Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.080
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Cell ; 178(3): 521-535.e23, 2019 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-31348885

RESUMEN

Intracellular accumulation of misfolded proteins causes toxic proteinopathies, diseases without targeted therapies. Mucin 1 kidney disease (MKD) results from a frameshift mutation in the MUC1 gene (MUC1-fs). Here, we show that MKD is a toxic proteinopathy. Intracellular MUC1-fs accumulation activated the ATF6 unfolded protein response (UPR) branch. We identified BRD4780, a small molecule that clears MUC1-fs from patient cells, from kidneys of knockin mice and from patient kidney organoids. MUC1-fs is trapped in TMED9 cargo receptor-containing vesicles of the early secretory pathway. BRD4780 binds TMED9, releases MUC1-fs, and re-routes it for lysosomal degradation, an effect phenocopied by TMED9 deletion. Our findings reveal BRD4780 as a promising lead for the treatment of MKD and other toxic proteinopathies. Generally, we elucidate a novel mechanism for the entrapment of misfolded proteins by cargo receptors and a strategy for their release and anterograde trafficking to the lysosome.


Asunto(s)
Benzamidas/metabolismo , Compuestos Bicíclicos con Puentes/farmacología , Heptanos/farmacología , Lisosomas/efectos de los fármacos , Proteínas de Transporte Vesicular/metabolismo , Factor de Transcripción Activador 6/metabolismo , Animales , Benzamidas/química , Benzamidas/farmacología , Compuestos Bicíclicos con Puentes/uso terapéutico , Células Epiteliales/citología , Células Epiteliales/metabolismo , Femenino , Mutación del Sistema de Lectura , Heptanos/uso terapéutico , Humanos , Receptores de Imidazolina/antagonistas & inhibidores , Receptores de Imidazolina/genética , Receptores de Imidazolina/metabolismo , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Riñón/citología , Riñón/metabolismo , Riñón/patología , Enfermedades Renales/metabolismo , Enfermedades Renales/patología , Lisosomas/metabolismo , Masculino , Ratones , Ratones Transgénicos , Mucina-1/química , Mucina-1/genética , Mucina-1/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Respuesta de Proteína Desplegada/efectos de los fármacos , Proteínas de Transporte Vesicular/química
2.
J Chem Inf Model ; 64(12): 4759-4772, 2024 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-38857305

RESUMEN

The accurate experimental estimation of protein-ligand systems' residence time (τ) has become very relevant in drug design projects due to its importance in the last stages of refinement of the drug's pharmacodynamics and pharmacokinetics. It is now well-known that it is not sufficient to estimate the affinity of a protein-drug complex in the thermodynamic equilibrium process in in vitro experiments (closed systems), where the concentrations of the drug and protein remain constant. On the contrary, it is mandatory to consider the conformational dynamics of the system in terms of the binding and unbinding processes between protein and drugs in in vivo experiments (open systems), where their concentrations are in constant flux. This last model has been proven to dictate much of several drugs' pharmacological activities in vivo. At the atomistic level, molecular dynamics simulations can explain why some drugs are more effective than others or unveil the molecular aspects that make some drugs work better in one molecular target. Here, the protein kinases Aurora A/B, complexed with its inhibitor Danusertib, were studied using conventional and enhanced molecular dynamics (MD) simulations to estimate the dissociation paths and, therefore, the computational τ values and their comparison with experimental ones. Using classical molecular dynamics (cMD), three differential residues within the Aurora A/B active site, which seems to play an essential role in the observed experimental Danusertib's residence time against these kinases, were characterized. Then, using WT-MetaD, the relative Danusertib's residence times against Aurora A/B kinases were measured in a nanosecond time scale and were compared to those τ values observed experimentally. In addition, the potential dissociation paths of Danusertib in Aurora A and B were characterized, and differences that might be explained by the differential residues in the enzyme's active sites were found. In perspective, it is expected that this computational protocol can be applied to other protein-ligand complexes to understand, at the molecular level, the differences in residence times and amino acids that may contribute to it.


Asunto(s)
Aurora Quinasa A , Aurora Quinasa B , Simulación de Dinámica Molecular , Aurora Quinasa B/metabolismo , Aurora Quinasa B/química , Aurora Quinasa B/antagonistas & inhibidores , Aurora Quinasa A/metabolismo , Aurora Quinasa A/química , Aurora Quinasa A/antagonistas & inhibidores , Pirazoles/química , Pirazoles/metabolismo , Conformación Proteica , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/metabolismo , Unión Proteica , Humanos , Benzamidas/química , Benzamidas/metabolismo , Benzamidas/farmacología , Termodinámica
3.
Environ Sci Technol ; 58(6): 2859-2869, 2024 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-38289638

RESUMEN

2,6-Dichlorobenzamide (BAM) is an omnipresent micropollutant in European groundwaters. Aminobacter niigataensis MSH1 is a prime candidate for biologically treating BAM-contaminated groundwater since this organism is capable of utilizing BAM as a carbon and energy source. However, detailed information on the BAM degradation kinetics by MSH1 at trace concentrations is lacking, while this knowledge is required for predicting and optimizing the degradation process. Contaminating assimilable organic carbon (AOC) in media makes the biodegradation experiment a mixed-substrate assay and hampers exploration of pollutant degradation at trace concentrations. In this study, we examined how the BAM concentration affects MSH1 growth and BAM substrate utilization kinetics in a AOC-restricted background to avoid mixed-substrate conditions. Conventional Monod kinetic models were unable to predict kinetic parameters at low concentrations from kinetics determined at high concentrations. Growth yields on BAM were concentration-dependent and decreased substantially at trace concentrations; i.e., growth of MSH1 diminished until undetectable levels at BAM concentrations below 217 µg-C/L. Nevertheless, BAM degradation continued. Decreasing growth yields at lower BAM concentrations might relate to physiological adaptations to low substrate availability or decreased expression of downstream steps of the BAM catabolic pathway beyond 2,6-dichlorobenzoic acid (2,6-DCBA) that ultimately leads to Krebs cycle intermediates for growth and energy conservation.


Asunto(s)
Benzamidas , Carbono , Phyllobacteriaceae , Biodegradación Ambiental , Benzamidas/metabolismo , Carbono/metabolismo
4.
Plant J ; 112(3): 664-676, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36069460

RESUMEN

Vacuolar H+ -ATPase (V-ATPase) has diverse functions related to plant development and growth. It creates the turgor pressure that drives cell growth by generating the energy needed for the active transport of solutes across the tonoplast. V-ATPase is a large protein complex made up of multiheteromeric subunits, some of which have unknown functions. In this study, a forward genetics-based strategy was employed to identify the vab3 mutant, which displayed resistance to isoxaben, a cellulose synthase inhibitor that could induce excessive transverse cell expansion. Map-based cloning and genetic complementary assays demonstrated that V-ATPase B subunit 3 (VAB3) is associated with the observed insensitivity of the mutant to isoxaben. Analysis of the vab3 mutant revealed defective ionic homeostasis and hypersensitivity to salt stress. Treatment with a V-ATPase inhibitor exacerbated ionic tolerance and cell elongation defects in the vab3 mutant. Notably, exogenous low-dose Ca2+ or Na+ could partially restore isoxaben resistance of the vab3 mutant, suggesting a relationship between VAB3-regulated cell growth and ion homeostasis. Taken together, the results of this study suggest that the V-ATPase subunit VAB3 is required for cell growth and ion homeostasis in Arabidopsis.


Asunto(s)
Arabidopsis , ATPasas de Translocación de Protón Vacuolares , Arabidopsis/metabolismo , ATPasas de Translocación de Protón Vacuolares/genética , ATPasas de Translocación de Protón Vacuolares/metabolismo , Benzamidas/farmacología , Benzamidas/metabolismo , Homeostasis
5.
Arch Biochem Biophys ; 748: 109782, 2023 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-37839789

RESUMEN

The efflux pump mechanism contributes to the antibiotic resistance of widely distributed strains of Staphylococcus aureus. Therefore, in the present work, the ability of the riparins N-(4-methoxyphenethyl)benzamide (I), 2-hydroxy-N-[2-(4-methoxyphenyl)ethyl]benzamide (II), 2, 6-dihydroxy-N-[ 2-(4-methoxyphenyl)ethyl]benzamide (III), and 3,4,5-trimethoxy-N-[2-(4-methoxyphenethyl)benzamide (IV) as potential inhibitors of the MepA efflux pump in S. aureus K2068 (fluoroquinolone-resistant). In addition, we performed checkerboard assays to obtain more information about the activity of riparins as potential inhibitors of MepA efflux and also analyzed the ability of riparins to act on the permeability of the bacterial membrane of S. aureus by the fluorescence method with SYTOX Green. A molecular coupling assay was performed to characterize the interaction between riparins and MepA, and ADMET (absorption, distribution, metabolism, and excretion) properties were analyzed. We observed that I-IV riparins did not show direct antibacterial activity against S. aureus. However, combination assays with substrates of MepA, ciprofloxacin, and ethidium bromide (EtBr) revealed a potentiation of the efficacy of these substrates by reducing the minimum inhibitory concentration (MIC). Furthermore, increased EtBr fluorescence emission was observed for all riparins. The checkerboard assay showed synergism between riparins I, II, and III, ciprofloxacin, and EtBr. Furthermore, riparins III and IV exhibited permeability in the S. aureus membrane at a concentration of 200 µg/mL. Molecular docking showed that riparins I, II, and III bound in a different region from the binding site of chlorpromazine (standard pump inhibitor), indicating a possible synergistic effect with the reference inhibitor. In contrast, riparin IV binds in the same region as the chlorpromazine binding site. From the in silico ADMET prediction based on MPO, it could be concluded that the molecules of riparin I-IV present their physicochemical properties within the ideal pharmacological spectrum allowing their preparation as an oral drug. Furthermore, the prediction of cytotoxicity in liver cell lines showed a low cytotoxic effect for riparins I-IV.


