Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
J Recept Signal Transduct Res ; 42(4): 325-337, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34323638

RESUMO

Non-steroidal anti-inflammatory drugs (NSAIDs) are widely prescribed to treat inflammatory-related diseases, pain and fever. However, the prolong use of traditional NSAIDs leads to undesirable side effects such as gastric, ulceration, and renal toxicity due to lack of selectivity toward respective targets for COX-2, 5-LOX, and PDE4B. Thus, targeting multiple sites can reduce these adverse effects of the drugs and increase its potency. A series of methoxyflavones (F1-F5) were synthesized and investigated for their anti-inflammatory properties through molecular docking and inhibition assays. Among these flavones, only F2 exhibited selectivity toward COX-2 (Selectivity Index, SI: 3.90, COX-2 inhibition: 98.96 ± 1.47%) in comparison with celecoxib (SI: 7.54, COX-2 inhibition: 98.20 ± 2.55%). For PDEs, F3 possessed better selectivity to PDE4B (SI: 4.67) than rolipram (SI: 0.78). F5 had the best 5-LOX inhibitory activity among the flavones (33.65 ± 4.74%) but less than zileuton (90.81 ± 0.19%). Docking analysis indicated that the position of methoxy group and the substitution of halogen play role in determining the bioactivities of flavones. Interestingly, F1-F5 displayed favorable pharmacokinetic profiles and acceptable range of toxicity (IC50>70 µM) in cell lines with the exception for F1 (IC50: 16.02 ± 1.165 µM). This study generated valuable insight in designing new anti-inflammatory drug based on flavone scaffold. The newly synthesized flavones can be further developed as future therapeutic agents against inflammation.


Assuntos
Araquidonato 5-Lipoxigenase , Flavonas , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios não Esteroides/farmacologia , Araquidonato 5-Lipoxigenase/genética , Araquidonato 5-Lipoxigenase/metabolismo , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4/genética , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4/metabolismo , Ciclo-Oxigenase 2/genética , Inibidores de Ciclo-Oxigenase 2/farmacologia , Flavonas/farmacologia , Inibidores de Lipoxigenase/farmacologia , Simulação de Acoplamento Molecular , Estrutura Molecular , Inibidores de Fosfodiesterase/farmacologia , Relação Estrutura-Atividade
2.
Molecules ; 24(13)2019 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-31261881

RESUMO

Herein a novel series of histone deacetylases (HDACs) and epidermal growth factor receptor (EGFR) dual inhibitors were designed and synthesized based on the structure of the approved EGFR inhibitor osimertinib (AZD9291). Among them, four compounds 5D, 5E, 9D and 9E exhibited more potent total HDAC inhibition than the approved HDAC inhibitor SAHA. However, these compounds only showed moderate to low inhibitory potency towards EGFR with compounds 5E and 9E possessing IC50 values against EGFRWT and EGFRT790M in the micromolar range. 3-[4,5-dimethyl-2-thiazolyl]-2,5-diphenyl-2H-tetrazolium bromide (MTT) assay revealed the potent antiproliferative activities of compounds 5D, 5E, 9D and 9E, among which 9E was even more potent against HeLa, MDA-MB-231, MDA-MB-468, HT-29 and KG-1 cell lines than SAHA and AZD9291. Further selectivity profile of 9E showed that this compound was not active against other 13 cancer-related kinases and two epigenetic targets lysine specific demethylase 1 (LSD1) and bromodomain-containing protein 4 (BRD4). These results support further structural modification of 9E to improve its EGFR inhibitory activity, which will lead to more potent and balanced HDAC and EGFR dual inhibitors as anticancer agents.


Assuntos
Acrilamidas/química , Compostos de Anilina/síntese química , Inibidores de Histona Desacetilases/síntese química , Inibidores de Proteínas Quinases/síntese química , Compostos de Anilina/química , Compostos de Anilina/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/genética , Células HT29 , Células HeLa , Inibidores de Histona Desacetilases/química , Inibidores de Histona Desacetilases/farmacologia , Humanos , Estrutura Molecular , Mutação , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/farmacologia
3.
Bioorg Med Chem Lett ; 27(22): 5046-5052, 2017 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-29033233

RESUMO

A series of 2-acetyl-5-O-(amino-alkyl)phenol derivatives was designed, synthesized and evaluated as multi-function inhibitors for the treatment of Alzheimer's disease (AD). The results revealed that compound TM-3 indicated selective AChE inhibitory potency (eeAChE, IC50 = 0.69 µM, selective index (SI) = 32.7). Both kinetic analysis of AChE inhibition and molecular modeling study suggested that TM-3 could simultaneously bind to the catalytic active site and peripheral anionic site of AChE. And TM-3 was also a highly selective MAO-B inhibitor (IC50 = 6.8 µM). Moreover, TM-3 could act as antioxidant (ORAC value was 1.5eq) and neuroprotectant, as well as a selective metal chelating agent. More interestingly, compound TM-3 could cross the blood-brain barrier (BBB) in vitro and abided by Lipinski's rule of five. Therefore, compound TM-3, a promising multi-targeted active molecule, offers an attractive starting point for further lead optimization in the drug-discovery process against AD.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Antioxidantes/síntese química , Benzofenonas/síntese química , Desenho de Fármacos , Fármacos Neuroprotetores/uso terapêutico , Fenóis/química , Piperazinas/síntese química , Acetilcolinesterase/química , Acetilcolinesterase/metabolismo , Animais , Antioxidantes/farmacologia , Antioxidantes/uso terapêutico , Benzofenonas/farmacologia , Benzofenonas/uso terapêutico , Sítios de Ligação , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Inibidores da Colinesterase/síntese química , Inibidores da Colinesterase/farmacologia , Inibidores da Colinesterase/uso terapêutico , Humanos , Peróxido de Hidrogênio/toxicidade , Concentração Inibidora 50 , Simulação de Acoplamento Molecular , Monoaminoxidase/química , Monoaminoxidase/metabolismo , Fármacos Neuroprotetores/síntese química , Fármacos Neuroprotetores/farmacologia , Células PC12 , Permeabilidade/efeitos dos fármacos , Fenóis/farmacologia , Fenóis/uso terapêutico , Piperazinas/farmacologia , Piperazinas/uso terapêutico , Estrutura Terciária de Proteína , Ratos , Relação Estrutura-Atividade
4.
Mol Divers ; 21(3): 511-523, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28194627

RESUMO

Breast cancer is the most frequent cancer reported in women, being responsible for hundreds of thousands of deaths. Chemotherapy has proven to be effective against this malignant neoplasm depending on different biological factors such as the histopathology, grade, and stage, among others. However, breast cancer cells have become resistant to current chemotherapeutic regimens, urging the discovery of new anti-breast cancer drugs. Computational approaches have the potential to offer promising alternatives to accelerate the search for potent and versatile anti-breast cancer agents. In the present work, we introduce the first multitasking (mtk) computational model devoted to the in silico fragment-based design of new molecules with high inhibitory activity against 19 different proteins involved in breast cancer. The mtk-computational model was created from a dataset formed by 24,285 cases, and it exhibited accuracy around 93% in both training and prediction (test) sets. Several molecular fragments were extracted from the molecules present in the dataset, and their quantitative contributions to the inhibitory activities against all the proteins under study were calculated. The combined use of the fragment contributions and the physicochemical interpretations of the different molecular descriptors in the mtk-computational model allowed the design of eight new molecular entities not reported in our dataset. These molecules were predicted as potent multi-target inhibitors against all the proteins, and they exhibited a desirable druglikeness according to the Lipinski's rule of five and its variants.


Assuntos
Antineoplásicos/química , Neoplasias da Mama/metabolismo , Biologia Computacional/métodos , Proteínas de Neoplasias/antagonistas & inibidores , Algoritmos , Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Linhagem Celular Tumoral , Simulação por Computador , Bases de Dados de Compostos Químicos , Desenho de Fármacos , Feminino , Humanos , Modelos Moleculares , Relação Quantitativa Estrutura-Atividade
5.
Int J Mol Sci ; 16(6): 13548-60, 2015 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-26075871

RESUMO

Bis-(2,3-dibromo-4,5-dihydroxy-phenyl)-methane (BDDPM) is a bromophenol first isolated from Rhodomelaceae confervoides. Our previous studies showed that BDDPM exerts PTP1B-inhibiting activity and anti-cancer activity against a wide range of tumor cells while it also showed lower cytotoxicity against normal cells. In the present study, we found that BDDPM exhibits significant activities toward angiogenesis in vitro. BDDPM inhibits multiple angiogenesis processes, including endothelial cell sprouting, migration, proliferation, and tube formation. Further kinase assays investigations found that BDDPM is a potent selective, but multi-target, receptor tyrosine kinase (RTKs) inhibitor. BDDPM (10 µM) inhibits the activities of fibroblast growth factor receptor 2 and 3 (FGFR2, 3), vascular endothelial growth factor receptor 2 (VEGFR2) and platelet-derived growth factor receptor α (PDGFRα) (inhibition rate: 57.7%, 78.6%, 78.5% and 71.1%, respectively). Moreover, BDDPM also decreases the phosphorylation of protein kinase B (PKB/Akt) and endothelial nitric oxide synthase (eNOS), as well as nitric oxide (NO) production in a dose dependent manner. These results indicate that BDDPM can be exploited as an anti-angiogenic drug, or as a lead compound for the development of novel multi-target RTKs inhibitors.


Assuntos
Inibidores da Angiogênese/farmacologia , Hidrocarbonetos Bromados/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/antagonistas & inibidores , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/antagonistas & inibidores , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/antagonistas & inibidores , Rodófitas/química , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Inibidores da Angiogênese/química , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Hidrocarbonetos Bromados/química , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Inibidores de Proteínas Quinases/química , Proteínas Proto-Oncogênicas c-akt/metabolismo
6.
Eur J Med Chem ; 266: 116127, 2024 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-38224650

RESUMO

The occurrence of cancer is closely related to metabolism and epigenetics. Histone deacetylases (HDACs) play a crucial role in the regulation of gene expression as epigenetic regulators, while nicotinamide phosphoribosyltransferase (NAMPT) is significantly involved in maintaining cellular metabolism. In this study, we rationally designed a series of novel HDAC/NAMPT dual inhibitors based on the structural similarity between HDAC and NAMPT inhibitors. The representative compounds 39a and 39h exhibit significant selective inhibitory activity on HDAC1-3 with IC50 values of 0.71-25.1 nM, while displaying modest activity against NAMPT. Compound 39h did not exhibit inhibitory activity against 370 kinases, demonstrating its target specificity. These two compounds exhibit potent anti-proliferative activity in multiple leukemia cell lines with low nanomolar IC50s. It is worth noticing that the dual inhibitors 39a and 39h overcome the primary resistance of HDAC or NAMPT single target inhibitor in p53-null AML cell lines, with the induction of apoptosis-related cell death. NMN recovers the cell death induced by HDAC/NAMPT dual inhibitors, which indicates the lethal effects are caused by the inhibition of NAD biosynthesis pathway as well as HDAC. This research provides an effective strategy to overcome the limitations of HDAC inhibitors in treating p53-null leukemia.


Assuntos
Inibidores de Histona Desacetilases , Leucemia , Humanos , Inibidores de Histona Desacetilases/farmacologia , Inibidores de Histona Desacetilases/química , Proteína Supressora de Tumor p53 , Nicotinamida Fosforribosiltransferase/metabolismo , Linhagem Celular Tumoral , Leucemia/tratamento farmacológico , Leucemia/metabolismo
7.
J Diabetes Metab Disord ; 22(1): 571-580, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37255789

RESUMO

Purpose: The study was undertaken to evaluate the anti-diabetic potential of p-propoxybenzoic acid (p-PBA). Methods: 36 Sprague-Dawley rats of either sex were utilized for the study. Animals were injected with nicotinamide (230 mg/kg) followed by streptozotocin (65 mg/kg) to induce Type-2 Diabetes (T2DM). Animals with blood glucose levels (BGL) over 200 mg/kg were allocated in six groups. Three groups were treated with p-PBA dose of 100 mg/kg, 200 mg/kg and 300 mg/kg respectively; standard control group was treated with 5 mg/kg glibenclamide, while the other two groups were considered as normal control and disease control group. Body weight (BW) and BGL were recorded on Day 0, Day 7, Day 14, and Day 28. Glycosylated hemoglobin (HbA1c), serum insulin levels and lipid profile were recorded on Day 28. Animals were euthanized on Day 28 and the pancreas was isolated for histopathological examination. Results: Diabetic animals treated with p-PBA showed significant improvements in BW (P < 0.05) and BGL (P < 0.001) over 28 days. Levels of HbA1c (P < 0.05) and serum insulin (P < 0.001) were significantly regulated in animals treated with p-PBA. A significant decrease (P < 0.001) was observed in elevated levels of TC, TG, LDL cholesterol and VLDL cholesterol in animals treated with p-PBA. p-PBA significantly regulated the levels of HDL cholesterol (P < 0.001). A notable protective effect of p-PBA was observed through the histopathological examination of pancreas. Conclusion: p-PBA can be characterized as a multi-target inhibiting anti-diabetic agent which can be evaluated against diabetic complications. Supplementary Information: The online version contains supplementary material available at 10.1007/s40200-022-01177-y.

8.
J Biomol Struct Dyn ; : 1-14, 2023 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-37732349

RESUMO

The emergence and immune evasion ability of SARS-CoV-2 Omicron strains, mainly BA.5.2 and BF.7 and other variants of concern have raised global apprehensions. With this context, the discovery of multitarget inhibitors may be proven more comprehensive paradigm than its one-drug-to-one target counterpart. In the current study, a library of 271 phytochemicals from 25 medicinal plants from the Indian Himalayan Region has been virtually screened against SARS-CoV-2 by targeting nine virus proteins, viz., papain-like protease, main protease, nsp12, helicase, nsp14, nsp15, nsp16, envelope, and nucleocapsid for screening of a multi-target inhibitor against the viral replication. Initially, 94 phytochemicals were screened by a hybrid machine learning model constructed by combining 6 confirmatory bioassays against SARS-CoV-2 replication using an instance-based learner lazy k-nearest neighbour classifier. Further, 25 screened compounds with excellent drug-like properties were subjected to molecular docking. The phytochemical Cepharadione A from the plant Piper longum showed binding potential against four proteins with the highest binding energy of -10.90 kcal/mol. The compound has acceptable absorption, distribution, metabolism, excretion, and toxicity properties and exhibits stable binding behaviour in terms of root mean square deviation (0.068 ± 0.05 nm), root-mean-square fluctuation, hydrogen bonds, solvent accessible surface area (83.88-161.89 nm2), and molecular mechanics Poisson-Boltzmann surface area during molecular dynamics simulation of 200 ns with selected target proteins. Concerning the utility of natural compounds in the therapeutics formulation, Cepharadione A could be further investigated as a remarkable lead candidate for the development of therapeutic drugs against SARS-CoV-2.Communicated by Ramaswamy H. Sarma.

9.
J Biomol Struct Dyn ; : 1-18, 2023 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-37691453

RESUMO

Multi-target inhibitors are currently trending in the pharmaceutical research, as they possess increased efficacy and reduced toxicity. In this study multi-target inhibitors for breast cancer are explored from a curated list of natural products, i.e. 4,670 phytochemicals belonging to 360 medicinal plants. In-silico screening of phytochemicals using SeeSAR and AutoDock Vina resulted in identification of Stearyl Palmitate as a potential drug molecule that inhibits three drug targets, i.e. HER-2, MEK-1 and PARP-1 proteins. Molecular Dynamics Simulation for 100 ns each for these three protein-ligand complexes using Desmond, Maestro platform also confirmed the prediction of multi-target inhibition by Stearyl Palmitate. Further in-vitro MTT assay demonstrated that Stearyl Palmitate has a significant IC50 value of 40 µM against MCF-7 cells and >1000 µM against L929 cells. This confirmed that Stearyl Palmitate is having selective cytotoxicity towards breast cancer cells in comparison to non-cancerous cells. Fluorescence staining and flow cytometry analysis confirmed that, Stearyl Palmitate is inducing apoptosis in MCF-7 cells at IC50 concentration. Finally, in-vivo efficacy and toxicity studies were performed using zebrafishes (Danio rerio). It was observed that the fishes treated with IC50 concentration of Stearyl Palmitate demonstrated 2x folds reduction in tumour size, while double dose resulted in 4x folds reduction in tumour size. Stearyl Palmitate did not demonstrate any toxicity or side effects in the zebrafishes. It is concluded that, Stearyl Palmitate, a phytochemical reported to be present in Althea officinalis is a potential anti-breast cancer agent, with ability to inhibit multiple targets such as HER-2, MEK-1 and PARP-2 proteins.Communicated by Ramaswamy H. Sarma.

10.
J Biomol Struct Dyn ; 41(3): 1085-1097, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-34913837

RESUMO

The PI3K/Akt/mTOR signaling pathway plays a pivotal role in cellular metabolism, growth and survival. PI3Kα hyperactivation impairs downstream signaling, including mTOR regulation, and are linked to poor prognosis and refractory cancer treatment. To support multi-target drug discovery, we took advantage from existing PI3Kα and mTOR crystallographic structures to map similarities and differences in their ATP-binding pockets in the presence of selective or dual inhibitors. Molecular dynamics and MM/PBSA calculations were employed to study the binding profile and identify the relative contribution of binding site residues. Our analysis showed that while varying parameters of solute and solvent dielectric constant interfered in the absolute binding free energy, it had no effect in the relative per residue contribution. In all complexes, the most important interactions were observed within 3-3.5 Å from inhibitors, responding for ∼75-100% of the total calculated interaction energy. While closest residues are essential for the strength of the binding of all ligands, more distant residues seem to have a larger impact on the binding of the dual inhibitor, as observed for PI3Kα residues Phe934, Lys802 and Asp805 and, mTOR residues Leu2192, Phe2358, Leu2354, Lys2187 and Tyr2225. A detailed description of individual residue contribution in the presence of selective or dual inhibitors is provided as an effort to improve the understanding of molecular mechanisms controlling multi-target inhibition. This work provides key information to support further studies seeking the rational design of potent PI3K/mTOR dual inhibitors for cancer treatment.Communicated by Ramaswamy H. Sarma.


Assuntos
Fosfatidilinositol 3-Quinases , Serina-Treonina Quinases TOR , Inibidores de Fosfoinositídeo-3 Quinase , Fosfatidilinositol 3-Quinases/química , Fosfatidilinositol 3-Quinases/metabolismo , Serina-Treonina Quinases TOR/química , Sítios de Ligação , Trifosfato de Adenosina/metabolismo
11.
J Biomol Struct Dyn ; 39(6): 1965-1974, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32174270

RESUMO

Cell-division protein kinases (CDKs) are gorgeous examples of targets for the helpful treatment of cancer by using multi-target inhibitors. Specifically, targeting cell-division protein kinase1/cyclin B (CDK1/Cyclin B), cell-division protein kinase 2/cyclin A (CDK2/Cyclin A) and cell-division protein kinase 4/cyclin D1 (CDK4/Cyclin D1) are considered a safe strategy to over the toxicity complications which are emerging from low specificity. In this work, we conducted the double docking and molecular dynamics to explicate the effect of amygdalin upon conformational modifications of selected targets. Moreover, the principal component analysis (PCA) was employed to inspect the effect of amygdalin on the fundamental motions of the each protein as target. Docking results illustrated that the binding free energies of amygdalin (AMY) to CDK1/Cyclin B, CDK 2/Cyclin A and CDK 4/Cyclin D1 were to be -9.41, -9.02 and -10.6 kcal/mol, respectively. The PCA results disclosed that binding of the AMY minimized the fundamental dynamics of CDK1/Cyclin B and CDK2/Cyclin A. The obtained results can give an insight into inhibitory activity of amygdalin that could help in designing of potential inhibitors. In the other word, it can be used AMY to inhibit other mechanisms and/or hallmarks of cancer.Communicated by Ramaswamy H. Sarma.


Assuntos
Amigdalina , Antineoplásicos , Quinases Ciclina-Dependentes , Amigdalina/farmacologia , Antineoplásicos/farmacologia , Ciclo Celular , Quinases Ciclina-Dependentes/metabolismo , Quinases Ciclina-Dependentes/farmacologia , Humanos , Simulação de Dinâmica Molecular
12.
Front Chem ; 9: 634663, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33777898

RESUMO

Parasitic diseases remain as unresolved health issues worldwide. While for some parasites the treatments involve drug combinations with serious side effects, for others, chemical therapies are inefficient due to the emergence of drug resistance. This urges the search for novel antiparasitic agents able to act through multiple mechanisms of action. Here, we report the first multi-target model based on quantitative structure-activity relationships and a multilayer perceptron neural network (mt-QSAR-MLP) to virtually design and predict versatile inhibitors of proteins involved in the survival and/or infectivity of different pathogenic parasites. The mt-QSAR-MLP model exhibited high accuracy (>80%) in both training and test sets for the classification/prediction of protein inhibitors. Several fragments were directly extracted from the physicochemical and structural interpretations of the molecular descriptors in the mt-QSAR-MLP model. Such interpretations enabled the generation of four molecules that were predicted as multi-target inhibitors against at least three of the five parasitic proteins reported here with two of the molecules being predicted to inhibit all the proteins. Docking calculations converged with the mt-QSAR-MLP model regarding the multi-target profile of the designed molecules. The designed molecules exhibited drug-like properties, complying with Lipinski's rule of five, as well as Ghose's filter and Veber's guidelines.

13.
Comput Methods Programs Biomed ; 195: 105660, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32726718

RESUMO

BACKGROUND AND OBJECTIVE: Historically, amygdalin has been used as alternative medicine or in vitro and in vivo studies, but no single study exists which discusses the structural mechanism of amygdalin at a molecular level. This paper inquiries into the inhibitory actions of amygdalin on the selected targets: AKT1, FAK, and ILK, which are regulators for various mediated signaling pathways, and are associated with cell adhesion, migration, and differentiation. In order to get details at the molecular level of amygdalin's inhibitory activities against chosen proteins, molecular modeling and simulation techniques including double docking, molecular dynamics simulation, free energy landscape analysis, and binding free energy calculation were exerted. METHODS: To get molecular level details of amygdalin inhibitory effects against the relevant proteins; here the utilized tools are the following: the double docking, molecular dynamics simulation, free energy landscape analysis, g_mmpbsa, and interaction entropy were used to evaluate the inhibitory activity against targeted proteins. RESULTS: The computational calculations revealed that amygdalin inhibits the selected targets via block the ATP-binding pocket of AKT1, FAK, and ILK by forming stable hydrogen bonds. Moreover, free energy landscape, FEL exposed that amygdalin stabilized the global conformations of both FAK and ILK proteins to the minimum global energy besides it reduced the essential dynamics of FAK and ILK proteins. MMPBSA computations provided further evidence for amygdalin's stability inside the ATP-binding pocket of AKT1, FAK, and ILK with a binding free energy of 45.067, -13.033, 13.109 kJ/mol, respectively. The binding free energies are lastly consistent with the hydrogen bonding and pairs within 0.35 nm results. The decomposition of binding energy shows the pivotal amino acid residues responsible for the stability of amygdalin's interactions inside the ATP-binding sites by forming hydrogen bonds. CONCLUSIONS: Before this work, it was enigmatic to make predictions about how amygdalin inhibits metastasis of cancer. But the computational results contribute in several ways to our understanding of amygdalin activity and provide a basic insight into the activity of amygdalin as a multi-target drug in the metastasis and invasion of cancer.


Assuntos
Amigdalina , Neoplasias , Sítios de Ligação , Entropia , Humanos , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Ligação Proteica
14.
Eur J Med Chem ; 163: 671-689, 2019 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-30572178

RESUMO

Although lung adenocarcinoma patients have benefited from the development of targeted therapy, patients with lung squamous cell carcinoma (SqCC) have no effective treatment due to the complexity and heterogeneity of the disease. Therefore, basing on the genetic analysis of mutations in lung squamous cell carcinoma to design multi-target inhibitors represents a potential strategy for the medical treatment. In this study, through screening an in-house focused library, we identified an interesting indazole scaffold. And following with binding analysis, we elaborated the structure-activity relationship of this hit compound by optimizing four parts guided by the DDR2 enzymatic assay, which resulted in a potent lead compound 10a. We conducted further optimization of dual enzymatic inhibitions towards FGFR1 and DDR2, two important kinases in lung squamous cell carcinoma. Finally, from the cellular antiproliferative activity tests and in vivo pharmacokinetic test, 3-substituted indazole derivative 11k was found to be a promising candidate and subjected to in vivo pharmacology study with the mouse xenograft models, demonstrating profound anti-tumor efficacy. Additional in vitro druglike assessment reinforced that compound 11k could be valuable for SqCC drug development.


Assuntos
Antineoplásicos/síntese química , Carcinoma de Células Escamosas/tratamento farmacológico , Descoberta de Drogas , Indazóis/síntese química , Neoplasias Pulmonares/tratamento farmacológico , Inibidores de Proteínas Quinases/química , Animais , Carcinoma de Células Escamosas/enzimologia , Receptor com Domínio Discoidina 2/antagonistas & inibidores , Xenoenxertos , Humanos , Indazóis/química , Indazóis/farmacologia , Neoplasias Pulmonares/enzimologia , Camundongos , Inibidores de Proteínas Quinases/uso terapêutico , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/antagonistas & inibidores , Relação Estrutura-Atividade
15.
Front Pharmacol ; 10: 797, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31379572

RESUMO

Introduction: Lipid mediators (LMs) comprise bioactive metabolites of polyunsaturated fatty acids, including pro-inflammatory prostaglandins (PGs), thromboxanes (TXs), and leukotrienes (LTs), as well as specialized pro-resolving mediators (SPMs). They are essentially biosynthesized via cyclooxygenase (COX) and lipoxygenase (LO) pathways in complex networks and regulate the progression as well as the resolution of inflammatory disorders including inflammation-triggered cancer. Ginkgolic acid (GA) is a phenolic acid contained in Ginkgo biloba L. with neuroprotective, antimicrobial, and antitumoral properties. Although LMs regulate microbial infections and tumor progression, whether GA affects LM biosynthesis is unknown and was investigated here in detail. Methods: Pharmacophore-based virtual screening was performed along with docking simulations. Activity assays were conducted for isolated human recombinant 5-LO, cytosolic phospholipase (PLA)2α, COX-2, and ovine COX-1. The activity of human mPGES-1 and thromboxane A2 synthase (TXAS) was determined in crude cellular fractions. Cellular LM formation was studied using human monocytes, neutrophils, platelets, and M1- and M2-like macrophages. LMs were identified after (ultra)high-performance liquid chromatography by UV detection or ESI-tandem mass spectrometry. Results: GA was identified as virtual hit in an mPGES-1 pharmacophore-based virtual screening. Cell-free assays revealed potent suppression of mPGES-1 activity (IC50 = 0.7 µM) that is fully reversible and essentially independent of the substrate concentration. Moreover, cell-free assays revealed COX-1 and TXAS as additional targets of GA with lower affinity (IC50 = 8.1 and 5.2 µM). Notably, 5-LO, the key enzyme in LT biosynthesis, was potently inhibited by GA (IC50 = 0.2 µM) in a reversible and substrate-independent manner. Docking simulations support the molecular interaction of GA with mPGES-1 and 5-LO and suggest concrete binding sites. Interestingly, interference of GA with mPGES-1, COX-1, TXAS, and 5-LO was evident also in intact cells with IC50 values of 2.1-3.8 µM; no radical scavenging or cytotoxic properties were obvious. Analysis of LM profiles from bacteria-stimulated human M1- and M2-like macrophages confirmed the multi-target features of GA and revealed LM redirection towards the formation of 12-/15-LO products including SPM. Conclusions: We reveal GA as potent multi-target inhibitor of key enzymes in the biosynthesis of pro-inflammatory LMs that contribute to the complex pharmacological and toxicological properties of GA.

16.
Eur J Med Chem ; 167: 61-75, 2019 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-30763817

RESUMO

A series of 1,3,5-substituted indole derivatives was prepared to explore the anti-proliferative activity against a panel of human tumour cell lines. A 5-carboxamide derivative (27) emerged as the most potent compound of this series, inhibiting the HeLa cell growth at sub-micromolar concentrations. Target fishing of 27 using a combination of inverse virtual screening (IVS) approach and ligand-based shape similarity study identified the top-ranked targets for 27 as belonging to kinome. These results were further confirmed by in vitro binding assays, leading to the identification of 27 as multi-target kinase inhibitor. The compound 27 was further characterized for its antiproliferative activity by in cell studies, showing a mechanism of action involving modification of the cell cycle, increase in ROS release and caspase 3-expression and decrease in ERK expression.


Assuntos
Ensaios de Seleção de Medicamentos Antitumorais/métodos , Indóis/farmacologia , Inibidores de Proteínas Quinases/síntese química , Antineoplásicos/síntese química , Antineoplásicos/farmacologia , Caspase 3/metabolismo , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Células HeLa , Humanos , Indóis/síntese química , Sistema de Sinalização das MAP Quinases , Inibidores de Proteínas Quinases/farmacologia , Espécies Reativas de Oxigênio/metabolismo
17.
Am J Cancer Res ; 7(9): 1884-1898, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28979811

RESUMO

Non-Small Cell Lung Cancer (NSCLC) is driven by a variety of deregulated kinases and the development of multi-target inhibitor for multiple signaling pathways or multiple steps is required. Here, we reported that ZWM026, an indolocarbazoles analogue, derived from mangrove in coastal marine wetland, exhibited selectivity and reversibility against T790M mutant over wild-type EGFR in naturally occurring NSCLC cells and constructed NIH-3T3 cells. It simultaneously inhibited activities of HER2, HER3, HER4 and RET but was different from current multi-target kinase inhibitors. There was no activity in protein kinase C (PKC) family which is generally recognized as molecule target of indolocarbazoles. ZWM026 had more potent activities against gefitinib sensitizing, non-sensitizing and rare EGFR mutant NSCLC cells and constructed NIH-3T3 cells. ZWM026 induced apoptosis and exerted a synergistic effect by combining with cisplatin in NCI-H1975 cells. In summary, we identified a novel reversible multi-target inhibitor which could serve as a promising lead compound of drug development for NSCLC.

18.
Eur J Med Chem ; 95: 104-15, 2015 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-25800646

RESUMO

A novel series of macrocyclic compounds were designed and synthesized as multi-target inhibitors targeting HDAC, FLT3 and JAK2. Some of these compounds exhibited potent HDAC inhibition as well as FLT3 and JAK2 inhibition under both cell-free and cellular conditions. In vitro antiproliferative assay indicated that these compounds were interestingly more cytotoxic to MV4-11 cells bearing FLT3-ITD mutation and HEL cells bearing JAK2(V617F) mutation.


Assuntos
Antineoplásicos/farmacologia , Desenho de Fármacos , Histona Desacetilases/química , Janus Quinase 2/antagonistas & inibidores , Compostos Macrocíclicos/síntese química , Compostos Macrocíclicos/farmacologia , Tirosina Quinase 3 Semelhante a fms/antagonistas & inibidores , Antineoplásicos/síntese química , Apoptose/efeitos dos fármacos , Western Blotting , Proliferação de Células/efeitos dos fármacos , Células HeLa , Inibidores de Histona Desacetilases/síntese química , Inibidores de Histona Desacetilases/farmacologia , Humanos , Simulação de Acoplamento Molecular , Inibidores de Proteínas Quinases/síntese química , Inibidores de Proteínas Quinases/farmacologia , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA