Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Chem Inf Model ; 63(9): 2866-2880, 2023 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-37058135

RESUMEN

SARS-CoV-2 is the causative agent of COVID-19 and is responsible for the current global pandemic. The viral genome contains 5 major open reading frames of which the largest ORF1ab codes for two polyproteins, pp1ab and pp1a, which are subsequently cleaved into 16 nonstructural proteins (nsp) by two viral cysteine proteases encoded within the polyproteins. The main protease (Mpro, nsp5) cleaves the majority of the nsp's, making it essential for viral replication and has been successfully targeted for the development of antivirals. The first oral Mpro inhibitor, nirmatrelvir, was approved for treatment of COVID-19 in late December 2021 in combination with ritonavir as Paxlovid. Increasing the arsenal of antivirals and development of protease inhibitors and other antivirals with a varied mode of action remains a priority to reduce the likelihood for resistance emerging. Here, we report results from an artificial intelligence-driven approach followed by in vitro validation, allowing the identification of five fragment-like Mpro inhibitors with IC50 values ranging from 1.5 to 241 µM. The three most potent molecules (compounds 818, 737, and 183) were tested against SARS-CoV-2 by in vitro replication in Vero E6 and Calu-3 cells. Compound 818 was active in both cell models with an EC50 value comparable to its measured IC50 value. On the other hand, compounds 737 and 183 were only active in Calu-3, a preclinical model of respiratory cells, showing selective indexes twice as high as those for compound 818. We also show that our in silico methodology was successful in identifying both reversible and covalent inhibitors. For instance, compound 818 is a reversible chloromethylamide analogue of 8-methyl-γ-carboline, while compound 737 is an N-pyridyl-isatin that covalently inhibits Mpro. Given the small molecular weights of these fragments, their high binding efficiency in vitro and efficacy in blocking viral replication, these compounds represent good starting points for the development of potent lead molecules targeting the Mpro of SARS-CoV-2.


Asunto(s)
Antivirales , COVID-19 , Humanos , Antivirales/farmacología , Antivirales/química , SARS-CoV-2 , Inteligencia Artificial , Inhibidores de Proteasas/farmacología , Inhibidores de Proteasas/química , Simulación del Acoplamiento Molecular
2.
Bioorg Med Chem ; 95: 117498, 2023 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-37857256

RESUMEN

The SARS-CoV-2 papain-like protease (PLpro) and main protease (Mpro) are nucleophilic cysteine enzymes that catalyze hydrolysis of the viral polyproteins pp1a/1ab. By contrast with Mpro, PLpro is also a deubiquitinase (DUB) that accepts post-translationally modified human proteins as substrates. Here we report studies on the DUB activity of PLpro using synthetic Nε-lysine-branched oligopeptides as substrates that mimic post-translational protein modifications by ubiquitin (Ub) or Ub-like modifiers (UBLs), such as interferon stimulated gene 15 (ISG15). Mass spectrometry (MS)-based assays confirm the DUB activity of isolated recombinant PLpro. They reveal that the sequence of both the peptide fragment derived from the post-translationally modified protein and that derived from the UBL affects PLpro catalysis; the nature of substrate binding in the S sites appears to be more important for catalytic efficiency than binding in the S' sites. Importantly, the results reflect the reported cellular substrate selectivity of PLpro, i.e. human proteins conjugated to ISG15 are better substrates than those conjugated to Ub or other UBLs. The combined experimental and modelling results imply that PLpro catalysis is affected not only by the identity of the substrate residues binding in the S and S' sites, but also by the substrate fold and the conformational dynamics of the blocking loop 2 of the PLpro:substrate complex. Nε-Lysine-branched oligopeptides thus have potential to help the identification of PLpro substrates. More generally, the results imply that MS-based assays with Nε-lysine-branched oligopeptides have potential to monitor catalysis by human DUBs and hence to inform on their substrate preferences.


Asunto(s)
COVID-19 , Lisina , Humanos , Proteínas Virales/metabolismo , SARS-CoV-2 , Ubiquitina/metabolismo , Enzimas Desubicuitinizantes , Oligopéptidos
3.
Proc Natl Acad Sci U S A ; 114(32): 8544-8549, 2017 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-28739903

RESUMEN

Glycoproteins traversing the eukaryotic secretory pathway begin life in the endoplasmic reticulum (ER), where their folding is surveyed by the 170-kDa UDP-glucose:glycoprotein glucosyltransferase (UGGT). The enzyme acts as the single glycoprotein folding quality control checkpoint: it selectively reglucosylates misfolded glycoproteins, promotes their association with ER lectins and associated chaperones, and prevents premature secretion from the ER. UGGT has long resisted structural determination and sequence-based domain boundary prediction. Questions remain on how this single enzyme can flag misfolded glycoproteins of different sizes and shapes for ER retention and how it can span variable distances between the site of misfold and a glucose-accepting N-linked glycan on the same glycoprotein. Here, crystal structures of a full-length eukaryotic UGGT reveal four thioredoxin-like (TRXL) domains arranged in a long arc that terminates in two ß-sandwiches tightly clasping the glucosyltransferase domain. The fold of the molecule is topologically complex, with the first ß-sandwich and the fourth TRXL domain being encoded by nonconsecutive stretches of sequence. In addition to the crystal structures, a 15-Å cryo-EM reconstruction reveals interdomain flexibility of the TRXL domains. Double cysteine point mutants that engineer extra interdomain disulfide bridges rigidify the UGGT structure and exhibit impaired activity. The intrinsic flexibility of the TRXL domains of UGGT may therefore endow the enzyme with the promiscuity needed to recognize and reglucosylate its many different substrates and/or enable reglucosylation of N-linked glycans situated at variable distances from the site of misfold.


Asunto(s)
Glucosiltransferasas/química , Glucosiltransferasas/fisiología , Animales , Chaetomium/genética , Chaetomium/metabolismo , Cristalografía por Rayos X/métodos , Retículo Endoplásmico/metabolismo , Eucariontes/metabolismo , Células Eucariotas/metabolismo , Glucosiltransferasas/metabolismo , Glicoproteínas/metabolismo , Conformación Molecular , Dominios Proteicos/fisiología , Pliegue de Proteína , Transporte de Proteínas/fisiología , Especificidad por Sustrato
4.
Nature ; 501(7467): 385-90, 2013 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-23995689

RESUMEN

ß-barrel membrane proteins are essential for nutrient import, signalling, motility and survival. In Gram-negative bacteria, the ß-barrel assembly machinery (BAM) complex is responsible for the biogenesis of ß-barrel membrane proteins, with homologous complexes found in mitochondria and chloroplasts. Here we describe the structure of BamA, the central and essential component of the BAM complex, from two species of bacteria: Neisseria gonorrhoeae and Haemophilus ducreyi. BamA consists of a large periplasmic domain attached to a 16-strand transmembrane ß-barrel domain. Three structural features shed light on the mechanism by which BamA catalyses ß-barrel assembly. First, the interior cavity is accessible in one BamA structure and conformationally closed in the other. Second, an exterior rim of the ß-barrel has a distinctly narrowed hydrophobic surface, locally destabilizing the outer membrane. And third, the ß-barrel can undergo lateral opening, suggesting a route from the interior cavity in BamA into the outer membrane.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/biosíntesis , Proteínas de la Membrana Bacteriana Externa/química , Haemophilus/química , Neisseria gonorrhoeae/química , Proteínas de la Membrana Bacteriana Externa/genética , Membrana Celular/química , Membrana Celular/metabolismo , Cristalografía por Rayos X , Escherichia coli/química , Escherichia coli/genética , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Interacciones Hidrofóbicas e Hidrofílicas , Modelos Moleculares , Mutagénesis , Conformación Proteica , Homología Estructural de Proteína
5.
J Biol Chem ; 290(46): 27736-48, 2015 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-26370075

RESUMEN

Streptococcus pneumoniae is an important human pathogen that causes a range of disease states. Sialidases are important bacterial virulence factors. There are three pneumococcal sialidases: NanA, NanB, and NanC. NanC is an unusual sialidase in that its primary reaction product is 2-deoxy-2,3-didehydro-N-acetylneuraminic acid (Neu5Ac2en, also known as DANA), a nonspecific hydrolytic sialidase inhibitor. The production of Neu5Ac2en from α2-3-linked sialosides by the catalytic domain is confirmed within a crystal structure. A covalent complex with 3-fluoro-ß-N-acetylneuraminic acid is also presented, suggesting a common mechanism with other sialidases up to the final step of product formation. A conformation change in an active site hydrophobic loop on ligand binding constricts the entrance to the active site. In addition, the distance between the catalytic acid/base (Asp-315) and the ligand anomeric carbon is unusually short. These features facilitate a novel sialidase reaction in which the final step of product formation is direct abstraction of the C3 proton by the active site aspartic acid, forming Neu5Ac2en. NanC also possesses a carbohydrate-binding module, which is shown to bind α2-3- and α2-6-linked sialosides, as well as N-acetylneuraminic acid, which is captured in the crystal structure following hydration of Neu5Ac2en by NanC. Overall, the pneumococcal sialidases show remarkable mechanistic diversity while maintaining a common structural scaffold.


Asunto(s)
Proteínas Bacterianas/química , Inhibidores Enzimáticos/síntesis química , Neuraminidasa/química , Streptococcus pneumoniae/enzimología , Azúcares Ácidos/síntesis química , Proteínas Bacterianas/ultraestructura , Dominio Catalítico , Cristalografía por Rayos X , Neuraminidasa/ultraestructura
6.
Proc Natl Acad Sci U S A ; 109(25): 9857-62, 2012 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-22679291

RESUMEN

Bacterial pathogens are becoming increasingly resistant to antibiotics. As an alternative therapeutic strategy, phage therapy reagents containing purified viral lysins have been developed against gram-positive organisms but not against gram-negative organisms due to the inability of these types of drugs to cross the bacterial outer membrane. We solved the crystal structures of a Yersinia pestis outer membrane transporter called FyuA and a bacterial toxin called pesticin that targets this transporter. FyuA is a ß-barrel membrane protein belonging to the family of TonB dependent transporters, whereas pesticin is a soluble protein with two domains, one that binds to FyuA and another that is structurally similar to phage T4 lysozyme. The structure of pesticin allowed us to design a phage therapy reagent comprised of the FyuA binding domain of pesticin fused to the N-terminus of T4 lysozyme. This hybrid toxin kills specific Yersinia and pathogenic E. coli strains and, importantly, can evade the pesticin immunity protein (Pim) giving it a distinct advantage over pesticin. Furthermore, because FyuA is required for virulence and is more common in pathogenic bacteria, the hybrid toxin also has the advantage of targeting primarily disease-causing bacteria rather than indiscriminately eliminating natural gut flora.


Asunto(s)
Bacteriófagos/metabolismo , Bacterias Gramnegativas/virología , Mucoproteínas/metabolismo , Proteínas Bacterianas/química , Bacteriocinas/química , Bacteriófagos/fisiología , Membrana Celular/metabolismo , Microscopía por Crioelectrón , Modelos Moleculares , Mucoproteínas/química , Conformación Proteica , Ingeniería de Proteínas , Transporte de Proteínas , Receptores de Superficie Celular/química
7.
RSC Chem Biol ; 5(2): 117-130, 2024 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-38333195

RESUMEN

The SARS-CoV-2 papain-like protease (PLpro) is an antiviral drug target that catalyzes the hydrolysis of the viral polyproteins pp1a/1ab, so releasing the non-structural proteins (nsps) 1-3 that are essential for the coronavirus lifecycle. The LXGG↓X motif in pp1a/1ab is crucial for recognition and cleavage by PLpro. We describe molecular dynamics, docking, and quantum mechanics/molecular mechanics (QM/MM) calculations to investigate how oligopeptide substrates derived from the viral polyprotein bind to PLpro. The results reveal how the substrate sequence affects the efficiency of PLpro-catalyzed hydrolysis. In particular, a proline at the P2' position promotes catalysis, as validated by residue substitutions and mass spectrometry-based analyses. Analysis of PLpro catalyzed hydrolysis of LXGG motif-containing oligopeptides derived from human proteins suggests that factors beyond the LXGG motif and the presence of a proline residue at P2' contribute to catalytic efficiency, possibly reflecting the promiscuity of PLpro. The results will help in identifying PLpro substrates and guiding inhibitor design.

8.
Bull Chem Soc Jpn ; 97(5): uoae018, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38828441

RESUMEN

Due to their constrained conformations, cyclic ß2,3-amino acids (cßAA) are key building blocks that can fold peptides into compact and rigid structures, improving peptidase resistance and binding affinity to target proteins, due to their constrained conformations. Although the translation efficiency of cßAAs is generally low, our engineered tRNA, referred to as tRNAPro1E2, enabled efficient incorporation of cßAAs into peptide libraries using the flexible in vitro translation (FIT) system. Here we report on the design and application of a macrocyclic peptide library incorporating 3 kinds of cßAAs: (1R,2S)-2-aminocyclopentane carboxylic acid (ß1), (1S,2S)-2-aminocyclohexane carboxylic acid (ß2), and (1R,2R)-2-aminocyclopentane carboxylic acid. This library was applied to an in vitro selection against the SARS-CoV-2 main protease (Mpro). The resultant peptides, BM3 and BM7, bearing one ß2 and two ß1, exhibited potent inhibitory activities with IC50 values of 40 and 20 nM, respectively. BM3 and BM7 also showed remarkable serum stability with half-lives of 48 and >168 h, respectively. Notably, BM3A and BM7A, wherein the cßAAs were substituted with alanine, lost their inhibitory activities against Mpro and displayed substantially shorter serum half-lives. This observation underscores the significant contribution of cßAA to the activity and stability of peptides. Overall, our results highlight the potential of cßAA in generating potent and highly stable macrocyclic peptides with drug-like properties.

9.
Acta Crystallogr D Biol Crystallogr ; 69(Pt 5): 920-3, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23633603

RESUMEN

Crystal dehydration is a post-crystallization technique that can potentially improve the diffraction of macromolecular crystals. There are currently several ways of undertaking this process; however, dehydration experiments are often limited in their throughput and require prior manipulation of the samples. In the present study, a novel method is proposed that uses in situ plate screening to assess the effect of dehydration by combining the throughput of 96-well crystallization plates with direct X-ray feedback on crystal diffraction quality.


Asunto(s)
Ensayos Analíticos de Alto Rendimiento/métodos , Proteínas/química , Cristalización/instrumentación , Cristalización/métodos , Cristalografía por Rayos X
10.
J Med Chem ; 66(4): 2663-2680, 2023 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-36757959

RESUMEN

Nirmatrelvir (PF-07321332) is a nitrile-bearing small-molecule inhibitor that, in combination with ritonavir, is used to treat infections by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). Nirmatrelvir interrupts the viral life cycle by inhibiting the SARS-CoV-2 main protease (Mpro), which is essential for processing viral polyproteins into functional nonstructural proteins. We report studies which reveal that derivatives of nirmatrelvir and other Mpro inhibitors with a nonactivated terminal alkyne group positioned similarly to the electrophilic nitrile of nirmatrelvir can efficiently inhibit isolated Mpro and SARS-CoV-2 replication in cells. Mass spectrometric and crystallographic evidence shows that the alkyne derivatives inhibit Mpro by apparent irreversible covalent reactions with the active site cysteine (Cys145), while the analogous nitriles react reversibly. The results highlight the potential for irreversible covalent inhibition of Mpro and other nucleophilic cysteine proteases by alkynes, which, in contrast to nitriles, can be functionalized at their terminal position to optimize inhibition and selectivity, as well as pharmacodynamic and pharmacokinetic properties.


Asunto(s)
Antivirales , COVID-19 , Proteasas 3C de Coronavirus , Nitrilos , SARS-CoV-2 , Inhibidores de Proteasa Viral , Humanos , Antivirales/farmacología , Cisteína/química , SARS-CoV-2/metabolismo , Proteínas no Estructurales Virales/metabolismo , Inhibidores de Proteasa Viral/farmacología
11.
Nat Chem ; 15(7): 998-1005, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37217786

RESUMEN

γ-Amino acids can play important roles in the biological activities of natural products; however, the ribosomal incorporation of γ-amino acids into peptides is challenging. Here we report how a selection campaign employing a non-canonical peptide library containing cyclic γ2,4-amino acids resulted in the discovery of very potent inhibitors of the SARS-CoV-2 main protease (Mpro). Two kinds of cyclic γ2,4-amino acids, cis-3-aminocyclobutane carboxylic acid (γ1) and (1R,3S)-3-aminocyclopentane carboxylic acid (γ2), were ribosomally introduced into a library of thioether-macrocyclic peptides. One resultant potent Mpro inhibitor (half-maximal inhibitory concentration = 50 nM), GM4, comprising 13 residues with γ1 at the fourth position, manifests a 5.2 nM dissociation constant. An Mpro:GM4 complex crystal structure reveals the intact inhibitor spans the substrate binding cleft. The γ1 interacts with the S1' catalytic subsite and contributes to a 12-fold increase in proteolytic stability compared to its alanine-substituted variant. Knowledge of interactions between GM4 and Mpro enabled production of a variant with a 5-fold increase in potency.


Asunto(s)
Aminoácidos , COVID-19 , Aminoácidos/química , Antivirales/química , Ácidos Carboxílicos , Péptidos/química , Inhibidores de Proteasas/química , Inhibidores de Proteasas/farmacología , Conformación Proteica , SARS-CoV-2/metabolismo
12.
Sci Adv ; 9(25): eadg7865, 2023 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-37343087

RESUMEN

Inhibitor discovery for emerging drug-target proteins is challenging, especially when target structure or active molecules are unknown. Here, we experimentally validate the broad utility of a deep generative framework trained at-scale on protein sequences, small molecules, and their mutual interactions-unbiased toward any specific target. We performed a protein sequence-conditioned sampling on the generative foundation model to design small-molecule inhibitors for two dissimilar targets: the spike protein receptor-binding domain (RBD) and the main protease from SARS-CoV-2. Despite using only the target sequence information during the model inference, micromolar-level inhibition was observed in vitro for two candidates out of four synthesized for each target. The most potent spike RBD inhibitor exhibited activity against several variants in live virus neutralization assays. These results establish that a single, broadly deployable generative foundation model for accelerated inhibitor discovery is effective and efficient, even in the absence of target structure or binder information.


Asunto(s)
Anticuerpos Antivirales , COVID-19 , Humanos , Anticuerpos Antivirales/química , SARS-CoV-2/metabolismo , Unión Proteica , Secuencia de Aminoácidos
13.
Biochem Soc Trans ; 40(6): 1503-6, 2012 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-23176506

RESUMEN

Purified phage lysins present an alternative to traditional antibiotics and work by hydrolysing peptidoglycan. Phage lysins have been developed against Gram-positive pathogens such as Bacillus anthracis and Streptococcus pneumoniae, where the peptidoglycan layer is exposed on the cell surface. Addition of the lysin to a bacterial culture results in rapid death of the organism. Gram-negative bacteria are resistant to phage lysins because they contain an outer membrane that protects the peptidoglycan from degradation. We solved crystal structures of a Yersinia pestis outer-membrane protein and the bacteriocin that targets it, which informed engineering of a bacterial-phage hybrid lysin that can be transported across the outer membrane to kill specific Gram-negative bacteria. This work provides a template for engineering phage lysins against a wide variety of bacterial pathogens.


Asunto(s)
Antibacterianos/química , Bacteriocinas/química , Proteínas Virales/química , Yersinia pestis/efectos de los fármacos , Antibacterianos/farmacología , Proteínas Bacterianas/metabolismo , Bacteriocinas/genética , Bacteriófagos/enzimología , Diseño de Fármacos , Modelos Moleculares , Conformación Proteica , Ingeniería de Proteínas , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Receptores de Superficie Celular/metabolismo , Proteínas Virales/genética , Proteínas Virales/farmacología , Yersinia pestis/metabolismo
14.
Trends Parasitol ; 38(11): 930-932, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36041933

RESUMEN

Since its discovery, bovine theileriosis has caused major socioeconomic losses in sub-Saharan Africa. Acaricide resistance of the intermediate host, paucity of therapeutics, and lack of sufficiently cross-protective vaccines increase the risk of parasite spread due to global warming. Here, we highlight three important areas that require investigation to develop next-generation vaccines.


Asunto(s)
Acaricidas , Vacunas Antiprotozoos , Theileria parva , Theileriosis , Animales , Bovinos , Humanos , Theileriosis/parasitología , Theileriosis/prevención & control
15.
Acta Crystallogr D Struct Biol ; 78(Pt 6): 752-769, 2022 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-35647922

RESUMEN

In macromolecular crystallography, radiation damage limits the amount of data that can be collected from a single crystal. It is often necessary to merge data sets from multiple crystals; for example, small-wedge data collections from micro-crystals, in situ room-temperature data collections and data collection from membrane proteins in lipidic mesophases. Whilst the indexing and integration of individual data sets may be relatively straightforward with existing software, merging multiple data sets from small wedges presents new challenges. The identification of a consensus symmetry can be problematic, particularly in the presence of a potential indexing ambiguity. Furthermore, the presence of non-isomorphous or poor-quality data sets may reduce the overall quality of the final merged data set. To facilitate and help to optimize the scaling and merging of multiple data sets, a new program, xia2.multiplex, has been developed which takes data sets individually integrated with DIALS and performs symmetry analysis, scaling and merging of multi-crystal data sets. xia2.multiplex also performs analysis of various pathologies that typically affect multi-crystal data sets, including non-isomorphism, radiation damage and preferential orientation. After the description of a number of use cases, the benefit of xia2.multiplex is demonstrated within a wider autoprocessing framework in facilitating a multi-crystal experiment collected as part of in situ room-temperature fragment-screening experiments on the SARS-CoV-2 main protease.


Asunto(s)
COVID-19 , Cristalografía por Rayos X , Análisis de Datos , Humanos , Sustancias Macromoleculares/química , SARS-CoV-2
16.
J Med Chem ; 65(11): 7682-7696, 2022 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-35549342

RESUMEN

The SARS-CoV-2 main protease (Mpro) is a medicinal chemistry target for COVID-19 treatment. Given the clinical efficacy of ß-lactams as inhibitors of bacterial nucleophilic enzymes, they are of interest as inhibitors of viral nucleophilic serine and cysteine proteases. We describe the synthesis of penicillin derivatives which are potent Mpro inhibitors and investigate their mechanism of inhibition using mass spectrometric and crystallographic analyses. The results suggest that ß-lactams have considerable potential as Mpro inhibitors via a mechanism involving reaction with the nucleophilic cysteine to form a stable acyl-enzyme complex as shown by crystallographic analysis. The results highlight the potential for inhibition of viral proteases employing nucleophilic catalysis by ß-lactams and related acylating agents.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , Cisteína , Antivirales/química , Antivirales/farmacología , Proteasas 3C de Coronavirus , Cisteína Endopeptidasas/química , Humanos , Penicilinas , Inhibidores de Proteasas/química , Inhibidores de Proteasas/farmacología , SARS-CoV-2 , beta-Lactamas
17.
ChemMedChem ; 17(9): e202200016, 2022 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-35085423

RESUMEN

The two SARS-CoV-2 proteases, i. e. the main protease (Mpro ) and the papain-like protease (PLpro ), which hydrolyze the viral polypeptide chain giving functional non-structural proteins, are essential for viral replication and are medicinal chemistry targets. We report a high-throughput mass spectrometry (MS)-based assay which directly monitors PLpro catalysis in vitro. The assay was applied to investigate the effect of reported small-molecule PLpro inhibitors and selected Mpro inhibitors on PLpro catalysis. The results reveal that some, but not all, PLpro inhibitor potencies differ substantially from those obtained using fluorescence-based assays. Some substrate-competing Mpro inhibitors, notably PF-07321332 (nirmatrelvir) which is in clinical development, do not inhibit PLpro . Less selective Mpro inhibitors, e. g. auranofin, inhibit PLpro , highlighting the potential for dual PLpro /Mpro inhibition. MS-based PLpro assays, which are orthogonal to widely employed fluorescence-based assays, are of utility in validating inhibitor potencies, especially for inhibitors operating by non-covalent mechanisms.


Asunto(s)
COVID-19 , SARS-CoV-2 , Antivirales/química , Proteasas Similares a la Papaína de Coronavirus , Humanos , Lactamas , Leucina , Espectrometría de Masas , Nitrilos , Péptido Hidrolasas , Prolina , Inhibidores de Proteasas/farmacología
18.
PLoS One ; 16(10): e0256070, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34653190

RESUMEN

Nontypeable Haemophilus influenzae (NTHi) is a significant pathogen in respiratory disease and otitis media. Important for NTHi survival, colonization and persistence in vivo is the Sap (sensitivity to antimicrobial peptides) ABC transporter system. Current models propose a direct role for Sap in heme and antimicrobial peptide (AMP) transport. Here, the crystal structure of SapA, the periplasmic component of Sap, in a closed, ligand bound conformation, is presented. Phylogenetic and cavity volume analysis predicts that the small, hydrophobic SapA central ligand binding cavity is most likely occupied by a hydrophobic di- or tri- peptide. The cavity is of insufficient volume to accommodate heme or folded AMPs. Crystal structures of SapA have identified surface interactions with heme and dsRNA. Heme binds SapA weakly (Kd 282 µM) through a surface exposed histidine, while the dsRNA is coordinated via residues which constitute part of a conserved motif (estimated Kd 4.4 µM). The RNA affinity falls within the range observed for characterized RNA/protein complexes. Overall, we describe in molecular-detail the interactions of SapA with heme and dsRNA and propose a role for SapA in the transport of di- or tri-peptides.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Proteínas Portadoras/metabolismo , Haemophilus influenzae/metabolismo , Hemo/metabolismo , ARN Bicatenario/metabolismo , Transportadoras de Casetes de Unión a ATP/genética , Antibacterianos/farmacología , Proteínas Portadoras/genética , Cristalografía por Rayos X , Farmacorresistencia Bacteriana Múltiple/genética , Infecciones por Haemophilus/microbiología , Infecciones por Haemophilus/patología , Haemophilus influenzae/efectos de los fármacos , Haemophilus influenzae/genética , Otitis Media/microbiología , Otitis Media/patología , Conformación Proteica , Transporte de Proteínas/fisiología , ARN Bicatenario/genética , Motivos de Unión al ARN/genética , Factores de Virulencia/metabolismo
19.
Chem Commun (Camb) ; 57(12): 1430-1433, 2021 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-33462575

RESUMEN

The main viral protease (Mpro) of SARS-CoV-2 is a nucleophilic cysteine hydrolase and a current target for anti-viral chemotherapy. We describe a high-throughput solid phase extraction coupled to mass spectrometry Mpro assay. The results reveal some ß-lactams, including penicillin esters, are active site reacting Mpro inhibitors, thus highlighting the potential of acylating agents for Mpro inhibition.


Asunto(s)
Antivirales/farmacología , Cisteína Endopeptidasas/efectos de los fármacos , Espectrometría de Masas/métodos , Inhibidores de Proteasas/farmacología , SARS-CoV-2/efectos de los fármacos , beta-Lactamas/farmacología , Acilación , Antivirales/química , COVID-19/virología , Dominio Catalítico , Ensayos Analíticos de Alto Rendimiento , Humanos , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Inhibidores de Proteasas/química , SARS-CoV-2/enzimología , beta-Lactamas/química
20.
Cell Chem Biol ; 28(12): 1795-1806.e5, 2021 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-34174194

RESUMEN

Designing covalent inhibitors is increasingly important, although it remains challenging. Here, we present covalentizer, a computational pipeline for identifying irreversible inhibitors based on structures of targets with non-covalent binders. Through covalent docking of tailored focused libraries, we identify candidates that can bind covalently to a nearby cysteine while preserving the interactions of the original molecule. We found âˆ¼11,000 cysteines proximal to a ligand across 8,386 complexes in the PDB. Of these, the protocol identified 1,553 structures with covalent predictions. In a prospective evaluation, five out of nine predicted covalent kinase inhibitors showed half-maximal inhibitory concentration (IC50) values between 155 nM and 4.5 µM. Application against an existing SARS-CoV Mpro reversible inhibitor led to an acrylamide inhibitor series with low micromolar IC50 values against SARS-CoV-2 Mpro. The docking was validated by 12 co-crystal structures. Together these examples hint at the vast number of covalent inhibitors accessible through our protocol.


Asunto(s)
Diseño de Fármacos , Inhibidores de Proteínas Quinasas/química , SARS-CoV-2/enzimología , Proteínas de la Matriz Viral/antagonistas & inhibidores , Acrilamida/química , Acrilamida/metabolismo , Sitios de Unión , COVID-19/patología , COVID-19/virología , Dominio Catalítico , Biología Computacional/métodos , Bases de Datos de Proteínas , Humanos , Concentración 50 Inhibidora , Simulación del Acoplamiento Molecular , Inhibidores de Proteínas Quinasas/metabolismo , SARS-CoV-2/aislamiento & purificación , Proteínas de la Matriz Viral/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA