Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Heliyon ; 10(9): e30292, 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38711664

RESUMEN

Cancer is a broad classification of diseases that can affect any organ or body tissue due to aberrant cellular proliferation for unknown reasons. Many present chemotherapeutic drugs are highly toxic and have little selectivity. Additionally, they lead to the development of medication resistance. Therefore, developing tailored chemotherapeutic drugs with minimal side effects and good selectivity is crucial for cancer treatment. 2-(1H)-Quinazolinone is one of the vital scaffold and anticancer activity is one of the prominent biological activities of this class. Here we report the novel set of amide-enriched 2-(1H)-quinazolinone derivatives (7a-j) and their apoptotic activity with the help of MTT assay method against four human cancer cell lines: PC3 (prostate cancer), DU-145 (prostate cancer), A549 (lung cancer), and MCF7 (breast cancer). When compared to etoposide, every synthetic test compound (7a-j) exhibited moderate to excellent activity. The IC50 values of the new amide derivatives (7a-j) varied from 0.07 ± 0.0061 µM to 10.8 ± 0.69 µM. While the positive control, etoposide, exhibited 1.97 ± 0.45 µM to 3.08 ± 0.135 µM range. Among the novel amide derivatives (7a-j), in particular, 7i and 7j showed strong apoptotic activity against MCF7; 7h showed against PC3, and 7g showed against DU-145. Molecular docking studies of test compounds (7a-j) with the EGFR tyrosine kinase domain (PDB ID: 1M17) protein provided the significant docking scores for each test compound (7a-j) (-9.00 to -9.67 kcal/mol). Additionally, DFT investigations and MD simulations validated the predictions of molecular docking. According to the findings of the ADME analysis, oral absorption by humans is anticipated to be higher than 85 % for all test compounds.

2.
J Biomol Struct Dyn ; 41(21): 11394-11414, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36591700

RESUMEN

The dynamically active and inactive conformations of kinases play a crucial role in the activation of intracellular downstream signaling pathways. The all-atom molecular dynamics (MD) simulations at microsecond (µs) timescale and longer provide robust insights into the structural details of conformational alterations in kinases that contribute to their cellular metabolic activities and signaling pathways. Tyro3, Axl and Mer (TAM) receptor tyrosine kinases (RTKs) are overexpressed in several types of human cancers. Cabozantinib, a small molecule inhibitor constrains the activity of TAM kinases at nanomolar concentrations. The apo, complexes of ATP (active state) and cabozantinib (active and inactive states) with TAM RTKs were studied by 1 µs MD simulations followed by trajectory analyses. The dynamic mechanistic pathways intrinsic to the kinase activity and protein conformational landscape in the cabozantinib bound TAM kinases are revealed due to the alterations in the P-loop, α-helix and activation loop that result in breaking the regulatory (R) and catalytic (C) spines, while the active states of ATP bound TAM kinases are retained. The co-existence of dynamical states when bound to cabozantinib was observed and the long-lived kinetic transition states of distinct active and inactive structural models were deciphered from MD simulation trajectories that have not been revealed so far.Communicated by Ramaswamy H. Sarma.


Asunto(s)
Proteínas Tirosina Quinasas Receptoras , Transducción de Señal , Humanos , Proteínas Tirosina Quinasas Receptoras/química , Proteínas Tirosina Quinasas Receptoras/metabolismo , Fosforilación , Adenosina Trifosfato/metabolismo
3.
J Biomol Struct Dyn ; 41(9): 3741-3751, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-35333147

RESUMEN

The pandemic coronavirus disease (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has resulted in more than 5 million deaths globally. Currently there are no effective drugs available to treat COVID-19. The viral protease replication can be blocked by the inhibition of main protease that is encoded in polyprotein 1a and is therefore a potential protein target for drug discovery. We have carried out virtual screening of NCI natural compounds followed by molecular docking in order to identify hit molecules as probable SARS-CoV-2 main protease inhibitors. The molecular dynamics (MD) simulations of apo form in complex with N3, α-ketoamide and NCI natural products was used to validate the screened compounds. The MD simulations trajectories were analyzed using normal mode analysis and principal component analysis revealing dynamical nature of the protein. These findings aid in understanding the binding of natural products and molecular mechanisms of SARS-CoV-2 main protease inhibition.Communicated by Ramaswamy H. Sarma.


Asunto(s)
Productos Biológicos , COVID-19 , Humanos , Simulación del Acoplamiento Molecular , SARS-CoV-2 , Productos Biológicos/farmacología , Simulación de Dinámica Molecular , Péptido Hidrolasas , Inhibidores de Proteasas/farmacología
4.
Struct Chem ; 33(5): 1467-1487, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35811782

RESUMEN

COVID-19 disease caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV2) has resulted in tremendous loss of lives across the world and is continuing to do so. Extensive work is under progress to develop inhibitors which can prevent the disease by arresting the virus in its life cycle. One such way is by targeting the main protease of the virus which is crucial for the cleavage and conversion of polyproteins into functional units of polypeptides. In this endeavor, our effort was to identify hit molecule inhibitors for SARS-CoV2 main protease using fragment-based drug discovery (FBDD), based on the available crystal structure of chromene-based inhibitor (PDB_ID: 6M2N). The designed molecules were validated by molecular docking and molecular dynamics simulations. The stability of the complexes was further assessed by calculating their binding free energies, normal mode analysis, mechanical stiffness, and principal component analysis. Supplementary Information: The online version contains supplementary material available at 10.1007/s11224-022-01995-z.

5.
J Biomol Struct Dyn ; 40(23): 12642-12659, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34516356

RESUMEN

Congenital Zika virus syndrome has caused a public health emergency of international concern. So far, there are no drugs available to prevent or treat the infection caused by Zika virus. The Zika virus NS3 helicase is a potential protein target for drug discovery due to its vital role in viral genome replication. NS3 helicase unwinds the viral RNA to enable the reproduction of the viral genome by the NS5 protein. NS3 helicase has two crucial binding sites; the ATP binding site and the RNA binding site. We used molecular docking and molecular dynamics (MD) simulations to study the structural behavior of Zika virus NS3 helicase in its apo form and in the presence of ATP, single-stranded RNA, and both ATP-RNA to understand their potential implications in NS3 helicase activity. Further, we have carried out virtual screening of FDA approved drugs, followed by molecular docking to identify the ATP-competitive hit molecules as probable Zika virus NS3 helicase inhibitors. The MD simulations trajectories were analyzed using normal mode analysis and principal component analysis that reveals fluctuations in the R-loop. These findings aid in understanding the molecular mechanisms of the simultaneous binding of ATP and RNA, and guide the design and discovery of new inhibitors of the Zika virus NS3 helicase as a promising drug target to treat the Zika virus infection. Communicated by Ramaswamy H. Sarma.


Asunto(s)
Infección por el Virus Zika , Virus Zika , Humanos , Simulación de Dinámica Molecular , Simulación del Acoplamiento Molecular , Proteínas no Estructurales Virales/química , ARN Viral/química , Adenosina Trifosfato/metabolismo
6.
Chem Biol Drug Des ; 98(4): 522-538, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34148296

RESUMEN

PLK-4 kinase plays an essential role in the cell cycle from regulating centriole duplication till cytokinesis and is therefore an attractive drug target in cancers such as breast, lung, and central nervous system tumors. CFI-400945 is an efficient PLK-4 inhibitor and inhibits other non-PLK family proteins at nanomolar concentrations. We have compared PLK-4 with other kinases to understand its similarity based on multiple sequence alignments from protein sequences of primary structures, outer and buried residues, and compact active site conservation based on three-dimensional motifs. These in-depth studies provide information on known interface targets and design of more selective inhibitors to PLK-4. Further, pharmacophore features based on CFI-400945 bound to PLK-4 were used for searching library of compounds that were screened using deep learning methods to bind PLK-4. The shortlisted molecules were docked into PLK-4 active site and were validated using molecular docking and molecular dynamics simulations studies. MM-PBSA calculations revealed the stability of hit molecules and PLK-4 complexes in comparison with CFI-400945 and the contribution to binding from key active site residues.


Asunto(s)
Indazoles/química , Indoles/química , Inhibidores de Proteínas Quinasas/química , Proteínas Serina-Treonina Quinasas/química , Bibliotecas de Moléculas Pequeñas/química , Secuencia de Aminoácidos , Dominio Catalítico , Ciclo Celular , Citocinesis , Aprendizaje Profundo , Humanos , Modelos Moleculares , Unión Proteica , Conformación Proteica , Relación Estructura-Actividad
7.
J Biomol Struct Dyn ; 39(4): 1213-1235, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32070235

RESUMEN

Receptor tyrosine kinases (RTKs) are essential proteins in the regulation of cell signaling. Tyro3, Axl and Mer are members of TAM RTKs and are overexpressed in several cancer forms. Kinase inhibitors such as cabozantinib, foretinib are reported to inhibit TAM kinases at nanomolar concentrations. The atomistic details of structure and mechanism of functional regulation is required to understand their normal physiological process and when bound to an inhibitor. The docking of cabozantinib into the active state conformations of TAM kinases (crystal structure and computational models) revealed the best binding pose and the complex formation that is mediated through non-bonding interactions involving the hinge region residues. The alterations in the conformations and the regions of flexibility in apo and complexed TAM kinases as a course of time are studied using 250 ns molecular dynamics (MD) simulations. The post-MD trajectory analysis using Python libraries like ProDy, MDTraj and PyEMMA revealed encrypted protein dynamic motions in active kinetic metastable states. Comparison between Principal component analysis and Anisotropic mode analysis deciphered structural residue interactions and salt bridge contacts between apo and inhibitor bound TAM kinases. Various structural changes occurred in αC-helix and activation loop involving hydrogen bonding between residues from Lys-(ß3 sheet), Glu-(αC-helix) and Asp-(DFG-motif) resulting in higher RMSD. Mechanical stiffness plots revealed that similar regions in apo and cabozantinib bound Axl fluctuated during MD simulations whereas different regions in Tyro3 and Mer kinases. The residue interaction network plots revealed important salt bridges that lead to constrained domain motions in the TAM kinases.Communicated by Ramaswamy H. Sarma.


Asunto(s)
Piridinas , Proteínas Tirosina Quinasas Receptoras , Anilidas , Modelos Moleculares , Unión Proteica , Proteínas Tirosina Quinasas Receptoras/metabolismo
8.
Redox Biol ; 28: 101318, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31546169

RESUMEN

Aurora A kinase is a master mitotic regulator whose functions are controlled by several regulatory interactions and post-translational modifications. It is frequently dysregulated in cancer, making Aurora A inhibition a very attractive antitumor target. However, recently uncovered links between Aurora A, cellular metabolism and redox regulation are not well understood. In this study, we report a novel mechanism of Aurora A regulation in the cellular response to oxidative stress through CoAlation. A combination of biochemical, biophysical, crystallographic and cell biology approaches revealed a new and, to our knowledge, unique mode of Aurora A inhibition by CoA, involving selective binding of the ADP moiety of CoA to the ATP binding pocket and covalent modification of Cys290 in the activation loop by the thiol group of the pantetheine tail. We provide evidence that covalent CoA modification (CoAlation) of Aurora A is specific, and that it can be induced by oxidative stress in human cells. Oxidising agents, such as diamide, hydrogen peroxide and menadione were found to induce Thr 288 phosphorylation and DTT-dependent dimerization of Aurora A. Moreover, microinjection of CoA into fertilized mouse embryos disrupts bipolar spindle formation and the alignment of chromosomes, consistent with Aurora A inhibition. Altogether, our data reveal CoA as a new, rather selective, inhibitor of Aurora A, which locks this kinase in an inactive state via a "dual anchor" mechanism of inhibition that might also operate in cellular response to oxidative stress. Finally and most importantly, we believe that these novel findings provide a new rationale for developing effective and irreversible inhibitors of Aurora A, and perhaps other protein kinases containing appropriately conserved Cys residues.


Asunto(s)
Aurora Quinasa A/química , Aurora Quinasa A/metabolismo , Coenzima A/administración & dosificación , Animales , Coenzima A/química , Coenzima A/farmacología , Cristalografía por Rayos X , Células HEK293 , Células Hep G2 , Humanos , Ratones , Modelos Moleculares , Estrés Oxidativo , Fosforilación , Conformación Proteica , Huso Acromático/efectos de los fármacos , Huso Acromático/metabolismo
9.
J Biosci ; 44(2)2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31180048

RESUMEN

A Kunitz-type protease inhibitor (OPI, okra protease inhibitor) has been purified from okra (Abelmoschus esculentus) seeds by a combination of ammonium sulfate precipitation, anion-exchange chromatography and reverse-phase high-performance liquid chromatography. The protein shows an apparent mass of 21 kDa on sodium dodecyl sulfate-polyacrylamide gel electrophoresis under reducing condition. OPI exhibits inhibitory activity against trypsin. Analysis of the far-UV circular dichroism spectrum showed that the protein contains approx. 39% beta-sheets but only approx. 5% alpha-helices. The protein is thermally quite stable, and exhibits a cooperative thermal unfolding transition at approx. 70 degree C, as determined by circular dichroism spectroscopy and differential scanning fluorimetry. De novo sequencing of OPI by nanoESI-Q-ToF mass spectrometry (MS) allowed the assignment of about 83% of its primary structure, which indicated that the protein shares 43% sequence identity with a putative 21 kDa trypsin inhibitor from Theobroma bicolor. An intramolecular disulfide linkage between Cys149 and Cys156 was also detected. The protein showed approx 24 and approx 25% sequence identity with alpha-amylase/subtilisin inhibitor from barley and soybean (Kunitz) trypsin inhibitor, respectively. Comparative structure modeling of OPI revealed a structural fold similar to other Kunitz-type TIs. The presence of Cys149-Cys156 disulfide bond as detected by MS and a second disulfide bond connecting Cys44-Cys91, conserved in all Kunitz-type TIs, is also identified in the model.


Asunto(s)
Abelmoschus/química , Péptidos/química , Proteínas de Plantas/química , Semillas/química , Tripsina/química , Abelmoschus/metabolismo , Secuencia de Aminoácidos , Sulfato de Amonio/química , Sitios de Unión , Cromatografía/métodos , Electroforesis en Gel de Gradiente Desnaturalizante , Modelos Moleculares , Peso Molecular , Péptidos/aislamiento & purificación , Proteínas de Plantas/aislamiento & purificación , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Semillas/metabolismo , Alineación de Secuencia , Homología Estructural de Proteína , Termodinámica
10.
J Biomol Struct Dyn ; 37(13): 3410-3421, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30146942

RESUMEN

Polo box domain (PBD) from Polo-Like Kinase-1 (PLK-1) a cell cycle regulator is one of the important non-kinase targets implicated in various cancers. The crystal structure of PLK-1 PBD bound to phosphopeptide inhibitor is available and acylthiourea derivatives have been reported as potent PBD inhibitors. In this work, structure and ligand-based pharmacophore methods have been used to identify new PBD inhibitors. The binding of acylthiourea analogs and new inhibitors to PBD were assessed using molecular docking and molecular dynamics simulations to understand their binding interactions, investigate the complex stability and reveal the molecular basis for inhibition. This study provides the binding free energies and residue-wise contributions to decipher the essential interactions in the protein-inhibitor complementarity for complex formation and the design of new PBD inhibitors with better binding. Communicated by Ramaswamy H. Sarma.


Asunto(s)
Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/metabolismo , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/química , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/química , Proteínas Proto-Oncogénicas/metabolismo , Sitios de Unión , Ensayos Analíticos de Alto Rendimiento , Humanos , Modelos Moleculares , Conformación Proteica , Dominios Proteicos , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/metabolismo , Quinasa Tipo Polo 1
11.
J Phys Chem B ; 121(2): 365-378, 2017 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-28054779

RESUMEN

Helicobacter pylori is a primitive Gram-negative bacterium that resides in the acidic environment of the human gastrointestinal tract, and some strains of this bacterium cause gastric ulcers and cancer. DNA methyltransferases (MTases) are promising drug targets for the treatment of cancer and other diseases that are also caused by epigenetic alternations of the genome. The N6-adenine-specific DNA MTase from H. pylori (M. Hpy N6mA) catalyzes the transfer of a methyl group from the cofactor S-adenosyl-l-methionine (AdoMet) to the flipped adenine of the substrate DNA. In this work, we report the sequence analyses, three-dimensional structure modeling, and molecular dynamics simulations of M. Hpy N6mA, when complexed with AdoMet as well as DNA. We analyzed the protein-DNA interactions prominently established by the flipped cytosine and the interactions between protein cofactors in the active site. The comparable orientation of AdoMet in both systems confirms that AdoMet is in a catalytically competent orientation in the bimolecular system that is retained upon DNA binding in the termolecular system of M. Hpy N6mA. In both systems, AdoMet is stabilized in the binding pocket by hydrogen bonding (Thr84, Glu99, Asp122, and Phe123) as well as van der Waals (Ile100, Phe160, Arg104, and Cys76) interactions. We propose that the contacts made by flipped adenine DA6 with Asn138 (N6 and N1 atom of DA6) and Pro139 (N6) and π-stacking interactions with Phe141 and Phe219 play an important role in the methylation mechanism at the N6 position in our N6mA model. Specific recognition of DNA is mediated by residues 143-155, 183-189, 212-220, 280-293, and 308-325. These findings are further supported by alanine scanning mutagenesis studies. The conserved residues in distantly related sequences of the small domain are important in DNA binding. Results reported here elucidate the sequence, structure, and binding features necessary for the recognition between cofactor AdoMet and substrate DNA by the vital epigenetic enzyme, M. Hpy N6mA.


Asunto(s)
ADN/metabolismo , S-Adenosilmetionina/metabolismo , Metiltransferasa de ADN de Sitio Específico (Adenina Especifica)/metabolismo , Secuencia de Aminoácidos , Secuencia de Bases , Dominio Catalítico , ADN/química , Helicobacter pylori , Enlace de Hidrógeno , Simulación de Dinámica Molecular , Estructura Molecular , Mutagénesis Sitio-Dirigida , Unión Proteica , S-Adenosilmetionina/química , Alineación de Secuencia , Metiltransferasa de ADN de Sitio Específico (Adenina Especifica)/química , Metiltransferasa de ADN de Sitio Específico (Adenina Especifica)/genética
12.
Mol Biosyst ; 12(10): 3111-23, 2016 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-27470658

RESUMEN

Helicobacter pylori is a Gram-negative bacterium that inhabits the human gastrointestinal tract, and some strains of this bacterium cause gastric ulcers and cancer. DNA methyltransferases (MTases) are promising drug targets for the treatment of cancer and other diseases that are also caused by epigenetic alternations of the genome. The C5-cytosine specific DNA methyltransferase from H. pylori (M. Hpy C5mC) catalyzes the transfer of the methyl group from the cofactor S-adenosyl-l-methionine (AdoMet) to the flipped cytosine of the substrate DNA. Herein we report the sequence analyses, 3-D structure modeling and molecular dynamics simulations of M. Hpy C5mC, when complexed with AdoMet as well as DNA. We analyzed the protein-DNA interactions prominently established by the flipped cytosine and the interactions between the protein and cofactor in the active site. We propose that the contacts made by cytosine O2 with Arg155 and Arg157, and the water-mediated interactions with cytosine N3 may be essential for the activity of methyl transfer as well as the deprotonation at the C5 position in our C5mC model. Specific recognition of DNA was mediated mainly by residues from Ser221-Arg229 and Ser243-Gln246 of the target recognition domain (TRD) and some residues of the loop Ser75-Lys83 from the large domain. These findings are further supported by alanine scanning mutagenesis studies. The results reported here explain the sequence, structure and binding features necessary for the recognition between the cofactor and the substrate by the key epigenetic enzyme, M. Hpy C5mC.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/química , ADN/química , Conformación Molecular , Simulación de Dinámica Molecular , S-Adenosilmetionina/química , Secuencia de Aminoácidos , Sitios de Unión , Dominio Catalítico , ADN/metabolismo , ADN (Citosina-5-)-Metiltransferasas/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN , Helicobacter pylori/enzimología , Enlace de Hidrógeno , Sustancias Macromoleculares/química , Sustancias Macromoleculares/metabolismo , Mutación , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , S-Adenosilmetionina/metabolismo , Relación Estructura-Actividad
13.
Biochem J ; 473(8): 1047-61, 2016 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-26920025

RESUMEN

Oestrogen receptor-α (ERα) is a ligand-dependent transcription factor that primarily mediates oestrogen (E2)-dependent gene transcription required for mammary gland development. Coregulators critically regulate ERα transcription functions by directly interacting with it. In the present study, we report that ELF3, an epithelial-specific ETS transcription factor, acts as a transcriptional repressor of ERα. Co-immunoprecipitation (Co-IP) analysis demonstrated that ELF3 strongly binds to ERα in the absence of E2, but ELF3 dissociation occurs upon E2 treatment in a dose- and time-dependent manner suggesting that E2 negatively influences such interaction. Domain mapping studies further revealed that the ETS (E-twenty six) domain of ELF3 interacts with the DNA binding domain of ERα. Accordingly, ELF3 inhibited ERα's DNA binding activity by preventing receptor dimerization, partly explaining the mechanism by which ELF3 represses ERα transcriptional activity. Ectopic expression of ELF3 decreases ERα transcriptional activity as demonstrated by oestrogen response elements (ERE)-luciferase reporter assay or by endogenous ERα target genes. Conversely ELF3 knockdown increases ERα transcriptional activity. Consistent with these results, ELF3 ectopic expression decreases E2-dependent MCF7 cell proliferation whereas ELF3 knockdown increases it. We also found that E2 induces ELF3 expression in MCF7 cells suggesting a negative feedback regulation of ERα signalling in breast cancer cells. A small peptide sequence of ELF3 derived through functional interaction between ERα and ELF3 could inhibit DNA binding activity of ERα and breast cancer cell growth. These findings demonstrate that ELF3 is a novel transcriptional repressor of ERα in breast cancer cells. Peptide interaction studies further represent a novel therapeutic option in breast cancer therapy.


Asunto(s)
Neoplasias de la Mama/metabolismo , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Receptor alfa de Estrógeno/química , Receptor alfa de Estrógeno/metabolismo , Proteínas Proto-Oncogénicas c-ets/química , Proteínas Proto-Oncogénicas c-ets/metabolismo , Factores de Transcripción/química , Factores de Transcripción/metabolismo , Transcripción Genética/fisiología , Secuencia de Aminoácidos , Neoplasias de la Mama/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/fisiología , Proteínas de Unión al ADN/genética , Relación Dosis-Respuesta a Droga , Receptor alfa de Estrógeno/genética , Femenino , Células HeLa , Humanos , Células MCF-7 , Datos de Secuencia Molecular , Estructura Secundaria de Proteína , Proteínas Proto-Oncogénicas c-ets/genética , Tamoxifeno/metabolismo , Tamoxifeno/farmacología , Factores de Transcripción/genética , Transcripción Genética/efectos de los fármacos
14.
J Biomol Struct Dyn ; 33(8): 1710-9, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25245460

RESUMEN

Human male germ cell-associated kinase (hMAK) is an androgen-inducible gene in prostate epithelial cells, and it acts as a coactivator of androgen receptor signaling in prostate cancer. The 3D structure of the hMAK kinase was modeled based on the crystal structure of CDK2 kinase using comparative modeling methods, and the ATP-binding site was characterized. We have collected five inhibitors of hMAK from the literature and docked into the ATP-binding site of the kinase domain. Solvated interaction energies (SIE) of inhibitor binding are calculated from the molecular dynamics simulations trajectories of protein-inhibitor complexes. The contribution from each active site residue in hMAK toward inhibitor binding revealed the nature and extent of interactions between inhibitors and individual residues. The main chain atoms of Met79 invariably form hydrogen bonds with all five inhibitors. The amino acids Leu7, Val15, and Leu129 stabilize the inhibitors via CH-pi interactions. The Asp140 in the active site and Glu77 in hinge region show characteristic hydrogen bonding interactions with inhibitors. From SIE, the residue-wise interactions revealed the nature of non-bonding contacts and modifications required to increase the inhibitor activity. Our work provides 3D model structure of hMAK and molecular basis for the mechanisms of hMAK inhibition at atomic level that aid in designing new potent inhibitors.


Asunto(s)
Modelos Moleculares , Conformación Molecular , Inhibidores de Proteínas Quinasas/química , Proteínas Quinasas/química , Sitios de Unión , Dominio Catalítico , Humanos , Masculino , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Unión Proteica , Inhibidores de Proteínas Quinasas/metabolismo , Proteínas Quinasas/metabolismo
15.
PLoS One ; 9(12): e115020, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25549250

RESUMEN

The availability of complete genome sequences of H. pylori 26695 has provided a wealth of information enabling us to carry out in silico studies to identify new molecular targets for pharmaceutical treatment. In order to construe the structural and functional information of complete proteome, use of computational methods are more relevant since these methods are reliable and provide a solution to the time consuming and expensive experimental methods. Out of 1590 predicted protein coding genes in H. pylori, experimentally determined structures are available for only 145 proteins in the PDB. In the absence of experimental structures, computational studies on the three dimensional (3D) structural organization would help in deciphering the protein fold, structure and active site. Functional annotation of each protein was carried out based on structural fold and binding site based ligand association. Most of these proteins are uncharacterized in this proteome and through our annotation pipeline we were able to annotate most of them. We could assign structural folds to 464 uncharacterized proteins from an initial list of 557 sequences. Of the 1195 known structural folds present in the SCOP database, 411 (34% of all known folds) are observed in the whole H. pylori 26695 proteome, with greater inclination for domains belonging to α/ß class (36.63%). Top folds include P-loop containing nucleoside triphosphate hydrolases (22.6%), TIM barrel (16.7%), transmembrane helix hairpin (16.05%), alpha-alpha superhelix (11.1%) and S-adenosyl-L-methionine-dependent methyltransferases (10.7%).


Asunto(s)
Proteínas Bacterianas/genética , Bases de Datos de Proteínas , Helicobacter pylori/genética , Anotación de Secuencia Molecular/métodos , Proteoma/genética , Estructura Terciaria de Proteína , Relación Estructura-Actividad
16.
PLoS One ; 8(11): e78556, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24236021

RESUMEN

BCR-ABL kinase domain inhibition can be used to treat chronic myeloid leukemia. The inhibitors such as imatinib, dasatinib and nilotinib are effective drugs but are resistant to some BCR-ABL mutations. The pan-BCR-ABL kinase inhibitor ponatinib exhibits potent activity against native, T315I, and all other clinically relevant mutants, and showed better inhibition than the previously known inhibitors. We have studied the molecular dynamics simulations and calculated solvated interaction energies of native and fourteen mutant BCR-ABL kinases (M244V, G250E, Q252H, Y253F, Y253H, E255K, E255V, T315A, T315I, F317L, F317V, M351T, F359V and H396P) complexed with ponatinib. These studies revealed that the interactions between ponatinib and individual residues in BCR-ABL kinase are also affected due to the remote residue mutations. We report that some residues, Met244, Lys245, Gln252, Gly254, Leu370 and Leu298 do not undergo any conformational changes, while the fluctuations in residues from P-loop, ß3-, ß5- strands and αC- helix are mainly responsible for ponatinib binding to native and all mutant BCR-ABL kinases. Our work provides the molecular mechanisms of native and mutant BCR-ABL kinases inhibition by ponatinib at atomic level that has not been studied before.


Asunto(s)
Antineoplásicos/química , Proteínas de Fusión bcr-abl/antagonistas & inhibidores , Imidazoles/química , Simulación de Dinámica Molecular , Piridazinas/química , Dominio Catalítico , Proteínas de Fusión bcr-abl/química , Proteínas de Fusión bcr-abl/genética , Humanos , Enlace de Hidrógeno , Mutación Missense , Unión Proteica , Estructura Secundaria de Proteína , Solventes/química , Termodinámica , Agua/química
17.
Proc Natl Acad Sci U S A ; 110(51): 20467-72, 2013 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-24282299

RESUMEN

Resistin, a cysteine-rich adipocytokine, proposed as a link between obesity and diabetes in mice, was shown as a proinflammatory molecule in humans. We earlier reported that human resistin (hRes), a trimer, was resistant to heat and urea denaturation, existed in an oligomeric polydispersed state, and showed a concentration-dependent conformational change. These properties and an intimate correlation of hRes expression with cellular stress prompted us to investigate hRes as a possible chaperone. Here, we show that recombinant human resistin was able to protect the heat-labile enzymes citrate synthase and Nde1 from thermal aggregation and inactivation and was able to refold and restore their enzymatic activities after heat/guanidinium chloride denaturation. Furthermore, recombinant human resistin could bind misfolded proteins only. Molecular dynamics-based association-dissociation kinetics of hRes subunits pointed to resistin being a molecular chaperone. Bis-ANS, which blocks surface hydrophobicity, abrogated the chaperone activity of hRes, establishing the importance of surface hydrophobicity for chaperone activity. Replacement of Phe49 with Tyr (F49YhRes), a critical residue within the hydrophobic patch of hRes, although it could prevent thermal aggregation of citrate synthase and Nde1, was unable to refold and restore their activities. Treatment of U937 cells with tunicamycin/thapsigargin resulted in reduced hRes secretion and concomitant localization in the endoplasmic reticulum. Escherichia coli transformants expressing hRes could be rescued from thermal stress, pointing to hRes's chaperone-like function in vivo. HeLa cells transfected with hRes showed protection from thapsigargin-induced apoptosis. In conclusion, hRes, an inflammatory protein, additionally exhibited chaperone-like properties, suggesting a possible link between inflammation and cellular stress.


Asunto(s)
Citocinas/metabolismo , Respuesta al Choque Térmico/fisiología , Mediadores de Inflamación/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Chaperonas Moleculares/metabolismo , Resistina/metabolismo , Animales , Antibacterianos/farmacología , Citocinas/genética , Inhibidores Enzimáticos/farmacología , Células HeLa , Respuesta al Choque Térmico/efectos de los fármacos , Humanos , Inflamación/genética , Inflamación/metabolismo , Ratones , Proteínas Asociadas a Microtúbulos/genética , Chaperonas Moleculares/genética , Resistina/genética , Tapsigargina/farmacología , Tunicamicina/farmacología , Células U937
18.
Phytochemistry ; 96: 26-36, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24094275

RESUMEN

Kunitz trypsin inhibitor was purified from the seeds of Trigonella foenum-graecum (TfgKTI) belonging to fabaceae family by ammonium sulphate precipitation, cation exchange, gel filtration and hydrophobic chromatography. Purity of the protein was analyzed by RP-HPLC and native-PAGE. SDS-PAGE analysis under reducing and non-reducing conditions showed that protein consists of a single polypeptide chain with molecular mass of approximately 20 kDa. Mass spectroscopy analysis revealed that the intact mass of purified inhibitor is 19,842.154 Da. One dimensional SDS gel was tryptically digested, resulting peptides were subjected to MALDI-TOF-MS analysis, and peptide mass fingerprinting (PMF) analysis of TfgKTI shows sequence similarity with Kunitz trypsin inhibitor in database search. Two dimensional electrophoresis identified presence of four isoinhibitors (pI values of 5.1, 5.4, 5.7 and 6.1). Kinetic studies showed that the protein is a competitive inhibitor and has high binding affinity with trypsin (Ki 3.01×10(-9)M) and chymotrypsin (Ki 0.52×10(-9)M). The TfgKTI retained the inhibitory activity over a broad range of pH (pH 3-10), temperature (37-100°C) and salt concentration (up to 3.5%). Far-UV circular dichroism measurements revealed that TfgKTI is predominantly composed of ß-sheets (39%) and unordered structures (48%) with slight helical content (13%). TfgKTI retained over 90% trypsin inhibition upon storage at 4°C for over a period of six months.


Asunto(s)
Aprotinina/aislamiento & purificación , Aprotinina/farmacología , Fabaceae/química , Aprotinina/química , Quimotripsina/análisis , Dicroismo Circular , Electroforesis en Gel de Poliacrilamida , Peso Molecular , Semillas/química , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Tripsina/análisis
19.
Protein Pept Lett ; 20(11): 1246-56, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23848594

RESUMEN

Aurora-A, B and C are non-receptor serine/threonine kinases in Homo sapiens. In spite of high similarity in their sequences, they possess distinct binding partners. These kinases play an important role in cell division and overexpressed in certain cancers. It has been demonstrated that Gly198 in Aurora-A kinase is responsible for its basal kinase activity, the mutation G198N transforms Aurora-A to Aurora-B like function and localization by binding to Inner centromere protein (INCENP). The molecular mechanisms, structural determinants and the binding energetics of the Aurora-A - INCENP complex owing to a single amino acid G198N mutation are not studied. Therefore, we have docked INCENP into human Aurora-A kinase, mutated Gly198 to Asn, Leu and Ala. The wild type and mutant Aurora-A - INCENP complexes were subjected to 40 ns molecular dynamics (MD) simulations. The Asn198 is located in the amphipathic cavity comprising Leu869(IN), Glu868(IN), Thr872(IN), Tyr197(AurA) and Tyr199(AurA) and the interactions mediated via hydrogen bonds are important to stabilize the Aurora-A(G198N) - INCENP complex. The fluctuations in the secondary structural elements and the solvent accessible surface area of all the four complexes during the MD simulations were studied. We calculated the binding free energy upon mutation in the three mutant complexes. The Aurora-A(G198N) - INCENP complex with hydrophilic amino acid mutation has the negative free energy of solvation indicating favorable interactions with INCENP. Our results provide the structural basis and energetics of the human Aurora-A(G198N) - INCENP complex.


Asunto(s)
Aurora Quinasa A/química , Proteínas Cromosómicas no Histona/química , Mitosis , Estructura Terciaria de Proteína , Secuencia de Aminoácidos , Aurora Quinasa A/genética , Línea Celular , Proteínas Cromosómicas no Histona/genética , Metabolismo Energético , Humanos , Enlace de Hidrógeno , Modelos Moleculares , Simulación de Dinámica Molecular , Mutación , Fosforilación , Unión Proteica , Homología de Secuencia de Aminoácido
20.
Chem Biol Drug Des ; 79(1): 84-91, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21967481

RESUMEN

MEK-1 and MEK-2 are dual-specificity kinases and important components in the mitogen-activated protein kinase pathway. These enzymes are crucial for normal cell survival and are also expressed in several types of cancers, making them important targets for drug design. We have applied an integrated in silico approach that combines comparative molecular field analysis, comparative molecular similarity indices analysis, and molecular docking to study the structural determinants for the recognition of substituted isothiazole analogs as allosteric inhibitors against MEK-1 kinase. The best 3D-QSAR models for comparative molecular field analysis and comparative molecular similarity indices analysis were selected based on statistical parameters. 3D contour maps suggested that bulky or long-chain substitutions at the X position on the core part decrease the inhibitory activity, and the presence of a hydrogen bond donor substitution enhances the activity. The bulky and electronegative substitutions at the Y position on the core part enhance the activity of the inhibitors. Molecular docking studies reveal a large and hydrophobic pocket that accommodates the Y substitution and a polar pocket that accommodates substitutions on the X position and forms hydrogen bonding interactions with MEK-1 kinase. The results of the 3D-QSAR analysis corroborate with the molecular docking results, and our findings will serve as a basis for further development of better allosteric inhibitors of MEK-1 kinase against several cancers.


Asunto(s)
Quinasa 1 de Quinasa de Quinasa MAP/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/química , Relación Estructura-Actividad Cuantitativa , Tiazoles/química , Regulación Alostérica , Simulación por Computador , Activación Enzimática/efectos de los fármacos , Quinasa 1 de Quinasa de Quinasa MAP/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Estructura Terciaria de Proteína , Programas Informáticos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA