Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
J Chem Inf Model ; 64(8): 3548-3557, 2024 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-38587997

RESUMEN

Protein-DNA interactions are pivotal to various cellular processes. Precise identification of the hotspot residues for protein-DNA interactions holds great significance for revealing the intricate mechanisms in protein-DNA recognition and for providing essential guidance for protein engineering. Aiming at protein-DNA interaction hotspots, this work introduces an effective prediction method, ESPDHot based on a stacked ensemble machine learning framework. Here, the interface residue whose mutation leads to a binding free energy change (ΔΔG) exceeding 2 kcal/mol is defined as a hotspot. To tackle the imbalanced data set issue, the adaptive synthetic sampling (ADASYN), an oversampling technique, is adopted to synthetically generate new minority samples, thereby rectifying data imbalance. As for molecular characteristics, besides traditional features, we introduce three new characteristic types including residue interface preference proposed by us, residue fluctuation dynamics characteristics, and coevolutionary features. Combining the Boruta method with our previously developed Random Grouping strategy, we obtained an optimal set of features. Finally, a stacking classifier is constructed to output prediction results, which integrates three classical predictors, Support Vector Machine (SVM), XGBoost, and Artificial Neural Network (ANN) as the first layer, and Logistic Regression (LR) algorithm as the second one. Notably, ESPDHot outperforms the current state-of-the-art predictors, achieving superior performance on the independent test data set, with F1, MCC, and AUC reaching 0.571, 0.516, and 0.870, respectively.


Asunto(s)
ADN , Aprendizaje Automático , ADN/química , ADN/metabolismo , Unión Proteica , Redes Neurales de la Computación , Proteínas/química , Proteínas/metabolismo , Termodinámica , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/química , Máquina de Vectores de Soporte , Algoritmos
2.
J Biol Chem ; 295(13): 4079-4092, 2020 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-32019865

RESUMEN

Organophosphate (OP) intoxications from nerve agent and OP pesticide exposures are managed with pyridinium aldoxime-based therapies whose success rates are currently limited. The pyridinium cation hampers uptake of OPs into the central nervous system (CNS). Furthermore, it frequently binds to aromatic residues of OP-inhibited acetylcholinesterase (AChE) in orientations that are nonproductive for AChE reactivation, and the structural diversity of OPs impedes efficient reactivation. Improvements of OP antidotes need to include much better access of AChE reactivators to the CNS and optimized orientation of the antidotes' nucleophile within the AChE active-center gorge. On the basis of X-ray structures of a CNS-penetrating reactivator, monoxime RS194B, reversibly bound to native and venomous agent X (VX)-inhibited human AChE, here we created seven uncharged acetamido bis-oximes as candidate antidotes. Both oxime groups in these bis-oximes were attached to the same central, saturated heterocyclic core. Diverse protonation of the heterocyclic amines and oxime groups of the bis-oximes resulted in equilibration among up to 16 distinct ionization forms, including uncharged forms capable of diffusing into the CNS and multiple zwitterionic forms optimal for reactivation reactions. Conformationally diverse zwitterions that could act as structural antidote variants significantly improved in vitro reactivation of diverse OP-human AChE conjugates. Oxime group reorientation of one of the bis-oximes, forcing it to point into the active center for reactivation, was confirmed by X-ray structural analysis. Our findings provide detailed structure-activity properties of several CNS-directed, uncharged aliphatic bis-oximes holding promise for use as protonation-dependent, conformationally adaptive, "smart" accelerated antidotes against OP toxicity.


Asunto(s)
Acetilcolinesterasa/química , Antídotos/química , Sistema Nervioso Central/efectos de los fármacos , Inhibidores de la Colinesterasa/química , Reactivadores de la Colinesterasa/química , Acetamidas/química , Acetamidas/uso terapéutico , Antídotos/síntesis química , Antídotos/uso terapéutico , Sistema Nervioso Central/enzimología , Inhibidores de la Colinesterasa/síntesis química , Inhibidores de la Colinesterasa/uso terapéutico , Reactivadores de la Colinesterasa/síntesis química , Reactivadores de la Colinesterasa/uso terapéutico , Cristalografía por Rayos X , Humanos , Cinética , Organofosfatos/química , Organofosfatos/toxicidad , Compuestos Organofosforados/química , Compuestos Organofosforados/toxicidad , Oximas/síntesis química , Oximas/química , Oximas/farmacología , Oximas/uso terapéutico , Conformación Proteica/efectos de los fármacos , Relación Estructura-Actividad
3.
J Chem Inf Model ; 61(7): 3477-3494, 2021 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-34165949

RESUMEN

An androgen receptor (AR) is an intensively studied treatment target for castration-resistant prostate cancer that is irresponsive to conventional antiandrogen therapeutics. Binding function 3 (BF3) inhibitors with alternative modes of action have emerged as a promising approach to overcoming antiandrogen resistance. However, how these BF3 inhibitors modulate AR function remains elusive, hindering the development of BF3-targeting agents. Here, we performed an integrated computational study to interrogate the binding mechanism of several known BF3 inhibitors with ARs. Our results show that the inhibitory effect of the BF3 antagonists arises from their allosteric modulation of the activation function (AF2) site, which alters the dynamic coupling between the BF3 and AF2 sites as well as the AF2-coactivator (SRC2-3) interaction. Moreover, the per-residue binding energy analyses reveal the "anchor" role of the linker connecting the phenyl ring and benzimidazole/indole in these BF3 inhibitors. Furthermore, the allosteric driver-interacting residues are found to include both "positive", e.g., Phe673 and Asn833, and "negative" ones, e.g., Phe826, and the differential interactions with these residues provide an explanation why stronger binding does not necessarily result in higher inhibitory activities. Finally, our allosteric communication pathway analyses delineate how the allosteric signals triggered by BF3 binding are propagated to the AF2 pocket through multiple short- and/or long-ranged transmission pathways. Collectively, our combined computational study provides a comprehensive structural mechanism underlying how the selected set of BF3 inhibitors modulate AR function, which will help guide future development of BF3 antagonists.


Asunto(s)
Neoplasias de la Próstata , Receptores Androgénicos , Antagonistas de Andrógenos , Antagonistas de Receptores Androgénicos/farmacología , Sitios de Unión , Humanos , Masculino , Modelos Moleculares
4.
J Biol Chem ; 294(27): 10607-10618, 2019 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-31138650

RESUMEN

Exposure to organophosphorus compounds (OPs) may be fatal if untreated, and a clear and present danger posed by nerve agent OPs has become palpable in recent years. OPs inactivate acetylcholinesterase (AChE) by covalently modifying its catalytic serine. Inhibited AChE cannot hydrolyze the neurotransmitter acetylcholine leading to its build-up at the cholinergic synapses and creating an acute cholinergic crisis. Current antidotes, including oxime reactivators that attack the OP-AChE conjugate to free the active enzyme, are inefficient. Better reactivators are sought, but their design is hampered by a conformationally rigid portrait of AChE extracted exclusively from 100K X-ray crystallography and scarcity of structural knowledge on human AChE (hAChE). Here, we present room temperature X-ray structures of native and VX-phosphonylated hAChE with an imidazole-based oxime reactivator, RS-170B. We discovered that inhibition with VX triggers substantial conformational changes in bound RS-170B from a "nonproductive" pose (the reactive aldoxime group points away from the VX-bound serine) in the reactivator-only complex to a "semi-productive" orientation in the VX-modified complex. This observation, supported by concurrent molecular simulations, suggested that the narrow active-site gorge of hAChE may be significantly more dynamic than previously thought, allowing RS-170B to reorient inside the gorge. Furthermore, we found that small molecules can bind in the choline-binding site hindering approach to the phosphorous of VX-bound serine. Our results provide structural and mechanistic perspectives on the reactivation of OP-inhibited hAChE and demonstrate that structural studies at physiologically relevant temperatures can deliver previously overlooked insights applicable for designing next-generation antidotes.


Asunto(s)
Acetilcolinesterasa/química , Compuestos Organotiofosforados/química , Oximas/química , Acetilcolinesterasa/genética , Acetilcolinesterasa/metabolismo , Sitios de Unión , Dominio Catalítico , Inhibidores de la Colinesterasa/química , Inhibidores de la Colinesterasa/metabolismo , Cristalografía por Rayos X , Teoría Funcional de la Densidad , Humanos , Simulación de Dinámica Molecular , Compuestos Organotiofosforados/metabolismo , Oximas/metabolismo , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Temperatura
5.
J Chem Inf Model ; 59(2): 842-857, 2019 02 25.
Artículo en Inglés | MEDLINE | ID: mdl-30658039

RESUMEN

Androgen receptor (AR), as a member of the nuclear receptor (NR) superfamily, regulates the gene transcription in response to the sequential binding of diverse agonists and coactivators. Great progress has been made in studies on the pharmacology and structure of AR, but the atomic level mechanism of the bidirectional communications between the ligand-binding pocket (LBP) and the activation function-2 (AF2) region of AR remains poorly understood. Therefore, in this study, molecular dynamics (MD) simulations and free energy calculations were carried out to explore the interactions among water, agonist (DHT) or antagonist (HFT), AR, and coactivator (SRC3). Upon the binding of an agonist (DHT) or antagonist (HFT), the LBP structure would transform to the agonistic or antagonistic state, and the conformational changes of the LBP would regulate the structure of the AF2 surface. As a result, the binding of the androgen DHT could promote the recruitment of the coactivator SRC3 to the AF2, and on the contrary, the binding of the antagonist HFT would induce a perturbation to the shape of the AF2 and then weaken its accommodating capability of the coactivators with the LXXLL motif. The simulation results illustrated that the DHT-AR binding affinity was enhanced by the association of the coactivator SRC3, which would reduce the conformational fluctuation of the AR-LBD and expand the size of the AR LBP. On the other hand, the coactivator-to-HFT allosteric pathway, which involves the SRC3, helix 3 (H3), helix 4 (H4), the loop (L1-3) between helix 1 (H1) and H3, and HFT, was characterized. The HFT's skewness and different interactions between HFT and the LBP were observed in the SRC3-present AR. The mutual communications between the AF2 surface and LBP, together with the processes involving the interplay of the ligand binding and coactivator recruitment events, would help in understanding the association of coactivators and rationally develop potent drugs to inhibit the activity of AR.


Asunto(s)
Simulación de Dinámica Molecular , Receptores Androgénicos/química , Receptores Androgénicos/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Sitios de Unión , Ligandos , Coactivador 3 de Receptor Nuclear/metabolismo , Unión Proteica , Termodinámica
6.
Bioorg Med Chem ; 26(4): 875-883, 2018 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29395803

RESUMEN

A series of novel pyraclostrobin derivatives were designed and prepared as antifungal agents. Their antifungal activities were tested in vitro with five important phytopathogenic fungi, namely, Batrylis cinerea, Phytophthora capsici, Fusarium sulphureum, Gloeosporium pestis and Sclerotinia sclerotiorum using the mycelium growth inhibition method. Among these compounds, 5s displayed IC50 value of 0.57 µg/mL against Batrylis cinerea and 5k-II displayed IC50 value of 0.43 µg/mL against Sclerotinia sclerotiorum, which were close to that of the positive control pyraclostrobin (0.18 µg/mL and 0.15 µg/mL). Other compounds 5f, 5k-II, 5j, 5m and 5s also exhibited strong antifungal activity. Further enzymatic assay demonstrated compound 5s inhibited porcine bc1 complex with IC50 value of 0.95 µM. The statistical results from an integrated computational pipeline demonstrated the predicted total binding free energy for compound 5s is the highest. Consequently, compound 5s with the biphenyl-4-methoxyl side chain could serve as a new motif as inhibitors of bc1 complex and deserve to be further investigated.


Asunto(s)
Antifúngicos/síntesis química , Diseño de Fármacos , Estrobilurinas/química , Antifúngicos/química , Antifúngicos/farmacología , Sitios de Unión , Complejo III de Transporte de Electrones/química , Complejo III de Transporte de Electrones/metabolismo , Hongos/efectos de los fármacos , Fusarium/efectos de los fármacos , Pruebas de Sensibilidad Microbiana , Simulación del Acoplamiento Molecular , Estructura Terciaria de Proteína , Estrobilurinas/síntesis química , Estrobilurinas/farmacología , Relación Estructura-Actividad , Termodinámica
7.
Phys Chem Chem Phys ; 20(7): 4851-4863, 2018 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-29383359

RESUMEN

Anaplastic lymphoma kinase (ALK) has been regarded as a promising target for the therapy of various cancers. A large number of ALK inhibitors with diverse scaffolds have been discovered, and most of them belong to Type-I inhibitors that only occupy the ATP-binding pocket. Recently, we reported a series of novel and potent Type-I1/2 inhibitors of ALK with the 1-purine-3-piperidinecarboxamide scaffold, which can bind to both the ATP-binding site of ALK and the adjacent hydrophobic allosteric pocket. In this study, the binding mechanisms of these Type-I1/2 ALK inhibitors were elucidated by multiple molecular modeling techniques. The calculation results demonstrate that the ensemble docking based on multiple protein structures and the MM/PB(GB)SA calculations based on molecular dynamics (MD) simulations yield better predictions than conventional rigid receptor docking (Glide, Surflex-Dock, and Autodock Vina), highlighting the importance of incorporating receptor flexibility in the predictions of binding poses and binding affinities of Type-I1/2 ALK inhibitors. Furthermore, the umbrella sampling (US) simulations and MM/GBSA binding free energy decomposition analyses indicate that Leu1122, Leu1198, Gly1202 and Glu1210 in the hinge region and Glu1197, Ile1171, Phe1174, Ile1179, His1247, Ile1268, Asp1270 and Phe1271 in the allosteric pocket of ALK are the key residues for determining the relative binding strength of the studied inhibitors. Besides, we found that the most potent inhibitor (001-017) tends to form stronger transient interactions with residues along the dissociation channel due to the high electronegativity of its bulky 4-(trifluoromethoxy) phenylamine tail. As a whole, both the stronger binding affinity and the higher energetic barrier (which may prolong the drug-target residence time) of 001-017 contribute to its excellent anti-proliferation activity against ALK-positive cancer cells.


Asunto(s)
Simulación de Dinámica Molecular , Inhibidores de Proteínas Quinasas/química , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Sitio Alostérico , Secuencia de Aminoácidos , Quinasa de Linfoma Anaplásico , Antineoplásicos/química , Sitios de Unión , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Simulación del Acoplamiento Molecular , Unión Proteica , Conformación Proteica , Termodinámica
8.
Environ Sci Technol ; 51(11): 6452-6460, 2017 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-28466639

RESUMEN

Parabens have been widely used in packaged foods, pharmaceuticals, and personal-care products. Considering their potential hydrolysis, we herein investigated structural features leading to the disruption of human androgen receptor (AR) and whether hydrolysis could alleviate such effects using the recombinant yeast two-hybrid assay. Parabens with an aryloxy side chain such as benzyl paraben and phenyl paraben have the strongest antiandrogenic activity. The antiandrogenic activity of parabens with alkyloxyl side chains decreases as the side chain length increases from 1 to 4, and no antiandrogenic effect occurred for heptyl, octyl, and dodecyl parabens with the number of alkoxyl carbon atoms longer than 7. The antiandrogenic activity of parabens correlates significantly with their binding energies (R2 = 0.84, p = 0.01) and were completely diminished after the hydrolysis, particularly for parabens with aryloxy side chains. The Km for the hydrolysis of parabens with aromatic moiety side chain is 1 order of magnitude higher than that of the parabens with alkyl side chains. Both in vitro and in silico data, for the first time, suggest parabens with aromatic side chains are less prone to hydrolysis. Our results provide an insight into risk of various paraben and considerations for design of new paraben-related substitutes.


Asunto(s)
Antagonistas de Andrógenos , Simulación del Acoplamiento Molecular , Parabenos/química , Humanos , Hidrólisis
10.
Phys Chem Chem Phys ; 18(3): 2034-46, 2016 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-26686753

RESUMEN

Due to the high sequence identity of the binding pockets of cyclin-dependent kinases (CDKs), designing highly selective inhibitors towards a specific CDK member remains a big challenge. 4-(thiazol-5-yl)-2-(phenylamino) pyrimidine derivatives are effective inhibitors of CDKs, among which the most promising inhibitor 12u demonstrates high binding affinity to CDK9 and attenuated binding affinity to other homologous kinases, such as CDK2. In this study, in order to rationalize the principle of the binding preference towards CDK9 over CDK2 and to explore crucial information that may aid the design of selective CDK9 inhibitors, MM/GBSA calculations based on conventional molecular dynamics (MD) simulations and enhanced sampling simulations (umbrella sampling and steered MD simulations) were carried out on two representative derivatives (12u and 4). The calculation results show that the binding specificity of 12u to CDK9 is primarily controlled by conformational change of the G-loop and variation of the van der Waals interactions. Furthermore, the enhanced sampling simulations revealed the different reaction coordinates and transient interactions of inhibitors 12u and 4 as they dissociate from the binding pockets of CDK9 and CDK2. The physical principles obtained from this study may facilitate the discovery and rational design of novel and specific inhibitors of CDK9.


Asunto(s)
Quinasas Ciclina-Dependientes/metabolismo , Nitrilos/metabolismo , Simulación de Dinámica Molecular
11.
J Chem Inf Model ; 55(12): 2693-704, 2015 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-26618892

RESUMEN

Angiopoietin (ANG) ligands and their downstream TIE receptors have been validated as the second vascular signaling system involving vessel remodeling and maturation. Among them, the ANG/TIE-2 signaling pathway is involved in numerous life-threatening diseases and has become an attractive potential therapeutic target. Several large-molecule inhibitors targeting the ANG/TIE-2 axis have recently entered clinical phase for the therapy of various solid tumors, but selective small-molecule inhibitors of TIE-2 are still quite limited. In the present work, structure-based virtual screening was performed to search for type-I inhibitors of TIE-2. Of the only 41 compounds selected by our strategy, 8 molecules with the concentration of 25 µg/mL exhibit over 50% inhibitory rate against TIE-2 in in vitro enzymatic activity assay, and the IC50 values of 2 hits are lower than 1 µM. Further optimization and SAR analysis based on compound TP-S1-30 and 31 were carried out by using substructure searching strategy, leading to the discovery of several sub-100 nM inhibitors. Among them, the most potent compound, TP-S1-68, showed an inhibitory IC50 of 0.149 µM. These novel inhibitors of TIE-2 discovered in this study and the analogs of the active core scaffolds can serve as the starting points for further drug development.


Asunto(s)
Diseño de Fármacos , Simulación del Acoplamiento Molecular , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Receptor TIE-2/antagonistas & inhibidores , Bioensayo , Cristalografía por Rayos X , Relación Dosis-Respuesta a Droga , Activación Enzimática/efectos de los fármacos , Concentración 50 Inhibidora , Receptor TIE-2/química , Receptor TIE-2/metabolismo , Relación Estructura-Actividad
12.
Phys Chem Chem Phys ; 17(8): 6098-113, 2015 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-25644934

RESUMEN

Anaplastic lymphoma kinase (ALK) has gained increased attention as an attractive therapeutic target for the treatment of various cancers, especially non-small-cell lung cancer (NSCLC). Recently, piperidine carboxamides were reported as Type I1/2 inhibitors of ALK, which occupy both the ATP binding site and the back ATP hydrophobic cavity in DFG-in conformation. Due to the dynamic behavior of ALK in the binding of Type I1/2 inhibitors, the accurate predictions of the binding structures and relative binding potencies of these inhibitors are quite challenging. In this study, different modeling techniques, including molecular docking, ensemble docking based on multiple receptor conformations, molecular dynamics simulations and free energy calculations, were utilized to explore the binding mechanisms of piperidine carboxamides. Our predictions show that the conventional docking protocols are not sufficient to predict the relative binding potencies of the studied inhibitors with high accuracy, but incorporating protein flexibility before or after docking is quite effective to improve the prediction accuracy. Notably, the binding free energies predicted by MM/GBSA or MM/PBSA based on the MD simulations for the docked poses give the highest correlation with the experimental data, highlighting the importance of the inclusion of receptor flexibility for the accurate predictions of the binding potencies for Type I1/2 inhibitors of ALK. Furthermore, the comprehensive analysis of several pairs of representative inhibitors demonstrates the importance of hydrophobic interactions in improving the binding affinities of the inhibitors with the hot-spot residues surrounding the binding pocket. This work is expected to provide valuable clues for further rational design of novel and potent Type I1/2 ALK inhibitors.


Asunto(s)
Amidas/química , Piperidinas/química , Inhibidores de Proteínas Quinasas/química , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Amidas/metabolismo , Quinasa de Linfoma Anaplásico , Sitios de Unión , Entropía , Interacciones Hidrofóbicas e Hidrofílicas , Simulación del Acoplamiento Molecular , Unión Proteica , Inhibidores de Proteínas Quinasas/metabolismo , Estructura Terciaria de Proteína , Proteínas Tirosina Quinasas Receptoras/metabolismo , Relación Estructura-Actividad
13.
Chem Biol Drug Des ; 104(2): e14607, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39179521

RESUMEN

The process of developing new drugs is widely acknowledged as being time-intensive and requiring substantial financial investment. Despite ongoing efforts to reduce time and expenses in drug development, ensuring medication safety remains an urgent problem. One of the major problems involved in drug development is hepatotoxicity, specifically known as drug-induced liver injury (DILI). The popularity of new drugs often poses a significant barrier during development and frequently leads to their recall after launch. In silico methods have many advantages compared with traditional in vivo and in vitro assays. To establish a more precise and reliable prediction model, it is necessary to utilize an extensive and high-quality database consisting of information on drug molecule properties and structural patterns. In addition, we should also carefully select appropriate molecular descriptors that can be used to accurately depict compound characteristics. The aim of this study was to conduct a comprehensive investigation into the prediction of DILI. First, we conducted a comparative analysis of the physicochemical properties of extensively well-prepared DILI-positive and DILI-negative compounds. Then, we used classic substructure dissection methods to identify structural pattern differences between these two different types of chemical molecules. These findings indicate that it is not feasible to establish property or substructure-based rules for distinguishing between DILI-positive and DILI-negative compounds. Finally, we developed quantitative classification models for predicting DILI using the naïve Bayes classifier (NBC) and recursive partitioning (RP) machine learning techniques. The optimal DILI prediction model was obtained using NBC, which combines 21 physicochemical properties, the VolSurf descriptors and the LCFP_10 fingerprint set. This model achieved a global accuracy (GA) of 0.855 and an area under the curve (AUC) of 0.704 for the training set, while the corresponding values were 0.619 and 0.674 for the test set, respectively. Moreover, indicative substructural fragments favorable or unfavorable for DILI were identified from the best naïve Bayesian classification model. These findings may help prioritize lead compounds in the early stage of drug development pipelines.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas , Aprendizaje Automático , Humanos , Preparaciones Farmacéuticas/química , Teorema de Bayes , Simulación por Computador
14.
Heliyon ; 10(14): e34846, 2024 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-39148993

RESUMEN

F. taipaiensis P. Y. Li represents a significant asset within traditional Chinese medicinal flora, though it confronts the challenge of germplasm deterioration during its cultivation phase. This study aimed to discern the implications of single strains or combinations of diverse growth-promoting actinomycetes on the growth metrics, antioxidant competence and pertinent gene expression in the leaves of F. taipaiensis. The result revealed that the malondialdehyde content within the plant's leaves notably diminished in the treatment groups compared to the CK group, with the S6 group showcasing the most pronounced malondialdehyde reduction, amounting to approximately one-third of the CK's value. Leaf area, length and width peaked in the S5 cohort, registering values 4.55, 2.46 and 1.85 times surpassing the CK group. Concurrently, plant height and stem thickness were maximal in the S6 group, being 2.29 and 1.75 times that of the CK group, whereas leaf thickness reached its zenith in the S7 group, marking a 2.17-fold elevation compared to the CK. Photosynthetic pigments, soluble sugars and soluble proteins in the leaves, exhibited augmentation across the inoculated groups to varying magnitudes. Specifically, the S5 group was superior in photosynthetic metrics and pigments, while the S6 group manifested the highest soluble sugar concentration, which was 1.35 times that of the CK. The S3 group demonstrated the pinnacle of soluble protein content, an impressive 5.86-fold increment relative to the CK group. The enzymatic activities of superoxide dismutase, peroxidase and catalase, along with their affiliated gene expressions, were observably augmented in the inoculated groups, with the S5 group standing out. To encapsulate, the actinomycete inoculation holds potential in fostering the growth and maturation of F. taipaiensis, amplifying its environmental resilience. The revelations from this study extend valuable insights for the judicious choice of microbial fertilizers in the cultivated propagation of Fritillaria taipaiensis P. Y. Li.

15.
Sci Adv ; 9(33): eadg7112, 2023 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-37595040

RESUMEN

FOXA1, a transcription factor involved in epigenetic reprogramming, is crucial for breast cancer progression. However, the mechanisms by which FOXA1 achieves its oncogenic functions remain elusive. Here, we demonstrate that the O-linked ß-N-acetylglucosamine modification (O-GlcNAcylation) of FOXA1 promotes breast cancer metastasis by orchestrating the transcription of numerous metastasis regulators. O-GlcNAcylation at Thr432, Ser441, and Ser443 regulates the stability of FOXA1 and promotes its assembly with chromatin. O-GlcNAcylation shapes the FOXA1 interactome, especially triggering the recruitment of the transcriptional repressor methyl-CpG binding protein 2 and consequently stimulating FOXA1 chromatin-binding sites to switch to chromatin loci of adhesion-related genes, including EPB41L3 and COL9A2. Site-specific depletion of O-GlcNAcylation on FOXA1 affects the expression of various downstream genes and thus inhibits breast cancer proliferation and metastasis both in vitro and in vivo. Our data establish the importance of aberrant FOXA1 O-GlcNAcylation in breast cancer progression and indicate that targeting O-GlcNAcylation is a therapeutic strategy for metastatic breast cancer.


Asunto(s)
Neoplasias de la Mama , Cromatina , Humanos , Sitios de Unión , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Epigenómica , Proteínas de Microfilamentos
16.
Protein Sci ; 31(12): e4499, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36335585

RESUMEN

As a key regulator for hormone activity, human aldo-keto reductase family 1 member C3 (AKR1C3) plays crucial roles in the occurrence of various hormone-dependent or independent malignancies. It is a promising target for treating castration-resistant prostate cancer (CRPC). However, the development of AKR1C3 specific inhibitors remains challenging due to the high sequence similarity to its isoform AKR1C2. Here, we performed a combined in silico study to illuminate the inhibitory preference of 3-(3,4-dihydroisoquinolin-2(1H)-ylsulfonyl)benzoic acids for AKR1C3 over AKR1C2, of which compound 38 can achieve up to 5000-fold anti-AKR1C3 selectivity. Our umbrella sampling (US) simulations together with end-point binding free energy calculation MM/GBSA uncover that the high inhibition selectivity originates from the different binding modes, namely "Inward" and "Outward," of this compound series in AKR1C3 and AKR1C2, respectively. In AKR1C3/38, the tetrahydroquinoline moiety of 38 is accommodated inside the SP1 pocket and interacts favorably with surrounding residues, while, in AKR1C2/38, the SP1 pocket is too small to hold the bulky tetrahydroquinoline group that instead moves out of the pocket with 38 transitioning from an "Inward" to an "Outward" state. Further 3D-QSAR and energy decomposition analyses suggest that SP1 in AKR1C3 prefers to bind with a rigid bicyclic moiety and the modification of the R3 group has important implication for the compound's activity. This work is the first attempt to elucidate the selectivity mechanism of inhibitors toward AKR1C3 at the atomic level, which is anticipated to propel the development of next-generation AKR1C3 inhibitors with enhanced efficacy and reduced "off-target" effect for CRPC therapy.


Asunto(s)
Hidroxiprostaglandina Deshidrogenasas , Neoplasias de la Próstata Resistentes a la Castración , Masculino , Humanos , Miembro C3 de la Familia 1 de las Aldo-Ceto Reductasas/metabolismo , Hidroxiprostaglandina Deshidrogenasas/metabolismo , 3-Hidroxiesteroide Deshidrogenasas/metabolismo , Benzoatos/química , Simulación por Computador , Isoformas de Proteínas , Hormonas
17.
ACS Pharmacol Transl Sci ; 4(5): 1628-1638, 2021 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-34661079

RESUMEN

The bromodomain and extra-terminal (BET) domain family of proteins, which include its prototypical member Brd4, is implicated in a variety of cancers and viral infections due to their interaction with cellular and viral proteins. BET proteins contain two bromodomains, a common protein motif that selectively binds acetylated lysine on histones. However, they are structurally distinct from other bromodomain-containing proteins because they encode a unique C-terminal extra-terminal (ET) domain that is important for the protein-protein interactions including jumonji C-domain-containing protein 6 (JMJD6) and histone-lysine N-methyltransferase NSD3 (NSD3). Brd4 functions primarily during transcription as a passive scaffold linking cellular and viral proteins to chromatin. The rapid development of clinical inhibitors targeting Brd4 highlights the importance of this protein as an anticancer target. Current therapeutic approaches focus on the development of small molecule acetylated lysine mimics of histone marks that block the ability of the bromodomains to bind their chromatin marks. Thus far, bromodomain-targeted agents have shown dose-limiting toxicities due to off-target effects on other bromodomain-containing proteins. Here, we exploited a viral-host protein interaction interface to design peptides for the disruption of BET protein function. A murine leukemia virus (MLV) integrase-derived peptide (ET binding motif, EBM) and its shorter minimal binding motif (pentapeptide LKIRL) were sufficient to directly bind the Brd4 ET domain and reduce cellular proliferation of an acute myeloid leukemia cell line. Using computational and biochemical approaches, we identified the minimal essential contacts between EBM and LKIRL peptides and the Brd4 ET domain. Our findings provide a structural foundation for inhibiting BET/Brd4-mediated cancers by targeting the ET domain with small peptide-based inhibitors.

18.
J Med Chem ; 62(24): 10927-10954, 2019 12 26.
Artículo en Inglés | MEDLINE | ID: mdl-31419130

RESUMEN

As a receptor tyrosine kinase of insulin receptor (IR) subfamily, anaplastic lymphoma kinase (ALK) has been validated to play important roles in various cancers, especially anaplastic large cell lymphoma (ALCL), nonsmall cell lung cancer (NSCLC), and neuroblastomas. Currently, five small-molecule inhibitors of ALK, including Crizotinib, Ceritinib, Alectinib, Brigatinib, and Lorlatinib, have been approved by the U.S. Food and Drug Administration (FDA) against ALK-positive NSCLCs. Novel type-I1/2 and type-II ALK inhibitors with improved kinase selectivity and enhanced capability to combat drug resistance have also been reported. Moreover, the "proteolysis targeting chimera" (PROTAC) technique has been successfully applied in developing ALK degraders, which opened a new avenue for targeted ALK therapies. This review provides an overview of the physiological and biological functions of ALK, the discovery and development of drugs targeting ALK by focusing on their chemotypes, activity, selectivity, and resistance as well as potential therapeutic strategies to overcome drug resistance.


Asunto(s)
Quinasa de Linfoma Anaplásico/antagonistas & inhibidores , Antineoplásicos/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Descubrimiento de Drogas , Neoplasias Pulmonares/tratamiento farmacológico , Terapia Molecular Dirigida , Inhibidores de Proteínas Quinasas/uso terapéutico , Animales , Carcinoma de Pulmón de Células no Pequeñas/enzimología , Carcinoma de Pulmón de Células no Pequeñas/patología , Humanos , Neoplasias Pulmonares/enzimología , Neoplasias Pulmonares/patología
19.
Sci Rep ; 7(1): 9088, 2017 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-28831147

RESUMEN

Janus kinase 2 (JAK2) has been regarded as an essential target for the treatment of myeloproliferative neoplasms (MPNs). BBT594 and CHZ868, Type-II inhibitors of JAK2, illustrate satisfactory efficacy in preclinical MPNs and acute lymphoblastic leukemia (ALL) models. However, the L884P mutation of JAK2 abrogates the suppressive effects of BBT594 and CHZ868. In this study, conventional molecular dynamics (MD) simulations, umbrella sampling (US) simulations and MM/GBSA free energy calculations were employed to explore how the L884P mutation affects the binding of BBT594 and CHZ868 to JAK2 and uncover the resistance mechanism induced by the L884P mutation. The results provided by the US and MD simulations illustrate that the L884P mutation enhances the flexibility of the allosteric pocket and alters their conformations, which amplify the conformational entropy change (-TΔS) and weaken the interactions between the inhibitors and target. Additionally, the structural analyses of BBT594 and CHZ868 in complex with the WT JAK2 illustrate that the drug tail with strong electronegativity and small size located in the allosteric pocket of JAK2 may enhance anti-resistance capability. In summary, our results highlight that both of the changes of the conformational entropies and enthalpies contribute to the L884P-induced resistance in the binding of two Type-II inhibitors into JAK2 kinase.


Asunto(s)
Resistencia a Antineoplásicos , Janus Quinasa 2/química , Janus Quinasa 2/metabolismo , Mutación , Inhibidores de Proteínas Quinasas/farmacología , Sitio Alostérico , Aminopiridinas/farmacología , Bencimidazoles/farmacología , Entropía , Humanos , Janus Quinasa 2/genética , Modelos Moleculares , Simulación de Dinámica Molecular , Piperidinas/farmacología , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Unión Proteica , Conformación Proteica , Estructura Secundaria de Proteína , Pirimidinas/farmacología
20.
ACS Cent Sci ; 3(11): 1208-1220, 2017 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-29202023

RESUMEN

Targeted inhibition of anaplastic lymphoma kinase (ALK) dramatically improved therapeutic outcomes in the treatment of ALK-positive cancers, but unfortunately patients invariably progressed due to acquired resistance mutations in ALK. Currently available drugs are all type-I inhibitors bound to the ATP-binding pocket and are most likely to be resistant in patients harboring genetic mutations surrounding the ATP pocket. To overcome drug resistance, we rationally designed a novel kind of "bridge" inhibitor, which specially bind into an extended hydrophobic back pocket adjacent to the ATP-binding site of ALK. The novel type-I1/2 inhibitors display excellent antiproliferation activity against ALK-positive cancer cells and appear superior to two clinically used drugs, crizotinib and ceritinib. Structural and molecular modeling analyses indicate that the inhibitor induces dramatic conformational transition and stabilizes unique DFG-shifted loop conformation, enabling persistent sensitivity to different genetic mutations in ALK. These data highlight a rationale for further development of next-generation ALK inhibitors to combat drug resistance.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA