Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 12(1): 5328, 2021 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-34493725

RESUMO

Aliphatic esters are essential constituents of biologically active compounds and versatile chemical intermediates for the synthesis of drugs. However, their preparation from readily available olefins remains challenging. Here, we report a strategy to access aliphatic esters from olefins through a photocatalyzed alkoxycarbonylation reaction. Alkyloxalyl chlorides, generated in situ from the corresponding alcohols and oxalyl chloride, are engaged as alkoxycarbonyl radical fragments under photoredox catalysis. This transformation tolerates a broad scope of electron-rich and electron-deficient olefins and provides the corresponding ß-chloro esters in good yields. Additionally, a formal ß-selective alkene alkoxycarbonylation is developed. Moreover, a variety of oxindole-3-acetates and furoindolines are prepared in good to excellent yields. A more concise formal synthesis of (±)-physovenine is accomplished as well. With these strategies, a wide range of natural-product-derived olefins and alkyloxalyl chlorides are also successfully employed.


Assuntos
Alcenos/química , Técnicas de Química Sintética , Ésteres/química , Indóis/síntese química , Oxindóis/síntese química , Álcoois/química , Catálise , Cloretos/química , Humanos , Estrutura Molecular , Oxalatos/química , Oxirredução , Processos Fotoquímicos , Fisostigmina/análogos & derivados , Fisostigmina/síntese química , Estereoisomerismo
2.
Biotechnol Appl Biochem ; 64(2): 153-164, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26790544

RESUMO

NS3 protease plays a vital role in the replication of the hepatitis C virus (HCV). BMS-605339 is a novel linear tetra-peptide α-ketoamide inhibitor of NS3 protease and shows specificity for HCV NS3 protease genotype 1a and genotype 1b. Mutation at the key site 168 of the HCV NS3 protease can induce resistance to BMS-605339, which greatly affects the antiviral therapy efficacy to hepatitis C. In the present study, we employed molecular dynamics simulations, free energy calculations, and free energy decomposition to explore the drug resistance mechanism of BMS-605339 due to the three representative mutations D168C/Y/V. The free energy decomposition analysis indicates that the decrease in the binding affinity is mainly attributed to the decrease in both van der Waals and electrostatic interactions. After detailed analysis of our calculated results, we observed that the break of the salt bridge between residues 155 and 168 caused by the mutations D168C/Y/V is the original reason for the decrease in the binding ability between BMS-605339 and the mutant NS3 proteases. The obtained results will reveal the drug resistance mechanism between BMS-605339 and the mutant NS3 proteases, and provide valuable clue for designing novel and more potent drugs to HCV NS3 protease.


Assuntos
Resistência a Medicamentos/genética , Hepatite C/tratamento farmacológico , Inibidores de Proteases/química , Proteínas não Estruturais Virais/genética , Antivirais/química , Antivirais/uso terapêutico , Biologia Computacional , Hepacivirus/efeitos dos fármacos , Hepacivirus/patogenicidade , Hepatite C/virologia , Humanos , Isoquinolinas/química , Isoquinolinas/uso terapêutico , Simulação de Dinâmica Molecular , Mutação , Inibidores de Proteases/uso terapêutico , Sulfonamidas/química , Sulfonamidas/uso terapêutico , Proteínas não Estruturais Virais/química
3.
Biochem Cell Biol ; 94(2): 147-58, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26836778

RESUMO

NS5B polymerase plays an important role in viral replication machinery. TMC647055 (TMC) is a novel and potent non-nucleoside inhibitor of the HCV NS5B polymerase. However, mutations that result in drug resistance to TMC have been reported. In this study, we used molecular dynamics (MD) simulations, binding free energy calculations, and free energy decomposition to investigate the drug resistance mechanism of HCV to TMC resulting from L392I, P495T, P495S, and P495L mutations in NS5B polymerase. From the calculated results we determined that the decrease in the binding affinity between TMC and NS5B(L392I) polymerase is mainly caused by the extra methyl group at the CB atom of Ile. The polarity of the side-chain of residue 495 has no distinct influence on residue 495 binding with TMC, whereas the smaller size of the side-chain of residue 495 causes a substantial decrease in the van der Walls interaction between TMC and residue 495. Moreover, the longer length of the side-chain of residue 495 has a significant effect on the electrostatic interaction between TMC and Arg-503. Finally, we performed the same calculations and detailed analysis on other 3 mutations (L392V, P495V, and P495I). The results further confirmed our conclusions. The computational results not only reveal the drug resistance mechanism between TMC647055 and NS5B polymerase, but also provide valuable information for the rational design of more potent non-nucleoside inhibitors targeting HCV NS5B polymerase.


Assuntos
Antivirais/farmacologia , Farmacorresistência Viral/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Sulfonamidas/farmacologia , Proteínas não Estruturais Virais/antagonistas & inibidores , Antivirais/química , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/química , Compostos Heterocíclicos de 4 ou mais Anéis/química , Modelos Moleculares , Mutação , Relação Estrutura-Atividade , Sulfonamidas/química , Termodinâmica , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/metabolismo
4.
Biochim Biophys Acta ; 1861(3): 149-55, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26680361

RESUMO

Fatty acid synthase (FAS) is a multifunctional homodimeric protein, and is the key enzyme required for the anabolic conversion of dietary carbohydrates to fatty acids. FAS synthesizes long-chain fatty acids from three substrates: acetyl-CoA as a primer, malonyl-CoA as a 2 carbon donor, and NADPH for reduction. The entire reaction is composed of numerous sequential steps, each catalyzed by a specific functional domain of the enzyme. FAS comprises seven different functional domains, among which the ß-ketoacyl synthase (KS) domain carries out the key condensation reaction to elongate the length of fatty acid chain. Acyl tail length controlled fatty acid synthesis in eukaryotes is a classic example of how a chain building multienzyme works. Different hypotheses have been put forward to explain how those sub-units of FAS are orchestrated to produce fatty acids with proper molecular weight. In the present study, molecular dynamic simulation based binding free energy calculation and access tunnels analysis showed that the C16 acyl tail fatty acid, the major product of FAS, fits to the active site on KS domain better than any other substrates. These simulations supported a new hypothesis about the mechanism of fatty acid production ratio: the geometric shape of active site on KS domain might play a determinate role.


Assuntos
Ácido Graxo Sintases/metabolismo , Ácidos Graxos/biossíntese , Acetilcoenzima A/metabolismo , Sítios de Ligação , Catálise , Domínio Catalítico , Ácido Graxo Sintases/química , Ácidos Graxos/química , Malonil Coenzima A/metabolismo , Simulação de Dinâmica Molecular , NADP/metabolismo , Oxirredução , Ligação Proteica , Estrutura Terciária de Proteína , Relação Estrutura-Atividade , Especificidade por Substrato
5.
Antiviral Res ; 113: 79-92, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25449363

RESUMO

Filibuvir, a potent non-nucleoside inhibitor of the hepatitis C virus (HCV) NS5B RNA-dependent RNA polymerase (RdRp), has shown great promise in phase IIb clinical trial. However, drug resistant mutations towards Filibuvir have been identified. In the present study, the drug resistance mechanism of wild-type (WT) and mutant NS5B polymerases (including V494I, V494A, M426A, and M423T) toward Filibuvir was investigated by molecular modeling methods. The predicted binding free energy of these five complexes is highly consistent with the experimental EC50 values of Filibuvir to the wild-type and mutant NS5B RdRps, V494I

Assuntos
Antivirais/farmacologia , Farmacorresistência Viral/efeitos dos fármacos , Farmacorresistência Viral/genética , Hepacivirus/efeitos dos fármacos , Mutação , Pironas/farmacologia , RNA Polimerase Dependente de RNA/genética , Triazóis/farmacologia , Antivirais/química , Antivirais/metabolismo , Hepacivirus/enzimologia , Hepacivirus/genética , Simulação de Dinâmica Molecular , Pironas/química , Pironas/metabolismo , RNA Polimerase Dependente de RNA/metabolismo , Termodinâmica , Triazóis/química , Triazóis/metabolismo
6.
Nanoscale ; 6(21): 12828-37, 2014 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-25225777

RESUMO

There are growing concerns about the biosafety of nanomaterials such as carbon nanotubes (CNTs) as their applications become more widespread. We report here a theoretical and experimental study of the binding of various sizes of CNTs [CNT (4,4), (5,5), (6,6) and (7,7)] to calmodulin (CaM) protein and, in particular, their impact on the Ca(2+)-dependent dynamic properties of CaM. Our simulations show that all the CNTs can plug into the hydrophobic binding pocket of Ca(2+)-bound CaM with binding affinities comparable with the native substrate M13 peptide. Even though CNT (4,4) shows a similar behavior to the M13 peptide in its dissociation from Ca(2+)-free CaM, wider CNTs still bind firmly to CaM, indicating a potential failure of Ca(2+) regulation. Such a size-dependent impact of CNTs on the dynamic properties of CaM is a result of the excessively strong hydrophobic interactions between the wider CNTs and CaM. These simulation results were confirmed by circular dichroism spectroscopy, which showed that the secondary structures of CaM become insensitive to Ca(2+) concentrations after the addition of CNTs. Our findings indicate that the cytotoxicity of nanoparticles to proteins arises not only from the inhibition of static protein structures (binding pockets), but also from impacts on their dynamic properties.


Assuntos
Calmodulina/química , Nanotecnologia/métodos , Nanotubos de Carbono/química , Motivos de Aminoácidos , Sítios de Ligação , Cálcio/química , Dicroísmo Circular , Simulação por Computador , Interações Hidrofóbicas e Hidrofílicas , Íons , Ligantes , Simulação de Dinâmica Molecular , Tamanho da Partícula , Peptídeos/química , Ligação Proteica , Estrutura Secundária de Proteína
7.
Biochem Cell Biol ; 92(5): 357-69, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25178998

RESUMO

Narlaprevir is a novel NS3/4A protease inhibitor of hepatitis C virus (HCV), and it has been tested in a phase II clinical trial recently. However, distinct drug-resistance of Narlaprevir has been discovered. In our study, the molecular mechanisms of drug-resistance of Narlaprevir due to the mutations V36M, R155K, V36M+R155K, T54A, and A156T of NS3/4A protease have been investigated by molecular dynamics (MD) simulations, free energy calculations, and free energy decomposition analysis. The predicted binding free energies of Narlaprevir towards the wild-type and five mutants show that the mutations V36M, R155K, and T54A lead to low-level drug resistance and the mutations V36M+R155K and A156T lead to high-level drug resistance, which is consistent with the experimental data. The analysis of the individual energy terms indicates that the van der Waals contribution is important for distinguishing the binding affinities of these six complexes. These findings again show that the combination of different molecular modeling techniques is an efficient way to interpret the molecular mechanism of drug-resistance. Our work mainly elaborates the molecular mechanism of drug-resistance of Narlaprevir and further provides valuable information for developing novel, safer, and more potent HCV antiviral drugs in the near future.


Assuntos
Antivirais/farmacologia , Dipeptídeos/farmacologia , Farmacorresistência Viral , Hepacivirus/efeitos dos fármacos , Hepacivirus/genética , Sulfonas/farmacologia , Proteínas não Estruturais Virais/genética , Ciclopropanos , Farmacorresistência Viral/genética , Genes Virais , Leucina/análogos & derivados , Simulação de Dinâmica Molecular , Mutação , Prolina/análogos & derivados , Ureia , Proteínas não Estruturais Virais/antagonistas & inibidores
8.
J Mol Model ; 19(12): 5187-98, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24122410

RESUMO

Human dihydrofolate reductase-like 1 (DHFRL1) has been identified as a second human dihydrofolate reductase (DHFR) enzyme. Although DHFRL1 have high sequence homology with human DHFR, dihydrofolate (DHF) exhibits a lowered binding affinity to DHFRL1 and the corresponding molecular mechanism is still unknown. To address this question, we studied the binding of DHF to DHFRL1 and DHFR by using molecular dynamics simulation. Moreover, to investigate the role the 24th residue of DHFR/DHFRL1 plays in DHF binding, R24W DHFRL1 mutant was also studied. The van der Waals interaction are more crucial for the total DHF binding energies, while the difference between the DHF binding energies of human DHFR and DHFRL1 can be attributed to the electrostatic interaction and the polar desolvation free energy.More specifically, lower DHF affinity to DHFRL1 can be mainly attributed to the reduction of net electrostatic interactions of residues Arg32 and Gln35 of DHFRL1 with DHF as being affected by Arg24. The side chain of Arg24 in DHFRL1 can extend deeply into the binding sites of DHF and NADPH, and disturb the DHF binding by steric effect, which rarely happens in human DHFR and R24W DHFRL1 mutant. Additionally, the conformation of loop I in DHFRL1 was also studied in this work. Interestingly, the loop conformation resemble to normal closed state of Escherichia coli DHFR other than the closed state of human DHFR. We hope this work will be useful to understand the general characteristics of DHFRL1.


Assuntos
Ácido Fólico/análogos & derivados , Simulação de Dinâmica Molecular , Tetra-Hidrofolato Desidrogenase/química , Sítios de Ligação , Escherichia coli/enzimologia , Ácido Fólico/química , Humanos , Cinética , Ligação Proteica
9.
J Chem Inf Model ; 53(5): 1157-67, 2013 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-23621621

RESUMO

Protein tyrosine phosphatase 1B (PTP1B) is a promising target for the treatment of obesity and type II diabetes. Allosteric inhibitors can stabilize an active conformation of PTP1B by hindering the conformational transition of the WPD loop of PTP1B from the open to the closed state. Here, the umbrella sampling molecular dynamics (MD) simulations were employed to compute the reaction path of the conformational transition of PTP1B, and the snapshots extracted from the MD trajectory were clustered into 58 conformational groups based on the key conformational parameter. Then, the impact of the conformational change of the WPD loop on the interactions between the allosteric site of PTP1B and an allosteric inhibitor BB3 was explored by using the MM/GBSA binding free energy calculations and free energy decomposition analysis. The simulation results show that the binding free energy of BB3 increases gradually from the open to the closed conformation of the WPD loop, providing the molecular mechanism of allosteric inhibition. Correlation analysis of the different energy terms indicates that the allosteric inhibitor with more negative van der Waals contribution cannot only exhibit stronger binding affinity but also hinder the swing of the WPD loop more effectively. Besides, it is found that the energy contribution of Lys292 in the α7 helix undergoes significant change, which reveals that Lys292 is not only the key residue for ligand binding but also plays an important role in hindering the conformational change of the WPD loop.


Assuntos
Inibidores Enzimáticos/farmacologia , Simulação de Dinâmica Molecular , Proteína Tirosina Fosfatase não Receptora Tipo 1/antagonistas & inibidores , Proteína Tirosina Fosfatase não Receptora Tipo 1/química , Regulação Alostérica/efeitos dos fármacos , Movimento , Estrutura Secundária de Proteína , Proteína Tirosina Fosfatase não Receptora Tipo 1/metabolismo , Termodinâmica
10.
J Comput Aided Mol Des ; 26(12): 1327-42, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23224743

RESUMO

In this work, selectivity mechanism of APP-IP inhibitor (ß-amyloid precursor protein-derived inhibitory peptide) over matrix metalloproteinases (MMPs including MMP-2, MMP-7, MMP-9 and MMP-14) was investigated by molecular modeling methods. Among MMPs, MMP-2 is the most favorable one for APP-IP interacting based on our calculations. The predicted binding affinities can give a good explanation of the activity difference of inhibitor APP-IP. In Comparison with MMP-2/APP-IP complex, the side chain of Tyr214(MMP-7) makes the binding pocket so shallow that the whole side chain of Tyr3(APP-IP) can not be fully embraced, thus unfavorable for the N-terminal of APP-IP binding to MMP-7. The poor selectivity of APP-IP toward MMP-9 is mainly related with the decrease of interaction between the APP-IP C-terminal and MMP-9 due to the bulky side chains of Pro193 and Gln199, which is in agreement with experiment. The mutations at residues P193A and Q199G of MMP-9 alternate the binding pattern of the C-terminal of APP-IP by forming two new hydrogen bonds and hydrophobic interactions with MMP-9. The mutants favor the binding affinity of MMP-9 largely. For MMP-14/APP-IP, the large steric effect of Phe204(MMP-14) and the weak contributions of the polar residues Asn231(MMP-14) and Thr190(MMP-14) could explain why MMP-14 is non-selective for APP-IP interacting. Here, the molecular modeling methods were successfully employed to explore the selective inhibitor of MMPs, and our work gives valuable information for future rational design of selective peptide inhibitors toward individual MMP.


Assuntos
Metaloproteinases da Matriz/metabolismo , Proteínas/metabolismo , Sequência de Aminoácidos , Sítios de Ligação , Domínio Catalítico , Simulação por Computador , Cristalografia por Raios X , Humanos , Metaloproteinases da Matriz/química , Modelos Moleculares , Dados de Sequência Molecular , Ligação Proteica , Estrutura Secundária de Proteína , Proteínas/química , Homologia de Sequência de Aminoácidos , Especificidade por Substrato
11.
Int J Mol Sci ; 13(6): 7057-7079, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22837679

RESUMO

Mitogen-activated protein kinase-activated protein kinase 2 (MK-2) has been identified as a drug target for the treatment of inflammatory diseases. Currently, a series of thiourea analogs as potent MK-2 inhibitors were studied using comprehensive computational methods by 3D-QSAR, molecular docking and molecular dynamics simulations for a further improvement in activities. The optimal 3D models exhibit high statistical significance of the results, especially for the CoMFA results with r(2) (ncv), q(2) values of 0.974, 0.536 for the internal validation, and r(2) (pred), r(2) (m) values of 0.910, 0.723 for the external validation and Roy's index, respectively. In addition, more rigorous validation criteria suggested by Tropsha were also employed to check the built models. Graphic representation of the results, as contoured 3D coefficient plots, also provides a clue to the reasonable modification of molecules: (i) The substituent with a bulky size and electron-rich group at the C5 position of the pyrazine ring is required to enhance the potency; (ii) The H-bond acceptor group in the C3 position of the pyrazine ring is likely to be helpful to increase MK-2 inhibition; (iii) The small and electropositive substituent as a hydrogen bond donor of the C2 position in the oxazolone ring is favored; In addition, several important amino acid residues were also identified as playing an important role in MK-2 inhibition. The agreement between 3D-QSAR, molecular docking and molecular dynamics simulations also proves the rationality of the developed models. These results, we hope, may be helpful in designing novel and potential MK-2 inhibitors.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Tioureia/análogos & derivados , Tioureia/química , Algoritmos , Eletroquímica , Elétrons , Humanos , Ligação de Hidrogênio , Imageamento Tridimensional , Concentração Inibidora 50 , Ligantes , Modelos Estatísticos , Simulação de Dinâmica Molecular , Oxazolona/química , Análise de Componente Principal , Ligação Proteica , Pirazinas/química , Relação Quantitativa Estrutura-Atividade , Software
12.
Int J Mol Sci ; 12(11): 8161-80, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22174657

RESUMO

Fructose 1,6-bisphosphatase (FBPase) has been identified as a drug discovery target for lowering glucose in type 2 diabetes mellitus. In this study, a large series of 105 FBPase inhibitors were studied using a combinational method by 3D-QSAR, molecular docking and molecular dynamics simulations for a further improvement in potency. The optimal 3D models exhibit high statistical significance of the results, especially for the CoMFA results with r(ncv) (2), q(2) values of 0.986, 0.514 for internal validation, and r(pred) (2), r(m) (2) statistics of 0.902, 0.828 statistics for external validation. Graphic representation of the results, as contoured 3D coefficient plots, also provides a clue to the reasonable modification of molecules. (1) Substituents with a proper length and size at the C5 position of the thiazole core are required to enhance the potency; (2) A small and electron-withdrawing group at the C2 position linked to the thiazole core is likely to help increase the FBPase inhibition; (3) Substituent groups as hydrogen bond acceptors at the C2 position of the furan ring are favored. In addition, the agreement between 3D-QSAR, molecular docking and molecular dynamics simulation proves the rationality of the developed models. These results, we hope, may be helpful in designing novel and potential FBPase inhibitors.


Assuntos
Simulação por Computador , Inibidores Enzimáticos/química , Frutose-Bifosfatase/antagonistas & inibidores , Oxazóis/química , Tiazóis/química , Diabetes Mellitus Tipo 2/tratamento farmacológico , Inibidores Enzimáticos/farmacologia , Ligação de Hidrogênio , Modelos Moleculares , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Oxazóis/farmacologia , Relação Quantitativa Estrutura-Atividade , Alinhamento de Sequência , Tiazóis/farmacologia
13.
J Chem Inf Model ; 51(10): 2626-35, 2011 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-21919471

RESUMO

In the present study, the impacts of G198N and W128F mutations on the recognition between Aurora A and targeting protein of Xenopus kinesin-like protein 2 (TPX2) were investigated using molecular dynamics (MD) simulations, free energy calculations, and free energy decomposition analysis. The predicted binding free energy of the wild-type complex is more favorable than those of three mutants, indicating that both single and double mutations are unfavorable for the Aurora A and TPX2 binding. It is also observed that the mutations alternate the binding pattern between Aurora A and TPX2, especially the downstream of TPX2. An intramolecular hydrogen bond between the atom OD of Asp11(TPX2) and the atom HE1 of Trp34(TPX2) disappear in three mutants and thus lead to the instability of the secondary structure of TPX2. The combination of different molecular modeling techniques is an efficient way to understand how mutation has impacts on the protein-protein binding and our work gives valuable information for the future design of specific peptide inhibitors for Aurora A.


Assuntos
Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/metabolismo , Proteínas Associadas aos Microtúbulos/química , Proteínas Associadas aos Microtúbulos/metabolismo , Simulação de Dinâmica Molecular , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Fosfoproteínas/química , Fosfoproteínas/metabolismo , Proteínas Serina-Treonina Quinases/química , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas de Xenopus/química , Proteínas de Xenopus/metabolismo , Aurora Quinases , Mutação , Peptídeos/farmacologia , Ligação Proteica , Conformação Proteica , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/genética , Estabilidade Proteica , Especificidade por Substrato , Termodinâmica
14.
J Comput Aided Mol Des ; 25(2): 171-80, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21222017

RESUMO

Aurora kinase family is one of the emerging targets in oncology drug discovery and several small molecules targeting aurora kinases have been discovered and evaluated under early phase I/II trials. Among them, PHA-739358 (compound 1r) is a 3-aminopyrazole derivative with strong activity against Aurora A under early phase II trial. Inhibitory potency of compound 1r (the benzylic substituent at the pro-R position) is 30 times over that of compound 1s (the benzylic substituent at the pro-S position). In present study, the mechanism of how different configurations influence the binding affinity was investigated using molecular dynamics (MD) simulations, free energy calculations and free energy decomposition analysis. The predicted binding free energies of these two complexes are consistent with the experimental data. The analysis of the individual energy terms indicates that although the van der Waals contribution is important for distinguishing the binding affinities of these two inhibitors, the electrostatic contribution plays a more crucial role in that. Moreover, it is observed that different configurations of the benzylic substituent could form different binding patterns with protein, thus leading to variant inhibitory potency of compounds 1r and 1s. The combination of different molecular modeling techniques is an efficient way to interpret the chirality effects of inhibitors and our work gives valuable information for the chiral drug design in the near future.


Assuntos
Benzamidas/química , Simulação de Dinâmica Molecular , Inibidores de Proteínas Quinases/química , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/química , Pirazóis/química , Aurora Quinases , Desenho de Fármacos , Ligação de Hidrogênio , Ligação Proteica , Conformação Proteica , Mapeamento de Interação de Proteínas , Estereoisomerismo , Relação Estrutura-Atividade
15.
J Mol Model ; 17(4): 795-803, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20559856

RESUMO

Glycogen synthase kinase 3 (GSK-3) is an attractive target for the treatment of diabetes, and paullones have been reported to be effective inhibitors of GSK-3. However, it is still a challenging task to improve selectivity among protein kinases, especially cyclin-dependent kinases (CDKs). Here we investigated the mechanism that enables paullones to selectively inhibit GSK-3 rather than cyclin-dependent kinase 5 (CDK5) using sequence alignment, molecular dynamics simulations, free-energy calculations and free-energy decomposition analysis. The results indicate that the interaction between paullones and Val135 of GSK-3 is obviously stronger than that between paullones and Cys83 of CDK5, suggesting that paullones could be utilized as potent selective inhibitors. Meanwhile, we observed that the decrease in the interaction between paullones and the Asp86 of CDK5 favors their selectivity towards GSK-3 rather than CDK5, as demonstrated using 1-azakenpaullone as an example. Although substitution at position 9 and replacement at position 2 may influence the activity of GSK-3, they only have a minor effect on the selectivity. We expect that the information obtained here could prove useful for developing specific paullone inhibitors of GSK-3.


Assuntos
Benzazepinas/química , Benzazepinas/farmacologia , Quinase 5 Dependente de Ciclina/química , Inibidores Enzimáticos/química , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Quinase 3 da Glicogênio Sintase/química , Simulação de Dinâmica Molecular , Sequência de Aminoácidos , Quinase 5 Dependente de Ciclina/metabolismo , Inibidores Enzimáticos/farmacologia , Ligação de Hidrogênio , Modelos Químicos , Dados de Sequência Molecular , Ligação Proteica/efeitos dos fármacos , Alinhamento de Sequência
16.
Free Radic Biol Med ; 49(12): 2010-8, 2010 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-20951799

RESUMO

Thioredoxin (Trx) and thioredoxin reductase (TrxR) function as antioxidant and anti-apoptotic proteins, which are often up-regulated in drug-resistant cancer cells. (-)-epigallocatechin-3-gallate (EGCG) is a naturally occurring antioxidant in green tea, but also exhibits prooxidant and apoptosis-inducing properties. We have previously showed a linkage between EGCG-induced inactivation of TrxR and decreased cell survival, revealing TrxR as a new target of EGCG. However, the molecular events underlying the importance of Trx/TrxR in EGCG-induced cytotoxicity remain unclear. Here, we show that the crosstalk between EGCG and Trx/TrxR occurred in a redox-dependent manner, and EGCG induced inactivation of Trx/TrxR in parallel with increased ROS levels in HeLa cells. Moreover, EGCG displayed great reactivity with Cys/Sec residues that have low pK(a) values. The structure of EGCG suggests that its quinone form would readily react with thiolate and selenolate nucleophiles. Using mass spectrometry, we have demonstrated the formation of EGCG-Trx1 (Cys(32)) and EGCG-TrxR (Cys/Sec) conjugates, confirming that EGCG quinone specifically conjugates with active-site Cys(32) in Trx or C-terminal Cys/Selenocysteine (Sec) couple in TrxR under conditions where Trx/TrxR are reduced. Non-reduced form of Trx/TrxR could escape from EGCG inhibition. These data reveal a potential mechanism for enhancing EGCG-induced cancer cell death by the NADPH-dependent reduction of Trx/TrxR.


Assuntos
Catequina/análogos & derivados , Espécies Reativas de Oxigênio/farmacologia , Tiorredoxina Dissulfeto Redutase/antagonistas & inibidores , Tiorredoxinas/metabolismo , Sequência de Aminoácidos , Animais , Domínio Catalítico , Catequina/farmacologia , Bovinos , Cisteína/metabolismo , Células HeLa , Humanos , Oxirredução , Ligação Proteica , Ratos , Proteínas Recombinantes/antagonistas & inibidores , Proteínas Recombinantes/metabolismo , Tiorredoxina Dissulfeto Redutase/metabolismo , Tiorredoxinas/química
17.
Bioorg Med Chem Lett ; 20(11): 3329-37, 2010 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-20452766

RESUMO

Hologram quantitative structure-activity relationships (HQSAR) analysis were conducted on two series of PTP1B inhibitors, 39 2-(oxalylamino) benzoic acid (OBA) analogues and 60 benzofuran and benzothiophene biphenyls (BBB) analogues. The optimal HQSAR model of the OBA analogue has q(2)=0.592 and r(2)=0.940, while the optimal HQSAR model for the BBB analogues shows q(2)=0.667 and r(2)=0.863. Two models were employed to predict the biological activities of two test sets. For OBA analogues, the optimal model was validated by an external test set of six compounds with satisfactory predictive r(2) value of 0.786. For BBB analogues, the optimal model shows satisfactory predictive r(2) value of 0.866 for an external test set of 10 compounds. The contribution maps derived from the optimal HQSAR models are consistent with the biological activities of the studied compounds. Two virtual combinatorial libraries were designed and screened by the optimal HQSAR models and potential candidates with high predictive biological activities were discovered. This work may provide valuable information for future design of more promising inhibitors for PTP1B.


Assuntos
Diabetes Mellitus Tipo 2/tratamento farmacológico , Inibidores Enzimáticos/farmacologia , Proteína Tirosina Fosfatase não Receptora Tipo 1/antagonistas & inibidores , Inibidores Enzimáticos/química , Inibidores Enzimáticos/uso terapêutico , Modelos Moleculares , Relação Quantitativa Estrutura-Atividade
18.
J Chem Inf Model ; 50(3): 380-7, 2010 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-20180532

RESUMO

Herein, we report a successful application of molecular modeling techniques to design two novel peptides with cytotoxicity on tumor cells. First, the interactions between the nuclear transport factor 2 (NTF2)-like domain of G3BP and the SH3 domain of RasGAP were studied by a well-designed protocol, which combines homology modeling, protein/protein docking, molecular dynamics simulations, molecular mechanics/generalized born surface area (MM/GBSA) free energy calculations, and MM/GBSA free energy decomposition analysis together. Then, based on the theoretical predictions, two novel peptides were designed and synthesized for biological assays, and they showed an obvious sensitizing effect on cis-platin. Furthermore, the designed peptides had no significant effects on normal cells, while cis-platin did. Our results demonstrate that it is feasible to use the peptides to enhance the efficacy of clinical drugs and to kill cancer cells selectively. We believe that our work should be very useful for finding new therapies for cancers.


Assuntos
Antineoplásicos/química , Antineoplásicos/farmacologia , Proteínas de Transporte/antagonistas & inibidores , Citotoxinas/química , Citotoxinas/farmacologia , Peptídeos/química , Peptídeos/farmacologia , Sequência de Aminoácidos , Proteínas de Transporte/química , Proteínas de Transporte/metabolismo , DNA Helicases , Células HeLa , Humanos , Simulação de Dinâmica Molecular , Dados de Sequência Molecular , Proteínas de Ligação a Poli-ADP-Ribose , RNA Helicases , Proteínas com Motivo de Reconhecimento de RNA , Termodinâmica , Domínios de Homologia de src/efeitos dos fármacos
19.
J Comput Aided Mol Des ; 23(10): 737-45, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19672560

RESUMO

4-(Phenylamino)-pyrrolo[2,1-f][1,2,4]triazines have been discovered as inhibitors of p38alpha. Experimental assays have proven that the configuration of alpha-Me-benzyl connected with amide at C6 is essential for the binding affinity. The S-configured inhibitor (11j) displays 80 times more potency than the R-configured one (11k). Here we investigated the mechanism how different configurations influence the binding affinity using molecular dynamics simulations, free energy calculations and free energy decomposition analysis. We found that the van der Waals interactions play the most important role in differentiating the activities between 11j and 11k with p38alpha. The difference of the van der Waals interactions is primarily determined by two residues, LEU108 and LEU167. Consequently stabilization of pyrrolo[2,1-f][1,2,4]triazine ring is important for the activities of inhibitors. Meanwhile we observed that the different configuration of the alpha-Me-benzyl group leads to the difference of binding between 11j and 11k. In conclusion, our work shows that it is feasible to analyze the chirality effect of inhibitors with different configurations by molecular dynamics simulations and free energy calculations, and provides useful information for drug design.


Assuntos
Simulação de Dinâmica Molecular , Pirróis/química , Triazinas/química , Proteínas Quinases p38 Ativadas por Mitógeno/química , Conformação Proteica , Estereoisomerismo , Difração de Raios X
20.
Biochimie ; 91(3): 434-44, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19059456

RESUMO

Black tea is recently reported to have anti-carcinogenic effects through pro-oxidant property, but the underlying mechanisms remain unclear. Mammalian cytosolic thioredoxin reductase (TrxR1) is well -known for its anti-oxidation activity. In this study, we found that black tea extract (BTE) and theaflavins (TFs), the major black tea polyphenols, inhibited the purified TrxR1 with IC(50) 44 microg/ml and 21+/-1 microg/ml, respectively. Kinetics of TFs exhibited a mixed type of competitive and non-competitive inhibition, with K(is) 4+/-1 microg/ml and K(ii) 26+/-5 microg/ml against coenzyme NADPH, and with K(is) 12+/-3 microg/ml and K(ii) 27+/-5 microg/ml against substrate DTNB. In addition, TFs inhibited TrxR1 in a time-dependent manner. In an equilibrium step, a reversible TrxR1-TFs complex (E*I) forms, which is followed by a slow irreversible first-order inactivation step. Rate constant of the inactivation was 0.7 min(-1), and dissociation constant of E*I was 51.9 microg/ml. Treatment of NADPH-reduced TrxR1 with TFs decreased 5-(Iodoacetamido) fluorescein incorporation, a fluorescent thiol-reactive reagent, suggesting that Sec/Cys residue(s) in the active site may be involved in the binding of TFs. The inhibitory capacity of TFs depends on their structure. Among the TFs tested, gallated forms had strong inhibitory effects. The interactions between TFs and TrxR1 were investigated by molecular docking, which revealed important features of the binding mechanism of theaflavins. An inhibitory effect of BTE on viability of HeLa cells was observed with IC(50) 29 microg/ml. At 33 microg/ml of BTE, TrxR1 activity in HeLa cells was decreased by 73% at 22 h after BTE treatment. TFs inhibited cell viability with IC(50) 10+/-4 microg/ml for HeLa cells and with IC(50) 20+/-5 microg/ml for EAhy926 cells. The cell susceptibility to TFs was inversely correlated to cellular levels of TrxR1. The inhibitory actions of TFs on TrxR1 may be an important mechanism of their anti-cancer properties.


Assuntos
Anticarcinógenos/farmacologia , Antioxidantes/metabolismo , Chá/metabolismo , Tiorredoxina Redutase 1/antagonistas & inibidores , Tiorredoxina Dissulfeto Redutase/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Citosol/química , Relação Dose-Resposta a Droga , Flavonoides/química , Fluoresceínas/metabolismo , Corantes Fluorescentes/metabolismo , Células HeLa , Humanos , Concentração Inibidora 50 , Cinética , Modelos Biológicos , Modelos Moleculares , Estrutura Molecular , NADPH Oxidases/metabolismo , Fenóis/química , Extratos Vegetais/farmacologia , Polifenóis , Tiorredoxina Redutase 1/isolamento & purificação , Tiorredoxina Dissulfeto Redutase/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA