by consumption of [14C]sphingomyelin and ceramide was quantified by a kinase method. We found that entry of pp-VSV-SARS-CoV-2 spike required activation of acid sphingomyelinase and release of ceramide, events that were all prevented by pretreatment with ambroxol. We also obtained nasal epithelial cells from human volunteers prior to and after inhalation of ambroxol. Inhalation of ambroxol reduced acid sphingomyelinase activity in nasal epithelial cells and prevented pp-VSV-SARS-CoV-2 spike-induced acid sphingomyelinase activation, ceramide release, and entry of pp-VSV-SARS-CoV-2 spike ex vivo. The addition of purified acid sphingomyelinase or C16 ceramide restored entry of pp-VSV-SARS-CoV-2 spike into ambroxol-treated epithelial cells. We propose that ambroxol might be suitable for clinical studies to prevent coronavirus disease 2019.


Subject(s)
Ambroxol/pharmacology , Antiviral Agents/pharmacology , SARS-CoV-2/drug effects , Sphingomyelin Phosphodiesterase/genetics , Vesiculovirus/drug effects , Virus Internalization/drug effects , Administration, Inhalation , Animals , Biological Transport , Ceramides/metabolism , Chlorocebus aethiops , Drug Repositioning , Epithelial Cells/drug effects , Epithelial Cells/enzymology , Epithelial Cells/virology , Expectorants , Gene Expression , Humans , Primary Cell Culture , Reassortant Viruses/drug effects , Reassortant Viruses/physiology , SARS-CoV-2/physiology , Sphingomyelin Phosphodiesterase/antagonists & inhibitors , Sphingomyelin Phosphodiesterase/metabolism , Sphingomyelins/metabolism , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/metabolism , Vero Cells , Vesiculovirus/physiology
2.
Virology ; 550: 51-60, 2020 11.
Article in English | MEDLINE | ID: mdl-32882637

ABSTRACT

Influenza virus neuraminidase (NA) contains a universally conserved epitope (NAe, NA222-230). However, no studies have reported vaccines targeting this NA conserved epitope and inducing antibodies recognizing NAe. The extracellular domain of M2 (M2e) is considered as an attractive target for a universal influenza vaccine. We generated recombinant influenza H1N1 viruses expressing conserved epitopes in hemagglutinin (HA) molecules: NAe (NAe-HA) or M2e (M2e-HA) within the HA head domain. Inactivated recombinant NAe-HA and M2e-HA viruses were more effective in inducing IgG antibodies specific for an inserted conserved epitope than live recombinant virus. Recombinant inactivated M2e-HA virus vaccination induced cross protection against H3N2 virus with less weight loss compared to NAe-HA and was more effective in inducing humoral and cellular M2e immune responses. This study provides insight into developing recombinant influenza virus vaccines compatible with current platforms to induce antibody responses to conserved poorly immunogenic epitopes.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza A Virus, H1N1 Subtype/immunology , Influenza Vaccines/biosynthesis , Neuraminidase/immunology , Orthomyxoviridae Infections/prevention & control , Reassortant Viruses/immunology , Viral Matrix Proteins/immunology , Animals , Antibodies, Viral/biosynthesis , Cross Protection , Epitopes/chemistry , Epitopes/immunology , Female , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Humans , Immunity, Cellular/drug effects , Immunity, Humoral/drug effects , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H3N2 Subtype/drug effects , Influenza A Virus, H3N2 Subtype/genetics , Influenza A Virus, H3N2 Subtype/immunology , Influenza A Virus, H5N1 Subtype/drug effects , Influenza A Virus, H5N1 Subtype/genetics , Influenza A Virus, H5N1 Subtype/immunology , Influenza A Virus, H9N2 Subtype/drug effects , Influenza A Virus, H9N2 Subtype/genetics , Influenza A Virus, H9N2 Subtype/immunology , Influenza Vaccines/administration & dosage , Influenza Vaccines/genetics , Mice , Mice, Inbred BALB C , Neuraminidase/genetics , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Reassortant Viruses/drug effects , Reassortant Viruses/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Vaccination/methods , Vaccines, Inactivated , Viral Matrix Proteins/genetics
3.
J Virol ; 94(21)2020 10 14.
Article in English | MEDLINE | ID: mdl-32817214

ABSTRACT

The "shock-and-kill" human immunodeficiency virus type 1 (HIV-1) cure strategy involves latency reversal followed by immune-mediated clearance of infected cells. We have previously shown that activation of the noncanonical NF-κB pathway using an inhibitor of apoptosis (IAP), AZD5582, reverses HIV/simian immunodeficiency virus (SIV) latency. Here, we combined AZD5582 with bispecific HIVxCD3 DART molecules to determine the impact of this approach on persistence. Rhesus macaques (RMs) (n = 13) were infected with simian/human immunodeficiency virus SHIV.C.CH505.375H.dCT, and triple antiretroviral therapy (ART) was initiated after 16 weeks. After 42 weeks of ART, 8 RMs received a cocktail of 3 HIVxCD3 DART molecules having human A32, 7B2, or PGT145 anti-HIV-1 envelope (Env) specificities paired with a human anti-CD3 specificity that is rhesus cross-reactive. The remaining 5 ART-suppressed RMs served as controls. For 10 weeks, a DART molecule cocktail was administered weekly (each molecule at 1 mg/kg of body weight), followed 2 days later by AZD5582 (0.1 mg/kg). DART molecule serum concentrations were well above those considered adequate for redirected killing activity against Env-expressing target cells but began to decline after 3 to 6 weekly doses, coincident with the development of antidrug antibodies (ADAs) against each of the DART molecules. The combination of AZD5582 and the DART molecule cocktail did not increase on-ART viremia or cell-associated SHIV RNA in CD4+ T cells and did not reduce the viral reservoir size in animals on ART. The lack of latency reversal in the model used in this study may be related to low pre-ART viral loads (median, <105 copies/ml) and low preintervention reservoir sizes (median, <102 SHIV DNA copies/million blood CD4+ T cells). Future studies to assess the efficacy of Env-targeting DART molecules or other clearance agents to reduce viral reservoirs after latency reversal may be more suited to models that better minimize immunogenicity and have a greater viral burden.IMPORTANCE The most significant barrier to an HIV-1 cure is the existence of the latently infected viral reservoir that gives rise to rebound viremia upon cessation of ART. Here, we tested a novel combination approach of latency reversal with AZD5582 and clearance with bispecific HIVxCD3 DART molecules in SHIV.C.CH505-infected, ART-suppressed rhesus macaques. We demonstrate that the DART molecules were not capable of clearing infected cells in vivo, attributed to the lack of quantifiable latency reversal in this model with low levels of persistent SHIV DNA prior to intervention as well as DART molecule immunogenicity.


Subject(s)
Alkynes/pharmacology , Anti-Retroviral Agents/pharmacology , Antibodies, Neutralizing/pharmacology , Antibodies, Viral/pharmacology , HIV Infections/drug therapy , Oligopeptides/pharmacology , Simian Acquired Immunodeficiency Syndrome/drug therapy , Viremia/drug therapy , Animals , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , Female , Gene Expression Regulation , HIV Infections/genetics , HIV Infections/immunology , HIV Infections/virology , HIV-1/drug effects , HIV-1/growth & development , HIV-1/immunology , Humans , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Inhibitor of Apoptosis Proteins/genetics , Inhibitor of Apoptosis Proteins/immunology , Macaca mulatta , NF-kappa B/genetics , NF-kappa B/immunology , Reassortant Viruses/drug effects , Reassortant Viruses/growth & development , Reassortant Viruses/immunology , Simian Acquired Immunodeficiency Syndrome/genetics , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/drug effects , Simian Immunodeficiency Virus/growth & development , Simian Immunodeficiency Virus/immunology , Viral Load/drug effects , Viremia/genetics , Viremia/immunology , Viremia/virology , Virus Latency/drug effects , Virus Replication/drug effects
4.
Virology ; 549: 77-84, 2020 10.
Article in English | MEDLINE | ID: mdl-32853849

ABSTRACT

Human infections with highly pathogenic avian influenza (HPAI) H7N9 virus were detected in late 2016. We examined the drug resistance profile of 30 HPAI H7N9 isolates from Mainland of China (2016-2019). Altogether, 23% (7/30) carried neuraminidase inhibitors (NAIs) - resistance mutations, and 13% (4/30) displayed reduced susceptibility to NAIs in neuraminidase (NA) inhibition test. An HPAI H7N9 reassortment virus we prepared was passaged with NAIs for 10 passages. Passage with zanamivir induced an E119G substitution in NA, whereas passage with oseltamivir induced R292K and E119V substitutions that simulated that seen in oseltamivir -treated HPAI H7N9 cases, indicating that the high frequency of resistant strains in the HPAI H7N9 isolates is related to NAIs use. In presence of NAIs, R238I, A146E, G151E and G234T substitutions were found in HA1 region of HA. No amino acid mutations were found in the internal genes of the recombinant virus.


Subject(s)
Drug Resistance, Viral/genetics , Influenza A Virus, H7N9 Subtype/genetics , Mutation , Neuraminidase/genetics , Reassortant Viruses/genetics , Viral Proteins/genetics , Amino Acid Substitution , Animals , Antiviral Agents/pharmacology , Birds/virology , Enzyme Inhibitors/pharmacology , Gene Expression , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Humans , Influenza A Virus, H7N9 Subtype/drug effects , Influenza A Virus, H7N9 Subtype/metabolism , Influenza A Virus, H7N9 Subtype/pathogenicity , Influenza in Birds/pathology , Influenza in Birds/transmission , Influenza in Birds/virology , Influenza, Human/pathology , Influenza, Human/transmission , Influenza, Human/virology , Microbial Sensitivity Tests , Models, Molecular , Neuraminidase/metabolism , Oseltamivir/pharmacology , Protein Conformation , Reassortant Viruses/drug effects , Reassortant Viruses/metabolism , Reassortant Viruses/pathogenicity , Viral Proteins/metabolism , Zanamivir/pharmacology
5.
Antiviral Res ; 179: 104807, 2020 07.
Article in English | MEDLINE | ID: mdl-32343991

ABSTRACT

Baloxavir marboxil (BXM) is a potent inhibitor of the polymerase acidic (PA) protein of influenza viruses. However, clinical trials predominantly involving influenza A(H1N1) and A(H3N2) infections showed that BXM exhibited a low barrier of resistance. Contrasting with influenza A viruses, BXM-resistant influenza B variants remain poorly documented. We evaluated the impact of I38 T/M and E23K PA substitutions, previously reported in influenza A viruses, on in vitro properties and virulence of contemporary influenza B recombinant viruses. Influenza B/Phuket/3073/2013 recombinant wild-type (WT) virus and the I38T, I38M and E23K PA mutants were assessed for their susceptibility to baloxavir acid (BXA), the active metabolite of BXM, by plaque reduction assays in ST6GalI-MDCK cells. Luciferase-based minigenome tests were performed to determine polymerase activity. Replication kinetics and genetic stability were evaluated in ST6GalI-MDCK cells. Virulence was evaluated in BALB/c mice. The I38T, I38M and E23K substitutions increased BXA IC50s values by 12.6-, 5.5-, and 2.6-fold, respectively, compared to the WT. Minigenome assays revealed a 46% loss of polymerase activity for the E23K substitution vs the WT while the I38T and I38M PA variants retained ≈80% of activity. Peak viral titers were comparable for the WT, I38T and I38M recombinants (7.95 ± 0.5, 7.45 ± 0.25 and 8.11 ± 0.28 logTCID50/mL), respectively, whereas it was significantly lower for the E23K mutant (6.28 ± 0.28 logTCID50/mL;P < 0.05 vs the WT). In mice, the WT, I38T and I38M recombinants induced mortality rates of 60%, 40% and 100%, respectively and similar lung viral titers were obtained for the three groups at days 3 and 6 p.i. In conclusion, the fitness of BXA-resistant I38T and I38M PA mutants appears unaltered in contemporary influenza B viruses warranting surveillance for their emergence.


Subject(s)
Antiviral Agents/pharmacology , Dibenzothiepins/pharmacology , Drug Resistance, Viral/genetics , Influenza B virus/drug effects , Influenza B virus/genetics , Morpholines/pharmacology , Pyridones/pharmacology , Triazines/pharmacology , Animals , Clinical Trials as Topic , Dogs , Female , Genome, Viral , Inhibitory Concentration 50 , Madin Darby Canine Kidney Cells , Mice , Mice, Inbred BALB C , Mutation , Orthomyxoviridae Infections/drug therapy , Orthomyxoviridae Infections/virology , Reassortant Viruses/drug effects , Recombination, Genetic , Virus Replication
6.
Article in English | MEDLINE | ID: mdl-32117796

ABSTRACT

Influenza A virus (IAV) is a threat to public health due to its high mutation rate and resistance to existing drugs. In this investigation, 15 targets selected from an influenza virus-host interaction network were successfully constructed as a multitarget virtual screening system for new drug discovery against IAV using Naïve Bayesian, recursive partitioning, and CDOCKER methods. The predictive accuracies of the models were evaluated using training sets and test sets. The system was then used to predict active constituents of Compound Yizhihao (CYZH), a Chinese medicinal compound used to treat influenza. Twenty-eight compounds with multitarget activities were selected for subsequent in vitro evaluation. Of the four compounds predicted to be active on neuraminidase (NA), chlorogenic acid, and orientin showed inhibitory activity in vitro. Linarin, sinensetin, cedar acid, isoliquiritigenin, sinigrin, luteolin, chlorogenic acid, orientin, epigoitrin, and rupestonic acid exhibited significant effects on TNF-α expression, which is almost consistent with predicted results. Results from a cytopathic effect (CPE) reduction assay revealed acacetin, indirubin, tryptanthrin, quercetin, luteolin, emodin, and apigenin had protective effects against wild-type strains of IAV. Quercetin, luteolin, and apigenin had good efficacy against resistant IAV strains in CPE reduction assays. Finally, with the aid of Gene Ontology biological process analysis, the potential mechanisms of CYZH action were revealed. In conclusion, a compound-protein interaction-prediction system was an efficient tool for the discovery of novel compounds against influenza, and the findings from CYZH provide important information for its usage and development.


Subject(s)
Antiviral Agents/pharmacology , Drug Discovery/methods , Drugs, Chinese Herbal/metabolism , Drugs, Chinese Herbal/pharmacology , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H3N2 Subtype/drug effects , A549 Cells , Animals , Antiviral Agents/chemistry , Antiviral Agents/metabolism , Computer Simulation , Cytopathogenic Effect, Viral , Dogs , Drugs, Chinese Herbal/chemistry , Genes, Viral , Host-Pathogen Interactions , Humans , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/physiology , Influenza A Virus, H3N2 Subtype/growth & development , Influenza A Virus, H3N2 Subtype/physiology , Ligands , Madin Darby Canine Kidney Cells , Neuraminidase/antagonists & inhibitors , Quantitative Structure-Activity Relationship , Reassortant Viruses/drug effects , Tumor Necrosis Factor-alpha/metabolism , Viral Proteins/antagonists & inhibitors
7.
Virus Res ; 247: 40-46, 2018 03 02.
Article in English | MEDLINE | ID: mdl-29382551

ABSTRACT

In the course of previous works, it was observed that the virus laboratory stock (T3DS) differs in sequence from the virus encoded by the ten plasmids currently in use in many laboratories (T3DK), and derived from a different original virus stock. Seven proteins are affected by these sequence differences. In the present study, replication of T3DK was shown to be more sensitive to the antiviral effect of interferon. Infection by the T3DK virus was also shown to induce the production of higher amount of ß and α-interferons compared to T3DS. Two proteins, the µ2 and λ2 proteins, were found to be responsible for increased sensitivity to interferon while both µ2 and λ1 are responsible for increased interferon secretion. Altogether this supports the idea that multiple reovirus proteins are involved in the control of induction of interferon and virus sensitivity to the interferon-induced response. While interrelated, interferon induction and sensitivity can be separated by defined gene combinations. While both µ2 and λ2 were previously suspected of a role in the control of the interferon response, other proteins are also likely involved, as first shown here for λ1. This also further stresses that due caution should be exerted when comparing different virus isolates with different genetic background.


Subject(s)
Capsid Proteins/genetics , DNA-Binding Proteins/genetics , Interferon-alpha/pharmacology , Interferon-beta/pharmacology , Nucleotidyltransferases/genetics , Orthoreovirus, Mammalian/drug effects , RNA-Binding Proteins/genetics , Reassortant Viruses/drug effects , Viral Core Proteins/genetics , Viral Proteins/genetics , Animals , Antiviral Agents/pharmacology , Capsid Proteins/chemistry , Capsid Proteins/metabolism , Cell Line , Cricetulus , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Epithelial Cells/drug effects , Epithelial Cells/immunology , Epithelial Cells/virology , Fibroblasts/drug effects , Fibroblasts/immunology , Fibroblasts/virology , Gene Expression Regulation, Viral , Host-Pathogen Interactions , Interferon-alpha/biosynthesis , Interferon-beta/biosynthesis , Mice , Models, Molecular , Nucleotidyltransferases/chemistry , Nucleotidyltransferases/metabolism , Orthoreovirus, Mammalian/genetics , Orthoreovirus, Mammalian/immunology , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism , Reassortant Viruses/genetics , Reassortant Viruses/immunology , Reverse Genetics , Viral Core Proteins/chemistry , Viral Core Proteins/metabolism , Viral Proteins/chemistry , Viral Proteins/metabolism
8.
BMC Res Notes ; 10(1): 243, 2017 Jul 05.
Article in English | MEDLINE | ID: mdl-28679441

ABSTRACT

BACKGROUND: In this work, we investigated the genetic diversity of HIV-1 and the presence of mutations conferring antiretroviral drug resistance in 50 drug-naïve infected persons in the Republic of Congo (RoC). Samples were obtained before large-scale access to HAART in 2002 and 2004. METHODS: To assess the HIV-1 genetic recombination, the sequencing of the pol gene encoding a protease and partial reverse transcriptase was performed and analyzed with updated references, including newly characterized CRFs. The assessment of drug resistance was conducted according to the WHO protocol. RESULTS: Among the 50 samples analyzed for the pol gene, 50% were classified as intersubtype recombinants, charring complex structures inside the pol fragment. Five samples could not be classified (noted U). The most prevalent subtypes were G with 10 isolates and D with 11 isolates. One isolate of A, J, H, CRF05, CRF18 and CRF37 were also found. Two samples (4%) harboring the mutations M230L and Y181C associated with the TAMs M41L and T215Y, respectively, were found. CONCLUSION: This first study in the RoC, based on WHO classification, shows that the threshold of transmitted drug resistance before large-scale access to antiretroviral therapy is 4%.


Subject(s)
Drug Resistance, Viral/genetics , Genetic Variation , Genotype , HIV Infections/virology , HIV-1/genetics , Reassortant Viruses/genetics , Adolescent , Adult , Anti-HIV Agents/pharmacology , Child , Child, Preschool , Congo , Female , Gene Expression , Genes, pol , HIV-1/classification , HIV-1/drug effects , HIV-1/isolation & purification , Humans , Male , Molecular Typing , Mutation , Reassortant Viruses/classification , Reassortant Viruses/drug effects , Reassortant Viruses/isolation & purification , Recombination, Genetic
9.
J Hepatol ; 67(4): 680-686, 2017 10.
Article in English | MEDLINE | ID: mdl-28619439

ABSTRACT

BACKGROUND & AIMS: Little is known about the epidemiology and frequency of recombinant HCV genotype 2/1 strains, which may represent a challenge for direct antiviral therapy (DAA). This study aims to identify the epidemiology and phylogeny of HCV genotype 2/1 strains and encourages genotype screening, to select the DAA-regimen that achieves the optimal sustained virologic response. METHODS: Consecutive samples from HCV genotype 2 infected patients, according to commercial genotyping, from Germany, Italy and Israel were re-genotyped by Sanger-based sequencing. Virologic, epidemiological, and phylogenetic analyses including other published chimeras were performed. RESULTS: Sequence analysis of 442 supposed HCV genotype 2 isolates revealed 61 (genotype 2k/1b (n=59), 2a/1b (n=1) or 2b/1a (n=1)) chimeras. No chimeras were observed in Italy, but the frequency was 14% and 25% in Germany and Israel. Treatment of viral chimera with sofosbuvir/ribavirin led to virologic relapse in 25/27 patients (93%). Nearly all patients treated with genotype 1-based DAA-regimens initially (n=8/9), or after relapse (n=13/13), achieved a sustained virologic response. Most patients with 2k/1b chimeras (88%) were originally from eight different areas of the former Soviet Union. All known 2k/1b chimeras harbour the same recombination breakpoint and build one phylogenetic cluster, while all other chimeras have different phylogenies. CONCLUSIONS: The HCV genotype 2k/1b variant derives from one single recombination event most likely in the former Soviet Union, while other chimeras are unique and develop independently. A relatively high frequency has been observed along the migration flows, in Germany and Israel. In countries with little migration from the former Soviet Union the prevalence of 2k/1b chimeras is expected to be low. Treatment with sofosbuvir plus ribavirin is insufficient, but genotype 1-based regimens seem to be effective. Lay summary: The frequency of recombinant HCV is higher than expected. A novel recombinant variant (HCV genotype 2a/1b) was identified. Screening for recombinant viruses would contribute to increased response rates to direct antiviral therapy.


Subject(s)
Hepacivirus/drug effects , Hepacivirus/genetics , Hepatitis C, Chronic/drug therapy , Hepatitis C, Chronic/virology , Amino Acid Substitution , Antiviral Agents/therapeutic use , Chimera/genetics , Drug Resistance, Viral/genetics , Evolution, Molecular , Genotype , Germany/epidemiology , Hepacivirus/classification , Hepatitis C, Chronic/epidemiology , Humans , Israel/epidemiology , Italy/epidemiology , Molecular Epidemiology , Phylogeny , Polymorphism, Single Nucleotide , Prevalence , Reassortant Viruses/drug effects , Reassortant Viruses/genetics , Recombination, Genetic , Sustained Virologic Response
10.
Antivir Ther ; 22(4): 365-368, 2017.
Article in English | MEDLINE | ID: mdl-28085002

ABSTRACT

Direct-acting antiviral (DAA) combinations are potent and effective drugs currently recommended for treatment of chronic HCV infection. Difficult to treat genotypes are the most important predictors of treatment failure. We report a case of DAA treatment failure in an HCV-infected patient carrying a recombinant genotype 2k/1b. This strain, first isolated from a Russian patient in 2002, has now been observed for the first time in Italy.


Subject(s)
Drug Resistance, Viral/genetics , Hepacivirus/genetics , Hepatitis C, Chronic/drug therapy , Liver Cirrhosis/drug therapy , Reassortant Viruses/genetics , Alanine Transaminase/blood , Antiviral Agents/therapeutic use , Drug Combinations , Female , Genotype , Hepacivirus/classification , Hepacivirus/drug effects , Hepacivirus/growth & development , Hepatitis C, Chronic/diagnosis , Hepatitis C, Chronic/virology , Humans , Italy , Liver Cirrhosis/diagnosis , Liver Cirrhosis/virology , Middle Aged , Phylogeny , Reassortant Viruses/classification , Reassortant Viruses/drug effects , Reassortant Viruses/growth & development , Ribavirin/therapeutic use , Sequence Analysis, RNA , Sofosbuvir/therapeutic use , Treatment Failure
11.
Arch Virol ; 162(2): 457-467, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27787680

ABSTRACT

Two hundred forty million people are chronically infected with hepatitis B virus (HBV) worldwide. The rise of globalization has facilitated the emergence of novel HBV recombinants and genotypes. We evaluated HBV genotypes and recombinants, mutations associated with resistance to antivirals (AVs), progression of hepatic illness, and inefficient hepatitis B vaccination responses in chronically infected individuals in the city of São Paulo, Brazil. Forty-five full-length and 24 partial-length sequences were obtained. The genotype distribution was as follows: A (66.7%), D (15.9%), F (11.6%) and C (4.3%). We describe a new recombinant (D2/D3), confirmed through next-generation sequencing (NGS) and reconstruction of the quasispecies sequences in silico. Primary resistance and major vaccine escape mutations were not found. We did, however, find mutations in the S region that might may be related to HBV antigenicity changes, as well as Pre-S deletions. The precore/core mutations A1762T + G1764A (40.9%) were found mostly in genotypes A and D, and G1896A (29.55%) was more frequent in genotype D than in genotype A. The genotypic distribution reflects the history of Brazilian immigration. This is the first description of recombination between genotypes D2 and D3 in Brazil. It is also the first confirmation through NGS and reconstruction of the quasispecies in silico. However, little is known about the response to treatment of recombinants. This demonstrates the need for molecular epidemiology studies involving the analysis of full-length HBV sequences.


Subject(s)
Drug Resistance, Viral/genetics , Hepatitis B virus/genetics , Hepatitis B, Chronic/epidemiology , Reassortant Viruses/genetics , Recombination, Genetic , Adenine/analogs & derivatives , Adenine/therapeutic use , Adult , Aged , Antiviral Agents/therapeutic use , Brazil/epidemiology , Female , Genotype , Hepatitis B virus/classification , Hepatitis B virus/drug effects , Hepatitis B virus/isolation & purification , Hepatitis B, Chronic/drug therapy , Hepatitis B, Chronic/transmission , Hepatitis B, Chronic/virology , High-Throughput Nucleotide Sequencing , Humans , Lamivudine/therapeutic use , Male , Middle Aged , Molecular Epidemiology , Mutation , Organophosphonates/therapeutic use , Phylogeny , Reassortant Viruses/classification , Reassortant Viruses/drug effects , Reassortant Viruses/isolation & purification , Retrospective Studies
12.
Ter Arkh ; 88(6): 101-105, 2016.
Article in Russian | MEDLINE | ID: mdl-27489903

ABSTRACT

The review gives the data available in the literature in the efficiency of treatment in patients with chronic hepatitis C infected with hepatitis C virus.(HCV) recombinants, by applying various antiviral therapy regimens. The low efficiency of treatment with- pegylated interferons (PEG IFN) + ribavirin (RIB) and sofosburin (SOF) +RIB in this patient group (a sustained virologic response was achieved in 22 and 30.7%, respectively) compared with the efficiency of treatment (87-97 and 83-97%, respectively) inpatients infected with HCV genotype 2 does not allow the 2015 EASL HCV genotype 2 treatment regimens to be used in. such patients. In this connection, subtyping genotype 2 isolates by NS5B sequencing should be introduced into clinical laboratory practice to successfully detect recombinant HCVs and to enhance the efficiency of antiviral therapy.


Subject(s)
Antiviral Agents/pharmacology , Hepacivirus , Hepatitis C, Chronic , Reassortant Viruses , Antiviral Agents/classification , Drug Therapy, Combination , Hepacivirus/drug effects , Hepacivirus/genetics , Hepatitis C, Chronic/drug therapy , Hepatitis C, Chronic/virology , Humans , Reassortant Viruses/drug effects , Reassortant Viruses/genetics , Recombination, Genetic , Treatment Outcome
13.
Antimicrob Agents Chemother ; 60(11): 6679-6691, 2016 11.
Article in English | MEDLINE | ID: mdl-27572398

ABSTRACT

T-705 (favipiravir) is a new antiviral agent in advanced clinical development for influenza therapy. It is supposed to act as an alternative substrate for the viral polymerase, causing inhibition of viral RNA synthesis or virus mutagenesis. These mechanisms were also proposed for ribavirin, an established and broad antiviral drug that shares structural similarity with T-705. We here performed a comparative analysis of the effects of T-705 and ribavirin on influenza virus and host cell functions. Influenza virus-infected cell cultures were exposed to T-705 or ribavirin during single or serial virus passaging. The effects on viral RNA synthesis and infectious virus yield were determined and mutations appearing in the viral genome were detected by whole-genome virus sequencing. In addition, the cellular nucleotide pools as well as direct inhibition of the viral polymerase enzyme were quantified. We demonstrate that the anti-influenza virus effect of ribavirin is based on IMP dehydrogenase inhibition, which results in fast and profound GTP depletion and an imbalance in the nucleotide pools. In contrast, T-705 acts as a potent and GTP-competitive inhibitor of the viral polymerase. In infected cells, viral RNA synthesis is completely inhibited by T-705 or ribavirin at ≥50 µM, whereas exposure to lower drug concentrations induces formation of noninfectious particles and accumulation of random point mutations in the viral genome. This mutagenic effect is 2-fold higher for T-705 than for ribavirin. Hence, T-705 and ribavirin both act as purine pseudobases but profoundly differ with regard to the mechanism behind their antiviral and mutagenic effects on influenza virus.


Subject(s)
Amides/pharmacology , Antiviral Agents/pharmacology , Gene Expression Regulation, Viral , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H3N2 Subtype/drug effects , Pyrazines/pharmacology , Reassortant Viruses/drug effects , Ribavirin/pharmacology , A549 Cells , Amides/chemistry , Animals , Antiviral Agents/chemistry , Chick Embryo , DNA-Directed RNA Polymerases/antagonists & inhibitors , DNA-Directed RNA Polymerases/genetics , DNA-Directed RNA Polymerases/metabolism , Dogs , Humans , IMP Dehydrogenase/antagonists & inhibitors , IMP Dehydrogenase/genetics , IMP Dehydrogenase/metabolism , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/growth & development , Influenza A Virus, H1N1 Subtype/metabolism , Influenza A Virus, H3N2 Subtype/genetics , Influenza A Virus, H3N2 Subtype/growth & development , Influenza A Virus, H3N2 Subtype/metabolism , Madin Darby Canine Kidney Cells , Mutation/drug effects , Pyrazines/chemistry , RNA, Viral/antagonists & inhibitors , RNA, Viral/biosynthesis , Reassortant Viruses/genetics , Reassortant Viruses/growth & development , Reassortant Viruses/metabolism , Ribavirin/chemistry , Sequence Analysis, RNA , Structure-Activity Relationship , Viral Proteins/antagonists & inhibitors , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Replication/drug effects
14.
PLoS One ; 11(7): e0159332, 2016.
Article in English | MEDLINE | ID: mdl-27428377

ABSTRACT

Women need multipurpose prevention products (MPTs) that protect against sexually transmitted infections (STIs) and provide contraception. The Population Council has developed a prototype intravaginal ring (IVR) releasing the non-nucleoside reverse transcriptase inhibitor (NNRTI) MIV-150 (M), zinc acetate (ZA), carrageenan (CG) and levonorgestrel (LNG) (MZCL IVR) to protect against HIV, HSV-2, HPV and unintended pregnancy. Our objective was to evaluate the anti-SHIV-RT activity of MZCL IVR in genital mucosa. First, macaque vaginal tissues were challenged with SHIV-RT in the presence of (i) MIV-150 ± LNG or (ii) vaginal fluids (VF); available from studies completed earlier) collected at various time points post insertion of MZCL and MZC IVRs. Then, (iii) MZCL IVRs (vs. LNG IVRs) were inserted in non-Depo Provera-treated macaques for 24h and VF, genital biopsies, and blood were collected and tissues were challenged with SHIV-RT. Infection was monitored with one step SIV gag qRT-PCR or p27 ELISA. MIV-150 (LCMS/MS, RIA), LNG (RIA) and CG (ELISA) were measured in different compartments. Log-normal generalized mixed linear models were used for analysis. LNG did not affect the anti-SHIV-RT activity of MIV-150 in vitro. MIV-150 in VF from MZC/MZCL IVR-treated macaques inhibited SHIV-RT in vaginal mucosa in a dose-dependent manner (p<0.05). MIV-150 in vaginal tissue from MZCL IVR-treated animals inhibited ex vivo infection relative to baseline (96%; p<0.0001) and post LNG IVR group (90%, p<0.001). No MIV-150 dose-dependent protection was observed, likely because of high MIV-150 concentrations in all vaginal tissue samples. In cervical tissue, MIV-150 inhibited infection vs. baseline (99%; p<0.05). No cervical tissue was available for MIV-150 measurement. Exposure to LNG IVR did not change tissue infection level. These observations support further development of MZCL IVR as a multipurpose prevention technology to improve women's sexual and reproductive health.


Subject(s)
Anti-Infective Agents/pharmacology , Contraceptive Agents, Female/pharmacology , Levonorgestrel/pharmacology , Pyridines/pharmacology , Reassortant Viruses/drug effects , Reverse Transcriptase Inhibitors/pharmacology , Urea/analogs & derivatives , Vagina/drug effects , Animals , Carrageenan/pharmacology , Contraceptive Devices, Female , Drug Combinations , Female , HIV/drug effects , HIV/genetics , HIV/growth & development , Macaca mulatta , Mucous Membrane/drug effects , Mucous Membrane/virology , Reassortant Viruses/genetics , Reassortant Viruses/growth & development , Simian Immunodeficiency Virus/drug effects , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/growth & development , Treatment Outcome , Urea/pharmacology , Vagina/virology , Zinc Acetate/pharmacology
15.
J Antimicrob Chemother ; 71(11): 3036-3045, 2016 11.
Article in English | MEDLINE | ID: mdl-27432605

ABSTRACT

OBJECTIVES: While subtype-specific substitutions linked to neuraminidase (NA) inhibitor resistance are well described in human N1 and N2 influenza NAs, little is known about other NA subtypes. The aim of this study was to determine whether the R292K and E119V ±â€ŠI222L substitutions could be associated with oseltamivir resistance in all group 2 NAs and had an impact on virus fitness. METHODS: Reassortant viruses with WT NA or variant N2, N3, N6, N7 or N9 NAs, bearing R292K or E119V ±â€ŠI222L substitutions, were produced by reverse genetics. The antiviral susceptibility, activity, Km of the NA, mutation stability and in vitro virus fitness in MDCK cells were determined. RESULTS: NA activities could be ranked as follows regardless of the substitution: N3 ≥ N6 > N2 ≥ N9 > N7. Using NA inhibitor resistance interpretation criteria used for human N1 or N2, the NA-R292K substitution conferred highly reduced inhibition by oseltamivir and the N6- or N9-R292K substitution conferred reduced inhibition by zanamivir and laninamivir. Viruses with the N3- or N6-E119V substitution showed normal inhibition by oseltamivir, while those with the N2-, N7- or N9-E119V substitution showed reduced inhibition by oseltamivir. Viruses with NA-E119V + I222L substitutions showed reduced inhibition (N3 and N6) or highly reduced inhibition (N2, N7 and N9) by oseltamivir. Viruses bearing the NA-R292K substitution had lower affinity and viruses bearing the NA-E119V substitution had higher affinity for the MUNANA substrate than viruses with corresponding WT NA. CONCLUSIONS: NA-R292K and E119V + I222L substitutions conferred reduced inhibition by oseltamivir for all group 2 NAs. Surveillance of NA inhibitor resistance for zoonotic and human influenza viruses and the development of novel antiviral agents with different targets should be continued.


Subject(s)
Antiviral Agents/pharmacology , Drug Resistance, Viral , Influenza A virus/drug effects , Influenza A virus/genetics , Mutation, Missense , Neuraminidase/genetics , Oseltamivir/pharmacology , Humans , Reassortant Viruses/drug effects , Reassortant Viruses/genetics , Reverse Genetics
16.
Antimicrob Agents Chemother ; 60(9): 5182-8, 2016 09.
Article in English | MEDLINE | ID: mdl-27297486

ABSTRACT

Ebola outbreaks occur on a frequent basis, with the 2014-2015 outbreak in West Africa being the largest one ever recorded. This outbreak has resulted in over 11,000 deaths in four African countries and has received international attention and intervention. Although there are currently no approved therapies or vaccines, many promising candidates are undergoing clinical trials, and several have had success in promoting recovery from Ebola. However, these prophylactics and therapeutics have been designed and tested only against the same species of Ebola virus as the one causing the current outbreak. Future outbreaks involving other species would require reformulation and possibly redevelopment. Therefore, a broad-spectrum alternative is highly desirable. We have found that a flavonoid derivative called quercetin 3-ß-O-d-glucoside (Q3G) has the ability to protect mice from Ebola even when given as little as 30 min prior to infection. Furthermore, we have demonstrated that this compound targets the early steps of viral entry. Most promisingly, antiviral activity against two distinct species of Ebola virus was seen. This study serves as a proof of principle that Q3G has potential as a prophylactic against Ebola virus infection.


Subject(s)
Antiviral Agents/pharmacology , Ebolavirus/drug effects , Hemorrhagic Fever, Ebola/prevention & control , Quercetin/analogs & derivatives , Reassortant Viruses/drug effects , Virus Replication/drug effects , Animals , Chlorocebus aethiops , Dose-Response Relationship, Drug , Ebolavirus/growth & development , Female , Hemorrhagic Fever, Ebola/mortality , Hemorrhagic Fever, Ebola/pathology , Hemorrhagic Fever, Ebola/virology , Injections, Intraperitoneal , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Quercetin/pharmacology , Reassortant Viruses/growth & development , Survival Analysis , Toxicity Tests, Chronic , Treatment Outcome , Vero Cells
17.
Vopr Virusol ; 61(1): 16-20, 2016.
Article in Russian | MEDLINE | ID: mdl-27145595

ABSTRACT

This work is devoted to the research of the live attenuated influenza vaccine (LAIV) comprising two reassortant B/USSR/60/69-based vaccine influenza viruses Victoria and Yamagata. The intranasal immunization of the CBA mice with both Victoria and Yamagata strains induced 100% lung protection against the subsequent infection with the wild-type influenza B viruses of any antigen lineage. The quadrivalent LAIV (qLAIV) comprising both reassortant influenza B viruses Victoria and Yamagata were safe and areactogenic in adult volunteers. Following qLAIV administration the immune response was achieved to both Victoria and Yamagata lineages.


Subject(s)
Antibodies, Viral/blood , Immunity, Humoral/drug effects , Influenza Vaccines/administration & dosage , Influenza, Human/prevention & control , Vaccination , Administration, Intranasal , Adolescent , Adult , Animals , Female , Hemagglutination Inhibition Tests , Humans , Influenza B virus/drug effects , Influenza B virus/genetics , Influenza B virus/immunology , Influenza Vaccines/biosynthesis , Influenza Vaccines/immunology , Influenza, Human/blood , Influenza, Human/immunology , Influenza, Human/virology , Male , Mice , Mice, Inbred CBA , Middle Aged , Reassortant Viruses/drug effects , Reassortant Viruses/genetics , Reassortant Viruses/immunology , Vaccines, Attenuated , Vaccines, Subunit
18.
PLoS One ; 11(3): e0150595, 2016.
Article in English | MEDLINE | ID: mdl-26937954

ABSTRACT

Eleven new furostanol saponins, typaspidosides B-L (1-11), one new spirostanol saponin, typaspidoside M (12), and five known spirostanol saponins, 25S-atropuroside (13), neoaspidistrin (14), (25S)-pratioside D1 (15), 25S-aspidistrin (16) and 25S-neosibiricoside (17) were isolated from the rhizomes of Aspidistra typica Baill. The structures of the new compounds were established using 1D and 2D NMR (1H-1H COSY, HMQC, HMBC and ROESY) spectroscopy, high resolution mass spectrometry, and chemical methods. The aglycones of 1-3 (unusual furostanol saponins with opened E ring type), 9 and 10 (the methoxyl substituent at C-23 position) were found, identified from natural products for the first time. Moreover, the anti-HIV activities of the isolated steroidal glycosides were assessed, and compounds 13, 14, 16 and 17 exhibited high active against HIV-1.


Subject(s)
Anti-HIV Agents/pharmacology , HIV-1/drug effects , Liliaceae/chemistry , Reassortant Viruses/drug effects , Rhizome/chemistry , Saponins/pharmacology , Sterols/pharmacology , Anti-HIV Agents/isolation & purification , Capsid Proteins/antagonists & inhibitors , Capsid Proteins/genetics , Capsid Proteins/metabolism , Gene Expression , Genes, Reporter , HEK293 Cells , HIV-1/genetics , HIV-1/growth & development , Humans , Luciferases/antagonists & inhibitors , Luciferases/genetics , Luciferases/metabolism , Magnetic Resonance Spectroscopy , Plant Extracts/chemistry , Reassortant Viruses/genetics , Reassortant Viruses/growth & development , Saponins/isolation & purification , Spirostans/isolation & purification , Spirostans/pharmacology , Steroids/isolation & purification , Steroids/pharmacology , Sterols/isolation & purification , Structure-Activity Relationship , Vesiculovirus/genetics , Virus Replication/drug effects
19.
J Virol ; 90(8): 4215-4231, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26865709

ABSTRACT

UNLABELLED: Adeno-associated viruses (AAVs) currently are being developed to efficiently transduce the retina following noninvasive, intravitreal (Ivt) injection. However, a major barrier encountered by intravitreally delivered AAVs is the inner limiting membrane (ILM), a basement membrane rich in heparan sulfate (HS) proteoglycan. The goal of this study was to determine the impact of HS binding on retinal transduction by Ivt-delivered AAVs. The heparin affinities of AAV2-based tyrosine-to-phenylalanine (Y-F) and threonine-to-valine (T-V) capsid mutants, designed to avoid proteasomal degradation during cellular trafficking, were established. In addition, the impact of grafting HS binding residues onto AAV1, AAV5, and AAV8(Y733F) as well as ablation of HS binding by AAV2-based vectors on retinal transduction was investigated. Finally, the potential relationship between thermal stability of AAV2-based capsids and Ivt-mediated transduction was explored. The results show that the Y-F and T-V AAV2 capsid mutants bind heparin but with slightly reduced affinity relative to that of AAV2. The grafting of HS binding increased Ivt transduction by AAV1 but not by AAV5 or AAV8(Y733F). The substitution of any canonical HS binding residues ablated Ivt-mediated transduction by AAV2-based vectors. However, these same HS variant vectors displayed efficient retinal transduction when delivered subretinally. Notably, a variant devoid of canonical HS binding residues, AAV2(4pMut)ΔHS, was remarkably efficient at transducing photoreceptors. The disparate AAV phenotypes indicate that HS binding, while critical for AAV2-based vectors, is not the sole determinant for transduction via the Ivt route. Finally, Y-F and T-V mutations alter capsid stability, with a potential relationship existing between stability and improvements in retinal transduction by Ivt injection. IMPORTANCE: AAV has emerged as the vector of choice for gene delivery to the retina, with attention focused on developing vectors that can mediate transduction following noninvasive, intravitreal injection. HS binding has been postulated to play a role in intravitreally mediated transduction of retina. Our evaluation of the HS binding of AAV2-based variants and other AAV serotype vectors and the correlation of this property with transduction points to HS affinity as a factor controlling retinal transduction following Ivt delivery. However, HS binding is not the only requirement for improved Ivt-mediated transduction. We show that AAV2-based vectors lacking heparin binding transduce retina by subretinal injection and display a remarkable ability to transduce photoreceptors, indicating that other receptors are involved in this phenotype.


Subject(s)
Dependovirus/physiology , Genetic Vectors , Heparitin Sulfate/pharmacology , Retina/metabolism , Transduction, Genetic , Animals , Capsid/metabolism , Dependovirus/drug effects , Dependovirus/genetics , Genetic Vectors/drug effects , Genetic Vectors/genetics , Genetic Vectors/metabolism , HEK293 Cells , Humans , Injections, Intraocular , Injections, Intravenous , Liver/metabolism , Mice, Inbred BALB C , Mice, Inbred C57BL , Mutation , Photoreceptor Cells, Vertebrate/metabolism , Reassortant Viruses/drug effects , Reassortant Viruses/genetics , Reassortant Viruses/physiology , Vitreous Body/metabolism
20.
J Acquir Immune Defic Syndr ; 69(4): 385-94, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-25886925

ABSTRACT

BACKGROUND: Preexposure prophylaxis (PrEP) for HIV prevention is a novel biomedical prevention method. We have previously modeled PrEP during rectal SHIV exposures in macaques and identified that Simian/Human Immunodeficiency Virus chimera (SHIV)-specific T-cell responses were induced in the presence of antiretroviral drugs, an observation previously termed T-cell chemo-vaccination. This report expands those initial findings by examining a larger group of macaques that were given oral or topical PrEP during repeated vaginal virus exposure. METHODS: Thirty-six female pigtail macaques received up to 20 repeat low-dose vaginal inoculations with wild-type (WT) SHIVSF162P3 (n = 24) or a clonal derivative with the tenofovir (TFV) K65R drug-resistant mutation (n = 12). PrEP consisted of oral Truvada (n = 6, WT), TFV vaginal gel (n = 6, K65R), or TFV intravaginal ring (n = 6, WT). The remaining animals were PrEP-inexperienced controls (n = 12, WT; n = 6, K65R). SHIV-specific T cells were identified and characterized using interferon γ Enzyme-Linked ImmunoSpot (ELISPOT) and multiparameter flow cytometry. RESULTS: Of 9 animals that were on PrEP and remained uninfected during WT SHIV vaginal challenges, 8 (88.9%) developed virus-specific T-cell responses. T cells were in CD4 and CD8 compartments, reached up to 4900 interferon γ-producing cells per million peripheral blood mononuclear cells, and primarily pol directed. In contrast, the replication-impaired K65R virus did not induce detectable T-cell responses, likely reflecting the need for adequate replication. CONCLUSIONS: Virus-specific T-cell responses occur frequently in oral or topical PrEP-protected pigtail macaques after vaginal exposure to WT SHIV virus. The contribution of such immune responses to protection from infection during and after PrEP warrants further investigation.


Subject(s)
Anti-HIV Agents/therapeutic use , Deoxycytidine/analogs & derivatives , HIV Infections/prevention & control , Macaca nemestrina , Organophosphorus Compounds , Simian Acquired Immunodeficiency Syndrome/prevention & control , T-Lymphocytes/physiology , Administration, Oral , Administration, Topical , Animals , Drug Combinations , Drug Resistance, Viral , Emtricitabine, Tenofovir Disoproxil Fumarate Drug Combination , Female , Pre-Exposure Prophylaxis , Reassortant Viruses/drug effects , Simian Immunodeficiency Virus/drug effects , Simian Immunodeficiency Virus/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...