Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 88
Filtrar
1.
Nat Commun ; 15(1): 1164, 2024 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-38326375

RESUMEN

The NACHT-, leucine-rich-repeat-, and pyrin domain-containing protein 3 (NLRP3) is a critical intracellular inflammasome sensor and an important clinical target against inflammation-driven human diseases. Recent studies have elucidated its transition from a closed cage to an activated disk-like inflammasome, but the intermediate activation mechanism remains elusive. Here we report the cryo-electron microscopy structure of NLRP3, which forms an open octamer and undergoes a ~ 90° hinge rotation at the NACHT domain. Mutations on open octamer's interfaces reduce IL-1ß signaling, highlighting its essential role in NLRP3 activation/inflammasome assembly. The centrosomal NIMA-related kinase 7 (NEK7) disrupts large NLRP3 oligomers and forms NEK7/NLRP3 monomers/dimers which is a critical step preceding the assembly of the disk-like inflammasome. These data demonstrate an oligomeric cooperative activation of NLRP3 and provide insight into its inflammasome assembly mechanism.


Asunto(s)
Inflamasomas , Proteína con Dominio Pirina 3 de la Familia NLR , Humanos , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Inflamasomas/metabolismo , Microscopía por Crioelectrón , Quinasas Relacionadas con NIMA/genética , Quinasas Relacionadas con NIMA/metabolismo , Proteínas
2.
Int J Mol Sci ; 24(22)2023 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-38003312

RESUMEN

Artificial intelligence (AI) has gained significant traction in the field of drug discovery, with deep learning (DL) algorithms playing a crucial role in predicting protein-ligand binding affinities. Despite advancements in neural network architectures, system representation, and training techniques, the performance of DL affinity prediction has reached a plateau, prompting the question of whether it is truly solved or if the current performance is overly optimistic and reliant on biased, easily predictable data. Like other DL-related problems, this issue seems to stem from the training and test sets used when building the models. In this work, we investigate the impact of several parameters related to the input data on the performance of neural network affinity prediction models. Notably, we identify the size of the binding pocket as a critical factor influencing the performance of our statistical models; furthermore, it is more important to train a model with as much data as possible than to restrict the training to only high-quality datasets. Finally, we also confirm the bias in the typically used current test sets. Therefore, several types of evaluation and benchmarking are required to understand models' decision-making processes and accurately compare the performance of models.


Asunto(s)
Inteligencia Artificial , Redes Neurales de la Computación , Algoritmos , Unión Proteica , Ligandos
3.
J Chem Theory Comput ; 19(21): 7437-7458, 2023 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-37902715

RESUMEN

Membrane proteins have diverse functions within cells and are well-established drug targets. The advances in membrane protein structural biology have revealed drug and lipid binding sites on membrane proteins, while computational methods such as molecular simulations can resolve the thermodynamic basis of these interactions. Particularly, alchemical free energy calculations have shown promise in the calculation of reliable and reproducible binding free energies of protein-ligand and protein-lipid complexes in membrane-associated systems. In this review, we present an overview of representative alchemical free energy studies on G-protein-coupled receptors, ion channels, transporters as well as protein-lipid interactions, with emphasis on best practices and critical aspects of running these simulations. Additionally, we analyze challenges and successes when running alchemical free energy calculations on membrane-associated proteins. Finally, we highlight the value of alchemical free energy calculations calculations in drug discovery and their applicability in the pharmaceutical industry.


Asunto(s)
Proteínas de la Membrana , Simulación de Dinámica Molecular , Entropía , Termodinámica , Ligandos , Lípidos , Unión Proteica
4.
J Chem Theory Comput ; 19(15): 5058-5076, 2023 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-37487138

RESUMEN

Binding free energy calculations predict the potency of compounds to protein binding sites in a physically rigorous manner and see broad application in prioritizing the synthesis of novel drug candidates. Relative binding free energy (RBFE) calculations have emerged as an industry-standard approach to achieve highly accurate rank-order predictions of the potency of related compounds; however, this approach requires that the ligands share a common scaffold and a common binding mode, restricting the methods' domain of applicability. This is a critical limitation since complex modifications to the ligands, especially core hopping, are very common in drug design. Absolute binding free energy (ABFE) calculations are an alternate method that can be used for ligands that are not congeneric. However, ABFE suffers from a known problem of long convergence times due to the need to sample additional degrees of freedom within each system, such as sampling rearrangements necessary to open and close the binding site. Here, we report on an alternative method for RBFE, called Separated Topologies (SepTop), which overcomes the issues in both of the aforementioned methods by enabling large scaffold changes between ligands with a convergence time comparable to traditional RBFE. Instead of only mutating atoms that vary between two ligands, this approach performs two absolute free energy calculations at the same time in opposite directions, one for each ligand. Defining the two ligands independently allows the comparison of the binding of diverse ligands without the artificial constraints of identical poses or a suitable atom-atom mapping. This approach also avoids the need to sample the unbound state of the protein, making it more efficient than absolute binding free energy calculations. Here, we introduce an implementation of SepTop. We developed a general and efficient protocol for running SepTop, and we demonstrated the method on four diverse, pharmaceutically relevant systems. We report the performance of the method, as well as our practical insights into the strengths, weaknesses, and challenges of applying this method in an industrial drug design setting. We find that the accuracy of the approach is sufficiently high to rank order ligands with an accuracy comparable to traditional RBFE calculations while maintaining the additional flexibility of SepTop.

5.
J Biol Chem ; 299(6): 104794, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37164155

RESUMEN

Clinical development of γ-secretases, a family of intramembrane cleaving proteases, as therapeutic targets for a variety of disorders including cancer and Alzheimer's disease was aborted because of serious mechanism-based side effects in the phase III trials of unselective inhibitors. Selective inhibition of specific γ-secretase complexes, containing either PSEN1 or PSEN2 as the catalytic subunit and APH1A or APH1B as supporting subunits, does provide a feasible therapeutic window in preclinical models of these disorders. We explore here the pharmacophoric features required for PSEN1 versus PSEN2 selective inhibition. We synthesized a series of brain penetrant 2-azabicyclo[2,2,2]octane sulfonamides and identified a compound with low nanomolar potency and high selectivity (>250-fold) toward the PSEN1-APH1B subcomplex versus PSEN2 subcomplexes. We used modeling and site-directed mutagenesis to identify critical amino acids along the entry part of this inhibitor into the catalytic site of PSEN1. Specific targeting one of the different γ-secretase complexes might provide safer drugs in the future.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide , Complejos Multiproteicos , Presenilina-1 , Sulfonamidas , Humanos , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/enzimología , Enfermedad de Alzheimer/metabolismo , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Presenilina-1/antagonistas & inhibidores , Presenilina-1/metabolismo , Complejos Multiproteicos/antagonistas & inhibidores , Complejos Multiproteicos/metabolismo , Sulfonamidas/farmacología , Especificidad por Sustrato , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Neoplasias/metabolismo
6.
J Chem Inf Model ; 63(9): 2857-2865, 2023 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-37093848

RESUMEN

Affinity ranking of structurally diverse small-molecule ligands is a challenging problem with important applications in structure-based drug discovery. Absolute binding free energy methods can model diverse ligands, but the high computational cost of the current methods limits application to data sets with few ligands. We recently developed MELD-Bracket, a Molecular Dynamics method for efficient affinity ranking of ligands [ JCTC 2022, 18 (1), 374-379]. It utilizes a Bayesian framework to guide sampling to relevant regions of phase space, and it couples this with a bracket-like competition on a pool of ligands. Here we find that 6-competitor MELD-Bracket can rank dozens of diverse ligands that have low structural similarity and different net charges. We benchmark it on four protein systems─PTB1B, Tyk2, BACE, and JAK3─having varied modes of interactions. We also validated 8-competitor and 12-competitor protocols. The MELD-Bracket protocols presented here may have the appropriate balance of accuracy and computational efficiency to be suitable for ranking diverse ligands from typical drug discovery campaigns.


Asunto(s)
Simulación de Dinámica Molecular , Proteínas , Unión Proteica , Teorema de Bayes , Proteínas/química , Ligandos
7.
J Chem Inf Model ; 63(6): 1776-1793, 2023 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-36878475

RESUMEN

Drug discovery is accelerated with computational methods such as alchemical simulations to estimate ligand affinities. In particular, relative binding free energy (RBFE) simulations are beneficial for lead optimization. To use RBFE simulations to compare prospective ligands in silico, researchers first plan the simulation experiment, using graphs where nodes represent ligands and graph edges represent alchemical transformations between ligands. Recent work demonstrated that optimizing the statistical architecture of these perturbation graphs improves the accuracy of the predicted changes in the free energy of ligand binding. Therefore, to improve the success rate of computational drug discovery, we present the open-source software package High Information Mapper (HiMap)─a new take on its predecessor, Lead Optimization Mapper (LOMAP). HiMap removes heuristics decisions from design selection and instead finds statistically optimal graphs over ligands clustered with machine learning. Beyond optimal design generation, we present theoretical insights for designing alchemical perturbation maps. Some of these results include that for n number of nodes, the precision of perturbation maps is stable at n·ln(n) edges. This result indicates that even an "optimal" graph can result in unexpectedly high errors if a plan includes too few alchemical transformations for the given number of ligands and edges. And, as a study compares more ligands, the performance of even optimal graphs will deteriorate with linear scaling of the edge count. In this sense, ensuring an A- or D-optimal topology is not enough to produce robust errors. We additionally find that optimal designs will converge more rapidly than radial and LOMAP designs. Moreover, we derive bounds for how clustering reduces cost for designs with a constant expected relative error per cluster, invariant of the size of the design. These results inform how to best design perturbation maps for computational drug discovery and have broader implications for experimental design.


Asunto(s)
Simulación de Dinámica Molecular , Termodinámica , Ligandos , Estudios Prospectivos , Entropía , Unión Proteica
8.
Artículo en Inglés | MEDLINE | ID: mdl-36382113

RESUMEN

Free energy calculations are rapidly becoming indispensable in structure-enabled drug discovery programs. As new methods, force fields, and implementations are developed, assessing their expected accuracy on real-world systems (benchmarking) becomes critical to provide users with an assessment of the accuracy expected when these methods are applied within their domain of applicability, and developers with a way to assess the expected impact of new methodologies. These assessments require construction of a benchmark-a set of well-prepared, high quality systems with corresponding experimental measurements designed to ensure the resulting calculations provide a realistic assessment of expected performance when these methods are deployed within their domains of applicability. To date, the community has not yet adopted a common standardized benchmark, and existing benchmark reports suffer from a myriad of issues, including poor data quality, limited statistical power, and statistically deficient analyses, all of which can conspire to produce benchmarks that are poorly predictive of real-world performance. Here, we address these issues by presenting guidelines for (1) curating experimental data to develop meaningful benchmark sets, (2) preparing benchmark inputs according to best practices to facilitate widespread adoption, and (3) analysis of the resulting predictions to enable statistically meaningful comparisons among methods and force fields. We highlight challenges and open questions that remain to be solved in these areas, as well as recommendations for the collection of new datasets that might optimally serve to measure progress as methods become systematically more reliable. Finally, we provide a curated, versioned, open, standardized benchmark set adherent to these standards (PLBenchmarks) and an open source toolkit for implementing standardized best practices assessments (arsenic) for the community to use as a standardized assessment tool. While our main focus is free energy methods based on molecular simulations, these guidelines should prove useful for assessment of the rapidly growing field of machine learning methods for affinity prediction as well.

9.
J Chem Inf Model ; 62(23): 6094-6104, 2022 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-36433835

RESUMEN

Force fields form the basis for classical molecular simulations, and their accuracy is crucial for the quality of, for instance, protein-ligand binding simulations in drug discovery. The huge diversity of small-molecule chemistry makes it a challenge to build and parameterize a suitable force field. The Open Force Field Initiative is a combined industry and academic consortium developing a state-of-the-art small-molecule force field. In this report, industry members of the consortium worked together to objectively evaluate the performance of the force fields (referred to here as OpenFF) produced by the initiative on a combined public and proprietary dataset of 19,653 relevant molecules selected from their internal research and compound collections. This evaluation was important because it was completely blind; at most partners, none of the molecules or data were used in force field development or testing prior to this work. We compare the Open Force Field "Sage" version 2.0.0 and "Parsley" version 1.3.0 with GAFF-2.11-AM1BCC, OPLS4, and SMIRNOFF99Frosst. We analyzed force-field-optimized geometries and conformer energies compared to reference quantum mechanical data. We show that OPLS4 performs best, and the latest Open Force Field release shows a clear improvement compared to its predecessors. The performance of established force fields such as GAFF-2.11 was generally worse. While OpenFF researchers were involved in building the benchmarking infrastructure used in this work, benchmarking was done entirely in-house within industrial organizations and the resulting assessment is reported here. This work assesses the force field performance using separate benchmarking steps, external datasets, and involving external research groups. This effort may also be unique in terms of the number of different industrial partners involved, with 10 different companies participating in the benchmark efforts.


Asunto(s)
Proteínas , Termodinámica , Ligandos , Proteínas/química , Fenómenos Físicos
10.
J Chem Theory Comput ; 18(10): 6259-6270, 2022 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-36148968

RESUMEN

Drug discovery can be thought of as a search for a needle in a haystack: searching through a large chemical space for the most active compounds. Computational techniques can narrow the search space for experimental follow up, but even they become unaffordable when evaluating large numbers of molecules. Therefore, machine learning (ML) strategies are being developed as computationally cheaper complementary techniques for navigating and triaging large chemical libraries. Here, we explore how an active learning protocol can be combined with first-principles based alchemical free energy calculations to identify high affinity phosphodiesterase 2 (PDE2) inhibitors. We first calibrate the procedure using a set of experimentally characterized PDE2 binders. The optimized protocol is then used prospectively on a large chemical library to navigate toward potent inhibitors. In the active learning cycle, at every iteration a small fraction of compounds is probed by alchemical calculations and the obtained affinities are used to train ML models. With successive rounds, high affinity binders are identified by explicitly evaluating only a small subset of compounds in a large chemical library, thus providing an efficient protocol that robustly identifies a large fraction of true positives.


Asunto(s)
Bibliotecas de Moléculas Pequeñas , Vuelo Espacial , Hidrolasas Diéster Fosfóricas , Unión Proteica , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología , Termodinámica
11.
Sci Rep ; 12(1): 10433, 2022 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-35729177

RESUMEN

Optimization of binding affinities for compounds to their target protein is a primary objective in drug discovery. Herein we report on a collaborative study that evaluates a set of compounds binding to ROS1 kinase. We use ESMACS (enhanced sampling of molecular dynamics with approximation of continuum solvent) and TIES (thermodynamic integration with enhanced sampling) protocols to rank the binding free energies. The predicted binding free energies from ESMACS simulations show good correlations with experimental data for subsets of the compounds. Consistent binding free energy differences are generated for TIES and ESMACS. Although an unexplained overestimation exists, we obtain excellent statistical rankings across the set of compounds from the TIES protocol, with a Pearson correlation coefficient of 0.90 between calculated and experimental activities.


Asunto(s)
Proteínas Tirosina Quinasas , Proteínas Proto-Oncogénicas , Simulación de Dinámica Molecular , Unión Proteica , Termodinámica
12.
J Chem Inf Model ; 62(5): 1172-1177, 2022 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-35191702

RESUMEN

Nowadays, drug design projects benefit from highly accurate protein-ligand binding free energy predictions based on molecular dynamics simulations. While such calculations have been computationally expensive in the past, we now demonstrate that workflows built on open source software packages can efficiently leverage pre-exascale computing resources to screen hundreds of compounds in a matter of days. We report our results of free energy calculations on a large set of pharmaceutically relevant targets assembled to reflect industrial drug discovery projects.


Asunto(s)
Diseño de Fármacos , Simulación de Dinámica Molecular , Ligandos , Unión Proteica , Programas Informáticos , Termodinámica
13.
ACS Med Chem Lett ; 13(1): 76-83, 2022 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-35059126

RESUMEN

We recently disclosed a set of heteroaryl-fused piperazine inhibitors of BACE1 that combined nanomolar potency with good intrinsic permeability and low Pgp-mediated efflux. Herein we describe further work on two prototypes of this family of inhibitors aimed at modulating their basicity and reducing binding to the human ether-a-go-go-related gene (hERG) channel. This effort has led to the identification of compound 36, a highly potent (hAß42 cell IC50 = 1.3 nM), cardiovascularly safe, and orally bioavailable compound that elicited sustained Aß42 reduction in mouse and dog animal models.

14.
J Chem Inf Model ; 62(3): 703-717, 2022 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-35061383

RESUMEN

The accurate prediction of binding affinity between protein and small molecules with free energy methods, particularly the difference in binding affinities via relative binding free energy calculations, has undergone a dramatic increase in use and impact over recent years. The improvements in methodology, hardware, and implementation can deliver results with less than 1 kcal/mol mean unsigned error between calculation and experiment. This is a remarkable achievement and beckons some reflection on the significance of calculation approaching the accuracy of experiment. In this article, we describe a statistical analysis of the implications of variance (standard deviation) of both experimental and calculated binding affinities with respect to the unknown true binding affinity. We reveal that plausible ratios of standard deviation in experiment and calculation can lead to unexpected outcomes for assessing the performance of predictions. The work extends beyond the case of binding free energies to other affinity or property prediction methods.


Asunto(s)
Proteínas , Entropía , Ligandos , Unión Proteica , Proteínas/química , Termodinámica
15.
J Chem Inf Model ; 62(3): 533-543, 2022 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-35041430

RESUMEN

The existence of a druggable binding pocket is a prerequisite for computational drug-target interaction studies including virtual screening. Retrospective studies have shown that extended sampling methods like Markov State Modeling and mixed-solvent simulations can identify cryptic pockets relevant for drug discovery. Here, we apply a combination of mixed-solvent molecular dynamics (MD) and time-structure independent component analysis (TICA) to four retrospective case studies: NPC2, the CECR2 bromodomain, TEM-1, and MCL-1. We compare previous experimental and computational findings to our results. It is shown that the successful identification of cryptic pockets depends on the system and the cosolvent probes. We used alternative TICA internal features such as the unbiased backbone coordinates or backbone dihedrals versus biased interatomic distances. We found that in the case of NPC2, TEM-1, and MCL-1, the use of unbiased features is able to identify cryptic pockets, although in the case of the CECR2 bromodomain, more specific features are required to properly capture a pocket opening. In the perspective of virtual screening applications, it is shown how docking studies with the parent ligands depend critically on the conformational state of the targets.


Asunto(s)
Descubrimiento de Drogas , Simulación de Dinámica Molecular , Sitios de Unión , Ligandos , Simulación del Acoplamiento Molecular , Estudios Retrospectivos , Solventes/química
16.
Heliyon ; 8(12): e12392, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36590518

RESUMEN

Malic enzymes (ME1, ME2, and ME3) are involved in cellular energy regulation, redox homeostasis, and biosynthetic processes, through the production of pyruvate and reducing agent NAD(P)H. Recent studies have implicated the third and least well-characterized isoform, mitochondrial NADP+-dependent malic enzyme 3 (ME3), as a therapeutic target for pancreatic cancers. Here, we utilized an integrated structure approach to determine the structures of ME3 in various ligand-binding states at near-atomic resolutions. ME3 is captured in the open form existing as a stable tetramer and its dynamic Domain C is critical for activity. Catalytic assay results reveal that ME3 is a non-allosteric enzyme and does not require modulators for activity while structural analysis suggests that the inner stability of ME3 Domain A relative to ME2 disables allostery in ME3. With structural information available for all three malic enzymes, the foundation has been laid to understand the structural and biochemical differences of these enzymes and could aid in the development of specific malic enzyme small molecule drugs.

18.
J Chem Theory Comput ; 17(10): 6262-6280, 2021 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-34551262

RESUMEN

We present a methodology for defining and optimizing a general force field for classical molecular simulations, and we describe its use to derive the Open Force Field 1.0.0 small-molecule force field, codenamed Parsley. Rather than using traditional atom typing, our approach is built on the SMIRKS-native Open Force Field (SMIRNOFF) parameter assignment formalism, which handles increases in the diversity and specificity of the force field definition without needlessly increasing the complexity of the specification. Parameters are optimized with the ForceBalance tool, based on reference quantum chemical data that include torsion potential energy profiles, optimized gas-phase structures, and vibrational frequencies. These quantum reference data are computed and are maintained with QCArchive, an open-source and freely available distributed computing and database software ecosystem. In this initial application of the method, we present essentially a full optimization of all valence parameters and report tests of the resulting force field against compounds and data types outside the training set. These tests show improvements in optimized geometries and conformational energetics and demonstrate that Parsley's accuracy for liquid properties is similar to that of other general force fields, as is accuracy on binding free energies. We find that this initial Parsley force field affords accuracy similar to that of other general force fields when used to calculate relative binding free energies spanning 199 protein-ligand systems. Additionally, the resulting infrastructure allows us to rapidly optimize an entirely new force field with minimal human intervention.


Asunto(s)
Benchmarking , Petroselinum , Ecosistema , Humanos , Ligandos , Conformación Molecular
19.
ACS Omega ; 6(35): 22997-23006, 2021 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-34514269

RESUMEN

Glutamate hyperfunction is implicated in multiple neurological and psychiatric diseases. Activation of the mGlu2 receptor results in reduced glutamate release and decreased excitability representing a promising novel therapeutic agent for the treatment of disorders such as epilepsy, schizophrenia, mood, anxiety, and other neuropsychiatric disorders. We have previously reported substantial efforts leading to potent and selective mGlu2 PAMs from different chemical series. Herein, the discovery and optimization of a novel series of imidazopyrazinone mGlu2 PAMs are reported. This new scaffold originated from computational searching of fragment databases and comparison with our previously explored scaffolds. Optimization guided by our robust understanding of SAR from former series led to potent, selective, and brain-penetrant compounds.

20.
Sci Rep ; 11(1): 15319, 2021 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-34321581

RESUMEN

Inhibition of the NACHT, LRR and PYD domains-containing protein 3 (NLRP3) inflammasome has recently emerged as a promising therapeutic target for several inflammatory diseases. After priming and activation by inflammation triggers, NLRP3 forms a complex with apoptosis-associated speck-like protein containing a CARD domain (ASC) followed by formation of the active inflammasome. Identification of inhibitors of NLRP3 activation requires a well-validated primary high-throughput assay followed by the deployment of a screening cascade of assays enabling studies of structure-activity relationship, compound selectivity and efficacy in disease models. We optimized a NLRP3-dependent fluorescent tagged ASC speck formation assay in murine immortalized bone marrow-derived macrophages and utilized it to screen a compound library of 81,000 small molecules. Our high-content screening assay yielded robust assay metrics and identified a number of inhibitors of NLRP3-dependent ASC speck formation, including compounds targeting HSP90, JAK and IKK-ß. Additional assays to investigate inflammasome priming or activation, NLRP3 downstream effectors such as caspase-1, IL-1ß and pyroptosis form the basis of a screening cascade to identify NLRP3 inflammasome inhibitors in drug discovery programs.


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Ensayos Analíticos de Alto Rendimiento/métodos , Inflamasomas/efectos de los fármacos , Macrófagos/efectos de los fármacos , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Animales , Proteínas Adaptadoras de Señalización CARD/metabolismo , Caspasa 1/biosíntesis , Células Cultivadas , Dimetilsulfóxido/farmacología , Descubrimiento de Drogas , Furanos/farmacología , Genes Reporteros , Indenos/farmacología , Interleucina-1beta/biosíntesis , Lipopolisacáridos/farmacología , Ratones , Nigericina/farmacología , Fenotipo , Piroptosis/efectos de los fármacos , Proteínas Recombinantes/metabolismo , Bibliotecas de Moléculas Pequeñas , Sulfonamidas/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...