Asunto(s)
Clorpromazina , Staphylococcus aureus , Staphylococcus aureus/metabolismo , Simulación del Acoplamiento Molecular , Clorpromazina/metabolismo , Clorpromazina/farmacología , Antibacterianos/química , Ciprofloxacina/farmacología , Etidio , Benzamidas/farmacología , Benzamidas/química , Benzamidas/metabolismo , Proteínas Bacterianas/metabolismo , Pruebas de Sensibilidad Microbiana
6.
Nature ; 546(7657): 312-315, 2017 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-28514449

RESUMEN

The glucagon-like peptide-1 receptor (GLP-1R) and the glucagon receptor (GCGR) are members of the secretin-like class B family of G-protein-coupled receptors (GPCRs) and have opposing physiological roles in insulin release and glucose homeostasis. The treatment of type 2 diabetes requires positive modulation of GLP-1R to inhibit glucagon secretion and stimulate insulin secretion in a glucose-dependent manner. Here we report crystal structures of the human GLP-1R transmembrane domain in complex with two different negative allosteric modulators, PF-06372222 and NNC0640, at 2.7 and 3.0 Å resolution, respectively. The structures reveal a common binding pocket for negative allosteric modulators, present in both GLP-1R and GCGR and located outside helices V-VII near the intracellular half of the receptor. The receptor is in an inactive conformation with compounds that restrict movement of the intracellular tip of helix VI, a movement that is generally associated with activation mechanisms in class A GPCRs. Molecular modelling and mutagenesis studies indicate that agonist positive allosteric modulators target the same general region, but in a distinct sub-pocket at the interface between helices V and VI, which may facilitate the formation of an intracellular binding site that enhances G-protein coupling.


Asunto(s)
Receptor del Péptido 1 Similar al Glucagón/química , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Regulación Alostérica/efectos de los fármacos , Sitio Alostérico/efectos de los fármacos , Secuencia de Aminoácidos , Aminopiridinas/química , Aminopiridinas/metabolismo , Aminopiridinas/farmacología , Benzamidas/química , Benzamidas/metabolismo , Benzamidas/farmacología , Cristalografía por Rayos X , Receptor del Péptido 1 Similar al Glucagón/agonistas , Humanos , Modelos Moleculares , Compuestos de Fenilurea/química , Compuestos de Fenilurea/metabolismo , Compuestos de Fenilurea/farmacología , Dominios Proteicos
7.
Nature ; 546(7657): 259-264, 2017 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-28514451

RESUMEN

The human glucagon receptor, GCGR, belongs to the class B G-protein-coupled receptor family and plays a key role in glucose homeostasis and the pathophysiology of type 2 diabetes. Here we report the 3.0 Å crystal structure of full-length GCGR containing both the extracellular domain and transmembrane domain in an inactive conformation. The two domains are connected by a 12-residue segment termed the stalk, which adopts a ß-strand conformation, instead of forming an α-helix as observed in the previously solved structure of the GCGR transmembrane domain. The first extracellular loop exhibits a ß-hairpin conformation and interacts with the stalk to form a compact ß-sheet structure. Hydrogen-deuterium exchange, disulfide crosslinking and molecular dynamics studies suggest that the stalk and the first extracellular loop have critical roles in modulating peptide ligand binding and receptor activation. These insights into the full-length GCGR structure deepen our understanding of the signalling mechanisms of class B G-protein-coupled receptors.


Asunto(s)
Receptores de Glucagón/química , Receptores de Glucagón/clasificación , Sitio Alostérico/efectos de los fármacos , Benzamidas/química , Benzamidas/metabolismo , Benzamidas/farmacología , Membrana Celular/metabolismo , Reactivos de Enlaces Cruzados/química , Cristalografía por Rayos X , Medición de Intercambio de Deuterio , Disulfuros/química , Humanos , Ligandos , Modelos Moleculares , Simulación de Dinámica Molecular , Compuestos de Fenilurea/química , Compuestos de Fenilurea/metabolismo , Compuestos de Fenilurea/farmacología , Dominios Proteicos , Estabilidad Proteica , Receptores de Glucagón/agonistas , Receptores de Glucagón/metabolismo
8.
Nature ; 547(7663): 364-368, 2017 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-28693035

RESUMEN

Polymodal thermo- and mechanosensitive two-pore domain potassium (K2P) channels of the TREK subfamily generate 'leak' currents that regulate neuronal excitability, respond to lipids, temperature and mechanical stretch, and influence pain, temperature perception and anaesthetic responses. These dimeric voltage-gated ion channel (VGIC) superfamily members have a unique topology comprising two pore-forming regions per subunit. In contrast to other potassium channels, K2P channels use a selectivity filter 'C-type' gate as the principal gating site. Despite recent advances, poor pharmacological profiles of K2P channels limit mechanistic and biological studies. Here we describe a class of small-molecule TREK activators that directly stimulate the C-type gate by acting as molecular wedges that restrict interdomain interface movement behind the selectivity filter. Structures of K2P2.1 (also known as TREK-1) alone and with two selective K2P2.1 (TREK-1) and K2P10.1 (TREK-2) activators-an N-aryl-sulfonamide, ML335, and a thiophene-carboxamide, ML402-define a cryptic binding pocket unlike other ion channel small-molecule binding sites and, together with functional studies, identify a cation-π interaction that controls selectivity. Together, our data reveal a druggable K2P site that stabilizes the C-type gate 'leak mode' and provide direct evidence for K2P selectivity filter gating.


Asunto(s)
Canales de Potasio de Dominio Poro en Tándem/agonistas , Canales de Potasio de Dominio Poro en Tándem/química , Animales , Ácido Araquidónico/química , Ácido Araquidónico/metabolismo , Ácido Araquidónico/farmacología , Benzamidas/química , Benzamidas/metabolismo , Benzamidas/farmacología , Sitios de Unión/efectos de los fármacos , Células HEK293 , Humanos , Activación del Canal Iónico/efectos de los fármacos , Lípidos , Ratones , Modelos Moleculares , Pichia , Canales de Potasio de Dominio Poro en Tándem/metabolismo , Conformación Proteica/efectos de los fármacos , Sulfonamidas/química , Sulfonamidas/metabolismo , Sulfonamidas/farmacología , Tiofenos/química , Tiofenos/metabolismo , Tiofenos/farmacología , Xenopus laevis
9.
Nat Chem Biol ; 16(3): 327-336, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32080624

RESUMEN

The retrograde transport inhibitor Retro-2 has a protective effect on cells and in mice against Shiga-like toxins and ricin. Retro-2 causes toxin accumulation in early endosomes and relocalization of the Golgi SNARE protein syntaxin-5 to the endoplasmic reticulum. The molecular mechanisms by which this is achieved remain unknown. Here, we show that Retro-2 targets the endoplasmic reticulum exit site component Sec16A, affecting anterograde transport of syntaxin-5 from the endoplasmic reticulum to the Golgi. The formation of canonical SNARE complexes involving syntaxin-5 is not affected in Retro-2-treated cells. By contrast, the interaction of syntaxin-5 with a newly discovered binding partner, the retrograde trafficking chaperone GPP130, is abolished, and we show that GPP130 must indeed bind to syntaxin-5 to drive Shiga toxin transport from the endosomes to the Golgi. We therefore identify Sec16A as a druggable target and provide evidence for a non-SNARE function for syntaxin-5 in interaction with GPP130.


Asunto(s)
Benzamidas/metabolismo , Proteínas Qa-SNARE/metabolismo , Tiofenos/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Benzamidas/farmacología , Transporte Biológico , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/metabolismo , Endosomas/metabolismo , Aparato de Golgi/metabolismo , Células HeLa , Humanos , Transporte de Proteínas , Ricina/metabolismo , Toxina Shiga/metabolismo , Toxinas Shiga/metabolismo , Tiofenos/farmacología , Proteínas de Transporte Vesicular/fisiología
10.
Bioorg Med Chem ; 74: 117069, 2022 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-36283251

RESUMEN

C-Abl is involved in various biological processes and plays an important role in neurodegenerative diseases, especially Parkinson's disease (PD). Previous studies have found that nilotinib shows a neuroprotective effect in cell and animal models of PD by inhibiting the activation of c-Abl. But the low blood-brain barrier permeability and potential toxicity limit the further use of nilotinib in PD. Based on molecular modeling studies, a series of 4-methyl-3-(pyridin-2-ylamino)benzamide derivatives were designed and synthesized. In particular, compound 9a exhibited significant inhibitory activity against c-Abl and a potent neuroprotective effect against MPP+-induced SH-SY5Y cell death. Moreover, 9a not only displayed lower cell toxicity compared with nilotinib, but also showed higher oral bioavailability and proper permeability of the blood-brain barrier. This paper provides 4-methyl-3-(pyridin-2-ylamino)benzamide derivatives as a new scaffold for c-Abl inhibitor with potential neuroprotective effect.


Asunto(s)
Neuroblastoma , Fármacos Neuroprotectores , Enfermedad de Parkinson , Animales , Humanos , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/metabolismo , Neuroblastoma/metabolismo , Barrera Hematoencefálica/metabolismo , Enfermedad de Parkinson/metabolismo , Benzamidas/farmacología , Benzamidas/metabolismo , Línea Celular Tumoral
11.
Nature ; 532(7600): 527-30, 2016 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-27042935

RESUMEN

The human σ1 receptor is an enigmatic endoplasmic-reticulum-resident transmembrane protein implicated in a variety of disorders including depression, drug addiction, and neuropathic pain. Recently, an additional connection to amyotrophic lateral sclerosis has emerged from studies of human genetics and mouse models. Unlike many transmembrane receptors that belong to large, extensively studied families such as G-protein-coupled receptors or ligand-gated ion channels, the σ1 receptor is an evolutionary isolate with no discernible similarity to any other human protein. Despite its increasingly clear importance in human physiology and disease, the molecular architecture of the σ1 receptor and its regulation by drug-like compounds remain poorly defined. Here we report crystal structures of the human σ1 receptor in complex with two chemically divergent ligands, PD144418 and 4-IBP. The structures reveal a trimeric architecture with a single transmembrane domain in each protomer. The carboxy-terminal domain of the receptor shows an extensive flat, hydrophobic membrane-proximal surface, suggesting an intimate association with the cytosolic surface of the endoplasmic reticulum membrane in cells. This domain includes a cupin-like ß-barrel with the ligand-binding site buried at its centre. This large, hydrophobic ligand-binding cavity shows remarkable plasticity in ligand recognition, binding the two ligands in similar positions despite dissimilar chemical structures. Taken together, these results reveal the overall architecture, oligomerization state, and molecular basis for ligand recognition by this important but poorly understood protein.


Asunto(s)
Receptores sigma/química , Benzamidas/química , Benzamidas/metabolismo , Sitios de Unión , Cristalografía por Rayos X , Retículo Endoplásmico/metabolismo , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Membranas Intracelulares/metabolismo , Isoxazoles/química , Isoxazoles/metabolismo , Ligandos , Modelos Moleculares , Piperidinas/química , Piperidinas/metabolismo , Estructura Terciaria de Proteína , Piridinas/química , Piridinas/metabolismo , Receptores sigma/metabolismo , Especificidad por Sustrato , Receptor Sigma-1
12.
Biochem J ; 478(15): 2977-2997, 2021 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-34259310

RESUMEN

SBI-0206965, originally identified as an inhibitor of the autophagy initiator kinase ULK1, has recently been reported as a more potent and selective AMP-activated protein kinase (AMPK) inhibitor relative to the widely used, but promiscuous inhibitor Compound C/Dorsomorphin. Here, we studied the effects of SBI-0206965 on AMPK signalling and metabolic readouts in multiple cell types, including hepatocytes, skeletal muscle cells and adipocytes. We observed SBI-0206965 dose dependently attenuated AMPK activator (991)-stimulated ACC phosphorylation and inhibition of lipogenesis in hepatocytes. SBI-0206965 (≥25 µM) modestly inhibited AMPK signalling in C2C12 myotubes, but also inhibited insulin signalling, insulin-mediated/AMPK-independent glucose uptake, and AICA-riboside uptake. We performed an extended screen of SBI-0206965 against a panel of 140 human protein kinases in vitro, which showed SBI-0206965 inhibits several kinases, including members of AMPK-related kinases (NUAK1, MARK3/4), equally or more potently than AMPK or ULK1. This screen, together with molecular modelling, revealed that most SBI-0206965-sensitive kinases contain a large gatekeeper residue with a preference for methionine at this position. We observed that mutation of the gatekeeper methionine to a smaller side chain amino acid (threonine) rendered AMPK and ULK1 resistant to SBI-0206965 inhibition. These results demonstrate that although SBI-0206965 has utility for delineating AMPK or ULK1 signalling and cellular functions, the compound potently inhibits several other kinases and critical cellular functions such as glucose and nucleoside uptake. Our study demonstrates a role for the gatekeeper residue as a determinant of the inhibitor sensitivity and inhibitor-resistant mutant forms could be exploited as potential controls to probe specific cellular effects of SBI-0206965.


Asunto(s)
Proteínas Quinasas Activadas por AMP/antagonistas & inhibidores , Homólogo de la Proteína 1 Relacionada con la Autofagia/antagonistas & inhibidores , Benzamidas/farmacología , Pirimidinas/farmacología , Proteínas Recombinantes/metabolismo , Células 3T3-L1 , Proteínas Quinasas Activadas por AMP/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Animales , Homólogo de la Proteína 1 Relacionada con la Autofagia/genética , Homólogo de la Proteína 1 Relacionada con la Autofagia/metabolismo , Benzamidas/metabolismo , Línea Celular , Línea Celular Tumoral , Células Cultivadas , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Simulación del Acoplamiento Molecular , Mutación Missense , Unión Proteica/efectos de los fármacos , Multimerización de Proteína , Pirimidinas/metabolismo , Ratas Sprague-Dawley , Proteínas Recombinantes/química , Proteínas Recombinantes/genética
13.
Biochem J ; 478(14): 2811-2823, 2021 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-34190988

RESUMEN

The human protein kinase ULK3 regulates the timing of membrane abscission, thus being involved in exosome budding and cytokinesis. Herein, we present the first high-resolution structures of the ULK3 kinase domain. Its unique features are explored against the background of other ULK kinases. An inhibitor fingerprint indicates that ULK3 is highly druggable and capable of adopting a wide range of conformations. In accordance with this, we describe a conformational switch between the active and an inactive ULK3 conformation, controlled by the properties of the attached small-molecule binder. Finally, we discuss a potential substrate-recognition mechanism of the full-length ULK3 protein.


Asunto(s)
Dominio Catalítico , Conformación Proteica , Dominios Proteicos , Proteínas Serina-Treonina Quinasas/química , Compuestos de Anilina/metabolismo , Compuestos de Anilina/farmacología , Benzamidas/metabolismo , Benzamidas/farmacología , Biocatálisis/efectos de los fármacos , Humanos , Modelos Moleculares , Nitrilos/metabolismo , Nitrilos/farmacología , Proteínas Oncogénicas/química , Proteínas Oncogénicas/metabolismo , Unión Proteica , Inhibidores de Proteínas Quinasas/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Pirimidinas/metabolismo , Pirimidinas/farmacología , Quinolinas/metabolismo , Quinolinas/farmacología , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Especificidad por Sustrato
14.
J Basic Microbiol ; 62(6): 711-720, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35417042

RESUMEN

Fluopyram (FLP) containing benzamidic genesis utilized for seed detoxification and as a foliar application is associated with low profound toxicity in mammals but long-term toxicology investigations have revealed that FLP can stimulate tumor growth. FLP attenuation has been the first time scrutinized employing microorganisms originally identified from soils. Biodegrative assays of four fungal strains; Aspergillus fumigatus (AFu), Aspergillus terreus (AT), Aspergillus flavus (AF), Aspergillus niger (AN), and three bacterial strains: Streptococcus pneumoniae (SP) Streptococcus pyogenes (SPy), and Escherichia coli (EC), were employed. Ten milligrams per liter FLP concentration was made employing separately microbe and analyzed for 35 days. The analytical technique was inclusive of ultraviolet-visible spectrophotometric and high-performance liquid chromatography procedure endeavored to test FLP biodegradation. SP and AT exhibited maximal potentiality to metabolize FLP. HPLC is indicative of several metabolites formations. FLP degradation by AFu, EC, SPy, AN, AF, AT, SP was observed to be 24.2%, 82.7%, 89.8%, 90.7%, 91.3%, 95.4%, and 99.3%, explicating the efficacy of all strains employed in FLP degradation. Current investigations are indicative of significant bioremediation strategies for xenobiotic mitigation. Furthermore, the current examinations are inclusive of the augmentation of biodegradative assays to be utilized on a large scale for efficient environmental management cost-effectively and sustainably.


Asunto(s)
Bacterias , Benzamidas , Animales , Aspergillus , Aspergillus flavus , Aspergillus niger/metabolismo , Benzamidas/metabolismo , Biodegradación Ambiental , Mamíferos
15.
Proteins ; 89(11): 1399-1412, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34156100

RESUMEN

The Receptor for Advanced Glycation End products (RAGE) is a pattern recognition receptor that signals for inflammation via the NF-κB pathway. RAGE has been pursued as a potential target to suppress symptoms of diabetes and is of interest in a number of other diseases associated with chronic inflammation, such as inflammatory bowel disease and bronchopulmonary dysplasia. Screening and optimization have previously produced small molecules that inhibit the activity of RAGE in cell-based assays, but efforts to develop a therapeutically viable direct-binding RAGE inhibitor have yet to be successful. Here, we show that a fragment-based approach can be applied to discover fundamentally new types of RAGE inhibitors that specifically target the ligand-binding surface. A series of systematic assays of structural stability, solubility, and crystallization were performed to select constructs of the RAGE ligand-binding domain and optimize conditions for NMR-based screening and co-crystallization of RAGE with hit fragments. An NMR-based screen of a highly curated ~14 000-member fragment library produced 21 fragment leads. Of these, three were selected for elaboration based on structure-activity relationships generated through cycles of structural analysis by X-ray crystallography, structure-guided design principles, and synthetic chemistry. These results, combined with crystal structures of the first linked fragment compounds, demonstrate the applicability of the fragment-based approach to the discovery of RAGE inhibitors.


Asunto(s)
Benzamidas/química , Diseño de Fármacos/métodos , Imidazoles/química , Receptor para Productos Finales de Glicación Avanzada/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas/química , Benzamidas/metabolismo , Benzamidas/farmacología , Sitios de Unión , Clonación Molecular , Cristalografía por Rayos X , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Humanos , Imidazoles/metabolismo , Imidazoles/farmacología , Ligandos , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Resonancia Magnética Nuclear Biomolecular , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Receptor para Productos Finales de Glicación Avanzada/química , Receptor para Productos Finales de Glicación Avanzada/genética , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Bibliotecas de Moléculas Pequeñas/metabolismo , Bibliotecas de Moléculas Pequeñas/farmacología , Relación Estructura-Actividad
16.
Plant Cell Physiol ; 61(12): 2139-2152, 2021 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-33104193

RESUMEN

Endosidin20 (ES20) was recently identified as a cellulose biosynthesis inhibitor (CBI) that targets the catalytic domain of CELLULOSE SYNTHASE 6 (CESA6) and thus inhibits the growth of Arabidopsis thaliana. Here, we characterized the effects of ES20 on the growth of other plant species and found that ES20 is a broad-spectrum plant growth inhibitor. We tested the inhibitory effects of previously characterized CBIs (isoxaben, indaziflam and C17) on the growth of Arabidopsis cesa6 mutants that have reduced sensitivity to ES20. We found that most of these mutants are sensitive to isoxaben, indaziflam and C17, indicating that these tested CBIs have a different mode of action than ES20. ES20 also has a synergistic inhibitory effect on plant growth when jointly applied with other CBIs, further confirming that ES20 has a different mode of action than isoxaben, indaziflam and C17. We demonstrated that plants carrying two missense mutations conferring resistance to ES20 and isoxaben can tolerate the dual inhibitory effects of these CBIs when combined. ES20 inhibits Arabidopsis growth in growth medium and in soil following direct spraying. Therefore, our results pave the way for using ES20 as a broad-spectrum herbicide, and for the use of gene-editing technologies to produce ES20-resistant crop plants.


Asunto(s)
Proteínas de Arabidopsis/metabolismo , Celulosa/biosíntesis , Arabidopsis/crecimiento & desarrollo , Arabidopsis/metabolismo , Arabidopsis/fisiología , Proteínas de Arabidopsis/fisiología , Benzamidas/metabolismo , Glucosiltransferasas/metabolismo
17.
Biochem Biophys Res Commun ; 534: 995-1002, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-33168189

RESUMEN

Esophageal squamous cell carcinoma (ESCC) is a common type of human oral malignancy with poor survival. Presently, it is necessary to find new and effective drugs for clinical therapy. This study aimed to identify the potential anti-tumor effects of ACP-5862, a major metabolite of acalabrutinib, on human ESCC progression, and to reveal the underlying mechanisms. Our findings suggested that ACP-5862 treatments markedly reduced the cell proliferation of ESCC cell lines in a time- and dose-dependent manner, while had no significant cytotoxicity to normal cells. Cell cycle arrest in G2/M phase was markedly induced by ACP-5862 in ESCC cells. Furthermore, apoptosis and endoplasmic reticulum (ER) stress were detected in ESCC cells treated with ACP-5862. Intriguingly, ACP-5862-induced apoptotic cell death was partly dependent on ER stress. Moreover, reactive oxygen species (ROS) was greatly triggered in ACP-5862-incubated ESCC cells, which was closely involved in apoptosis and ER stress mediated by ACP-5862. In addition, we showed that the expression of nuclear factor-erythroid 2-related factor-2 (Nrf-2) was considerably reduced in ACP-5862-treated cells. Importantly, ACP-5862 combined with Nrf-2 knockdown could further induce apoptosis and ER stress in ESCC cells compared with the ACP-5862 single group. Animal studies confirmed that repressing Nrf-2 promoted the anti-tumor effect of ACP-5862 on ESCC growth. Taken together, these findings demonstrated that ACP-5862 exerted anti-cancer effects on ESCC through inducing ER stress-mediated apoptosis via the ROS production. Meanwhile, ACP-5862 co-treated with Nrf-2 inhibitors may supply new and effective therapeutic strategies for ESCC treatment in future.


Asunto(s)
Antineoplásicos/uso terapéutico , Benzamidas/uso terapéutico , Estrés del Retículo Endoplásmico/efectos de los fármacos , Neoplasias Esofágicas/tratamiento farmacológico , Carcinoma de Células Escamosas de Esófago/tratamiento farmacológico , Pirazinas/uso terapéutico , Animales , Antineoplásicos/metabolismo , Apoptosis/efectos de los fármacos , Benzamidas/metabolismo , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/patología , Carcinoma de Células Escamosas de Esófago/metabolismo , Carcinoma de Células Escamosas de Esófago/patología , Humanos , Masculino , Ratones Endogámicos BALB C , Pirazinas/metabolismo , Especies Reactivas de Oxígeno/metabolismo
18.
Int J Mol Sci ; 22(24)2021 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-34948390

RESUMEN

Since the start of the COVID-19 outbreak, pharmaceutical companies and research groups have focused on the development of vaccines and antiviral drugs against SARS-CoV-2. Here, we apply a drug repurposing strategy to identify drug candidates that are able to block the entrance of the virus into human cells. By combining virtual screening with in vitro pseudovirus assays and antiviral assays in Human Lung Tissue (HLT) cells, we identify entrectinib as a potential antiviral drug.


Asunto(s)
Benzamidas/farmacología , Tratamiento Farmacológico de COVID-19 , Indazoles/farmacología , SARS-CoV-2/efectos de los fármacos , Animales , Antivirales/farmacología , Benzamidas/metabolismo , COVID-19/metabolismo , Línea Celular , Chlorocebus aethiops , Evaluación Preclínica de Medicamentos , Reposicionamiento de Medicamentos/métodos , Humanos , Indazoles/metabolismo , Pulmón/patología , Pulmón/virología , Simulación del Acoplamiento Molecular , SARS-CoV-2/metabolismo , SARS-CoV-2/patogenicidad , Células Vero , Acoplamiento Viral/efectos de los fármacos
19.
J Integr Plant Biol ; 63(2): 340-352, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32678930

RESUMEN

Fusarium head blight (FHB) caused by Fusarium graminearum Schwabe (teleomorph Gibberella zeae (Schw.) Perch) results in large yield losses in annual global wheat production. Although studies have identified a number of wheat FHB resistance genes, a deeper understanding of the mechanisms underlying host plant resistance to F. graminearum is required for the control of FHB. Here, an integrated metabolomics and transcriptomics analysis of infected wheat plants (Triticum aestivum L.) enabled identification of 789 differentially accumulated metabolites, including flavonoids, phenolamides, tryptamine derivatives, and phytohormones, and revealed altered expression of more than 100 genes that function in the biosynthesis or regulation of these pathways. Our data regarding the effects of F. graminearum infection on flavonoids and auxin signaling led to follow-up experiments that showed that exogenous kaempferide and apigenin application on spikes increased wheat resistance to FHB, while exogenous auxin treatment increased FHB susceptibility. RNAi-mediated knockdown of the gene encoding the auxin receptor, TaTIR1, increased FHB resistance. Our data supported the use of TaTIR1 knockdown in controlling FHB. Our study provides insights on the wheat response to F. graminearum infection and its FHB resistance mechanisms while illustrating the potential of TaTIR1 knockdown in increasing FHB resistance during crop improvement programs.


Asunto(s)
Fusarium/fisiología , Ácidos Indolacéticos/metabolismo , Metabolómica , Proteínas de Plantas/genética , Proteínas de Plantas/metabolismo , Transcriptoma/genética , Triticum/inmunología , Triticum/microbiología , Benzamidas/metabolismo , Vías Biosintéticas/genética , Resistencia a la Enfermedad/genética , Flavonoides/metabolismo , Silenciador del Gen/efectos de los fármacos , Enfermedades de las Plantas/inmunología , Enfermedades de las Plantas/microbiología , Reguladores del Crecimiento de las Plantas/metabolismo , Reproducibilidad de los Resultados , Transducción de Señal , Triticum/genética , Triticum/metabolismo , Triptaminas/metabolismo
20.
Psychol Med ; 50(5): 799-807, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-30935427

RESUMEN

BACKGROUND: The 22q11.2 deletion syndrome (22q11DS) is caused by a deletion on chromosome 22 locus q11.2. This copy number variant results in haplo-insufficiency of the catechol-O-methyltransferase (COMT) gene, and is associated with a significant increase in the risk for developing cognitive impairments and psychosis. The COMT gene encodes an enzyme that primarily modulates clearance of dopamine (DA) from the synaptic cleft, especially in the prefrontal cortical areas. Consequently, extracellular DA levels may be increased in prefrontal brain areas in 22q11DS, which may underlie the well-documented susceptibility for cognitive impairments and psychosis in affected individuals. This study aims to examine DA D2/3 receptor binding in frontal brain regions in adults with 22q11DS, as a proxy of frontal DA levels. METHODS: The study was performed in 14 non-psychotic, relatively high functioning adults with 22q11DS and 16 age- and gender-matched healthy controls (HCs), who underwent DA D2/3 receptor [18F]fallypride PET imaging. Frontal binding potential (BPND) was used as the main outcome measure. RESULTS: BPND was significantly lower in adults with 22q11DS compared with HCs in the prefrontal cortex and the anterior cingulate gyrus. After Bonferroni correction significance remained for the anterior cingulate gyrus. There were no between-group differences in BPND in the orbitofrontal cortex and anterior cingulate cortex. CONCLUSIONS: This study is the first to demonstrate lower frontal D2/3 receptor binding in adults with 22q11DS. It suggests that a 22q11.2 deletion affects frontal dopaminergic neurotransmission.


Asunto(s)
Benzamidas/metabolismo , Síndrome de DiGeorge/diagnóstico por imagen , Tomografía de Emisión de Positrones , Corteza Prefrontal/diagnóstico por imagen , Receptores de Dopamina D2/metabolismo , Adulto , Mapeo Encefálico , Catecol O-Metiltransferasa/genética , Síndrome de DiGeorge/metabolismo , Femenino , Humanos , Masculino , Persona de Mediana Edad , Corteza Prefrontal/metabolismo , Trastornos Psicóticos/complicaciones , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA