Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 89
Filtrar
1.
Plast Reconstr Surg ; 150(6): 1264e-1274e, 2022 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-36112847

RESUMEN

BACKGROUND: Using immunomodulatory methods to address the challenging issue of craniofacial bone repair may be a potentially effective approach. The protease inhibitor saquinavir has been shown to inhibit the inflammatory response by targeting the toll-like receptor 4/myeloid differentiation primary response complex. Independently, inhibition of toll-like receptor 4 or myeloid differentiation primary response led to enhanced skull bone repair. Therefore, the authors aimed to investigate the effects of saquinavir on skull bone healing. METHODS: The effects of saquinavir on skull bone healing were assessed by means of gene expression, histology, immunohistochemistry, and tomography in a mouse calvarial defect model. Subsequently, the role of saquinavir in cell viability, migration, and osteogenic and osteoclastogenic differentiation was also evaluated in vitro. RESULTS: One-week saquinavir administration improved skull bone healing based on micro-computed tomographic and histomorphometric analyses. Compared to the vehicle control, 1-week saquinavir treatment (1) enhanced osteoclast infiltration (tartrate-resistant acid phosphatase staining) at day 7, but not at days 14 and 28; (2) induced more CD206 + M2 macrophage infiltration, but not F4/80 + M0 macrophages at days 7, 14, and 28; and (3) elevated osteoclastogenic gene RANKL (quantitative polymerase chain reaction) expression and other osteogenic and cytokine expression. Furthermore, in vitro data showed that saquinavir administration did not influence MC3T3-E1 cell migration or mineralization, whereas higher concentrations of saquinavir inhibited cell viability. Saquinavir treatment also enhanced the osteoclastic differentiation of bone marrow-derived precursors, and partially reversed high-mobility group box 1-driven osteoclastogenesis inhibition and elevated proinflammatory cytokine expression. CONCLUSION: The improved skull bone repair following short-term saquinavir treatment may involve enhanced osteoclastogenesis and modulated inflammatory response following skull injury. CLINICAL RELEVANCE STATEMENT: The authors' work demonstrates improved skull bone healing by short-term application of saquinavir, a drug traditionally used in the treatment of acquired immunodeficiency syndrome. As such, saquinavir may be repurposed for skeletal repair.


Asunto(s)
Inhibidores de la Proteasa del VIH , Saquinavir , Ratones , Animales , Saquinavir/farmacología , Saquinavir/metabolismo , Saquinavir/uso terapéutico , Inhibidores de la Proteasa del VIH/farmacología , Inhibidores de la Proteasa del VIH/metabolismo , Inhibidores de la Proteasa del VIH/uso terapéutico , Receptor Toll-Like 4/fisiología , Osteogénesis , Cráneo/lesiones
2.
Sci Rep ; 10(1): 16986, 2020 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-33046764

RESUMEN

We performed molecular dynamics simulation of the dimeric SARS-CoV-2 (severe acute respiratory syndrome corona virus 2) main protease (Mpro) to examine the binding dynamics of small molecular ligands. Seven HIV inhibitors, darunavir, indinavir, lopinavir, nelfinavir, ritonavir, saquinavir, and tipranavir, were used as the potential lead drugs to investigate access to the drug binding sites in Mpro. The frequently accessed sites on Mpro were classified based on contacts between the ligands and the protein, and the differences in site distributions of the encounter complex were observed among the ligands. All seven ligands showed binding to the active site at least twice in 28 simulations of 200 ns each. We further investigated the variations in the complex structure of the active site with the ligands, using microsecond order simulations. Results revealed a wide variation in the shapes of the binding sites and binding poses of the ligands. Additionally, the C-terminal region of the other chain often interacted with the ligands and the active site. Collectively, these findings indicate the importance of dynamic sampling of protein-ligand complexes and suggest the possibilities of further drug optimisations.


Asunto(s)
Betacoronavirus/efectos de los fármacos , Infecciones por Coronavirus/tratamiento farmacológico , Cisteína Endopeptidasas/metabolismo , Reposicionamiento de Medicamentos/métodos , Inhibidores de la Proteasa del VIH/farmacología , Neumonía Viral/tratamiento farmacológico , Proteínas no Estructurales Virales/metabolismo , Betacoronavirus/metabolismo , Sitios de Unión/efectos de los fármacos , Fenómenos Biofísicos , COVID-19 , Dominio Catalítico/efectos de los fármacos , Biología Computacional , Proteasas 3C de Coronavirus , Darunavir/metabolismo , Darunavir/farmacología , Inhibidores de la Proteasa del VIH/metabolismo , Humanos , Indinavir/metabolismo , Indinavir/farmacología , Lopinavir/metabolismo , Lopinavir/farmacología , Simulación de Dinámica Molecular , Nelfinavir/metabolismo , Nelfinavir/farmacología , Pandemias , Ritonavir/metabolismo , Ritonavir/farmacología , SARS-CoV-2 , Saquinavir/metabolismo , Saquinavir/farmacología
3.
J Chem Inf Model ; 60(12): 5771-5780, 2020 12 28.
Artículo en Inglés | MEDLINE | ID: mdl-32530282

RESUMEN

The novel coronavirus (SARS-CoV-2) has infected several million people and caused thousands of deaths worldwide since December 2019. As the disease is spreading rapidly all over the world, it is urgent to find effective drugs to treat the virus. The main protease (Mpro) of SARS-CoV-2 is one of the potential drug targets. Therefore, in this context, we used rigorous computational methods, including molecular docking, fast pulling of ligand (FPL), and free energy perturbation (FEP), to investigate potential inhibitors of SARS-CoV-2 Mpro. We first tested our approach with three reported inhibitors of SARS-CoV-2 Mpro, and our computational results are in good agreement with the respective experimental data. Subsequently, we applied our approach on a database of ∼4600 natural compounds, as well as 8 available HIV-1 protease (PR) inhibitors and an aza-peptide epoxide. Molecular docking resulted in a short list of 35 natural compounds, which was subsequently refined using the FPL scheme. FPL simulations resulted in five potential inhibitors, including three natural compounds and two available HIV-1 PR inhibitors. Finally, FEP, the most accurate and precise method, was used to determine the absolute binding free energy of these five compounds. FEP results indicate that two natural compounds, cannabisin A and isoacteoside, and an HIV-1 PR inhibitor, darunavir, exhibit a large binding free energy to SARS-CoV-2 Mpro, which is larger than that of 13b, the most reliable SARS-CoV-2 Mpro inhibitor recently reported. The binding free energy largely arises from van der Waals interaction. We also found that Glu166 forms H-bonds to all of the inhibitors. Replacing Glu166 by an alanine residue leads to ∼2.0 kcal/mol decreases in the affinity of darunavir to SARS-CoV-2 Mpro. Our results could contribute to the development of potential drugs inhibiting SARS-CoV-2.


Asunto(s)
Antivirales/química , Tratamiento Farmacológico de COVID-19 , Inhibidores de la Proteasa del VIH/química , Proteasa del VIH/metabolismo , SARS-CoV-2/efectos de los fármacos , Secuencia de Aminoácidos , Antivirales/metabolismo , Antivirales/farmacología , Sitios de Unión , Productos Biológicos/química , Productos Biológicos/farmacología , Darunavir/química , Darunavir/farmacología , Bases de Datos Factuales , Diseño de Fármacos , Glucósidos/química , Glucósidos/farmacología , Inhibidores de la Proteasa del VIH/metabolismo , Inhibidores de la Proteasa del VIH/farmacología , Humanos , Simulación del Acoplamiento Molecular , Péptidos/química , Fenoles/química , Fenoles/farmacología , Unión Proteica , Relación Estructura-Actividad , Termodinámica
4.
J Med Chem ; 63(9): 4867-4879, 2020 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-32348139

RESUMEN

We describe here design, synthesis, and biological evaluation of a series of highly potent HIV-1 protease inhibitors containing stereochemically defined and unprecedented tricyclic furanofuran derivatives as P2 ligands in combination with a variety of sulfonamide derivatives as P2' ligands. These inhibitors were designed to enhance the ligand-backbone binding and van der Waals interactions in the protease active site. A number of inhibitors containing the new P2 ligand, an aminobenzothiazole as the P2' ligand and a difluorophenylmethyl as the P1 ligand, displayed very potent enzyme inhibitory potency and also showed excellent antiviral activity against a panel of highly multidrug-resistant HIV-1 variants. The tricyclic P2 ligand has been synthesized efficiently in an optically active form using enzymatic desymmetrization of meso-1,2-(dihydroxymethyl)cyclohex-4-ene as the key step. We determined high-resolution X-ray structures of inhibitor-bound HIV-1 protease. These structures revealed extensive interactions with the backbone atoms of HIV-1 protease and provided molecular insights into the binding properties of these new inhibitors.


Asunto(s)
Furanos/farmacología , Inhibidores de la Proteasa del VIH/farmacología , VIH-1/efectos de los fármacos , Compuestos Heterocíclicos de Anillo en Puente/farmacología , Sustitución de Aminoácidos , Dominio Catalítico , Línea Celular Tumoral , Cristalografía por Rayos X , Diseño de Fármacos , Furanos/síntesis química , Furanos/metabolismo , Proteasa del VIH/química , Proteasa del VIH/genética , Proteasa del VIH/metabolismo , Inhibidores de la Proteasa del VIH/síntesis química , Inhibidores de la Proteasa del VIH/metabolismo , VIH-1/enzimología , Compuestos Heterocíclicos de Anillo en Puente/síntesis química , Compuestos Heterocíclicos de Anillo en Puente/metabolismo , Humanos , Pruebas de Sensibilidad Microbiana , Estructura Molecular , Unión Proteica , Estereoisomerismo
5.
ACS Chem Biol ; 14(11): 2441-2452, 2019 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-31361460

RESUMEN

Drug resistance continues to be a growing global problem. The efficacy of small molecule inhibitors is threatened by pools of genetic diversity in all systems, including antibacterials, antifungals, cancer therapeutics, and antivirals. Resistant variants often include combinations of active site mutations and distal "secondary" mutations, which are thought to compensate for losses in enzymatic activity. HIV-1 protease is the ideal model system to investigate these combinations and underlying molecular mechanisms of resistance. Darunavir (DRV) binds wild-type (WT) HIV-1 protease with a potency of <5 pM, but we have identified a protease variant that loses potency to DRV 150 000-fold, with 11 mutations in and outside the active site. To elucidate the roles of these mutations in DRV resistance, we used a multidisciplinary approach, combining enzymatic assays, crystallography, and molecular dynamics simulations. Analysis of protease variants with 1, 2, 4, 8, 9, 10, and 11 mutations showed that the primary active site mutations caused ∼50-fold loss in potency (2 mutations), while distal mutations outside the active site further decreased DRV potency from 13 nM (8 mutations) to 0.76 µM (11 mutations). Crystal structures and simulations revealed that distal mutations induce subtle changes that are dynamically propagated through the protease. Our results reveal that changes remote from the active site directly and dramatically impact the potency of the inhibitor. Moreover, we find interdependent effects of mutations in conferring high levels of resistance. These mechanisms of resistance are likely applicable to many other quickly evolving drug targets, and the insights may have implications for the design of more robust inhibitors.


Asunto(s)
Darunavir/metabolismo , Farmacorresistencia Viral/genética , Inhibidores de la Proteasa del VIH/metabolismo , Proteasa del VIH/genética , Proteasa del VIH/metabolismo , Biocatálisis , Dominio Catalítico/genética , Humanos , Simulación de Dinámica Molecular , Mutación , Unión Proteica , Conformación Proteica
6.
Plant Biotechnol J ; 17(5): 969-981, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30451369

RESUMEN

Betalains are plant pigments primarily produced by plants of the order Caryophyllales. Because betalain possesses anti-inflammatory and anticancer activities, it may be useful as a pharmaceutical agent and dietary supplement. Recent studies have identified the genes involved in the betalain biosynthesis of betanin. Amaranthin and celosianin II are abundant in the quinoa (Chenopodium quinoa Willd.) hypocotyl, and amaranthin comprises glucuronic acid bound to betanin; therefore, this suggests the existence of a glucuronyltransferase involved in the synthesis of amaranthin in the quinoa hypocotyl. To identify the gene involved in amaranthin biosynthesis, we performed a BLAST analysis and phylogenetic tree analysis based on sequences homologous to flavonoid glycosyltransferase, followed by expression analysis on the quinoa hypocotyl to obtain three candidate proteins. Production of amaranthin in a transient Nicotiana benthamiana expression system was evaluated for these candidates and one was identified as having the ability to produce amaranthin. The gene encoding this protein was quinoa amaranthin synthetase 1 (CqAmaSy1). We also created a transgenic tobacco bright yellow-2 (BY-2) cell line wherein four betalain biosynthesis genes were introduced to facilitate amaranthin production. This transgenic cell line produced 13.67 ± 4.13 µm (mean ± SEM) amaranthin and 26.60 ± 1.53 µm betanin, whereas the production of isoamaranthin and isobetanin could not be detected. Tests confirmed the ability of amaranthin and betanin to slightly suppress cancer cell viability. Furthermore, amaranthin was shown to significantly inhibit HIV-1 protease activity, whereas betanin did not.


Asunto(s)
Betacianinas/biosíntesis , Chenopodium quinoa/enzimología , Ligasas/aislamiento & purificación , Nicotiana/metabolismo , Proteínas de Plantas/aislamiento & purificación , Betacianinas/metabolismo , Reactores Biológicos , Células Cultivadas , Chenopodium quinoa/metabolismo , Clonación Molecular , Proteasa del VIH , Inhibidores de la Proteasa del VIH/metabolismo , Inhibidores de la Proteasa del VIH/farmacología , Ligasas/metabolismo , Redes y Vías Metabólicas , Proteínas de Plantas/metabolismo , Plantas Modificadas Genéticamente , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Nicotiana/citología , Nicotiana/enzimología
7.
Sci Rep ; 8(1): 5789, 2018 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-29636521

RESUMEN

HIV-2 protease (PR2) is naturally resistant to most FDA (Food and Drug Administration)-approved HIV-1 protease inhibitors (PIs), a major antiretroviral class. In this study, we compared the PR1 and PR2 binding pockets extracted from structures complexed with 12 ligands. The comparison of PR1 and PR2 pocket properties showed that bound PR2 pockets were more hydrophobic with more oxygen atoms and fewer nitrogen atoms than PR1 pockets. The structural comparison of PR1 and PR2 pockets highlighted structural changes induced by their sequence variations and that were consistent with these property changes. Specifically, substitutions at residues 31, 46, and 82 induced structural changes in their main-chain atoms that could affect PI binding in PR2. In addition, the modelling of PR1 mutant structures containing V32I and L76M substitutions revealed a cooperative mechanism leading to structural deformation of flap-residue 45 that could modify PR2 flexibility. Our results suggest that substitutions in the PR1 and PR2 pockets can modify PI binding and flap flexibility, which could underlie PR2 resistance against PIs. These results provide new insights concerning the structural changes induced by PR1 and PR2 pocket variation changes, improving the understanding of the atomic mechanism of PR2 resistance to PIs.


Asunto(s)
Inhibidores de la Proteasa del VIH/farmacología , Proteasa del VIH/efectos de los fármacos , VIH/enzimología , Modelos Moleculares , Secuencia de Aminoácidos , Descubrimiento de Drogas , Proteasa del VIH/química , Proteasa del VIH/metabolismo , Inhibidores de la Proteasa del VIH/metabolismo , VIH-1/enzimología , VIH-2/enzimología , Unión Proteica , Conformación Proteica , Análisis de Secuencia de Proteína
8.
Sci Rep ; 8(1): 4202, 2018 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-29523802

RESUMEN

Polygonumins A, a new compound, was isolated from the stem of Polygonum minus. Based on NMR results, the compound's structure is identical to that of vanicoside A, comprising four phenylpropanoid ester units and a sucrose unit. The structure differences were located at C-3″″'. The cytotoxic activity of polygonumins A was evaluated on several cancer cell lines by a cell viability assay using tetrazolium dye 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT). The compound showed the highest antiproliferative (p < 0.05) activities against K562 (Human Leukaemia Cell Line), MCF7 (Human breast adenocarcinoma cell line), and HCT116 (Colorectal cancer cells) cells. Cytotoxic studies against V79-4 cells were carried out and showed that polygonumins A was toxic at 50 µg/ml, suggesting that this compound may be used as an anticancer drug without affecting normal cells. Polygonumins A also showed promising activity as an HIV-1 protease inhibitor with 56% relative inhibition. Molecular docking results indicated that the compound possesses high binding affinity towards the HIV protease over the low binding free energy range of -10.5 to -11.3 kcal/mol. P. minus is used in Malaysian traditional medicine for the treatment of tumour cells. This is the first report on the use of P. minus as an HIV-1 protease inhibitor.


Asunto(s)
Antineoplásicos Fitogénicos/química , Antineoplásicos Fitogénicos/farmacología , Inhibidores de la Proteasa del VIH/química , Inhibidores de la Proteasa del VIH/farmacología , Tallos de la Planta/química , Polygonum/química , Antineoplásicos Fitogénicos/aislamiento & purificación , Antineoplásicos Fitogénicos/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Cinamatos/química , Proteasa del VIH/química , Proteasa del VIH/metabolismo , Inhibidores de la Proteasa del VIH/aislamiento & purificación , Inhibidores de la Proteasa del VIH/metabolismo , Humanos , Microscopía Electrónica de Transmisión de Rastreo , Simulación del Acoplamiento Molecular , Dominios Proteicos
9.
Spectrochim Acta A Mol Biomol Spectrosc ; 188: 362-371, 2018 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-28753530

RESUMEN

Darunavir (DRV), a second-generation HIV protease inhibitor, is widely used across the world as an important component of HIV therapy. The interaction of DRV with bovine serum albumin (BSA), a major carrier protein, has been studied under simulated physiological conditions (pH7.4) by multi-spectroscopic techniques in combination with molecular modeling. Fluorescence data revealed that the intrinsic fluorescence of BSA was quenched by DRV in terms of a static quenching procedure due to the formation of the DRV-BSA complex. The results indicated the presence of single weak affinity binding site (~103M-1, 310K) on protein. The thermodynamic parameters, namely enthalpy change (ΔH0), entropy change (ΔS0) and Gibbs free energy change (ΔG0) were calculated, which signified that the binding reaction was spontaneous, the main binding forces were hydrogen bonding and van der Waals forces. Importantly, competitive binding experiments with three site probes, phenylbutazone (in sub-domain IIA, site I), ibuprofen (in sub-domain IIIA, site II) and artemether (in the interface between sub-domain IIA and IIB, site II'), suggested that DRV was preferentially bound to the hydrophobic cavity in site II' of BSA, and this finding was validated by the docking results. Additionally, synchronous fluorescence, three-dimensional fluorescence and Resonance Rayleigh Scattering (RRS) spectroscopy gave qualitative information on the conformational changes of BSA upon adding DRV, while quantitative data were obtained with Fourier transform infrared spectroscopy (FT-IR).


Asunto(s)
Darunavir/metabolismo , Inhibidores de la Proteasa del VIH/metabolismo , Modelos Moleculares , Albúmina Sérica Bovina/metabolismo , Animales , Sitios de Unión , Bovinos , Darunavir/química , Transferencia de Energía , Inhibidores de la Proteasa del VIH/química , Interacciones Hidrofóbicas e Hidrofílicas , Cinética , Simulación del Acoplamiento Molecular , Unión Proteica , Estructura Secundaria de Proteína , Dispersión de Radiación , Albúmina Sérica Bovina/química , Espectrometría de Fluorescencia , Espectrofotometría Ultravioleta , Espectroscopía Infrarroja por Transformada de Fourier , Termodinámica
10.
J Biomol Struct Dyn ; 36(16): 4366-4377, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29237328

RESUMEN

HIV polyprotein Gag is increasingly found to contribute to protease inhibitor resistance. Despite its role in viral maturation and in developing drug resistance, there remain gaps in the knowledge of the role of certain Gag subunits (e.g. p6), and that of non-cleavage mutations in drug resistance. As p6 is flexible, it poses a problem for structural experiments, and is hence often omitted in experimental Gag structural studies. Nonetheless, as p6 is an indispensable component for viral assembly and maturation, we have modeled the full length Gag structure based on several experimentally determined constraints and studied its structural dynamics. Our findings suggest that p6 can mechanistically modulate Gag conformations. In addition, the full length Gag model reveals that allosteric communication between the non-cleavage site mutations and the first Gag cleavage site could possibly result in protease drug resistance, particularly in the absence of mutations in Gag cleavage sites. Our study provides a mechanistic understanding to the structural dynamics of HIV-1 Gag, and also proposes p6 as a possible drug target in anti-HIV therapy.


Asunto(s)
Farmacorresistencia Viral/genética , Proteasa del VIH/genética , VIH-1/genética , Mutación , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/genética , Regulación Alostérica , Sitios de Unión/genética , Farmacorresistencia Viral/efectos de los fármacos , Infecciones por VIH/prevención & control , Infecciones por VIH/virología , Proteasa del VIH/química , Proteasa del VIH/metabolismo , Inhibidores de la Proteasa del VIH/química , Inhibidores de la Proteasa del VIH/metabolismo , Inhibidores de la Proteasa del VIH/farmacología , VIH-1/efectos de los fármacos , VIH-1/fisiología , Humanos , Modelos Moleculares , Unión Proteica , Conformación Proteica , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/antagonistas & inhibidores , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/química , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/metabolismo
11.
Pharm Res ; 34(9): 1925-1933, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28616684

RESUMEN

PURPOSE: Although the prevalence of alcohol consumption is higher in HIV+ people than general public, limited information is available on how alcohol affects the metabolism and bioavailability of darunavir (DRV). METHODS: DRV was quantified by using LC-MS/MS method. All in vitro experiments were performed using human liver microsomes and HIV-infected monocytic cells. CYP3A4 and DRV/Ritonavir (RTV) docking was performed using GOLD suite 5.8. RESULTS: Ethanol (20 mM) significantly decreased apparent half-life and increased degradation rate constant of RTV-boosted DRV but not for DRV alone. Similarly, ethanol exposure increased hepatic intrinsic clearance for RTV-boosted DRV with no significant influence on DRV alone. Ethanol showed a limited influence on intracellular total DRV exposure in the presence of RTV without altering maximum concentration (Cmax) values in HIV-infected monocytic cells. Ethanol alone elevated HIV replication but this effect was nullified with the addition of DRV or DRV + RTV. Additionally, inhibitory potency of DRV was significantly reduced in the presence of ethanol. Our docking results projected that ethanol increases the average distance between DRV and CYP3A4 heme, and alter the orientation of DRV-CYP3A4 binding. CONCLUSIONS: Collectively these findings suggest that DRV metabolism is primarily influenced by ethanol in the liver, but has minor effect in HIV-residing monocytes.


Asunto(s)
Citocromo P-450 CYP3A/metabolismo , Darunavir/metabolismo , Etanol/metabolismo , Inhibidores de la Proteasa del VIH/metabolismo , Hígado/metabolismo , Monocitos/metabolismo , Línea Celular , Darunavir/farmacocinética , Darunavir/farmacología , VIH/efectos de los fármacos , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/metabolismo , Infecciones por VIH/virología , Inhibidores de la Proteasa del VIH/farmacocinética , Inhibidores de la Proteasa del VIH/farmacología , Humanos , Hígado/efectos de los fármacos , Hígado/virología , Microsomas Hepáticos/efectos de los fármacos , Microsomas Hepáticos/metabolismo , Microsomas Hepáticos/virología , Simulación del Acoplamiento Molecular , Monocitos/efectos de los fármacos , Monocitos/virología , Ritonavir/metabolismo , Ritonavir/farmacocinética , Ritonavir/farmacología , Replicación Viral/efectos de los fármacos
12.
Nat Commun ; 8(1): 39, 2017 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-28659616

RESUMEN

Only one out of 10 drugs in development passes clinical trials. Many fail because experimental animal models poorly predict human xenobiotic metabolism. Human liver chimeric mice are a step forward in this regard, as the human hepatocytes in chimeric livers generate human metabolites, but the remaining murine hepatocytes contain an expanded set of P450 cytochromes that form the major class of drug-metabolizing enzymes. We therefore generated a conditional knock-out of the NADPH-P450 oxidoreductase (Por) gene combined with Il2rg - /- /Rag2 - /- /Fah - /- (PIRF) mice. Here we show that homozygous PIRF mouse livers are readily repopulated with human hepatocytes, and when the murine Por gene is deleted (<5%), they predominantly use human cytochrome metabolism. When given the anticancer drug gefitinib or the retroviral drug atazanavir, the Por-deleted humanized PIRF mice develop higher levels of the major human metabolites than current models. Humanized, murine Por-deficient PIRF mice can thus predict human drug metabolism and should be useful for preclinical drug development.Human liver chimeric mice are increasingly used for drug testing in preclinical development, but express residual murine p450 cytochromes. Here the authors generate mice lacking the Por gene in the liver, and show that human cytochrome metabolism is used following repopulation with human hepatocytes.


Asunto(s)
Sulfato de Atazanavir/metabolismo , Sistema Enzimático del Citocromo P-450/metabolismo , Quinazolinas/metabolismo , Animales , Antineoplásicos/metabolismo , Quimera , Sistema Enzimático del Citocromo P-450/genética , Citocromos/metabolismo , Femenino , Gefitinib , Genotipo , Inhibidores de la Proteasa del VIH/metabolismo , Humanos , Hígado/enzimología , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos
13.
J Pharm Sci ; 105(2): 854-863, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26213315

RESUMEN

The aim of this work was to explore the contribution of the organic anion transporting polypeptide-1B (OATP1B) drug transporters in the hepatic clearance (Cl) of all marketed HIV protease inhibitors (PI) in humans. HIV PI uptake rates in OATP1B1/1B3-transfected Chinese hamster ovary cells were converted to uptake Cl values in human hepatocytes via a relative activity factor, which was determined by comparing uptake of known substrates between OATP1B1/3-transfected cells and human hepatocytes. Metabolic Cl values were determined in human liver microsomes. In vivo hepatic Cl values were calculated either by combining drug uptake and metabolism or based on one of these individual Cl processes and compared with published in vivo hepatic Cl values. Excellent in vitro-in vivo correlation (R(2) = 0.85) was observed when only uptake Cl values were used, but not when only metabolic Cl was used (R(2) = 0.40). The correlation did not improve when both processes were taken into account (R(2) = 0.85). PBPK models confirmed the remarkable sensitivity of predicted exposure to hepatic drug uptake, indicating a key role for OATP1B1/3 in hepatic disposition of several HIV PI in man. This may contribute to the interindividual variability in systemic and hepatic exposure to these drugs in the clinic.


Asunto(s)
Inhibidores de la Proteasa del VIH/metabolismo , Hepatocitos/metabolismo , Tasa de Depuración Metabólica/fisiología , Microsomas Hepáticos/metabolismo , Animales , Células CHO , Células Cultivadas , Cricetinae , Cricetulus , Predicción , Inhibidores de la Proteasa del VIH/farmacología , Hepatocitos/efectos de los fármacos , Humanos , Tasa de Depuración Metabólica/efectos de los fármacos , Microsomas Hepáticos/efectos de los fármacos
14.
Drug Des Devel Ther ; 9: 6055-65, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26622167

RESUMEN

Based on experimental data, the anticancer activity of nelfinavir (NFV), a US Food and Drug Administration (FDA)-approved HIV-1 protease inhibitor (PI), was reported. Nevertheless, the mechanism of action of NFV is yet to be verified. It was hypothesized that the anticancer activity of NFV is due to its inhibitory effect on heat shock protein 90 (Hsp90), a promising target for anticancer therapy. Such findings prompted us to investigate the potential anticancer activity of all other FDA-approved HIV-1 PIs against human Hsp90. To accomplish this, "loop docking" - an enhanced in-house developed molecular docking approach - followed by molecular dynamic simulations and postdynamic analyses were performed to elaborate on the binding mechanism and relative binding affinities of nine FDA-approved HIV-1 PIs against human Hsp90. Due to the lack of the X-ray crystal structure of human Hsp90, homology modeling was performed to create its 3D structure for subsequent simulations. Results showed that NFV has better binding affinity (ΔG =-9.2 kcal/mol) when compared with other PIs: this is in a reasonable accordance with the experimental data (IC50 3.1 µM). Indinavir, saquinavir, and ritonavir have close binding affinity to NFV (ΔG =-9.0, -8.6, and -8.5 kcal/mol, respectively). Per-residue interaction energy decomposition analysis showed that hydrophobic interaction (most importantly with Val534 and Met602) played the most predominant role in drug binding. To further validate the docking outcome, 5 ns molecular dynamic simulations were performed in order to assess the stability of the docked complexes. To our knowledge, this is the first account of detailed computational investigations aimed to investigate the potential anticancer activity and the binding mechanism of the FDA-approved HIV PIs binding to human Hsp90. Information gained from this study should also provide a route map toward the design, optimization, and further experimental investigation of potential derivatives of PIs to treat HER2+ breast cancer.


Asunto(s)
Antineoplásicos/farmacología , Reposicionamiento de Medicamentos , Inhibidores de la Proteasa del VIH/farmacología , Proteasa del VIH/metabolismo , VIH-1/efectos de los fármacos , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Nelfinavir/farmacología , Antineoplásicos/química , Antineoplásicos/metabolismo , Sitios de Unión , Diseño de Fármacos , Proteasa del VIH/química , Inhibidores de la Proteasa del VIH/química , Inhibidores de la Proteasa del VIH/metabolismo , VIH-1/enzimología , Proteínas HSP90 de Choque Térmico/química , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Nelfinavir/química , Nelfinavir/metabolismo , Unión Proteica , Conformación Proteica , Reproducibilidad de los Resultados , Relación Estructura-Actividad
15.
Int J Mol Sci ; 15(2): 2622-45, 2014 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-24534807

RESUMEN

Molecular docking has been extensively applied in virtual screening of small molecule libraries for lead identification and optimization. A necessary prerequisite for successful differentiation between active and non-active ligands is the accurate prediction of their binding affinities in the complex by use of docking scoring functions. However, many studies have shown rather poor correlations between docking scores and experimental binding affinities. Our work aimed to improve this correlation by implementing a multipose binding concept in the docking scoring scheme. Multipose binding, i.e., the property of certain protein-ligand complexes to exhibit different ligand binding modes, has been shown to occur in nature for a variety of molecules. We conducted a high-throughput docking study and implemented multipose binding in the scoring procedure by considering multiple docking solutions in binding affinity prediction. In general, improvement of the agreement between docking scores and experimental data was observed, and this was most pronounced in complexes with large and flexible ligands and high binding affinities. Further developments of the selection criteria for docking solutions for each individual complex are still necessary for a general utilization of the multipose binding concept for accurate binding affinity prediction by molecular docking.


Asunto(s)
Simulación del Acoplamiento Molecular , Proteasa del VIH/química , Proteasa del VIH/metabolismo , Inhibidores de la Proteasa del VIH/química , Inhibidores de la Proteasa del VIH/metabolismo , Ligandos , Péptidos/química , Péptidos/metabolismo , Análisis de Componente Principal , Unión Proteica , Proteínas/química , Proteínas/metabolismo , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/metabolismo , Programas Informáticos , Dominios Homologos src
16.
J Mass Spectrom ; 48(9): 1019-31, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24078243

RESUMEN

Atazanavir (Reyataz(®)) is an important member of the HIV protease inhibitor class. Because of the complexity of its chemical structure, metabolite identification and structural elucidation face serious challenges. So far, only seven non-conjugated metabolites in human plasma have been reported, and their structural elucidation is not complete, especially for the major metabolites produced by oxidations. To probe the exact sites of metabolism and to elucidate the relationship among in vivo metabolites of atazanavir, we designed and performed two sets of experiments. The first set of experiments was to determine atazanavir metabolites in human plasma by LC-MS, from which more than a dozen metabolites were discovered, including seven new ones that have not been reported. The second set involved deuterium labeling on potential metabolic sites to generate D-atazanavir analogs. D-atazanavir analogs were dosed to human in parallel with atazanavir. Metabolites of D-atazanavir were identified by the same LC-MS method, and the results were compared with those of atazanavir. A metabolite structure can be readily elucidated by comparing the results of the analogs and the pathway by which the metabolite is formed can be proposed with confidence. Experimental results demonstrated that oxidation is the most common metabolic pathway of atazanavir, resulting in the formation of six metabolites of monooxidation (M1, M2, M7, M8, M13, and M14) and four of dioxidation (M15, M16, M17, and M18). The second metabolic pathway is hydrolysis, and the third is N-dealkylation. Metabolites produced by hydrolysis include M3, M4, and M19. Metabolites formed by N-dealkylation are M5, M6a, and M6b.


Asunto(s)
Inhibidores de la Proteasa del VIH/sangre , Inhibidores de la Proteasa del VIH/metabolismo , Oligopéptidos/sangre , Oligopéptidos/metabolismo , Piridinas/sangre , Piridinas/metabolismo , Sulfato de Atazanavir , Cromatografía Líquida de Alta Presión/métodos , Deuterio/análisis , Deuterio/metabolismo , VIH/enzimología , Infecciones por VIH/tratamiento farmacológico , Inhibidores de la Proteasa del VIH/administración & dosificación , Inhibidores de la Proteasa del VIH/química , Humanos , Hidrólisis , Redes y Vías Metabólicas , Oligopéptidos/administración & dosificación , Oligopéptidos/química , Oxidación-Reducción , Piridinas/administración & dosificación , Piridinas/química , Espectrometría de Masas en Tándem/métodos
17.
Biochem Biophys Res Commun ; 438(4): 703-8, 2013 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-23921229

RESUMEN

Multidrug-resistant (MDR) clinical isolate-769, human immunodeficiency virus type-1 (HIV-1) protease (PDB ID: 1TW7), was shown to exhibit wide-open flaps and an expanded active site cavity, causing loss of contacts with protease inhibitors. In the current study, the expanded active site cavity of MDR769 HIV-1 protease was screened with a series of peptide-inhibitors that were designed to mimic the natural substrate cleavage site, capsid/p2. Scanning Ala/Phe chemical mutagenesis approach was incorporated into the design of the peptide series to mimic the substrate co-evolution. Among the peptides synthesized and evaluated, a lead peptide (6a) with potent activity (IC50: 4.4nM) was identified against the MDR769 HIV-1 protease. Isothermal titration calorimetry data showed favorable binding profile for 6a against both wild type and MDR769 HIV-1 protease variants. Nuclear magnetic resonance spectrum of (15)N-labeled MDR769 HIV-1 protease in complex with 6a showed some major perturbations in chemical shift, supporting the peptide induced conformational changes in protease. Modeling analysis revealed multiple contacts between 6a and MDR769 HIV-1 protease. The lead peptide-inhibitor, 6a, with high potency and good binding profile can be used as the basis for developing potent small molecule inhibitors against MDR variants of HIV.


Asunto(s)
Infecciones por VIH/tratamiento farmacológico , Inhibidores de la Proteasa del VIH/química , Inhibidores de la Proteasa del VIH/farmacología , Proteasa del VIH/metabolismo , VIH-1/enzimología , Péptidos/química , Péptidos/farmacología , Secuencia de Aminoácidos , Diseño de Fármacos , Farmacorresistencia Viral Múltiple , Infecciones por VIH/virología , Proteasa del VIH/genética , Inhibidores de la Proteasa del VIH/metabolismo , VIH-1/efectos de los fármacos , VIH-1/genética , Humanos , Simulación del Acoplamiento Molecular , Mutagénesis , Biblioteca de Péptidos , Péptidos/genética
18.
Drug Metab Dispos ; 41(10): 1843-51, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23913028

RESUMEN

Erlotinib is approved for the treatment of non-small cell lung and pancreatic cancers, and is metabolized by CYP3A4. Inducers and inhibitors of CYP3A enzymes such as ritonavir and efavirenz, respectively, may be used as part of the highly active antiretroviral therapy drugs to treat patients with human immunodeficiency virus (HIV). When HIV patients with a malignancy need treatment with erlotinib, there is a potential of as-yet-undefined drug-drug interaction. We evaluated these interactions using human hepatocytes benchmarked against the interaction of erlotinib with ketoconazole and rifampin, the archetype cytochrome P450 inhibitor and inducer, respectively. Hepatocytes were treated with vehicle [0.1% dimethylsulfoxide, ritonavir (10 µM)], ketoconazole (10 µM), efavirenz (10 µM), or rifampin (10 µM) for 4 days. On day 5, erlotinib (5 µM) was incubated with the above agents for another 24-48 hours. Concentrations of erlotinib and O-desmethyl erlotinib were quantitated in collected samples (combined lysate and medium) using liquid chromatography and tandem mass spectrometry. The half-life (t(½)) of erlotinib increased from 10.6 ± 2.6 to 153 ± 80 and 23.9 ± 4.8 hours, respectively, upon treatment with ritonavir and ketoconazole. The apparent intrinsic clearance (C(Lint, app)) of erlotinib was lowered 16-fold by ritonavir and 1.9-fold by ketoconazole. Efavirenz and rifampin decreased t1/2 of erlotinib from 10.3 ± 1.1 to 5.0 ± 1.5 and 3.4 ± 0.2 hours, respectively. Efavirenz and rifampin increased the C(Lint, app) of erlotinib by 2.2- and 2-fold, respectively. Our results suggest that to achieve desired drug exposure, the clinically used dose (150 mg daily) of erlotinib may have to be significantly reduced (25 mg every other day) or increased (300 mg daily), respectively, when ritonavir or efavirenz is coadministered.


Asunto(s)
Benzoxazinas/uso terapéutico , Interacciones Farmacológicas/fisiología , Infecciones por VIH/tratamiento farmacológico , Hepatocitos/metabolismo , Neoplasias/tratamiento farmacológico , Quinazolinas/uso terapéutico , Ritonavir/uso terapéutico , Inhibidores de 14 alfa Desmetilasa/uso terapéutico , Adulto , Anciano , Alquinos , Fármacos Anti-VIH/metabolismo , Fármacos Anti-VIH/uso terapéutico , Ciclopropanos , Clorhidrato de Erlotinib , Femenino , Infecciones por VIH/metabolismo , Infecciones por VIH/virología , Inhibidores de la Proteasa del VIH/metabolismo , Inhibidores de la Proteasa del VIH/uso terapéutico , Semivida , Hepatocitos/efectos de los fármacos , Humanos , Cetoconazol/uso terapéutico , Masculino , Persona de Mediana Edad , Neoplasias/metabolismo , Neoplasias/virología , Inhibidores de la Síntesis del Ácido Nucleico/uso terapéutico , Quinazolinas/metabolismo , Rifampin/uso terapéutico
19.
J Neuroinflammation ; 10: 58, 2013 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-23642074

RESUMEN

BACKGROUND: Active HIV infection within the central nervous system (CNS) is confined primarily to microglia. The glial cell compartment acts as a viral reservoir behind the blood-brain barrier. It provides an additional roadblock to effective pharmacological treatment via expression of multiple drug efflux transporters, including P-glycoprotein. HIV/AIDS patients frequently suffer bacterial and viral co-infections, leading to deregulation of glial cell function and release of pro-inflammatory mediators including cytokines, chemokines, and nitric oxide. METHODS: To better define the role of inflammation in decreased HIV drug accumulation into CNS targets, accumulation of the antiretroviral saquinavir was examined in purified cultures of rodent microglia exposed to the prototypical inflammatory mediator lipopolysaccharide (LPS). RESULTS: [(3)H]-Saquinavir accumulation by microglia was rapid, and was increased up to two-fold in the presence of the specific P-glycoprotein inhibitor, PSC833. After six or 24 hours of exposure to 10 ng/ml LPS, saquinavir accumulation was decreased by up to 45%. LPS did not directly inhibit saquinavir transport, and did not affect P-glycoprotein protein expression. LPS exposure did not alter RNA and/or protein expression of other transporters including multidrug resistance-associated protein 1 and several solute carrier uptake transporters. CONCLUSIONS: The decrease in saquinavir accumulation in microglia following treatment with LPS is likely multi-factorial, since drug accumulation was attenuated by inhibitors of NF-κß and the MEK1/2 pathway in the microglia cell line HAPI, and in primary microglia cultures from toll-like receptor 4 deficient mice. These data provide new pharmacological insights into why microglia act as a difficult-to-treat viral sanctuary site.


Asunto(s)
Infecciones por VIH/tratamiento farmacológico , Inhibidores de la Proteasa del VIH/farmacología , Activación de Macrófagos/efectos de los fármacos , Microglía/fisiología , Saquinavir/farmacología , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Animales , Western Blotting , Línea Celular , Interpretación Estadística de Datos , Femenino , Infecciones por VIH/virología , Inhibidores de la Proteasa del VIH/metabolismo , Lipopolisacáridos/farmacología , Nitritos/metabolismo , Embarazo , Cultivo Primario de Células , Ratas , Ratas Endogámicas F344 , Ratas Wistar , Reacción en Cadena en Tiempo Real de la Polimerasa , Saquinavir/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo
20.
Biopharm Drug Dispos ; 34(3): 155-64, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23280499

RESUMEN

Although HIV protease inhibitors (PIs) produce profound metabolic interactions through inactivation/inhibition of CYP3A enzymes, their role as victims of transporter-based drug-drug interactions (DDIs) is less well understood. Therefore, this study investigated if the PIs, nelfinavir (NFV), ritonavir (RTV), lopinavir (LPV) or amprenavir (APV) were transported into sandwich-cultured human hepatocytes (SCHH), and whether OATPs contributed to this transport. The findings showed that, except for (3) H-APV, no significant decrease in the total hepatocyte accumulation of the (3) H-PIs was detected in the presence of the corresponding unlabeled PI, indicating that the uptake of the other PIs was not mediated. Further, hepatocyte biliary efflux studies using (3) H-APV and unlabeled APV confirmed this decrease to be due to inhibition of sinusoidal influx transporter(s) and not the canalicular efflux transporters. Moreover, this sinusoidal transport of APV was not OATP-mediated. The results indicate that the hepatic uptake of NFV, RTV or LPV was primarily mediated by passive diffusion. The hepatic uptake of APV was mediated by an unidentified sinusoidal transporter(s). Therefore, NFV, RTV or LPV will not be victims of DDIs involving inhibition of hepatic influx transporters; however, the disposition of APV may be affected if its sinusoidal transport is inhibited.


Asunto(s)
Inhibidores de la Proteasa del VIH/metabolismo , Hepatocitos/metabolismo , Transportadores de Anión Orgánico/metabolismo , Carbamatos/metabolismo , Células Cultivadas , Interacciones Farmacológicas , Furanos , Humanos , Lopinavir/metabolismo , Nelfinavir/metabolismo , Ritonavir/metabolismo , Sulfonamidas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA