Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 167
Filter
Add more filters

Publication year range
2.
J Virol ; 94(5)2020 02 14.
Article in English | MEDLINE | ID: mdl-31801862

ABSTRACT

HIV diversification facilitates immune escape and complicates antiretroviral therapy. In this study, we take advantage of a humanized-mouse model to probe the contribution of APOBEC3 mutagenesis to viral evolution. Humanized mice were infected with isogenic HIV molecular clones (HIV-WT, HIV-45G, and HIV-ΔSLQ) that differ in their abilities to counteract APOBEC3G (A3G). Infected mice remained naive or were treated with the reverse transcriptase (RT) inhibitor lamivudine (3TC). Viremia, emergence of drug-resistant variants, and quasispecies diversification in the plasma compartment were determined throughout infection. While both HIV-WT and HIV-45G achieved robust infection, over time, HIV-45G replication was significantly reduced compared to that of HIV-WT in the absence of 3TC treatment. In contrast, treatment responses differed significantly between HIV-45G- and HIV-WT-infected mice. Antiretroviral treatment failed in 91% of HIV-45G-infected mice, while only 36% of HIV-WT-infected mice displayed a similar negative outcome. Emergence of 3TC-resistant variants and nucleotide diversity were determined by analyzing 155,462 single HIV reverse transcriptase gene (RT) and 6,985 vif sequences from 33 mice. Prior to treatment, variants with genotypic 3TC resistance (RT-M184I/V) were detected at low levels in over a third of all the animals. Upon treatment, the composition of the plasma quasispecies rapidly changed, leading to a majority of circulating viral variants encoding RT-184I. Interestingly, increased viral diversity prior to treatment initiation correlated with higher plasma viremia in HIV-45G-infected animals, but not in HIV-WT-infected animals. Taken together, HIV variants with suboptimal anti-A3G activity were attenuated in the absence of selection but displayed a fitness advantage in the presence of antiretroviral treatment.IMPORTANCE Both viral (e.g., RT) and host (e.g., A3G) factors can contribute to HIV sequence diversity. This study shows that suboptimal anti-A3G activity shapes viral fitness and drives viral evolution in the plasma compartment in humanized mice.


Subject(s)
APOBEC-3G Deaminase/metabolism , Drug Resistance, Viral/physiology , HIV Infections/immunology , HIV-1/immunology , Animals , Anti-HIV Agents/pharmacology , Disease Models, Animal , Drug Resistance, Viral/drug effects , Genetic Variation , HEK293 Cells , HIV-1/drug effects , Humans , Lamivudine/pharmacology , Mice , Virus Replication/drug effects
3.
Int J Mol Sci ; 22(10)2021 May 18.
Article in English | MEDLINE | ID: mdl-34069929

ABSTRACT

The success of antiretroviral treatment (ART) is threatened by the emergence of drug resistance mutations (DRM). Since Brazil presents the largest number of people living with HIV (PLWH) in South America we aimed at understanding the dynamics of DRM in this country. We analyzed a total of 20,226 HIV-1 sequences collected from PLWH undergoing ART between 2008-2017. Results show a mild decline of DRM over the years but an increase of the K65R reverse transcriptase mutation from 2.23% to 12.11%. This increase gradually occurred following alterations in the ART regimens replacing zidovudine (AZT) with tenofovir (TDF). PLWH harboring the K65R had significantly higher viral loads than those without this mutation (p < 0.001). Among the two most prevalent HIV-1 subtypes (B and C) there was a significant (p < 0.001) association of K65R with subtype C (11.26%) when compared with subtype B (9.27%). Nonetheless, evidence for K65R transmission in Brazil was found both for C and B subtypes. Additionally, artificial neural network-based immunoinformatic predictions suggest that K65R could enhance viral recognition by HLA-B27 that has relatively low prevalence in the Brazilian population. Overall, the results suggest that tenofovir-based regimens need to be carefully monitored particularly in settings with subtype C and specific HLA profiles.


Subject(s)
Drug Resistance, Viral/genetics , HIV Infections/genetics , Adenine/therapeutic use , Adult , Aged , Anti-HIV Agents/pharmacology , Brazil/epidemiology , Drug Resistance, Viral/physiology , Female , HIV Infections/drug therapy , HIV Reverse Transcriptase/genetics , HIV-1/drug effects , HIV-1/genetics , HIV-1/pathogenicity , Humans , Male , Middle Aged , Mutation/drug effects , Reverse Transcriptase Inhibitors/pharmacology , Tenofovir/therapeutic use , Treatment Failure , Viral Load/drug effects , Zidovudine/therapeutic use
4.
Appl Environ Microbiol ; 86(19)2020 09 17.
Article in English | MEDLINE | ID: mdl-32709728

ABSTRACT

We determined the disinfection efficacy and inactivation mechanisms of peracetic acid (PAA)-based sanitizer using pH values relevant for vegetable sanitation against rotavirus (RV) and Tulane virus (TV; a human norovirus surrogate). TV was significantly more resistant to PAA disinfection than RV: for a 2-log10 reduction of virus titer, RV required 1 mg/liter PAA for 3.5 min of exposure, while TV required 10 mg/liter PAA for 30 min. The higher resistance of TV can be explained, in part, by significantly more aggregation of TV in PAA solutions. The PAA mechanisms of virus inactivation were explored by quantifying (i) viral genome integrity and replication using reverse transcription-quantitative PCR (RT-qPCR) and (ii) virus-host receptor interactions using a cell-free binding assay with porcine gastric mucin conjugated with magnetic beads (PGM-MBs). We observed that PAA induced damage to both RV and TV genomes and also decreased virus-receptor interactions, with the latter suggesting that PAA damages viral proteins important for binding its host cell receptors. Importantly, the levels of genome-versus-protein damage induced by PAA were different for each virus. PAA inactivation correlated with higher levels of RV genome damage than of RV-receptor interactions. For PAA-treated TV, the opposite trends were observed. Thus, PAA inactivates each of these viruses via different molecular mechanisms. The findings presented here potentially contribute to the design of a robust sanitation strategy for RV and TV using PAA to prevent foodborne disease.IMPORTANCE In this study, we examined the inactivation mechanisms of peracetic acid (PAA), a sanitizer commonly used for postharvest vegetable washing, for two enteric viruses: Tulane virus (TV) as a human norovirus surrogate and rotavirus (RV). PAA disinfection mechanisms for RV were mainly due to genome damage. In contrast, PAA disinfection in TV was due to damage of the proteins important for binding to its host receptor. We also observed that PAA triggered aggregation of TV to a much greater extent than RV. These studies demonstrate that different viruses are inactivated via different PAA mechanisms. This information is important for designing an optimal sanitation practice for postharvest vegetable washing to minimize foodborne viral diseases.


Subject(s)
Caliciviridae/drug effects , Disinfectants/pharmacology , Drug Resistance, Viral/physiology , Peracetic Acid/pharmacology , Rotavirus/drug effects , Caliciviridae/physiology , Disinfection , Inactivation, Metabolic , Rotavirus/physiology
5.
Clin Infect Dis ; 68(2): 177-187, 2019 01 07.
Article in English | MEDLINE | ID: mdl-30052811

ABSTRACT

Background: Contemporary antiretroviral therapies (ART) and management strategies have diminished both human immunodeficiency virus (HIV) treatment failure and the acquired resistance to drugs in resource-rich regions, but transmission of drug-resistant viruses has not similarly decreased. In low- and middle-income regions, ART roll-out has improved outcomes, but has resulted in increasing acquired and transmitted resistances. Our objective was to review resistance to ART drugs and methods to detect it, and to provide updated recommendations for testing and monitoring for drug resistance in HIV-infected individuals. Methods: A volunteer panel of experts appointed by the International Antiviral (formerly AIDS) Society-USA reviewed relevant peer-reviewed data that were published or presented at scientific conferences. Recommendations were rated according to the strength of the recommendation and quality of the evidence, and reached by full panel consensus. Results: Resistance testing remains a cornerstone of ART. It is recommended in newly-diagnosed individuals and in patients in whom ART has failed. Testing for transmitted integrase strand-transfer inhibitor resistance is currently not recommended, but this may change as more resistance emerges with widespread use. Sanger-based and next-generation sequencing approaches are each suited for genotypic testing. Testing for minority variants harboring drug resistance may only be considered if treatments depend on a first-generation nonnucleoside analogue reverse transcriptase inhibitor. Different HIV-1 subtypes do not need special considerations regarding resistance testing. Conclusions: Testing for HIV drug resistance in drug-naive individuals and in patients in whom antiretroviral drugs are failing, and the appreciation of the role of testing, are crucial to the prevention and management of failure of ART.


Subject(s)
Anti-HIV Agents/pharmacology , Drug Resistance, Viral/physiology , HIV Infections/drug therapy , HIV Infections/virology , HIV-1/drug effects , Internationality , Developing Countries , Humans , Societies, Scientific , United States
6.
PLoS Pathog ; 13(1): e1006138, 2017 01.
Article in English | MEDLINE | ID: mdl-28046097

ABSTRACT

Asthma and chronic obstructive pulmonary disease (COPD) exacerbations are commonly associated with respiratory syncytial virus (RSV), rhinovirus (RV) and influenza A virus (IAV) infection. The ensuing airway inflammation is resistant to the anti-inflammatory actions of glucocorticoids (GCs). Viral infection elicits transforming growth factor-ß (TGF-ß) activity, a growth factor we have previously shown to impair GC action in human airway epithelial cells through the activation of activin-like kinase 5 (ALK5), the type 1 receptor of TGF-ß. In the current study, we examine the contribution of TGF-ß activity to the GC-resistance caused by viral infection. We demonstrate that viral infection of human bronchial epithelial cells with RSV, RV or IAV impairs GC anti-inflammatory action. Poly(I:C), a synthetic analog of double-stranded RNA, also impairs GC activity. Both viral infection and poly(I:C) increase TGF-ß expression and activity. Importantly, the GC impairment was attenuated by the selective ALK5 (TGFßRI) inhibitor, SB431542 and prevented by the therapeutic agent, tranilast, which reduced TGF-ß activity associated with viral infection. This study shows for the first time that viral-induced glucocorticoid-insensitivity is partially mediated by activation of endogenous TGF-ß.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Asthma/pathology , Glucocorticoids/pharmacology , Pulmonary Disease, Chronic Obstructive/pathology , Respiratory Mucosa/virology , Transforming Growth Factor beta/metabolism , Antiviral Agents/pharmacology , Asthma/virology , Benzamides/pharmacology , Cell Line , Dioxoles/pharmacology , Drug Resistance, Viral/physiology , Enzyme Activation , Epithelial Cells/virology , Humans , Influenza A virus , Influenza, Human/virology , Picornaviridae Infections/virology , Poly I-C/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Pulmonary Disease, Chronic Obstructive/virology , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Receptors, Transforming Growth Factor beta/metabolism , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Viruses , Rhinovirus , ortho-Aminobenzoates/pharmacology
7.
Pharm Stat ; 17(1): 74-89, 2018 02.
Article in English | MEDLINE | ID: mdl-29178367

ABSTRACT

The product limit or Kaplan-Meier (KM) estimator is commonly used to estimate the survival function in the presence of incomplete time to event. Application of this method assumes inherently that the occurrence of an event is known with certainty. However, the clinical diagnosis of an event is often subject to misclassification due to assay error or adjudication error, by which the event is assessed with some uncertainty. In the presence of such errors, the true distribution of the time to first event would not be estimated accurately using the KM method. We develop a method to estimate the true survival distribution by incorporating negative predictive values and positive predictive values, into a KM-like method of estimation. This allows us to quantify the bias in the KM survival estimates due to the presence of misclassified events in the observed data. We present an unbiased estimator of the true survival function and its variance. Asymptotic properties of the proposed estimators are provided, and these properties are examined through simulations. We demonstrate our methods using data from the Viral Resistance to Antiviral Therapy of Hepatitis C study.


Subject(s)
Computer Simulation/statistics & numerical data , Diagnostic Tests, Routine/statistics & numerical data , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Diagnostic Tests, Routine/standards , Drug Resistance, Viral/drug effects , Drug Resistance, Viral/physiology , Hepatitis C/drug therapy , Hepatitis C/epidemiology , Humans , Survival Analysis
8.
Nature ; 477(7362): 95-8, 2011 Aug 17.
Article in English | MEDLINE | ID: mdl-21849975

ABSTRACT

Latency and ongoing replication have both been proposed to explain the drug-insensitive human immunodeficiency virus (HIV) reservoir maintained during antiretroviral therapy. Here we explore a novel mechanism for ongoing HIV replication in the face of antiretroviral drugs. We propose a model whereby multiple infections per cell lead to reduced sensitivity to drugs without requiring drug-resistant mutations, and experimentally validate the model using multiple infections per cell by cell-free HIV in the presence of the drug tenofovir. We then examine the drug sensitivity of cell-to-cell spread of HIV, a mode of HIV transmission that can lead to multiple infection events per target cell. Infections originating from cell-free virus decrease strongly in the presence of antiretrovirals tenofovir and efavirenz whereas infections involving cell-to-cell spread are markedly less sensitive to the drugs. The reduction in sensitivity is sufficient to keep multiple rounds of infection from terminating in the presence of drugs. We examine replication from cell-to-cell spread in the presence of clinical drug concentrations using a stochastic infection model and find that replication is intermittent, without substantial accumulation of mutations. If cell-to-cell spread has the same properties in vivo, it may have adverse consequences for the immune system, lead to therapy failure in individuals with risk factors, and potentially contribute to viral persistence and hence be a barrier to curing HIV infection.


Subject(s)
Anti-Retroviral Agents/pharmacology , HIV Infections/virology , HIV-1/physiology , Virus Replication/physiology , Adenine/analogs & derivatives , Adenine/pharmacology , Cell Line , Drug Resistance, Viral/physiology , HEK293 Cells , HIV Infections/transmission , HIV-1/drug effects , Humans , Models, Biological , Organophosphonates/pharmacology , Tenofovir , Virus Replication/drug effects
9.
Biochemistry ; 55(10): 1529-40, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26892689

ABSTRACT

HIV-1 protease variants resist drugs by active and non-active-site mutations. The active-site mutations, which are the primary or first set of mutations, hamper the stability of the enzyme and resist the drugs minimally. As a result, secondary mutations that not only increase protein stability for unhindered catalytic activity but also resist drugs very effectively arise. While the mechanism of drug resistance of the active-site mutations is through modulating the active-site pocket volume, the mechanism of drug resistance of the non-active-site mutations is unclear. Moreover, how these allosteric mutations, which are 8-21 Å distant, communicate to the active site for drug efflux is completely unexplored. Results from molecular dynamics simulations suggest that the primary mechanism of drug resistance of the secondary mutations involves opening of the flexible protease flaps. Results from both residue- and community-based network analyses reveal that this precise action of protease is accomplished by the presence of robust communication paths between the mutational sites and the functionally relevant regions: active site and flaps. While the communication is more direct in the wild type, it traverses across multiple intermediate residues in mutants, leading to weak signaling and unregulated motions of flaps. The global integrity of the protease network is, however, maintained through the neighboring residues, which exhibit high degrees of conservation, consistent with clinical data and mutagenesis studies.


Subject(s)
Drug Resistance, Viral/physiology , HIV Protease/genetics , HIV Protease/metabolism , Mutation/physiology , Binding Sites/physiology , Drug Resistance, Viral/drug effects , Enzyme Activation/physiology , Gene Regulatory Networks/physiology , HIV Protease/chemistry , HIV Protease Inhibitors/chemistry , HIV Protease Inhibitors/metabolism , HIV Protease Inhibitors/pharmacology , HIV-1/drug effects , HIV-1/enzymology , Humans , Protein Structure, Secondary
10.
J Virol ; 89(17): 9068-79, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26109731

ABSTRACT

UNLABELLED: During HIV-1 infection of cells, the viral capsid plays critical roles in reverse transcription and nuclear entry of the virus. The capsid-targeting small molecule PF74 inhibits HIV-1 at early stages of infection. HIV-1 resistance to PF74 is complex, requiring multiple amino acid substitutions in the viral CA protein. Here we report the identification and analysis of a novel PF74-resistant mutant encoding amino acid changes in both domains of CA, three of which are near the pocket where PF74 binds. Interestingly, the mutant virus retained partial PF74 binding, and its replication was stimulated by the compound. The mutant capsid structure was not significantly perturbed by binding of PF74; rather, the mutations inhibited capsid interactions with CPSF6 and Nup153 and altered HIV-1 dependence on these host factors and on TNPO3. Moreover, the replication of the mutant virus was markedly impaired in activated primary CD4(+) T cells and macrophages. Our results suggest that HIV-1 escapes a capsid-targeting small molecule inhibitor by altering the virus's dependence on host factors normally required for entry into the nucleus. They further imply that clinical resistance to inhibitors targeting the PF74 binding pocket is likely to be strongly limited by functional constraints on HIV-1 evolution. IMPORTANCE: The HIV-1 capsid plays critical roles in early steps of infection and is an attractive target for therapy. Here we show that selection for resistance to a capsid-targeting small molecule inhibitor can result in viral dependence on the compound. The mutant virus was debilitated in primary T cells and macrophages--cellular targets of infection in vivo. The mutations also altered the virus's dependence on cellular factors that are normally required for HIV-1 entry into the nucleus. This work provides new information regarding mechanisms of HIV-1 resistance that should be useful in efforts to develop clinically useful drugs targeting the HIV-1 capsid.


Subject(s)
Capsid Proteins/genetics , Capsid/drug effects , Drug Resistance, Viral/physiology , HIV-1/drug effects , Indoles/pharmacology , Phenylalanine/analogs & derivatives , Amino Acid Substitution , Anti-HIV Agents/pharmacology , Binding Sites/genetics , CD4-Positive T-Lymphocytes/virology , Cell Line , HIV Infections/drug therapy , HIV Infections/metabolism , HIV-1/genetics , Host-Pathogen Interactions , Humans , Macrophages/virology , Molecular Chaperones/genetics , Nuclear Pore Complex Proteins/genetics , Nuclear Pore Complex Proteins/metabolism , Phenylalanine/pharmacology , Protein Binding/genetics , Protein Conformation , RNA Interference , RNA, Small Interfering , Virus Internalization/drug effects , Virus Replication/drug effects , beta Karyopherins/genetics , beta Karyopherins/metabolism , mRNA Cleavage and Polyadenylation Factors/metabolism
11.
BMC Infect Dis ; 16: 280, 2016 06 13.
Article in English | MEDLINE | ID: mdl-27296625

ABSTRACT

BACKGROUND: In resource-limited settings, where resistance testing is unavailable, confirmatory testing for patients with high viral loads (VL) delays antiretroviral therapy (ART) switches for persons with resistance. We developed a risk score algorithm to predict need for ART change by identifying resistance among persons with persistently elevated VL. METHODS: We analyzed data from a Phase IV open-label trial. Using logistic regression, we identified demographic and clinical characteristics predictive of need for ART change among participants with VLs ≥1000 copies/ml, and assigned model-derived scores to predictors. We designed three models, including only variables accessible in resource-limited settings. RESULTS: Among 290 participants with at least one VL ≥1000 copies/ml, 51 % (148/290) resuppressed and did not have resistance testing; among those who did not resuppress and had resistance testing, 47 % (67/142) did not have resistance and 53 % (75/142) had resistance (ART change needed for 25.9 % (75/290)). Need for ART change was directly associated with higher baseline VL and higher VL at time of elevated measure, and inversely associated with treatment duration. Other predictors included body mass index and adherence. Area under receiver operating characteristic curves ranged from 0.794 to 0.817. At a risk score ≥9, sensitivity was 14.7-28.0 % and specificity was 96.7-98.6 %. CONCLUSIONS: Our model performed reasonably well and may be a tool to quickly transition persons in need of ART change to more effective regimens when resistance testing is unavailable. Use of this algorithm may result in public health benefits and health system savings through reduced transmissions of resistant virus and costs on laboratory investigations.


Subject(s)
Algorithms , Anti-HIV Agents/therapeutic use , Drug Substitution/statistics & numerical data , HIV Infections/drug therapy , Risk Assessment , Viral Load , Adult , Aged , Area Under Curve , Drug Resistance, Viral/physiology , Female , HIV Infections/virology , HIV-1/physiology , Humans , Logistic Models , Male , Middle Aged , ROC Curve , Risk , Young Adult
12.
J Infect Dis ; 211(8): 1211-8, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25587020

ABSTRACT

BACKGROUND: Preexposure prophylaxis (PrEP) with emtricitabine plus tenofovir disoproxil fumarate (FTC/TDF) or TDF alone reduces the risk of human immunodeficiency virus (HIV) acquisition. Understanding the risk of antiretroviral resistance selected by PrEP during breakthrough infections is important because of the risk of treatment failure during subsequent antiretroviral use. METHODS: Within the largest randomized trial of FTC/TDF versus TDF as PrEP, plasma samples were tested for HIV with resistance mutations associated with FTC (K65R and M184IV) and TDF (K65R and K70E), using 454 sequencing. RESULTS: Of 121 HIV seroconverters, 25 received FTC/TDF, 38 received TDF, and 58 received placebo. Plasma drug levels in 26 individuals indicated PrEP use during or after HIV acquisition, of which 5 had virus with resistance mutations associated with their PrEP regimen. Among those with PrEP drug detected during infection, resistance was more frequent in the FTC/TDF arm (4 of 7 [57%]), compared with the TDF arm (1 of 19 [5.3%]; P = .01), owing to the FTC-associated mutation M184IV. Of these cases, 3 had unrecognized acute infection at PrEP randomization, and 2 were HIV negative at enrollment. CONCLUSIONS: These results suggest that resistance selected by PrEP is rare but can occur both with PrEP initiation during acute seronegative HIV infection and in PrEP breakthrough infections and that FTC is associated with a greater frequency of resistance mutations than TDF.


Subject(s)
Anti-HIV Agents/therapeutic use , Anti-Retroviral Agents/therapeutic use , Drug Resistance, Viral/physiology , HIV Infections/drug therapy , HIV-1/drug effects , Adenine/analogs & derivatives , Adenine/therapeutic use , Deoxycytidine/analogs & derivatives , Deoxycytidine/therapeutic use , Double-Blind Method , Emtricitabine , HIV Seropositivity/physiopathology , Humans , Organophosphonates/therapeutic use , Risk , Tenofovir
13.
Liver Int ; 35(2): 422-8, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25277773

ABSTRACT

BACKGROUND & AIMS: We evaluated the antiviral response of Asian or Pacific Islander (API) patients with chronic hepatitis B (CHB) who had baseline high viral load (HVL), defined as pre-treatment hepatitis B virus (HBV) DNA ≥9 log10 copies/ml, following up to 288 weeks of tenofovir disoproxil fumarate (TDF) treatment. METHODS: A total of 205 HBeAg-negative and HBeAg-positive self-described API patients received 48 weeks of TDF 300 mg (HVL n = 18) or adefovir dipivoxil 10 mg (HVL n = 15) in a blinded fashion, followed by open-label TDF for an additional 240 weeks. The proportions of HVL vs. non-HVL patients with HBV DNA <400 copies/ml were compared. Mean declines in HBV DNA were evaluated in API vs. non-API patients. RESULTS: Throughout the first 72 weeks of treatment, a smaller proportion of HVL API patients reached HBV DNA <400 copies/ml than non-HVL API patients. However, after this timepoint similar proportions of HVL and non-HVL API patients achieved HBV DNA <400 copies/ml (100% vs. 97%, respectively), which was maintained through week 288, where 92% of HVL patients and 99% of non-HVL API patients on treatment had HBV DNA <400 copies/ml. During the 288 weeks of treatment, API patients had similar mean HBV DNA declines as non-API patients, regardless of whether patients were HVL or non-HVL. No API HVL patient had persistent viremia at week 288. No resistance was detected among HVL or non-HVL patients. CONCLUSIONS: API patients with HVL CHB achieve HBV DNA <400 copies/ml with long-term TDF treatment; however, achieving viral suppression may take longer for HVL patients relative to non-HVL API patients.


Subject(s)
Adenine/analogs & derivatives , Hepatitis B, Chronic/drug therapy , Organophosphonates/therapeutic use , Adenine/pharmacology , Adenine/therapeutic use , Adult , Asian People , Drug Resistance, Viral/physiology , Humans , Native Hawaiian or Other Pacific Islander , Organophosphonates/pharmacology , Statistics, Nonparametric , Tenofovir , Viral Load/drug effects
14.
Mem Inst Oswaldo Cruz ; 109(7): 912-7, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25410995

ABSTRACT

After the World Health Organization officially declared the end of the first pandemic of the XXI century in August 2010, the influenza A(H1N1)pdm09 virus has been disseminated in the human population. In spite of its sustained circulation, very little on phylogenetic data or oseltamivir (OST) resistance is available for the virus in equatorial regions of South America. In order to shed more light on this topic, we analysed the haemagglutinin (HA) and neuraminidase (NA) genes of influenza A(H1N1)pdm09 positive samples collected during the pandemic period in the Pernambuco (PE), a northeastern Brazilian state. Complete HA sequences were compared and amino acid changes were related to clinical outcome. In addition, the H275Y substitution in NA, associated with OST resistance, was investigated by pyrosequencing. Samples from PE were grouped in phylogenetic clades 6 and 7, being clustered together with sequences from South and Southeast Brazil. The D222N/G HA gene mutation, associated with severity, was found in one deceased patient that was pregnant. Additionally, the HA mutation K308E, which appeared in Brazil in 2010 and was only detected worldwide the following year, was identified in samples from hospitalised cases. The resistance marker H275Y was not identified in samples tested. However, broader studies are needed to establish the real frequency of resistance in this Brazilian region.


Subject(s)
Hemagglutinins/genetics , Influenza A Virus, H1N1 Subtype/genetics , Influenza, Human/epidemiology , Neuraminidase/genetics , Pandemics , Antiviral Agents/therapeutic use , Biomarkers/analysis , Brazil/epidemiology , Drug Resistance, Viral/physiology , Female , Gene Frequency/genetics , Humans , Influenza A Virus, H1N1 Subtype/classification , Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza, Human/virology , Mutation/genetics , Oseltamivir/therapeutic use , Phylogeny , Pregnancy , RNA, Viral/analysis , Sequence Analysis, DNA/methods , Virulence , Virulence Factors/genetics
15.
Vopr Virusol ; 59(6): 32-5, 2014.
Article in Russian | MEDLINE | ID: mdl-25929034

ABSTRACT

The activity of the phosphite of acycloguanosine (P-ACG) and six antivirals was tested individually and in double and triple combinations on two strains of the herpes simplex virus (HSV) type 1 (sensitive to acyclovir and resistant to acyclovir) using the CPE inhibition method in the Vero E6 cell microcultures. These are: phosphite of acycloguanosine (P-ACG), Ara-A, cidofovir (CDV), ribavirin (Rib), phosphonoformate (PFA), glycyrrhizic acid (GLN) and alpha-interferon (alpha-IFN). All studied double combinations and triple combinations including P-ACV inhibited replication of both HSV strains more effectively than any drug alone. Various types of interactions depending on the virus type were observed in both viral models: synergistic (double combinations P-ACG with PFA, CDV, Rib, alpha-IFN and triple combinations P-ACG with alpha-IFN +PFA, alpha-IFN +AraA, alpha-IFN +CDV, PFA+CDV, PFA+Rib, CDV+AraA, CDV+Rib, CDV+GLN,PFA+AraA) and additive (P-ACG with AraA and PFA+GLN). Neither antagonism nor interference was noted for any combinations. Adduced results suggest that these combinations might be clinically useful for the treatment of certain herpes simplex virus type 1 infections.


Subject(s)
Acyclovir/pharmacology , Antiviral Agents/pharmacology , Herpesvirus 1, Human/drug effects , Animals , Chlorocebus aethiops , Cidofovir , Cytosine/analogs & derivatives , Cytosine/pharmacology , Drug Resistance, Viral/physiology , Drug Synergism , Drug Therapy, Combination , Foscarnet/pharmacology , Glycyrrhizic Acid/pharmacology , Herpesvirus 1, Human/physiology , Humans , Interferon-alpha/pharmacology , Organophosphonates/pharmacology , Phosphites , Ribavirin/pharmacology , Vero Cells , Vidarabine/pharmacology , Virus Replication/drug effects
16.
Biochemistry ; 52(45): 8150-64, 2013 Nov 12.
Article in English | MEDLINE | ID: mdl-24128064

ABSTRACT

The emergence of influenza drug resistance is a major public health concern. The molecular basis of resistance to oseltamivir (Tamiflu) is investigated using a computational assay involving multiple 500 ns unrestrained molecular dynamics (MD) simulations of oseltamivir complexed with mutants of H1N1-2009 influenza neuraminidase. The simulations, accelerated using graphics processors (GPUs), and using a fully explicit model of water, are of sufficient length to observe multiple drug unbinding and rebinding events. Drug unbinding occurs during simulations of known oseltamivir-resistant mutants of neuraminidase. Molecular-level rationalizations of drug resistance are revealed by analysis of these unbinding trajectories, with particular emphasis on the dynamics of the mutant residues. The results indicate that MD simulations can predict weakening of binding associated with drug resistance. In addition, visualization and analysis of binding site water molecules reveal their importance in stabilizing the binding mode of the drug. Drug unbinding is accompanied by conformational changes, driven by the mutant residues, which results in flooding of a key pocket containing tightly bound water molecules. This displaces oseltamivir, allowing the tightly bound water molecules to be released into bulk. In addition to the role of water, analysis of the trajectories reveals novel behavior of the structurally important 150-loop. Motion of the loop, which can move between an open and closed conformation, is intimately associated with drug unbinding and rebinding. Opening of the loop occurs coincidentally with drug unbinding, and interactions between oseltamivir and the loop seem to aid in the repositioning of the drug back into an approximation of its original binding mode on rebinding. The similarity of oseltamivir to a transition state analogue for neuraminidase suggests that the dynamics of the loop could play an important functional role in the enzyme, with loop closing aiding in binding of the substrate and loop opening aiding the release of the product.


Subject(s)
Antiviral Agents/pharmacology , Neuraminidase/metabolism , Oseltamivir/pharmacology , Binding Sites , Drug Resistance, Viral/genetics , Drug Resistance, Viral/physiology , Humans , Molecular Dynamics Simulation , Mutation , Neuraminidase/genetics , Protein Binding , Thermodynamics
17.
J Biol Chem ; 287(11): 8297-309, 2012 Mar 09.
Article in English | MEDLINE | ID: mdl-22235115

ABSTRACT

Peptides corresponding to N- and C-terminal heptad repeat regions (HR1 and HR2, respectively) of viral fusion proteins can block infection of viruses in a dominant negative manner by interfering with refolding of the viral HR1 and HR2 to form a six-helix bundle (6HB) that drives fusion between viral and host cell membranes. The 6HB of the HIV gp41 (endogenous bundle) consists of an HR1 coiled-coil trimer with grooves lined by antiparallel HR2 helices. HR1 peptides form coiled-coil oligomers that may bind to gp41 HR2 as trimers to form a heterologous 6HB (inhibitor bundle) or to gp41 HR1 as monomers or dimers to form a heterologous coiled coil. To gain insights into mechanisms of Env entry and inhibition by HR1 peptides, we compared resistance to a peptide corresponding to 36 residues in gp41 HR1 (N36) and the same peptide with a coiled-coil trimerization domain fused to its N terminus (IZN36) that stabilizes the trimer and increases inhibitor potency (Eckert, D. M., and Kim, P. S. (2001) Proc. Nat. Acad. Sci. U.S.A. 98, 11187-11192). Whereas N36 selected two genetic pathways with equal probability, each defined by an early mutation in either HR1 or HR2, IZN36 preferentially selected the HR1 pathway. Both pathways conferred cross-resistance to both peptides. Each HR mutation enhanced the thermostability of the endogenous 6HB, potentially allowing the virus to simultaneously escape inhibitors targeting either gp41 HR1 or HR2. These findings inform inhibitor design and identify regions of plasticity in the highly conserved gp41 that modulate virus entry and escape from HR1 peptide inhibitors.


Subject(s)
Drug Design , Drug Resistance, Viral/physiology , HIV Envelope Protein gp41/antagonists & inhibitors , HIV Fusion Inhibitors/chemistry , HIV-1/metabolism , Peptides/chemistry , Virus Internalization , Cell Line , Drug Resistance, Viral/drug effects , HIV Envelope Protein gp41/genetics , HIV Envelope Protein gp41/metabolism , HIV Fusion Inhibitors/pharmacology , HIV-1/genetics , Humans , Mutation , Peptides/genetics , Peptides/pharmacology , Protein Structure, Secondary
18.
Biochim Biophys Acta ; 1820(10): 1526-34, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22698669

ABSTRACT

BACKGROUND: ITMN-191 (RG7227, Danoprevir), as a potential inhibitor of the NS3/4A protease of hepatitis C virus, has been in phase 2 clinical trial. Unfortunately, several ITMN-191 resistance mutants including R155K, A156V, and D168A/E have been identified. METHODS: Molecular dynamics simulation, binding free energy calculation and per-residue energy decomposition were employed to explore the binding and resistance mechanism of hepatitis C virus NS3/4A protease to ITMN-191. RESULTS: Based on molecular dynamics simulation and per-residue energy decomposition, the nonpolar energy term was found to be the driving force for ITMN-191 binding. For the studied R155K, A156V, D168A/E mutants, the origin of resistance is mainly from the conformational changes of the S4 and extended S2 binding pocket induced by the studied mutants and further leading to the reduced binding ability to the extended P2 and P4 moieties of ITMN-191. CONCLUSIONS: Further structural analysis indicates that the destruction of conservative salt bridges between residues 168 and 155 should be responsible for the large conformation changes of the binding pocket in R155K and D168A/E mutants. For A156V mutation, the occurrence of drug resistance is mainly from the changed binding pocket by a replacement of one bulky residue Val. GENERAL SIGNIFICANCE: The obtained drug resistance mechanism of this study will provide useful guidance for the development of new and effective HCV NS3/4A inhibitors with low resistance.


Subject(s)
Drug Resistance, Viral/genetics , Hepacivirus/genetics , Lactams/pharmacology , Mutation, Missense/physiology , Sulfonamides/pharmacology , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/genetics , Alanine/chemistry , Alanine/genetics , Amino Acid Substitution/physiology , Antiviral Agents/pharmacology , Arginine/chemistry , Arginine/genetics , Aspartic Acid/chemistry , Aspartic Acid/genetics , Comprehension , Computational Biology , Cyclopropanes , Drug Resistance, Viral/physiology , Glutamic Acid/chemistry , Glutamic Acid/genetics , Hepacivirus/chemistry , Hepacivirus/enzymology , Isoindoles , Lactams, Macrocyclic , Lysine/chemistry , Lysine/genetics , Models, Molecular , Molecular Dynamics Simulation , Proline/analogs & derivatives , Valine/chemistry , Valine/genetics , Viral Nonstructural Proteins/physiology
19.
Gastroenterology ; 143(2): 429-38.e8, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22522091

ABSTRACT

BACKGROUND & AIMS: Hepatitis C virus (HCV) is a common cause of chronic liver disease. Many patients do not clear the viral infection; little is known about the mechanisms of HCV persistence or the frequent failure of interferon (IFN) to eliminate it. Better culture systems are needed to study viral replication in quiescent liver cells. METHODS: We used human hepatoma (Huh7.5) cells and those that had undergone proliferation arrest and differentiation (Huh7.5(dif)) to study the persistence of HCV infection following exposure of the cells to IFN-α and to compare the antiviral effects of IFN-α and IFN-λ. We validated these results with primary human hepatocytes and Huh7 cells that expressed an IFN-inducible fluorophore. RESULTS: Following infection of Huh7.5(dif) cells, HCV replicated persistently and released infectious particles. Long-term exposure of the cells to IFN-α reduced HCV replication ∼1000-fold but did not eliminate the virus; viral replication rebounded after withdrawal of IFN, as it does in patients with chronic HCV infection. HCV replicated at higher levels, but not exclusively, in cells that had a low level of response to IFN-α. Following incubation of cells with equipotent concentrations of IFN-α or IFN-λ, Huh7.5(dif) cells expressed a wider pattern of IFN-stimulated genes than undifferentiated Huh7.5 cells or primary human hepatocytes, indicating that the antiviral response depends on the differentiation status of the cells. CONCLUSIONS: We developed a cell culture system using hepatoma cells to study persistent HCV infection during the type I or type III IFN-induced antiviral response. The level and range of the antiviral responses were associated with the differentiation status of the cells. We propose that HCV exploits the stochastic nature of the response of hepatocytes to IFN to sustain persistence.


Subject(s)
Antiviral Agents/pharmacology , Drug Resistance, Viral/physiology , Hepacivirus/drug effects , Hepatocytes/virology , Interferon-alpha/pharmacology , Interleukins/pharmacology , Biomarkers/metabolism , Cell Differentiation , Cell Line, Tumor , GTP-Binding Proteins/metabolism , Hepacivirus/physiology , Hepatocytes/cytology , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Interferons , Myxovirus Resistance Proteins , Virus Replication
20.
J Antimicrob Chemother ; 68(1): 200-5, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22915463

ABSTRACT

OBJECTIVES: To explore the durability of three first-line tenofovir/emtricitabine-based regimens in combination with atazanavir/ritonavir, efavirenz or lopinavir/ritonavir in HIV-1-infected patients. PATIENTS AND METHODS: A retrospective, longitudinal, multicentre analysis of adult patients enrolled in the Antiretroviral Resistance Cohort Analysis (ARCA), a national prospective observational cohort of HIV-1-infected patients followed up at more than 100 clinical and laboratory units in Italy. Patients eligible were those starting first-line antiretroviral therapy between 1 June 2004 and 15 April 2011 and who were followed up for at least 6 months. The primary endpoint was durability, defined as the time from antiretroviral therapy initiation to first treatment modification. Time-dependent events were analysed by the Kaplan-Meier approach and the Cox proportional hazard model. RESULTS: There are 26,000 HIV-infected patients in the ARCA database, of whom 1654 met study inclusion criteria. Six hundred and thirty-nine (38.6%) received efavirenz, 321 (19.4%) received atazanavir/ritonavir and 694 (41.9%) received lopinavir/ritonavir as a first-line regimen. Over a total observation period of 88 months, equivalent to more than 2805 person-years of follow-up, 618 patients underwent treatment modification. Lopinavir/ritonavir, given twice daily, was associated with a higher discontinuation rate than efavirenz- and atazanavir-based regimens [hazard ratio (HR) 1.83, 95% confidence interval (CI) 1.56-2.15, P = 0.001]. Comparing the once-daily regimens, the rate of discontinuation of efavirenz was higher than that of atazanavir/ritonavir (HR 1.39, 95% CI 1.06-1.83, P = 0.016). CONCLUSIONS: Significant differences in treatment duration were observed among the three studied regimens. Once-daily regimens exhibited greater durability than the twice-daily regimen. Among the specific regimens examined, tenofovir/emtricitabine plus atazanavir/ritonavir showed the greatest durability.


Subject(s)
Adenine/analogs & derivatives , Benzoxazines/administration & dosage , Deoxycytidine/analogs & derivatives , Lopinavir/administration & dosage , Oligopeptides/administration & dosage , Organophosphonates/administration & dosage , Pyridines/administration & dosage , Ritonavir/administration & dosage , Adenine/administration & dosage , Alkynes , Anti-Retroviral Agents/administration & dosage , Atazanavir Sulfate , Cohort Studies , Cyclopropanes , Deoxycytidine/administration & dosage , Drug Resistance, Viral/drug effects , Drug Resistance, Viral/physiology , Drug Therapy, Combination , Emtricitabine , HIV Infections/drug therapy , HIV Infections/epidemiology , HIV-1/drug effects , Humans , Italy/epidemiology , Longitudinal Studies , Prospective Studies , Retrospective Studies , Tenofovir , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL