Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Eur J Med Chem ; 257: 115501, 2023 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-37244161

RESUMEN

Protease inhibitors are the most potent antivirals against HIV-1, but they still lose efficacy against resistant variants. Improving the resistance profile is key to developing more robust inhibitors, which may be promising candidates for simplified next-generation antiretroviral therapies. In this study, we explored analogs of darunavir with a P1 phosphonate modification in combination with increasing size of the P1' hydrophobic group and various P2' moieties to improve potency against resistant variants. The phosphonate moiety substantially improved potency against highly mutated and resistant HIV-1 protease variants, but only when combined with more hydrophobic moieties at the P1' and P2' positions. Phosphonate analogs with a larger hydrophobic P1' moiety maintained excellent antiviral potency against a panel of highly resistant HIV-1 variants, with significantly improved resistance profiles. The cocrystal structures indicate that the phosphonate moiety makes extensive hydrophobic interactions with the protease, especially with the flap residues. Many residues involved in these protease-inhibitor interactions are conserved, enabling the inhibitors to maintain potency against highly resistant variants. These results highlight the need to balance inhibitor physicochemical properties by simultaneous modification of chemical groups to further improve resistance profiles.


Asunto(s)
Inhibidores de la Proteasa del VIH , VIH-1 , Inhibidores de la Proteasa del VIH/farmacología , Inhibidores de la Proteasa del VIH/química , Darunavir/farmacología , Péptido Hidrolasas , Proteasa del VIH/genética , Cristalografía por Rayos X
2.
Elife ; 122023 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-36920025

RESUMEN

Darunavir (DRV) is exceptional among potent HIV-1 protease inhibitors (PIs) in high drug concentrations that are achieved in vivo. Little is known about the de novo resistance pathway for DRV. We selected for resistance to high drug concentrations against 10 PIs and their structural precursor DRV. Mutations accumulated through two pathways (anchored by protease mutations I50V or I84V). Small changes in the inhibitor P1'-equivalent position led to preferential use of one pathway over the other. Changes in the inhibitor P2'-equivalent position determined differences in potency that were retained in the resistant viruses and that impacted the selected mutations. Viral variants from the two pathways showed differential selection of compensatory mutations in Gag cleavage sites. These results reveal the high level of selective pressure that is attainable with fifth-generation PIs and how features of the inhibitor affect both the resistance pathway and the residual potency in the face of resistance.


Asunto(s)
Infecciones por VIH , Inhibidores de la Proteasa del VIH , VIH-1 , Humanos , Inhibidores de la Proteasa del VIH/química , VIH-1/genética , Darunavir/farmacología , Darunavir/uso terapéutico , Mutación , Farmacorresistencia Viral/genética , Infecciones por VIH/tratamiento farmacológico
3.
Bioorg Chem ; 131: 106269, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36446201

RESUMEN

Viruses from the Flavivirus genus infect millions of people worldwide and cause severe diseases, including recent epidemics of dengue virus (DENV), and Zika virus (ZIKV). There is currently no antiviral treatment against flavivirus infections, despite considerable efforts to develop inhibitors against essential viral enzymes including NS2B/NS3 protease. Targeting the flavivirus NS2B/NS3 protease proved to be challenging because of the conformational dynamics, topology, and electrostatic properties of the active site. Here, we report the identification of quinoxaline-based allosteric inhibitors by fragment-based drug discovery approach as a promising new drug-like scaffold to target the NS2B/NS3 protease. Enzymatic assays and mutational analysis of the allosteric site in ZIKV NS2B/NS3 protease support noncompetitive inhibition mechanism as well as engineered DENV protease construct indicating the compounds likely compete with the NS2B cofactor for binding to the protease domain. Furthermore, antiviral activity confirmed the therapeutic potential of this new inhibitor scaffold.


Asunto(s)
Flavivirus , Infección por el Virus Zika , Virus Zika , Humanos , Flavivirus/química , Flavivirus/metabolismo , Virus Zika/metabolismo , Péptido Hidrolasas , Quinoxalinas/farmacología , Proteínas no Estructurales Virales , Serina Endopeptidasas/metabolismo , Inhibidores de Proteasas/farmacología , Antivirales/química
4.
Biochemistry ; 60(39): 2925-2931, 2021 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-34506130

RESUMEN

Rupintrivir targets the 3C cysteine proteases of the picornaviridae family, which includes rhinoviruses and enteroviruses that cause a range of human diseases. Despite being a pan-3C protease inhibitor, rupintrivir activity is extremely weak against the homologous 3C-like protease of SARS-CoV-2. In this study, the crystal structures of rupintrivir were determined bound to enterovirus 68 (EV68) 3C protease and the 3C-like main protease (Mpro) from SARS-CoV-2. While the EV68 3C protease-rupintrivir structure was similar to previously determined complexes with other picornavirus 3C proteases, rupintrivir bound in a unique conformation to the active site of SARS-CoV-2 Mpro splitting the catalytic cysteine and histidine residues. This bifurcation of the catalytic dyad may provide a novel approach for inhibiting cysteine proteases.


Asunto(s)
Antivirales/metabolismo , Proteasas 3C de Coronavirus/metabolismo , Inhibidores de Cisteína Proteinasa/metabolismo , Isoxazoles/metabolismo , Fenilalanina/análogos & derivados , Pirrolidinonas/metabolismo , SARS-CoV-2/enzimología , Valina/análogos & derivados , Antivirales/química , Dominio Catalítico , Proteasas 3C de Coronavirus/antagonistas & inhibidores , Proteasas 3C de Coronavirus/química , Cristalografía por Rayos X , Inhibidores de Cisteína Proteinasa/química , Enterovirus Humano D/enzimología , Enlace de Hidrógeno , Isoxazoles/química , Fenilalanina/química , Fenilalanina/metabolismo , Unión Proteica , Pirrolidinonas/química , Electricidad Estática , Valina/química , Valina/metabolismo
5.
ACS Chem Biol ; 16(3): 529-538, 2021 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-33619959

RESUMEN

Human T-cell lymphotropic virus type 1 (HTLV-1) is a retrovirus that can cause severe paralytic neurologic disease and immune disorders as well as cancer. An estimated 20 million people worldwide are infected with HTLV-1, with prevalence reaching 30% in some parts of the world. In stark contrast to HIV-1, no direct acting antivirals (DAAs) exist against HTLV-1. The aspartyl protease of HTLV-1 is a dimer similar to that of HIV-1 and processes the viral polyprotein to permit viral maturation. We report that the FDA-approved HIV-1 protease inhibitor darunavir (DRV) inhibits the enzyme with 0.8 µM potency and provides a scaffold for drug design against HTLV-1. Analogs of DRV that we designed and synthesized achieved submicromolar inhibition against HTLV-1 protease and inhibited Gag processing in viral maturation assays and in a chronically HTLV-1 infected cell line. Cocrystal structures of these inhibitors with HTLV-1 protease highlight opportunities for future inhibitor design. Our results show promise toward developing highly potent HTLV-1 protease inhibitors as therapeutic agents against HTLV-1 infections.


Asunto(s)
Antivirales/química , Ácido Aspártico Endopeptidasas/antagonistas & inhibidores , Darunavir/análogos & derivados , Virus Linfotrópico T Tipo 1 Humano/efectos de los fármacos , Inhibidores de Proteasas/química , Secuencia de Aminoácidos , Antivirales/farmacología , Ácido Aspártico Endopeptidasas/química , Ácido Aspártico Endopeptidasas/genética , Darunavir/farmacología , Descubrimiento de Drogas , Escherichia coli/genética , Humanos , Simulación de Dinámica Molecular , Estructura Molecular , Terapia Molecular Dirigida , Inhibidores de Proteasas/farmacología , Unión Proteica , Conformación Proteica , Relación Estructura-Actividad
6.
Chem Rev ; 121(6): 3238-3270, 2021 03 24.
Artículo en Inglés | MEDLINE | ID: mdl-33410674

RESUMEN

Drug resistance is prevalent across many diseases, rendering therapies ineffective with severe financial and health consequences. Rather than accepting resistance after the fact, proactive strategies need to be incorporated into the drug design and development process to minimize the impact of drug resistance. These strategies can be derived from our experience with viral disease targets where multiple generations of drugs had to be developed to combat resistance and avoid antiviral failure. Significant efforts including experimental and computational structural biology, medicinal chemistry, and machine learning have focused on understanding the mechanisms and structural basis of resistance against direct-acting antiviral (DAA) drugs. Integrated methods show promise for being predictive of resistance and potency. In this review, we give an overview of this research for human immunodeficiency virus type 1, hepatitis C virus, and influenza virus and the lessons learned from resistance mechanisms of DAAs. These lessons translate into rational strategies to avoid resistance in drug design, which can be generalized and applied beyond viral targets. While resistance may not be completely avoidable, rational drug design can and should incorporate strategies at the outset of drug development to decrease the prevalence of drug resistance.


Asunto(s)
Antivirales/química , Inhibidores Enzimáticos/química , Preparaciones Farmacéuticas/química , Proteínas Virales/química , Virosis/tratamiento farmacológico , Antivirales/metabolismo , Antivirales/farmacología , Biología Computacional , Diseño de Fármacos , Farmacorresistencia Viral , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/farmacología , VIH-1/efectos de los fármacos , Hepacivirus/efectos de los fármacos , Humanos , Aprendizaje Automático , Mutación , Orthomyxoviridae/efectos de los fármacos , Preparaciones Farmacéuticas/metabolismo , Unión Proteica , Transducción de Señal , Relación Estructura-Actividad
7.
Viruses ; 13(2)2021 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-33503819

RESUMEN

Viral proteases are critical enzymes for the maturation of many human pathogenic viruses and thus are key targets for direct acting antivirals (DAAs). The current viral pandemic caused by SARS-CoV-2 is in dire need of DAAs. The Main protease (Mpro) is the focus of extensive structure-based drug design efforts which are mostly covalent inhibitors targeting the catalytic cysteine. ML188 is a non-covalent inhibitor designed to target SARS-CoV-1 Mpro, and provides an initial scaffold for the creation of effective pan-coronavirus inhibitors. In the current study, we found that ML188 inhibits SARS-CoV-2 Mpro at 2.5 µM, which is more potent than against SAR-CoV-1 Mpro. We determined the crystal structure of ML188 in complex with SARS-CoV-2 Mpro to 2.39 Å resolution. Sharing 96% sequence identity, structural comparison of the two complexes only shows subtle differences. Non-covalent protease inhibitors complement the design of covalent inhibitors against SARS-CoV-2 main protease and are critical initial steps in the design of DAAs to treat CoVID 19.


Asunto(s)
Antivirales/química , Proteasas 3C de Coronavirus/química , Inhibidores de Proteasas/química , SARS-CoV-2/enzimología , Secuencia de Aminoácidos , Antivirales/metabolismo , Dominio Catalítico , Proteasas 3C de Coronavirus/antagonistas & inhibidores , Proteasas 3C de Coronavirus/metabolismo , Cristalografía por Rayos X , Descubrimiento de Drogas , Concentración 50 Inhibidora , Modelos Moleculares , Inhibidores de Proteasas/metabolismo , Unión Proteica , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/enzimología
8.
J Med Chem ; 63(15): 8296-8313, 2020 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-32672965

RESUMEN

The design, synthesis, and X-ray structural analysis of hybrid HIV-1 protease inhibitors (PIs) containing bis-tetrahydrofuran (bis-THF) in a pseudo-C2-symmetric dipeptide isostere are described. A series of PIs were synthesized by incorporating bis-THF of darunavir on either side of the Phe-Phe isostere of lopinavir in combination with hydrophobic amino acids on the opposite P2/P2' position. Structure-activity relationship studies indicated that the bis-THF moiety can be attached at either the P2 or P2' position without significantly affecting potency. However, the group on the opposite P2/P2' position had a dramatic effect on potency depending on the size and shape of the side chain. Cocrystal structures of inhibitors with wild-type HIV-1 protease revealed that the bis-THF moiety retained similar interactions as observed in the darunavir-protease complex regardless of the position on the Phe-Phe isostere. Analyses of cocrystal structures and molecular dynamics simulations provide insights into optimizing HIV-1 PIs containing bis-THF in non-sulfonamide dipeptide isosteres.


Asunto(s)
Furanos/química , Furanos/farmacología , Inhibidores de la Proteasa del VIH/química , Inhibidores de la Proteasa del VIH/farmacología , Proteasa del VIH/metabolismo , VIH-1/enzimología , Cristalografía por Rayos X , Darunavir/análogos & derivados , Darunavir/farmacología , Dipéptidos/química , Dipéptidos/farmacología , Diseño de Fármacos , Células HEK293 , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/virología , Proteasa del VIH/química , VIH-1/efectos de los fármacos , Humanos , Modelos Moleculares , Relación Estructura-Actividad
9.
Biochemistry ; 58(35): 3711-3726, 2019 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-31386353

RESUMEN

Protease inhibitors have the highest potency among antiviral therapies against HIV-1 infections, yet the virus can evolve resistance. Darunavir (DRV), currently the most potent Food and Drug Administration-approved protease inhibitor, retains potency against single-site mutations. However, complex combinations of mutations can confer resistance to DRV. While the interdependence between mutations within HIV-1 protease is key for inhibitor potency, the molecular mechanisms that underlie this control remain largely unknown. In this study, we investigated the interdependence between the L89V and L90M mutations and their effects on DRV binding. These two mutations have been reported to be positively correlated with one another in HIV-1 patient-derived protease isolates, with the presence of one mutation making the probability of the occurrence of the second mutation more likely. The focus of our investigation is a patient-derived isolate, with 24 mutations that we call "KY"; this variant includes the L89V and L90M mutations. Three additional KY variants with back-mutations, KY(V89L), KY(M90L), and the KY(V89L/M90L) double mutation, were used to experimentally assess the individual and combined effects of these mutations on DRV inhibition and substrate processing. The enzymatic assays revealed that the KY(V89L) variant, with methionine at residue 90, is highly resistant, but its catalytic function is compromised. When a leucine to valine mutation at residue 89 is present simultaneously with the L90M mutation, a rescue of catalytic efficiency is observed. Molecular dynamics simulations of these DRV-bound protease variants reveal how the L90M mutation induces structural changes throughout the enzyme that undermine the binding interactions.


Asunto(s)
Sustitución de Aminoácidos/fisiología , Farmacorresistencia Viral/genética , Epistasis Genética/genética , Proteasa del VIH/genética , Sustitución de Aminoácidos/genética , Dominio Catalítico , Cristalografía por Rayos X , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/virología , Proteasa del VIH/química , Proteasa del VIH/metabolismo , Inhibidores de la Proteasa del VIH/farmacología , Inhibidores de la Proteasa del VIH/uso terapéutico , VIH-1/enzimología , VIH-1/genética , Humanos , Leucina/genética , Metionina/genética , Modelos Moleculares , Simulación de Dinámica Molecular , Mutación Missense/fisiología , Unión Proteica , Desnaturalización Proteica , Valina/genética
10.
J Med Chem ; 62(17): 8062-8079, 2019 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-31386368

RESUMEN

A structure-guided design strategy was used to improve the resistance profile of HIV-1 protease inhibitors by optimizing hydrogen bonding and van der Waals interactions with the protease while staying within the substrate envelope. Stereoisomers of 4-(1-hydroxyethyl)benzene and 4-(1,2-dihydroxyethyl)benzene moieties were explored as P2' ligands providing pairs of diastereoisomers epimeric at P2', which exhibited distinct potency profiles depending on the configuration of the hydroxyl group and size of the P1' group. While compounds with the 4-(1-hydroxyethyl)benzene P2' moiety maintained excellent antiviral potency against a panel of multidrug-resistant HIV-1 strains, analogues with the polar 4-(1,2-dihydroxyethyl)benzene moiety were less potent, and only the (R)-epimer incorporating a larger 2-ethylbutyl P1' group showed improved potency. Crystal structures of protease-inhibitor complexes revealed strong hydrogen bonding interactions of both (R)- and (S)-stereoisomers of the hydroxyethyl group with Asp30'. Notably, the (R)-dihydroxyethyl group was involved in a unique pattern of direct hydrogen bonding interactions with the backbone amides of Asp29' and Asp30'. The SAR data and analysis of crystal structures provide insights for optimizing these promising HIV-1 protease inhibitors.


Asunto(s)
Fármacos Anti-VIH/farmacología , Inhibidores de la Proteasa del VIH/farmacología , Proteasa del VIH/metabolismo , VIH-1/efectos de los fármacos , Fármacos Anti-VIH/síntesis química , Fármacos Anti-VIH/química , Línea Celular , Cristalografía por Rayos X , Relación Dosis-Respuesta a Droga , Células HEK293 , Proteasa del VIH/química , Inhibidores de la Proteasa del VIH/síntesis química , Inhibidores de la Proteasa del VIH/química , VIH-1/enzimología , Humanos , Enlace de Hidrógeno , Ligandos , Pruebas de Sensibilidad Microbiana , Modelos Moleculares , Estructura Molecular , Estereoisomerismo , Relación Estructura-Actividad , Especificidad por Sustrato
11.
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
13.
ACS Infect Dis ; 5(2): 316-325, 2019 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-30543749

RESUMEN

HIV-1 protease is one of the prime targets of agents used in antiretroviral therapy against HIV. However, under selective pressure of protease inhibitors, primary mutations at the active site weaken inhibitor binding to confer resistance. Darunavir (DRV) is the most potent HIV-1 protease inhibitor in clinic; resistance is limited, as DRV fits well within the substrate envelope. Nevertheless, resistance is observed due to hydrophobic changes at residues including I50, V82, and I84 that line the S1/S1' pocket within the active site. Through enzyme inhibition assays and a series of 12 crystal structures, we interrogated susceptibility of DRV and two potent analogues to primary S1' mutations. The analogues had modifications at the hydrophobic P1' moiety compared to DRV to better occupy the unexploited space in the S1' pocket where the primary mutations were located. Considerable losses of potency were observed against protease variants with I84V and I50V mutations for all three inhibitors. The crystal structures revealed an unexpected conformational change in the flap region of I50V protease bound to the analogue with the largest P1' moiety, indicating interdependency between the S1' subsite and the flap region. Collective analysis of protease-inhibitor interactions in the crystal structures using principle component analysis was able to distinguish inhibitor identity and relative potency solely based on van der Waals contacts. Our results reveal the complexity of the interplay between inhibitor P1' moiety and S1' mutations and validate principle component analyses as a useful tool for distinguishing resistance and inhibitor potency.


Asunto(s)
Darunavir/análogos & derivados , Inhibidores de la Proteasa del VIH/química , Inhibidores de la Proteasa del VIH/farmacología , Proteasa del VIH/genética , VIH-1/efectos de los fármacos , VIH-1/genética , Sitios de Unión , Dominio Catalítico , Cristalografía por Rayos X , Darunavir/química , VIH-1/enzimología , Humanos , Cinética , Modelos Moleculares , Mutación , Conformación Proteica , Especificidad por Sustrato
14.
J Chem Theory Comput ; 15(1): 637-647, 2019 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-30457868

RESUMEN

APOBEC3 (A3) proteins, a family of human cytidine deaminases, protect the host from endogenous retro-elements and exogenous viral infections by introducing hypermutations. However, overexpressed A3s can modify genomic DNA to promote tumorigenesis, especially A3B. Despite their overall similarity, A3 proteins have distinct deamination activity. Recently determined A3 structures have revealed the molecular determinants of nucleotide specificity and DNA binding. However, for A3B, the structural basis for regulation of deamination activity and the role of active site loops in coordinating DNA had remained unknown. Using advanced molecular modeling followed by experimental mutational analysis and dynamics simulations, we investigated the molecular mechanism of DNA binding by A3B-CTD. We modeled fully native A3B-DNA structure, and we identified Arg211 in loop 1 as the gatekeeper coordinating DNA and critical residue for nucleotide specificity. We also identified a unique autoinhibited conformation in A3B-CTD that restricts access and binding of DNA to the active site. Our results reveal the structural basis for DNA binding and relatively lower catalytic activity of A3B and provide opportunities for rational design of specific inhibitors to benefit cancer therapeutics.


Asunto(s)
Citidina Desaminasa/metabolismo , ADN de Cadena Simple/metabolismo , Antígenos de Histocompatibilidad Menor/metabolismo , Dominio Catalítico , Citidina Desaminasa/química , Citidina Desaminasa/genética , Humanos , Antígenos de Histocompatibilidad Menor/química , Antígenos de Histocompatibilidad Menor/genética , Simulación de Dinámica Molecular , Unión Proteica , Conformación Proteica , Especificidad por Sustrato
15.
J Mol Biol ; 430(24): 5182-5195, 2018 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-30414407

RESUMEN

Retroviral proteases (PRs) have a unique specificity that allows cleavage of sites with or without a P1' proline. A P1' proline is required at the MA/CA cleavage site due to its role in a post-cleavage conformational change in the capsid protein. However, the HIV-1 PR prefers to have large hydrophobic amino acids flanking the scissile bond, suggesting that PR recognizes two different classes of substrate sequences. We analyzed the cleavage rate of over 150 combinations of six different HIV-1 cleavage sites to explore rate determinants of cleavage. We found that cleavage rates are strongly influenced by the two amino acids flanking the amino acids at the scissile bond (P2-P1/P1'-P2'), with two complementary sets of rules. When P1' is proline, the P2 side chain interacts with a polar region in the S2 subsite of the PR, while the P2' amino acid interacts with a hydrophobic region of the S2' subsite. When P1' is not proline, the orientations of the P2 and P2' side chains with respect to the scissile bond are reversed; P2 residues interact with a hydrophobic face of the S2 subsite, while the P2' amino acid usually engages hydrophilic amino acids in the S2' subsite. These results reveal that the HIV-1 PR has evolved bi-functional S2 and S2' subsites to accommodate the steric effects imposed by a P1' proline on the orientation of P2 and P2' substrate side chains. These results also suggest a new strategy for inhibitor design to engage the multiple specificities in these subsites.


Asunto(s)
Aminoácidos/metabolismo , Proteasa del VIH/química , Proteasa del VIH/metabolismo , VIH-1/enzimología , Sitios de Unión , Interacciones Hidrofóbicas e Hidrofílicas , Cinética , Modelos Moleculares , Prolina/metabolismo , Conformación Proteica , Proteolisis , Especificidad por Sustrato
16.
ACS Infect Dis ; 2(10): 734-743, 2016 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-27657335

RESUMEN

Dengue virus (DENV), transmitted predominantly in tropical and subtropical regions by the mosquito Aedes aegypti, infects millions of people and leads to dengue fever and thousands of deaths each year. There are no direct-acting antivirals to combat DENV, and molecular and structural knowledge is required to develop such compounds. The dengue NS2B/NS3 protease is a promising target for direct-acting antivirals, as viral polyprotein cleavage during replication is required for the maturation of the viral particle. The NS2B/NS3 protease processes 8 of the 13 viral polyprotein cleavage sites to allow viral maturation. Although these sites share little sequence homology beyond the P1 and P2 positions, most are well conserved among the serotypes. How the other substrate residues, especially at the P' side, affect substrate recognition remains unclear. We exploited the tight-binding general serine protease inhibitor aprotinin to investigate protease-substrate interactions at the molecular level. We engineered aprotinin's binding loop with sequences mimicking the P' side of DENV substrates. P' residues significantly modulate substrate affinity to protease, with inhibition constants varying from nanomolar to sub-millimolar. Structural and dynamic analysis revealed the molecular basis of this modulation and allowed identifying optimal residues for each of the P' positions. In addition, isothermal titration calorimetry showed binding to be solely entropy driven for all constructs. Potential flaviviral P' side inhibitors could benefit from mimicking the optimal residues at P' positions and incorporate hydrophobicity and rigidity to maintain entropic advantage for potency.


Asunto(s)
Virus del Dengue/enzimología , Dengue/virología , Poliproteínas/metabolismo , Serina Endopeptidasas/química , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/metabolismo , Sitios de Unión , Virus del Dengue/química , Virus del Dengue/genética , Humanos , Poliproteínas/química , Poliproteínas/genética , Serina Endopeptidasas/genética , Serina Endopeptidasas/metabolismo , Especificidad por Sustrato , Proteínas no Estructurales Virales/genética
17.
Structure ; 23(5): 903-911, 2015 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-25914058

RESUMEN

Deaminase activity mediated by the human APOBEC3 family of proteins contributes to genomic instability and cancer. APOBEC3A is by far the most active in this family and can cause rapid cell death when overexpressed, but in general how the activity of APOBEC3s is regulated on a molecular level is unclear. In this study, the biochemical and structural basis of APOBEC3A substrate binding and specificity is elucidated. We find that specific binding of single-stranded DNA is regulated by the cooperative dimerization of APOBEC3A. The crystal structure elucidates this homodimer as a symmetric domain swap of the N-terminal residues. This dimer interface provides insights into how cooperative protein-protein interactions may affect function in the APOBEC3 enzymes and provides a potential scaffold for strategies aimed at reducing their mutation load.


Asunto(s)
Citidina Desaminasa/química , Citidina Desaminasa/metabolismo , ADN de Cadena Simple/metabolismo , Proteínas/química , Proteínas/metabolismo , Sitios de Unión , Cristalografía por Rayos X , Citidina Desaminasa/genética , Dimerización , Humanos , Modelos Moleculares , Mutación , Unión Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Proteínas/genética , Especificidad por Sustrato , Zinc/metabolismo
18.
J Am Chem Soc ; 136(34): 11956-63, 2014 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-25091085

RESUMEN

HIV-1 protease inhibitors are part of the highly active antiretroviral therapy effectively used in the treatment of HIV infection and AIDS. Darunavir (DRV) is the most potent of these inhibitors, soliciting drug resistance only when a complex combination of mutations occur both inside and outside the protease active site. With few exceptions, the role of mutations outside the active site in conferring resistance remains largely elusive. Through a series of DRV-protease complex crystal structures, inhibition assays, and molecular dynamics simulations, we find that single and double site mutations outside the active site often associated with DRV resistance alter the structure and dynamic ensemble of HIV-1 protease active site. These alterations correlate with the observed inhibitor binding affinities for the mutants, and suggest a network hypothesis on how the effect of distal mutations are propagated to pivotal residues at the active site and may contribute to conferring drug resistance.


Asunto(s)
Farmacorresistencia Viral/genética , Inhibidores de la Proteasa del VIH/farmacología , Proteasa del VIH/química , VIH-1/enzimología , Mutación , Sulfonamidas/farmacología , Sitios de Unión , Darunavir , Proteasa del VIH/genética , Proteasa del VIH/metabolismo , Modelos Moleculares , Unión Proteica , Conformación Proteica
19.
J Virol ; 87(8): 4176-84, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23365446

RESUMEN

Drug resistance occurs through a series of subtle changes that maintain substrate recognition but no longer permit inhibitor binding. In HIV-1 protease, mutations at I50 are associated with such subtle changes that confer differential resistance to specific inhibitors. Residue I50 is located at the protease flap tips, closing the active site upon ligand binding. Under selective drug pressure, I50V/L substitutions emerge in patients, compromising drug susceptibility and leading to treatment failure. The I50V substitution is often associated with amprenavir (APV) and darunavir (DRV) resistance, while the I50L substitution is observed in patients failing atazanavir (ATV) therapy. To explain how APV, DRV, and ATV susceptibility are influenced by mutations at residue 50 in HIV-1 protease, structural and binding thermodynamics studies were carried out on I50V/L-substituted protease variants in the compensatory mutation A71V background. Reduced affinity to both I50V/A71V and I50L/A71V double mutants is largely due to decreased binding entropy, which is compensated for by enhanced enthalpy for ATV binding to I50V variants and APV binding to I50L variants, leading to hypersusceptibility in these two cases. Analysis of the crystal structures showed that the substitutions at residue 50 affect how APV, DRV, and ATV bind the protease with altered van der Waals interactions and that the selection of I50V versus I50L is greatly influenced by the chemical moieties at the P1 position for APV/DRV and the P2 position for ATV. Thus, the varied inhibitor susceptibilities of I50V/L protease variants are largely a direct consequence of the interdependent changes in protease inhibitor interactions.


Asunto(s)
Fármacos Anti-VIH/farmacología , Farmacorresistencia Viral , Proteasa del VIH/química , VIH-1/efectos de los fármacos , Mutación Missense , Sulfato de Atazanavir , Carbamatos/farmacología , Cristalografía por Rayos X , Darunavir , Furanos , Proteasa del VIH/genética , VIH-1/genética , Humanos , Cinética , Modelos Moleculares , Proteínas Mutantes/química , Proteínas Mutantes/genética , Oligopéptidos/farmacología , Mutación Puntual , Unión Proteica , Conformación Proteica , Piridinas/farmacología , Sulfonamidas/farmacología , Termodinámica
20.
ACS Chem Biol ; 7(9): 1536-46, 2012 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-22712830

RESUMEN

The development of HIV-1 protease inhibitors has been the historic paradigm of rational structure-based drug design, where structural and thermodynamic analyses have assisted in the discovery of novel inhibitors. While the total enthalpy and entropy change upon binding determine the affinity, often the thermodynamics are considered in terms of inhibitor properties only. In the current study, profound changes are observed in the binding thermodynamics of a drug-resistant variant compared to wild-type HIV-1 protease, irrespective of the inhibitor bound. This variant (Flap+) has a combination of flap and active site mutations and exhibits extremely large entropy-enthalpy compensation compared to wild-type protease, 5-15 kcal/mol, while losing only 1-3 kcal/mol in total binding free energy for any of six FDA-approved inhibitors. Although entropy-enthalpy compensation has been previously observed for a variety of systems, never have changes of this magnitude been reported. The co-crystal structures of Flap+ protease with four of the inhibitors were determined and compared with complexes of both the wild-type protease and another drug-resistant variant that does not exhibit this energetic compensation. Structural changes conserved across the Flap+ complexes, which are more pronounced for the flaps covering the active site, likely contribute to the thermodynamic compensation. The finding that drug-resistant mutations can profoundly modulate the relative thermodynamic properties of a therapeutic target independent of the inhibitor presents a new challenge for rational drug design.


Asunto(s)
Farmacorresistencia Viral , Inhibidores de la Proteasa del VIH/química , Inhibidores de la Proteasa del VIH/farmacología , Proteasa del VIH/química , Proteasa del VIH/genética , VIH-1/enzimología , Dominio Catalítico , Cristalografía por Rayos X , Entropía , Proteasa del VIH/metabolismo , VIH-1/química , VIH-1/efectos de los fármacos , VIH-1/genética , Modelos Moleculares , Mutación , Unión Proteica , Termodinámica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA