Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.042
Filtrer
1.
Viruses ; 16(4)2024 03 22.
Article de Anglais | MEDLINE | ID: mdl-38675834

RÉSUMÉ

Tenofovir (TFV) is the active form of the prodrugs tenofovir disoproxil fumarate (TDF) and tenofovir alafenamide (TAF), both clinically prescribed as HIV reverse transcriptase inhibitors. The biophysical interactions between these compounds and human serum albumin (HSA), the primary carrier of exogenous compounds in the human bloodstream, have not yet been thoroughly characterized. Thus, the present study reports the interaction profile between HSA and TFV, TDF, and TAF via UV-Vis, steady-state, and time-resolved fluorescence techniques combined with isothermal titration calorimetry (ITC) and in silico calculations. A spontaneous interaction in the ground state, which does not perturb the microenvironment close to the Trp-214 residue, is classified as weak. In the case of HSA/TFV and HSA/TDF, the binding is both enthalpically and entropically driven, while for HSA/TAF, the binding is only entropically dominated. The binding constant (Ka) and thermodynamic parameters obtained via ITC assays agree with those obtained using steady-state fluorescence quenching measurements, reinforcing the reliability of the data. The small internal cavity known as site I is probably the main binding pocket for TFV due to the low steric volume of the drug. In contrast, most external sites (II and III) can better accommodate TAF due to the high steric volume of this prodrug. The cross-docking approach corroborated experimental drug-displacement assays, indicating that the binding affinity of TFV and TAF might be impacted by the presence of different compounds bound to albumin. Overall, the weak binding capacity of albumin to TFV, TDF, and TAF is one of the main factors for the low residence time of these antiretrovirals in the human bloodstream; however, positive cooperativity for TAF and TDF was detected in the presence of some drugs, which might improve their residence time (pharmacokinetic profile).


Sujet(s)
Agents antiVIH , Liaison aux protéines , Inhibiteurs de la transcriptase inverse , Sérum-albumine humaine , Ténofovir , Ténofovir/analogues et dérivés , Humains , Inhibiteurs de la transcriptase inverse/métabolisme , Inhibiteurs de la transcriptase inverse/composition chimique , Ténofovir/métabolisme , Ténofovir/composition chimique , Sérum-albumine humaine/métabolisme , Sérum-albumine humaine/composition chimique , Agents antiVIH/métabolisme , Thermodynamique , Calorimétrie , Sites de fixation , Infections à VIH/virologie , Infections à VIH/traitement médicamenteux , Alanine/métabolisme , Transcriptase inverse du VIH/métabolisme , Transcriptase inverse du VIH/composition chimique
2.
Chem Pharm Bull (Tokyo) ; 72(1): 41-47, 2024.
Article de Anglais | MEDLINE | ID: mdl-38171903

RÉSUMÉ

The capsid of human immunodeficiency virus type 1 (HIV-1) forms a conical structure by assembling oligomers of capsid (CA) proteins and is a virion shell that encapsulates viral RNA. The inhibition of the CA function could be an appropriate target for suppression of HIV-1 replication because the CA proteins are highly conserved among many strains of HIV-1, and the drug targeting CA, lenacapavir, has been clinically developed by Gilead Sciences, Inc. Interface hydrophobic interactions between two CA molecules via the Trp184 and Met185 residues in the CA sequence are indispensable for conformational stabilization of the CA multimer. Our continuous studies found two types of small molecules with different scaffolds, MKN-1 and MKN-3, designed by in silico screening as a dipeptide mimic of Trp184 and Met185 have significant anti-HIV-1 activity. In the present study, MKN-1 derivatives have been designed and synthesized. Their structure-activity relationship studies found some compounds having potent anti-HIV activity. The present results should be useful in the design of novel CA-targeting molecules with anti-HIV activity.


Sujet(s)
Agents antiVIH , VIH-1 (Virus de l'Immunodéficience Humaine de type 1) , Humains , Protéines de capside/composition chimique , Protéines de capside/génétique , Protéines de capside/métabolisme , Assemblage viral , Capside/métabolisme , Agents antiVIH/pharmacologie , Agents antiVIH/composition chimique , Agents antiVIH/métabolisme
3.
Viruses ; 15(11)2023 Oct 31.
Article de Anglais | MEDLINE | ID: mdl-38005879

RÉSUMÉ

Norovirus is the leading cause of viral gastroenteritis worldwide, and there are no approved vaccines or therapeutic treatments for chronic or severe norovirus infections. The structural characterisation of the norovirus protease and drug development has predominantly focused upon GI.1 noroviruses, despite most global outbreaks being caused by GII.4 noroviruses. Here, we determined the crystal structures of the GII.4 Sydney 2012 ligand-free norovirus protease at 2.79 Å and at 1.83 Å with a covalently bound high-affinity (IC50 = 0.37 µM) protease inhibitor (NV-004). We show that the active sites of the ligand-free protease structure are present in both open and closed conformations, as determined by their Arg112 side chain orientation. A comparative analysis of the ligand-free and ligand-bound protease structures reveals significant structural differences in the active site cleft and substrate-binding pockets when an inhibitor is covalently bound. We also report a second molecule of NV-004 non-covalently bound within the S4 substrate binding pocket via hydrophobic contacts and a water-mediated hydrogen bond. These new insights can guide structure-aided drug design against the GII.4 genogroup of noroviruses.


Sujet(s)
Agents antiVIH , Infections à Caliciviridae , Norovirus , Humains , Peptide hydrolases/métabolisme , Norovirus/métabolisme , Endopeptidases/métabolisme , Domaine catalytique , Agents antiVIH/métabolisme , Génotype , Phylogenèse
4.
Biomed Pharmacother ; 168: 115750, 2023 Dec.
Article de Anglais | MEDLINE | ID: mdl-37871555

RÉSUMÉ

BACKGROUND: Dual therapy (DT) has shown comparable results to triple therapy (TT) in efficacy and other immunological aspects. However, there are still some concerns about DT, including several immunological features. Therefore, we evaluated whether HIV-1-specific memory T-cell responses and exhaustion phenotypes are adversely influenced after simplification to DT. METHODS: HIV-1-specific CD4+ and CD8+ T-cell responses were assessed by intracellular cytokine and degranulation marker staining, and polyfunctionality indexes after stimulation with a Gag peptide pool. Exhaustion phenotypes were evaluated by PD-1, TIM-3, and LAG-3 expression in CD4+ and CD8+ T cells. RESULTS: Forty participants in the TRIDUAL trial (ClinicalTrials.gov: NCT03447873) who were randomized to continue integrase inhibitor-based TT (n = 20) or to switch to DT (dolutegravir or darunavir/cobicistat plus lamivudine) (n = 20). After 96 weeks, the magnitude of CD4+ and CD8+ T-cell responses was similar in both treatment arms (p = 0.221 and p = 0.602, respectively). The CD4+ polyfunctionality index decreased in the TT arm (p = 0.013) and remained stable in the DT arm, while the polyfunctionality of CD8+ T cells was unchanged in both arms. There was a significant decrease in the expression of PD-1, TIM-3, and the co-expression of PD-1+TIM-3+LAG-3+, and PD-1 +TIM-3 + in both CD4+ and CD8+ T cells. However, the decrease in the expression of exhaustion markers did not improve HIV-1-specific T-cell responses. CONCLUSIONS: Our results suggest that simplification to DT does not negatively influence the HIV-1-specific T-cell response or the exhaustion phenotype after 96 weeks of follow-up.


Sujet(s)
Agents antiVIH , Infections à VIH , VIH-1 (Virus de l'Immunodéficience Humaine de type 1) , Humains , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/génétique , Récepteur cellulaire-2 du virus de l'hépatite A/métabolisme , Lymphocytes T CD8+ , Inhibiteurs de l'intégrase/métabolisme , Inhibiteurs de l'intégrase/usage thérapeutique , Infections à VIH/traitement médicamenteux , Récepteur-1 de mort cellulaire programmée/métabolisme , Agents antiVIH/usage thérapeutique , Agents antiVIH/métabolisme
5.
Thromb Res ; 229: 53-68, 2023 09.
Article de Anglais | MEDLINE | ID: mdl-37413892

RÉSUMÉ

BACKGROUND: HIV-infected individuals are known to be at higher risk for thrombotic cardiovascular disease (CVD), which may also be differentially affected by components of anti-HIV drugs. To identify the effects of a series of FDA-approved anti-HIV drugs on platelet aggregation in humans, focusing on the novel pharmacological effects of rilpivirine (RPV), a reverse transcriptase inhibitor, on platelet function both in vitro and in vivo and the mechanisms involved. METHODS AND RESULTS: In vitro studies showed that RPV was the only anti-HIV reagent that consistently and efficiently inhibited aggregation elicited by different agonists, exocytosis, morphological extension on fibrinogen, and clot retraction. Treatment of mice with RPV significantly prevented thrombus formation in FeCl3-injured mesenteric vessels, postcava with stenosis surgery, and ADP -induced pulmonary embolism models without defects in platelet viability, tail bleeding, and coagulation activities. RPV also improved cardiac performance in mice with post-ischemic reperfusion. A mechanistic study revealed that RPV preferentially attenuated fibrinogen-stimulated Tyr773 phosphorylation of ß3-integrin by inhibiting Tyr419 autophosphorylation of c-Src. Molecular docking and surface plasmon resonance analyses showed that RPV can bind directly to c-Src. Further mutational analysis showed that the Phe427 residue of c-Src is critical for RPV interaction, suggesting a novel interaction site for targeting c-Src to block ß3-integrin outside-in signaling. CONCLUSION: These results demonstrated that RPV was able to prevent the progression of thrombotic CVDs by interrupting ß3-integrin-mediated outside-in signaling via inhibiting c-Src activation without hemorrhagic side effects, highlighting RPV as a promising reagent for the prevention and therapy of thrombotic CVDs.


Sujet(s)
Agents antiVIH , Thrombose , Humains , Souris , Animaux , Intégrine bêta3/métabolisme , Phosphorylation , Rilpivirine/métabolisme , Rilpivirine/pharmacologie , Complexe glycoprotéique IIb-IIIa de la membrane plaquettaire/métabolisme , Repositionnement des médicaments , Simulation de docking moléculaire , Plaquettes/métabolisme , Thrombose/traitement médicamenteux , Thrombose/prévention et contrôle , Thrombose/métabolisme , Agents antiVIH/métabolisme , Agents antiVIH/pharmacologie , Fibrinogène/métabolisme
6.
Viruses ; 15(5)2023 04 23.
Article de Anglais | MEDLINE | ID: mdl-37243126

RÉSUMÉ

Human immunodeficiency virus type 1 (HIV-1) is characterized by high variability and drug resistance. This has necessitated the development of antivirals with a new chemotype and therapy. We previously identified an artificial peptide with non-native protein sequence, AP3, with the potential to inhibit HIV-1 fusion through targeting hydrophobic grooves on the N-terminal heptad repeat trimer of viral glycoprotein gp41. Here, a small-molecule HIV-1 inhibitor targeting chemokine coreceptor CCR5 on the host cell was integrated into the AP3 peptide, producing a novel dual-target inhibitor with improved activity against multiple HIV-1 strains including those resistant to the currently used anti-HIV-1 drug enfuvirtide. Its superior antiviral potency in comparison with the respective pharmacophoric moieties is in consonance with the dual binding of viral gp41 and host factor CCR5. Therefore, our work provides a potent artificial peptide-based bifunctional HIV-1 entry inhibitor and highlights the multitarget-directed ligands approach in the development of novel therapeutic anti-HIV-1 agents.


Sujet(s)
Agents antiVIH , Inhibiteurs de fusion du VIH , VIH-1 (Virus de l'Immunodéficience Humaine de type 1) , Humains , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/métabolisme , Protéine d'enveloppe gp41 du VIH/composition chimique , Inhibiteurs de fusion du VIH/pharmacologie , Inhibiteurs de fusion du VIH/composition chimique , Peptides/pharmacologie , Peptides/métabolisme , Agents antiVIH/pharmacologie , Agents antiVIH/métabolisme , Glycoprotéines/métabolisme , Membrane cellulaire/métabolisme , Récepteurs CCR5/métabolisme
7.
J Comput Chem ; 44(13): 1291-1299, 2023 05 15.
Article de Anglais | MEDLINE | ID: mdl-36751051

RÉSUMÉ

The frequent outbreaks of the AIDS (Acquired Immune Deficiency Syndrome) pandemic and the limited availability of anti-Human Immunodeficiency Virus (HIV) drugs highlight the urgent need to develop new antiviral drugs. A detailed understanding of the interactions between TAR-Binding Proteins (TBP) and RNA will facilitate the discovery of new anti-AIDS drugs. In order to characterize and explore the key interactions between RNA and TBP, we focused on the wild type (WT) and three mutant TBPs (TBP6.9, TBP6.7, and TBP6.3) with RNA, multiple molecular dynamics simulation and energy computation were performed. The results showed that 12 key residues played a major role in the interaction between TBP and RNA. The mutated residues of TBP changed the interaction between their surrounding residues and RNA, thus affecting the binding of TBP to RNA. In addition, structural and energy analyses showed that in contrast with WT TBP-RNA complex, the mutated residues had little effect on the backbone structure of TBP, but changes in the van der Waals interactions and electrostatic interaction associated with the side chains are responsible for the altered the binding between three mutant TBPs and RNA complexes. The discovery of TBP-RNA recognition mechanism in our work provides some useful insights and new opportunities for the development of anti-aids drugs.


Sujet(s)
Agents antiVIH , VIH-1 (Virus de l'Immunodéficience Humaine de type 1) , Simulation de dynamique moléculaire , ARN/métabolisme , Agents antiVIH/métabolisme
8.
J Virol ; 96(20): e0114822, 2022 10 26.
Article de Anglais | MEDLINE | ID: mdl-36197106

RÉSUMÉ

Long interspersed element type 1 (LINE-1) is the only known type of retroelement that can replicate autonomously, and its retrotransposition activity can trigger interferon (IFN) production. IFN production suppresses the infectivity of exogenous viruses, such as human immunodeficiency virus (HIV). As a counteraction, HIV has been reported to use multiple proteins and mechanisms to suppress LINE-1 replication. However, the mechanisms of HIV-mediated LINE-1 regulation are not fully understood. In this study, we discovered that Nef protein, which is expressed by HIV and is important for HIV pathogenesis, inhibits LINE-1 retrotransposition. Two distinct mechanisms have been uncovered for Nef-induced LINE-1 suppression. Without direct interaction with LINE-1 DNA, Nef potently inhibits the promoter activity of the LINE-1 5'-untranslated region (5'-UTR) and reduces the expression levels of LINE-1 RNA and proteins. Alternatively, although Nef does not bind to the LINE-1 open reading frame 1 protein (ORF1p) or LINE-1 RNA, it significantly compromises the ORF1p-LINE-1 RNA interaction, which is essential for LINE-1 retrotransposition. Both mechanisms can be suppressed by the G2A mutation, which abolishes myristoylation of Nef, suggesting that membrane attachment is essential for Nef to suppress LINE-1. Consequently, through LINE-1 inhibition, Nef downregulates IFN production in host cells. Therefore, our data revealed that Nef is a potent LINE-1 suppressor and an effective innate immune regulator, which not only provides new information on the intricate interaction between HIV, LINE-1, and IFN signaling systems but also strengthens the importance of Nef in HIV infection and highlights the potential of designing novel Nef-targeting anti-HIV drugs. IMPORTANCE Human immunodeficiency viruses are pathogens of AIDS that were first discovered almost 40 years ago and continue to threaten human lives to date. While currently used anti-HIV drugs are sufficient to suppress viral loads in HIV-infected patients, both drug-resistant HIV strains and adverse side effects triggered by the long-term use of these drugs highlight the need to develop novel anti-HIV drugs targeting different viral proteins and/or different steps in viral replication. To achieve this, more information is required regarding HIV pathogenesis and especially its impact on cellular activities in host cells. In this study, we discovered that the Nef protein expressed by HIV potently inhibits LINE-1 retrotransposition. During our attempt to determine the mechanism of Nef-mediated LINE-1 suppression, two additional functions of Nef were uncovered. Nef effectively repressed the promoter activity of LINE-1 5'-UTR and destabilized the interaction between ORF1p and LINE-1 RNA. Consequently, Nef not only compromises LINE-1 replication but also reduces LINE-1-triggered IFN production. The reduction in IFN production, in theory, promotes HIV infectivity. Together with its previously known functions, these findings indicate that Nef is a potential target for the development of novel anti-HIV drugs. Notably, the G2 residue, which has been reported to be essential for most Nef functions, was found to be critical in the regulation of innate immune activation by Nef, suggesting that compromising myristoylation or membrane attachment of Nef may be a good strategy for the inhibition of HIV infection.


Sujet(s)
Agents antiVIH , Infections à VIH , VIH-1 (Virus de l'Immunodéficience Humaine de type 1) , Humains , Produits du gène nef du virus de l'immunodéficience humaine/génétique , Produits du gène nef du virus de l'immunodéficience humaine/métabolisme , Rétroéléments/génétique , Produits du gène nef/génétique , Agents antiVIH/métabolisme , Interférons/métabolisme , ARN/métabolisme , Régions non traduites
9.
J Med Chem ; 65(3): 2458-2470, 2022 02 10.
Article de Anglais | MEDLINE | ID: mdl-35061384

RÉSUMÉ

Here, we report the design, synthesis, structure-activity relationship studies, antiviral activity, enzyme inhibition, and druggability evaluation of dihydrofuro[3,4-d]pyrimidine derivatives as a potent class of HIV-1 non-nucleoside reverse transcriptase inhibitors (NNRTIs). Compounds 14b (EC50 = 5.79-28.3 nM) and 16c (EC50 = 2.85-18.0 nM) exhibited superior potency against a panel of HIV-1-resistant strains. Especially, for the changeling mutations F227L/V106A and K103N/Y181C, both compounds exhibited remarkably improved activity compared to those of etravirine and rilpivirine. Moreover, 14b and 16c showed moderate RT enzyme inhibition (IC50 = 0.14-0.15 µM), which demonstrated that they acted as HIV-1 NNRTIs. Furthermore, 14b and 16c exhibited favorable pharmacokinetic and safety properties, making them excellent leads for further development.


Sujet(s)
Agents antiVIH/pharmacologie , Furanes/pharmacologie , Transcriptase inverse du VIH/antagonistes et inhibiteurs , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/effets des médicaments et des substances chimiques , Pyrimidines/pharmacologie , Inhibiteurs de la transcriptase inverse/pharmacologie , Animaux , Agents antiVIH/synthèse chimique , Agents antiVIH/métabolisme , Agents antiVIH/pharmacocinétique , Conception de médicament , Femelle , Furanes/synthèse chimique , Furanes/métabolisme , Furanes/pharmacocinétique , Transcriptase inverse du VIH/génétique , Transcriptase inverse du VIH/métabolisme , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/enzymologie , Mâle , Souris , Simulation de docking moléculaire , Structure moléculaire , Mutation , Liaison aux protéines , Pyrimidines/synthèse chimique , Pyrimidines/métabolisme , Pyrimidines/pharmacocinétique , Rat Sprague-Dawley , Inhibiteurs de la transcriptase inverse/synthèse chimique , Inhibiteurs de la transcriptase inverse/métabolisme , Inhibiteurs de la transcriptase inverse/pharmacocinétique , Relation structure-activité
10.
Pharmacogenomics ; 23(3): 157-159, 2022 02.
Article de Anglais | MEDLINE | ID: mdl-35023374

RÉSUMÉ

Tweetable abstract Pharmacogenetic tests are a promising strategy to improve safety and effectiveness of HIV therapy. However, implementation can be challenging in populations with complex genetic structure.


Sujet(s)
Agents antiVIH/usage thérapeutique , Infections à VIH/traitement médicamenteux , Agents antiVIH/métabolisme , Infections à VIH/génétique , Humains , Test pharmacogénomique , Résultat thérapeutique
11.
J Med Chem ; 65(3): 2122-2138, 2022 02 10.
Article de Anglais | MEDLINE | ID: mdl-35073089

RÉSUMÉ

A series of novel heteroaromatic biphenyl-methyl-pyrimidine analogues were designed via hybridization of privileged structures of two HIV-1 inhibitors. Among them, compound 7a containing 4-pyridinyl-phenyl and methyl-pyrimidine fragments revealed excellent wild-type HIV-1 inhibitory activity with low cytotoxicity. 7a had favorable solubility and liver microsome stability; moreover, no apparent CYP enzymatic inhibitory activity or acute toxicity was observed. However, its inhibitory activity toward mutant strains and the pharmacokinetic (PK) profiles were still unsatisfactory. Further optimizations resulted in a highly potent compound 9d without methyl on the pyrimidine but a heteroaromatic dimethyl-biphenyl on the left rings of difluoro-pyridinyl-diarylpyrimidines (DAPYs). A broad-spectrum activity (EC50 = 2.0-57 nM) of 9d against resistant strains was revealed. This compound also exhibited good solubility and safety profiles and a good PK profile with an oral bioavailability of 59% in rats. Collectively, these novel heteroaromatic dimethyl-biphenyl-DAPYs represent promising drug candidates for HIV clinical therapy.


Sujet(s)
Agents antiVIH/composition chimique , Pyridines/composition chimique , Pyrimidines/composition chimique , Animaux , Agents antiVIH/métabolisme , Agents antiVIH/pharmacocinétique , Agents antiVIH/pharmacologie , Sites de fixation , Survie cellulaire/effets des médicaments et des substances chimiques , Cytochrome P-450 enzyme system/composition chimique , Cytochrome P-450 enzyme system/métabolisme , Conception de médicament , Résistance virale aux médicaments/effets des médicaments et des substances chimiques , Stabilité de médicament , Femelle , Transcriptase inverse du VIH/antagonistes et inhibiteurs , Transcriptase inverse du VIH/génétique , Transcriptase inverse du VIH/métabolisme , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/génétique , Période , Humains , Souris , Simulation de docking moléculaire , Mutation , Solubilité , Relation structure-activité
12.
AIDS Res Hum Retroviruses ; 38(3): 198-207, 2022 03.
Article de Anglais | MEDLINE | ID: mdl-34498948

RÉSUMÉ

It is unknown whether antiretroviral (ARV) drugs in women living with HIV (WLHIV) are associated with mitochondrial toxicity and altered fat oxidation and branched-chain amino acid metabolism in the placenta and fetus. Immediately after delivery, we froze placental biopsies from 20 WLHIV and 20 matched uninfected women. We analyzed global biochemical profiles using high-performance liquid chromatography/tandem mass spectrometry and gas chromatography/mass spectrometry. We used t-tests, principle component analysis, hierarchical clustering, and random forest analysis (RFA) in our analysis. Twelve WLHIV were on protease inhibitors, six on non-nucleoside reverse inhibitors, and two on integrase strand inhibitors with optimized backbone. Mean birth weight of HIV-exposed neonates was significantly lower than unexposed neonates (3,075 g vs. 3,498 g, p = .01) at similar gestational age. RFA identified 30 of 702 analytes that differentiated the placental profiles of WLHIV from uninfected women with 72.5% predictive accuracy. Placental profiles of non-nucleoside reverse transcriptase inhibitor (NNRTI)-treated WLHIV exhibited lower levels of amino acids, including essential and branched-chain amino acids, and some medium-chain acylcarnitines. Placental metabolism may be altered in WLHIV, possibly associated with ARV exposure. The lower birth weight among neonates of WLHIV suggests the need for further studies considering potential deleterious effects of altered placenta metabolism on fetal growth and development.


Sujet(s)
Agents antiVIH , Infections à VIH , Agents antiVIH/effets indésirables , Agents antiVIH/métabolisme , Antirétroviraux/usage thérapeutique , Femelle , Infections à VIH/traitement médicamenteux , Infections à VIH/métabolisme , Humains , Nouveau-né , Métabolomique , Placenta/métabolisme , Grossesse
13.
Eur J Med Chem ; 227: 113903, 2022 Jan 05.
Article de Anglais | MEDLINE | ID: mdl-34653770

RÉSUMÉ

Further clinical development of PF74, a lead compound targeting HIV-1 capsid, is impeded by low antiviral activity and inferior metabolic stability. By modifying the benzene (region I) and indole of PF74, we identified two potent compounds (7m and 7u) with significantly improved metabolic stability. Compared to PF74, 7u displayed greater metabolic stability in human liver microsomes (HLMs) with half-life (t1/2) 109-fold that of PF74. Moreover, mechanism of action (MOA) studies demonstrated that 7m and 7u effectively mirrored the MOA of compounds that interact within the PF74 interprotomer pocket, showing direct and robust interactions with recombinant CA, and 7u displaying antiviral effects in both the early and late stages of HIV-1 replication. Furthermore, MD simulation corroborated that 7u was bound to the PF74 binding site, and the results of the online molinspiration software predicted that 7m and 7u had desirable physicochemical properties. Unexpectedly, this series of compounds exhibited better antiviral activity than PF74 against HIV-2, represented by compound 7m whose anti-HIV-2 activity was almost 5 times increased potency over PF74. Therefore, we have rationally redesigned the PF74 chemotype to inhibitors with novel structures and enhanced metabolic stability in this study. We hope that these new compounds can serve as a blueprint for developing a new generation of HIV treatment regimens.


Sujet(s)
Agents antiVIH/pharmacologie , Benzothiazoles/pharmacologie , Protéines de capside/antagonistes et inhibiteurs , Conception de médicament , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/effets des médicaments et des substances chimiques , Phénylalanine/pharmacologie , Agents antiVIH/composition chimique , Agents antiVIH/métabolisme , Benzothiazoles/composition chimique , Benzothiazoles/métabolisme , Protéines de capside/métabolisme , Relation dose-effet des médicaments , Humains , Tests de sensibilité microbienne , Microsomes du foie/composition chimique , Microsomes du foie/métabolisme , Structure moléculaire , Phénylalanine/composition chimique , Phénylalanine/métabolisme , Relation structure-activité , Réplication virale/effets des médicaments et des substances chimiques
14.
Viruses ; 13(12)2021 12 06.
Article de Anglais | MEDLINE | ID: mdl-34960720

RÉSUMÉ

Fullerene derivatives with hydrophilic substituents have been shown to exhibit a range of biological activities, including antiviral ones. For a long time, the anti-HIV activity of fullerene derivatives was believed to be due to their binding into the hydrophobic pocket of HIV-1 protease, thereby blocking its activity. Recent work, however, brought new evidence of a novel, protease-independent mechanism of fullerene derivatives' action. We studied in more detail the mechanism of the anti-HIV-1 activity of N,N-dimethyl[70]fulleropyrrolidinium iodide fullerene derivatives. By using a combination of in vitro and cell-based approaches, we showed that these C70 derivatives inhibited neither HIV-1 protease nor HIV-1 maturation. Instead, our data indicate effects of fullerene C70 derivatives on viral genomic RNA packaging and HIV-1 cDNA synthesis during reverse transcription-without impairing reverse transcriptase activity though. Molecularly, this could be explained by a strong binding affinity of these fullerene derivatives to HIV-1 nucleocapsid domain, preventing its proper interaction with viral genomic RNA, thereby blocking reverse transcription and HIV-1 infectivity. Moreover, the fullerene derivatives' oxidative activity and fluorescence quenching, which could be one of the reasons for the inconsistency among reported anti-HIV-1 mechanisms, are discussed herein.


Sujet(s)
Agents antiVIH/pharmacologie , Fullerènes/métabolisme , Fullerènes/pharmacologie , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/effets des médicaments et des substances chimiques , Protéines nucléocapside/métabolisme , ARN viral/métabolisme , Encapsidation du génome viral/effets des médicaments et des substances chimiques , Agents antiVIH/métabolisme , Génome viral/effets des médicaments et des substances chimiques , Cellules HEK293 , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/génétique , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/métabolisme , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/physiologie , Humains , Liaison aux protéines , Transcription inverse , Virion/métabolisme , Décapsidation virale/effets des médicaments et des substances chimiques , Produits du gène gag du virus de l'immunodéficience humaine/métabolisme
15.
J Am Chem Soc ; 143(45): 19137-19148, 2021 11 17.
Article de Anglais | MEDLINE | ID: mdl-34739240

RÉSUMÉ

The assembly and maturation of human immunodeficiency virus type 1 (HIV-1) require proteolytic cleavage of the Gag polyprotein. The rate-limiting step resides at the junction between the capsid protein CA and spacer peptide 1, which assembles as a six-helix bundle (6HB). Bevirimat (BVM), the first-in-class maturation inhibitor drug, targets the 6HB and impedes proteolytic cleavage, yet the molecular mechanisms of its activity, and relatedly, the escape mechanisms of mutant viruses, remain unclear. Here, we employed extensive molecular dynamics (MD) simulations and free energy calculations to quantitatively investigate molecular structure-activity relationships, comparing wild-type and mutant viruses in the presence and absence of BVM and inositol hexakisphosphate (IP6), an assembly cofactor. Our analysis shows that the efficacy of BVM is directly correlated with preservation of 6-fold symmetry in the 6HB, which exists as an ensemble of structural states. We identified two primary escape mechanisms, and both lead to loss of symmetry, thereby facilitating helix uncoiling to aid access of protease. Our findings also highlight specific interactions that can be targeted for improved inhibitor activity and support the use of MD simulations for future inhibitor design.


Sujet(s)
Agents antiVIH/métabolisme , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/composition chimique , Succinates/métabolisme , Triterpènes/métabolisme , Produits du gène gag du virus de l'immunodéficience humaine/métabolisme , Simulation de dynamique moléculaire , Mutation , Acide phytique/métabolisme , Structure en hélice alpha/effets des médicaments et des substances chimiques , Dépliement des protéines/effets des médicaments et des substances chimiques , Produits du gène gag du virus de l'immunodéficience humaine/composition chimique , Produits du gène gag du virus de l'immunodéficience humaine/génétique
16.
SAR QSAR Environ Res ; 32(11): 889-915, 2021 Nov.
Article de Anglais | MEDLINE | ID: mdl-34551634

RÉSUMÉ

HIV-1 protease (PR) is thought to be efficient targets of anti-AIDS drug design. Molecular dynamics (MD) simulations and multiple post-processing analysis technologies were applied to decipher molecular mechanism underlying binding of three drugs Lopinavir (LPV), Nelfinavir (NFV) and Atazanavir (ATV) to the PR. Binding free energies calculated by molecular mechanics generalized Born surface area (MM-GBSA) suggest that compensation between binding enthalpy and entropy plays a vital role in binding of drugs to PR. Dynamics analyses show that binding of LPV, NFV and ATV highly affects structural flexibility, motion modes and dynamics behaviour of the PR, especially for two flaps. Computational alanine scanning and interaction network analysis verify that although three drugs have structural difference, they share similar binding modes to the PR and common interaction clusters with the PR. The current findings also confirm that residues located interaction clusters, such as Asp25/Asp25', Gly27/Gly27', Ala28/Ala28', Asp29, Ile47/Ile47', Gly49/Gly49', Ile50/Ile50', Val82/Val82' and Ile84/Ile84, can be used as efficient targets of clinically available inhibitors towards the PR.


Sujet(s)
Agents antiVIH/métabolisme , Sulfate d'atazanavir/métabolisme , Protéase du VIH/métabolisme , Lopinavir/métabolisme , Simulation de dynamique moléculaire , Nelfinavir/métabolisme , Sites de fixation
17.
J Med Chem ; 64(18): 13604-13621, 2021 09 23.
Article de Anglais | MEDLINE | ID: mdl-34496571

RÉSUMÉ

Two series of new pyridyl-bearing fused bicyclic analogues designed to target the dual-tolerant regions of the non-nucleoside reverse transcriptase inhibitor (NNRTI)-binding pocket were synthesized and evaluated for their anti-HIV activities. Several compounds, such as 6, 14, 15, 21, 30, and 33, were found to be potent inhibitors against the wild-type (WT) HIV-1 strain or multiple NNRTI-resistant strains at low nanomolar levels. Detailed structure-activity relationships were obtained by utilizing the variation of moieties within the corresponding pharmacophores. In vitro metabolic stability profiles and some drug-like properties of selected compounds were assessed, furnishing the preliminary structure-metabolic stability relationships. Furthermore, molecular modeling studies elucidated the binding modes of compounds 6, 15, 21, and 30 in the binding pocket of WT, E138K, K103N, or Y181C HIV-1 RTs. These promising compounds can be used as lead compounds and warrant further structural optimization to yield more active HIV-1 inhibitors.


Sujet(s)
Agents antiVIH/pharmacologie , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/effets des médicaments et des substances chimiques , Composés hétérobicycliques/pharmacologie , Pyridines/pharmacologie , Agents antiVIH/synthèse chimique , Agents antiVIH/métabolisme , Lignée cellulaire , Conception de médicament , Transcriptase inverse du VIH/composition chimique , Transcriptase inverse du VIH/génétique , Transcriptase inverse du VIH/métabolisme , Composés hétérobicycliques/synthèse chimique , Composés hétérobicycliques/métabolisme , Humains , Microsomes du foie/métabolisme , Simulation de dynamique moléculaire , Structure moléculaire , Mutation , Liaison aux protéines , Pyridines/synthèse chimique , Pyridines/métabolisme , Inhibiteurs de la transcriptase inverse/synthèse chimique , Inhibiteurs de la transcriptase inverse/métabolisme , Inhibiteurs de la transcriptase inverse/pharmacologie , Relation structure-activité
18.
PLoS One ; 16(8): e0255693, 2021.
Article de Anglais | MEDLINE | ID: mdl-34347839

RÉSUMÉ

A method for predicting HIV drug resistance by using genotypes would greatly assist in selecting appropriate combinations of antiviral drugs. Models reported previously have had two major problems: lack of information on the 3D protein structure and processing of incomplete sequencing data in the modeling procedure. We propose obtaining the 3D structural information of viral proteins by using homology modeling and molecular field mapping, instead of just their primary amino acid sequences. The molecular field potential parameters reflect the physicochemical characteristics associated with the 3D structure of the proteins. We also introduce the Bayesian conditional mutual information theory to estimate the probabilities of occurrence of all possible protein candidates from an incomplete sequencing sample. This approach allows for the effective use of uncertain information for the modeling process. We applied these data analysis techniques to the HIV-1 protease inhibitor dataset and developed drug resistance prediction models with reasonable performance.


Sujet(s)
Agents antiVIH/composition chimique , Agents antiVIH/métabolisme , Résistance virale aux médicaments/génétique , Infections à VIH/traitement médicamenteux , Inhibiteurs de protéase du VIH/composition chimique , Inhibiteurs de protéase du VIH/métabolisme , Protéase du VIH/composition chimique , Protéase du VIH/métabolisme , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/enzymologie , Séquence d'acides aminés , Théorème de Bayes , Analyse de données , Génotype , Infections à VIH/virologie , Protéase du VIH/génétique , Humains , Apprentissage machine , Modèles chimiques , Modèles moléculaires , Conformation des protéines , Analyse de séquence de protéine/méthodes
19.
ACS Chem Biol ; 16(8): 1377-1389, 2021 08 20.
Article de Anglais | MEDLINE | ID: mdl-34338505

RÉSUMÉ

Baculiferins are a group of marine sponge-derived polycyclic alkaloids with anti-HIV (human immunodeficiency virus) activities. To identify additional baculiferin-based congeners for SAR analysis and to investigate the mode of action, a total of 18 new baculiferin-type derivatives were synthesized. The inhibitory activities of the congeners against the HIV-1 virus were evaluated in vitro, and the relevant SAR was discussed. Compound 18 exerted the most potent activity toward VSV-G-pseudotyped HIV-1 (IC50 of 3.44 µM) and HIV-1 strain SF33 (IC50 of 2.80 µM) in vitro. To identify the cellular targets, three photoaffinity baculiferin probes were simultaneously synthesized. Photoaffinity labeling experiments together with LC-MS/MS data identified aspartate-tRNA ligase (DARS) as a putative target protein of 18. The overexpression and knockdown of DARS in HEK293T cells provided additional data to demonstrate that DARS is a potential target protein in the regulation of HIV virus infection. The modes of antiviral baculiferins 13 and 18 binding to DARS were determined by a molecular docking simulation. Thus, baculiferin 18 is considered a promising lead as a new molecular target for the development of anti-HIV agents.


Sujet(s)
Alcaloïdes/pharmacologie , Agents antiVIH/pharmacologie , Aspartate-tRNA ligase/antagonistes et inhibiteurs , Antienzymes/pharmacologie , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/effets des médicaments et des substances chimiques , Alcaloïdes/synthèse chimique , Alcaloïdes/métabolisme , Agents antiVIH/synthèse chimique , Agents antiVIH/métabolisme , Aspartate-tRNA ligase/composition chimique , Aspartate-tRNA ligase/métabolisme , Antienzymes/synthèse chimique , Antienzymes/métabolisme , Cellules HEK293 , Humains , Tests de sensibilité microbienne , Simulation de docking moléculaire , Structure moléculaire , Marqueurs de photoaffinité/synthèse chimique , Marqueurs de photoaffinité/métabolisme , Marqueurs de photoaffinité/pharmacologie , Liaison aux protéines , Relation structure-activité
20.
J Inorg Biochem ; 223: 111551, 2021 10.
Article de Anglais | MEDLINE | ID: mdl-34340058

RÉSUMÉ

G-quadruplex nucleic acids (G4s) are RNA and DNA secondary structures involved in the regulation of multiple key biological processes. They can be found in telomeres, oncogene promoters, RNAs, but also in viral genomes. Due to their unique structural features, very distinct from the canonical duplexes or single-strands, G4s represent promising pharmacological targets for small molecules, namely G4-ligands. Gold(III) penta-cationic porphyrins, as specific G4 ligands, are able to inhibit HIV-1 infectivity and their antiviral activity correlates with their affinity for G4s. Up to now, one of the best antiviral compounds is meso-5,10,15,20-tetrakis[4-(N-methyl-pyridinium-2-yl)phenyl]porphyrinato gold(III) (1). Starting from this compound, we report a structure/affinity relationship study of gold(III) cationic porphyrins to find out the best porphyrin candidate for functionalization, in order to study the antiviral mechanism of action of these gold(III) porphyrins.


Sujet(s)
Agents antiVIH/métabolisme , ADN/métabolisme , G-quadruplexes , Métalloporphyrines/métabolisme , Agents antiVIH/synthèse chimique , ADN/génétique , Or/composition chimique , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/composition chimique , Métalloporphyrines/synthèse chimique , Simulation de docking moléculaire , Structure moléculaire , Relation structure-activité
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...