Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Acta Pharmacol Sin ; 40(6): 850-858, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30796354

RESUMEN

Serine/threonine phosphatase (Stp1) is a member of the bacterial Mg2+- or Mn2+- dependent protein phosphatase/protein phosphatase 2C family, which is involved in the regulation of Staphylococcus aureus virulence. Aurintricarboxylic acid (ATA) is a known Stp1 inhibitor with an IC50 of 1.03 µM, but its inhibitory mechanism has not been elucidated in detail because the Stp1-ATA cocrystal structure has not been determined thus far. In this study, we performed 400 ns molecular dynamics (MD) simulations of the apo-Stp1 and Stp1-ATA complex models. During MD simulations, the flap subdomain of the Stp1-ATA complex experienced a clear conformational transition from an open state to a closed state, whereas the flap domain of apo-Stp1 changed from an open state to a semi-open state. In the Stp1-ATA complex model, the hydrogen bond (H-bond) between D137 and N142 disappeared, whereas critical H-bond interactions were formed between Q160 and H13, Q160/R161 and ATA, as well as N162 and D198. Finally, four residues (D137, N142, Q160, and R161) in Stp1 were mutated to alanine and the mutant enzymes were assessed using phosphate enzyme activity assays, which confirmed their important roles in maintaining Stp1 activity. This study indicated the inhibitory mechanism of ATA targeting Stp1 using MD simulations and sheds light on the future design of allosteric Stp1 inhibitors.


Asunto(s)
Ácido Aurintricarboxílico/metabolismo , Proteínas Bacterianas/antagonistas & inhibidores , Inhibidores Enzimáticos/metabolismo , Fosfoproteínas Fosfatasas/antagonistas & inhibidores , Staphylococcus aureus/enzimología , Secuencia de Aminoácidos , Ácido Aurintricarboxílico/química , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Dominio Catalítico , Inhibidores Enzimáticos/química , Enlace de Hidrógeno , Simulación de Dinámica Molecular , Mutación , Fosfoproteínas Fosfatasas/química , Fosfoproteínas Fosfatasas/genética , Fosfoproteínas Fosfatasas/metabolismo , Unión Proteica , Conformación Proteica , Alineación de Secuencia
2.
J Med Chem ; 62(3): 1677-1683, 2019 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-30562026

RESUMEN

We identify three submicromolar inhibitors with new chemical scaffolds for cystathionine γ-lyase (CSE) by a tandem-well-based high-throughput assay. NSC4056, the most potent inhibitor with an IC50 of 0.6 µM, which is also known as aurintricarboxylic acid, selectively binds to Arg and Tyr residues of CSE active site and preferably inhibits the CSE activity in cells rather than cystathionine ß-synthase (CBS), the other H2S-generating enzyme. Moreover, NSC4056 effectively rescues hypotension in hemorrhagic shock rats.


Asunto(s)
Ácido Aurintricarboxílico/farmacología , Cistationina gamma-Liasa/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Animales , Ácido Aurintricarboxílico/química , Ácido Aurintricarboxílico/metabolismo , Dominio Catalítico/efectos de los fármacos , Cistationina gamma-Liasa/química , Cistationina gamma-Liasa/metabolismo , Descubrimiento de Drogas , Inhibidores Enzimáticos/metabolismo , Células HEK293 , Humanos , Masculino , Ratones , Simulación del Acoplamiento Molecular , Estructura Molecular , Nitroquinolinas/farmacología , Unión Proteica , Células RAW 264.7 , Ratas Sprague-Dawley , Relación Estructura-Actividad
3.
SLAS Discov ; 23(10): 1083-1091, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-29958052

RESUMEN

CC-chemokine receptor 7 (CCR7) is a G protein-coupled receptor expressed on a variety of immune cells. CCR7 plays a critical role in the migration of lymphocytes into secondary lymphoid tissues. CCR7 expression, however, has been linked to numerous disease states. Due to its therapeutic relevance and absence of available CCR7 inhibitors, we undertook a high-throughput screen (HTS) to identify small-molecule antagonists of the receptor. Here, we describe a robust HTS approach using a commercially available ß-galactosidase enzyme fragment complementation system and confirmatory transwell chemotaxis assays. This work resulted in the identification of several compounds with activity against CCR7. The most potent of these was subsequently determined to be cosalane, a cholesterol derivative previously designed as a therapeutic for human immunodeficiency virus. Cosalane inhibited both human and murine CCR7 in response to both CCL19 and CCL21 agonists at physiologic concentrations. Furthermore, cosalane produced durable inhibition of the receptor following a cellular incubation period with subsequent washout. Overall, our work describes the development of an HTS-compatible assay, completion of a large HTS campaign, and demonstration for the first time that cosalane is a validated CCR7 antagonist. These efforts could pave the way for new approaches to address CCR7-associated disease processes.


Asunto(s)
Ácido Aurintricarboxílico/análogos & derivados , Ensayos Analíticos de Alto Rendimiento , Receptores CCR7/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Animales , Ácido Aurintricarboxílico/química , Ácido Aurintricarboxílico/farmacología , Línea Celular , Quimiotaxis/efectos de los fármacos , Diseño de Fármacos , Humanos , Ligandos , Ratones , Estructura Molecular , Receptores CCR7/química , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/química , Relación Estructura-Actividad , Linfocitos T/efectos de los fármacos , Linfocitos T/metabolismo
4.
Anticancer Res ; 37(9): 4799-4806, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28870898

RESUMEN

BACKGROUND: Rapidly-dividing cancer cells have higher requirement for iron compared to non-transformed cells, making iron chelating a potential anticancer strategy. In the present study we compared the anticancer activity of uncommon iron chelator aurintricarboxylic acid (ATA) with the known deferoxamine (DFO). MATERIALS AND METHODS: We investigated the impact of ATA and DFO on the viability and proliferation of MCF-7 cancer cells. Moreover we performed enzymatic activity assays and computational analysis of the ATA and DFO effects on pro-oncogenic phosphatases PTP1B and SHP2. RESULTS: ATA and DFO decrease the viability and proliferation of breast cancer cells, but only ATA considerably reduces the activity of PTP1B and SHP2 phosphatases. Our studies indicated that ATA strongly inactivates and binds in the PTP1B and SHP2 active site, interacting with arginine residue essential for enzyme activity. CONCLUSION: We confirmed that iron chelating can be considered as a potential strategy for the adjunctive treatment of breast cancer.


Asunto(s)
Ácido Aurintricarboxílico/farmacología , Neoplasias de la Mama/enzimología , Deferoxamina/farmacología , Quelantes del Hierro/farmacología , Proteína Tirosina Fosfatasa no Receptora Tipo 11/antagonistas & inhibidores , Proteína Tirosina Fosfatasa no Receptora Tipo 1/antagonistas & inhibidores , Ácido Aurintricarboxílico/química , Sitios de Unión , Neoplasias de la Mama/patología , Catalasa/metabolismo , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Deferoxamina/química , Femenino , Humanos , Concentración 50 Inhibidora , Células MCF-7 , Simulación del Acoplamiento Molecular , Proteína Tirosina Fosfatasa no Receptora Tipo 1/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo
5.
Oncotarget ; 8(7): 12234-12246, 2017 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-28103571

RESUMEN

The survival of patients diagnosed with glioblastoma (GBM), the most deadly form of brain cancer, is compromised by the proclivity for local invasion into the surrounding normal brain, which prevents complete surgical resection and contributes to therapeutic resistance. Tumor necrosis factor-like weak inducer of apoptosis (TWEAK), a member of the tumor necrosis factor (TNF) superfamily, can stimulate glioma cell invasion and survival via binding to fibroblast growth factor-inducible 14 (Fn14) and subsequent activation of the transcription factor NF-κB. To discover small molecule inhibitors that disrupt the TWEAK-Fn14 signaling axis, we utilized a cell-based drug-screening assay using HEK293 cells engineered to express both Fn14 and a NF-κB-driven firefly luciferase reporter protein. Focusing on the LOPAC1280 library of 1280 pharmacologically active compounds, we identified aurintricarboxylic acid (ATA) as an agent that suppressed TWEAK-Fn14-NF-κB dependent signaling, but not TNFα-TNFR-NF-κB driven signaling. We demonstrated that ATA repressed TWEAK-induced glioma cell chemotactic migration and invasion via inhibition of Rac1 activation but had no effect on cell viability or Fn14 expression. In addition, ATA treatment enhanced glioma cell sensitivity to both the chemotherapeutic agent temozolomide (TMZ) and radiation-induced cell death. In summary, this work reports a repurposed use of a small molecule inhibitor that targets the TWEAK-Fn14 signaling axis, which could potentially be developed as a new therapeutic agent for treatment of GBM patients.


Asunto(s)
Ácido Aurintricarboxílico/farmacología , Neoplasias Encefálicas/tratamiento farmacológico , Glioblastoma/tratamiento farmacológico , Receptores del Factor de Necrosis Tumoral/metabolismo , Transducción de Señal/efectos de los fármacos , Factores de Necrosis Tumoral/metabolismo , Animales , Antineoplásicos Alquilantes/farmacología , Ácido Aurintricarboxílico/química , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Supervivencia Celular/efectos de la radiación , Citocina TWEAK , Dacarbazina/análogos & derivados , Dacarbazina/farmacología , Sinergismo Farmacológico , Glioblastoma/genética , Glioblastoma/metabolismo , Células HEK293 , Humanos , Estimación de Kaplan-Meier , Ratones Desnudos , Estructura Molecular , Interferencia de ARN , Receptores del Factor de Necrosis Tumoral/genética , Transducción de Señal/genética , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología , Receptor de TWEAK , Temozolomida , Factores de Necrosis Tumoral/genética , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
6.
World J Microbiol Biotechnol ; 32(10): 163, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27562597

RESUMEN

Yersinia sp. bacteria owe their viability and pathogenic virulence to the YopH factor, which is a highly active bacterial protein tyrosine phosphatase. Inhibition of YopH phosphatase results in the lack of Yersinia sp. pathogenicity. We have previously described that aurintricarboxylic acid inhibits the activity of YopH at nanomolar concentrations and represents a unique mechanism of YopH inactivation due to a redox process. This work is a continuation of our previous studies. Here we show that modifications of the structure of aurintricarboxylic acid reduce the ability to inactivate YopH and lead to higher cytotoxicity. In the present paper we examine the inhibitory properties of aurintricarboxylic acid analogues, such as eriochrome cyanine R (ECR) and pararosaniline. Computational docking studies we report here indicate that ATA analogues are not precluded to bind in the YopH active site and in all obtained binding conformations ECR and pararosaniline bind to YopH active site. The free binding energy calculations show that ECR has a stronger binding affinity to YopH than pararosaniline, which was confirmed by experimental YopH enzymatic activity studies. We found that ATA analogues can reversibly reduce the enzymatic activity of YopH, but possess weaker inhibitory properties than ATA. The ATA analogues induced inactivation of YopH is probably due to oxidative mechanism, as pretreatment with catalase prevents from inhibition. We also found that ATA analogues significantly decrease the viability of macrophage cells, especially pararosaniline, while ATA reveals only slight effect on cell viability.


Asunto(s)
Ácido Aurintricarboxílico/análogos & derivados , Proteínas de la Membrana Bacteriana Externa/química , Bencenosulfonatos/química , Proteínas Tirosina Fosfatasas/química , Colorantes de Rosanilina/química , Toluidinas/química , Yersinia/efectos de los fármacos , Animales , Ácido Aurintricarboxílico/química , Ácido Aurintricarboxílico/farmacología , Proteínas de la Membrana Bacteriana Externa/antagonistas & inhibidores , Bencenosulfonatos/farmacología , Dominio Catalítico/efectos de los fármacos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Ratones , Modelos Moleculares , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Estructura Molecular , Unión Proteica , Proteínas Tirosina Fosfatasas/antagonistas & inhibidores , Colorantes de Rosanilina/farmacología , Toluidinas/farmacología , Yersinia/enzimología
7.
Cell Chem Biol ; 23(8): 1002-13, 2016 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-27499528

RESUMEN

The increasing threats of antibiotic resistance urge the need for developing new strategies against bacterial infections. Targeting eukaryotic-like Ser/Thr phosphatase Stp1-mediated virulence regulation represents a promising approach for combating staphylococcal infection yet to be explored. Here, we report the 2.32-Å resolution crystal structure of Stp1. Stp1 binds an unexpected fourth metal ion, which is important for Stp1's enzymatic activity as demonstrated by amino acid substitution studies. Inspired by the structural details of Stp1, we identified a potent and selective Stp1 inhibitor, aurintricarboxylic acid (ATA). Transcriptome analysis and biochemical studies supported Stp1 as the target of ATA inhibition within the pathogen, preventing upregulation of virulence genes. Notably, ATA did not affect in vitro growth of Staphylococcus aureus, while simultaneously attenuating staphylococcal virulence in mice. Our findings demonstrate that ATA is a potent anti-virulence compound against staphylococcal infection, laying the foundation for further developing new scaffolds for Stp1-targeted small molecules.


Asunto(s)
Ácido Aurintricarboxílico/farmacología , Monoéster Fosfórico Hidrolasas/antagonistas & inhibidores , Infecciones Estafilocócicas/tratamiento farmacológico , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/efectos de los fármacos , Staphylococcus aureus/patogenicidad , Animales , Ácido Aurintricarboxílico/química , Relación Dosis-Respuesta a Droga , Femenino , Ratones , Ratones Endogámicos BALB C , Modelos Moleculares , Estructura Molecular , Monoéster Fosfórico Hidrolasas/metabolismo , Staphylococcus aureus/enzimología , Relación Estructura-Actividad , Virulencia/efectos de los fármacos
8.
Oncotarget ; 6(21): 18364-73, 2015 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-26286963

RESUMEN

YopH is a bacterial protein tyrosine phosphatase, which is essential for the viability and pathogenic virulence of the plague-causing Yersinia sp. bacteria. Inactivation of YopH activity would lead to the loss of bacterial pathogenicity. We have studied the inhibitory properties of aurintricarboxylic acid (ATA) against YopH phosphatase and found that at nanomolar concentrations ATA reversibly decreases the activity of YopH. Computational docking studies indicated that in all binding poses ATA binds in the YopH active site. Molecular dynamics simulations showed that in the predicted binding pose, ATA binds to the essential Cys403 and Arg409 residues in the active site and has a stronger binding affinity than the natural substrate (pTyr). The cyclic voltammetry experiments suggest that ATA reacts remarkably strongly with molecular oxygen. Additionally, the electrochemical reduction of ATA in the presence of a negative potential from -2.0 to 2.5 V generates a current signal, which is observed for hydrogen peroxide. Here we showed that ATA indicates a unique mechanism of YopH inactivation due to a redox process. We proposed that the potent inhibitory properties of ATA are a result of its strong binding in the YopH active site and in situ generation of hydrogen peroxide near catalytic cysteine residue.


Asunto(s)
Ácido Aurintricarboxílico/química , Proteínas de la Membrana Bacteriana Externa/química , Proteínas Tirosina Fosfatasas/química , Factores de Virulencia/química , Algoritmos , Ácido Aurintricarboxílico/metabolismo , Ácido Aurintricarboxílico/farmacología , Proteínas de la Membrana Bacteriana Externa/antagonistas & inhibidores , Proteínas de la Membrana Bacteriana Externa/metabolismo , Humanos , Cinética , Conformación Molecular , Simulación de Dinámica Molecular , Estructura Molecular , Oxidación-Reducción , Peste/microbiología , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Tirosina Fosfatasas/antagonistas & inhibidores , Proteínas Tirosina Fosfatasas/metabolismo , Virulencia , Factores de Virulencia/antagonistas & inhibidores , Factores de Virulencia/metabolismo , Yersinia pestis/metabolismo , Yersinia pestis/patogenicidad , Yersinia pestis/fisiología
9.
J Biol Chem ; 290(3): 1623-38, 2015 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-25477506

RESUMEN

Pharmacological induction of proteotoxic stress is rapidly emerging as a promising strategy for cancer cell-directed chemotherapeutic intervention. Here, we describe the identification of a novel drug-like heat shock response inducer for the therapeutic induction of proteotoxic stress targeting malignant human melanoma cells. Screening a focused library of compounds containing redox-directed electrophilic pharmacophores employing the Stress & Toxicity PathwayFinder(TM) PCR Array technology as a discovery tool, a drug-like triphenylmethane-derivative (aurin; 4-[bis(p-hydroxyphenyl)methylene]-2,5-cyclohexadien-1-one) was identified as an experimental cell stress modulator that causes (i) heat shock factor transcriptional activation, (ii) up-regulation of heat shock response gene expression (HSPA6, HSPA1A, DNAJB4, HMOX1), (iii) early unfolded protein response signaling (phospho-PERK, phospho-eIF2α, CHOP (CCAAT/enhancer-binding protein homologous protein)), (iv) proteasome impairment with increased protein-ubiquitination, and (v) oxidative stress with glutathione depletion. Fluorescence polarization-based experiments revealed that aurin displays activity as a geldanamycin-competitive Hsp90α-antagonist, a finding further substantiated by molecular docking and ATPase inhibition analysis. Aurin exposure caused caspase-dependent cell death in a panel of human malignant melanoma cells (A375, G361, LOX-IMVI) but not in non-malignant human skin cells (Hs27 fibroblasts, HaCaT keratinocytes, primary melanocytes) undergoing the aurin-induced heat shock response without impairment of viability. Aurin-induced melanoma cell apoptosis depends on Noxa up-regulation as confirmed by siRNA rescue experiments demonstrating that siPMAIP1-based target down-regulation suppresses aurin-induced cell death. Taken together, our data suggest feasibility of apoptotic elimination of malignant melanoma cells using the quinone methide-derived heat shock response inducer aurin.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Apoptosis , Ácido Aurintricarboxílico/análogos & derivados , Proteínas de Choque Térmico/metabolismo , Melanoma/tratamiento farmacológico , Neoplasias Cutáneas/tratamiento farmacológico , Proteínas Adaptadoras Transductoras de Señales , Ácido Aurintricarboxílico/química , Línea Celular Tumoral , Supervivencia Celular , Ensayos de Selección de Medicamentos Antitumorales , Citometría de Flujo , Glutatión/metabolismo , Respuesta al Choque Térmico/genética , Humanos , Indolquinonas/química , Concentración 50 Inhibidora , Queratinocitos/efectos de los fármacos , Melanocitos/efectos de los fármacos , Potencial de la Membrana Mitocondrial , Modelos Moleculares , Estrés Oxidativo , Reacción en Cadena de la Polimerasa , ARN Interferente Pequeño/metabolismo , Regulación hacia Arriba
10.
J Biomol Screen ; 19(7): 1060-9, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24608100

RESUMEN

Methylated DNA binding proteins such as Methyl-CpG Binding Domain Protein 2 (MBD2) can transduce DNA methylation alterations into a repressive signal by recruiting transcriptional co-repressor complexes. Interfering with MBD2 could lead to reactivation of tumor suppressor genes and therefore represents an attractive strategy for epigenetic therapy. We developed and compared fluorescence polarization (FP) and time-resolved fluorescence resonance energy transfer (TR-FRET)-based high-throughput screening (HTS) assays to identify small-molecule inhibitors of the interaction between the methyl binding domain of MBD2 (MBD2-MBD) and methylated DNA. Although both assays performed well in 96-well format, the TR-FRET assay (Z' factor = 0.58) emerged as a superior screening strategy compared with FP (Z' factor = 0.08) when evaluated in an HTS 384-well plate format. Using TR-FRET, we screened the Sigma LOPAC library for MBD2-MBD inhibitors and identified four compounds that also validated in a dose-response series. This included two known DNA intercalators (mitoxantrone and idarubicin) among two other inhibitory compounds (NF449 and aurintricarboxylic acid). All four compounds also inhibited the binding of SP-1, a transcription factor with a GC-rich binding sequence, to a methylated oligonucleotide, demonstrating that the activity was nonspecific. Our results provide proof of principle for using TR-FRET-based HTS to identify small-molecule inhibitors of MBD2 and other DNA-protein interactions.


Asunto(s)
Proteínas de Unión al ADN/antagonistas & inhibidores , Transferencia Resonante de Energía de Fluorescencia/métodos , Ácido Aurintricarboxílico/química , Bencenosulfonatos/química , ADN/química , Metilación de ADN , Relación Dosis-Respuesta a Droga , Descubrimiento de Drogas/métodos , Evaluación Preclínica de Medicamentos/métodos , Colorantes Fluorescentes/química , Ensayos Analíticos de Alto Rendimiento/métodos , Humanos , Idarrubicina/química , Mitoxantrona/química , Oligonucleótidos/química , Unión Proteica , Factor de Transcripción Sp1/química
11.
Adv Protein Chem Struct Biol ; 87: 219-47, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22607757

RESUMEN

DNA methyltransferases (DNMTs) are promising epigenetic targets for the development of novel anticancer drugs and other diseases. Molecular modeling and experimental approaches are being used to identify and develop inhibitors of human DNMTs. Most of the computational efforts conducted so far with DNMT1 employ homology models of the enzyme. Recently, a crystallographic structure of the methyltransferase domain of human DNMT1 bound to unmethylated DNA was published. Following on our previous computational and experimental studies with DNMTs, we herein present molecular dynamics of the crystal structure of human DNMT1. Docking studies of established DNMT1 inhibitors with the crystal structure gave rise to a structure-based pharmacophore model that suggests key interactions of the inhibitors with the catalytic binding site. Results had a good agreement with the docking and pharmacophore models previously developed using a homology model of the catalytic domain of DNMT1. The docking protocol was able to distinguish active DNMT1 inhibitors from, for example, experimentally known inactive DNMT1 inhibitors. As part of our efforts to identify novel inhibitors of DNMT1, we conducted the experimental characterization of aurintricarboxylic acid (ATA) that in preliminary docking studies showed promising activity. ATA had a submicromolar inhibition (IC(50)=0.68 µM) against DNMT1. ATA was also evaluated for Dnmt3a inhibition showing an IC(50)=1.4 µM. This chapter illustrates the synergy from integrating molecular modeling and experimental methods to further advance the discovery of novel candidates for epigenetic therapies.


Asunto(s)
Ácido Aurintricarboxílico/química , Ácido Aurintricarboxílico/farmacología , ADN (Citosina-5-)-Metiltransferasas/antagonistas & inhibidores , ADN (Citosina-5-)-Metiltransferasas/química , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Cristalografía por Rayos X , ADN (Citosina-5-)-Metiltransferasa 1 , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Humanos , Modelos Moleculares , Simulación de Dinámica Molecular , Relación Estructura-Actividad
12.
J Photochem Photobiol B ; 106: 113-9, 2012 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-22115956

RESUMEN

The present paper reports the investigations on the spectroscopic behavior of the binary complexes of the dye aurintricarboxylic acid (ATA) with protein bovine serum albumin (BSA) and 18-crown 6 (CW) (ATA·BSA, ATA·CW) and the ternary complex ATA·CW·BSA by using UV-vis steady state and time resolved fluorescence spectroscopy. The primary aim of the work is to determine the protein (BSA) quantization by fluorescence enhancement method and investigate the 'enhancer' activity of crown ether (CW) on it to increase the resolution. Steady state and time resolved fluorescence measurements demonstrated how fluorescence intensity of ATA could be used for the determination of the protein BSA in aqueous solution. The binding of dye (probe/fluorescent medicinal molecule) with protein and the denaturing effect in the polar environment of acetonitrile of the dye protein complex act as drug binding as well as drug release activity. Apart from its basic research point of view, the present study also possesses significant importance and applications in the field of medicinal chemistry.


Asunto(s)
Albúmina Sérica Bovina/análisis , Espectrofotometría Ultravioleta , Animales , Ácido Aurintricarboxílico/química , Ácido Aurintricarboxílico/metabolismo , Bovinos , Éteres Corona/química , Éteres Corona/metabolismo , Unión Proteica , Albúmina Sérica Bovina/metabolismo , Factores de Tiempo
13.
J Mol Model ; 18(4): 1583-9, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21805123

RESUMEN

DNA methyltransferase 1 (DNMT1) is an emerging target for the treatment of cancer, brain disorders, and other diseases. Currently, there are only a few DNMT1 inhibitors with potential application as therapeutic agents or research tools. 5,5-Methylenedisalicylic acid is a novel scaffold previously identified by virtual screening with detectable although weak inhibitory activity of DNMT1 in biochemical assays. Herein, we report enzyme inhibition of a structurally related compound, trimethylaurintricarboxylic acid (NSC97317) that showed a low micromolar inhibition of DNMT1 (IC(50) = 4.79 µM). Docking studies of the new inhibitor with the catalytic domain of DNMT1 suggest that NSC97317 can bind into the catalytic site. Interactions with amino acid residues that participate in the mechanism of DNA methylation contribute to the binding recognition. In addition, NSC97317 had a good match with a structure-based pharmacophore model recently developed for inhibitors of DNMT1. Trimethylaurintricarboxylic acid can be a valuable biochemical tool to study DNMT1 inhibition in cancer and other diseases related to DNA methylation.


Asunto(s)
Ácido Aurintricarboxílico/análogos & derivados , ADN (Citosina-5-)-Metiltransferasas/antagonistas & inhibidores , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN/efectos de los fármacos , Ácido Aurintricarboxílico/química , Ácido Aurintricarboxílico/metabolismo , Ácido Aurintricarboxílico/farmacología , Dominio Catalítico/efectos de los fármacos , ADN (Citosina-5-)-Metiltransferasa 1 , ADN (Citosina-5-)-Metiltransferasas/química , Diseño de Fármacos , Humanos , Modelos Moleculares , Simulación de Dinámica Molecular , Estructura Terciaria de Proteína
14.
Mini Rev Med Chem ; 10(10): 966-76, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20540707

RESUMEN

Cosalane and related compounds are a peculiar group of anti-HIV agents with activities against a broad range of viral targets, such as viral entry and reverse transcriptase (RT). Cosalane and its analogues having anionic pharmacophore inhibit the binding of gp120 to CD4 as well as the fusion of the viral envelope with the cell membrane. The alkenyldiarylmethanes (ADAMs), characterized by the lack of the steroidal moiety of cosalane, are a unique class of non-nucleoside reverse transcriptase inhibitors (NNRTIs) that have potential value in the treatment of HIV infection. In this article, the structural modifications, structure-activity relationship (SAR) studies and/or crystallographic studies of cosalane related derivatives as potent antiviral agents were reviewed, which will be beneficial to the discovery of next generation cosalane derivatives with improved antiviral potency, metabolic stability and bioavailability.


Asunto(s)
Fármacos Anti-VIH/química , Ácido Aurintricarboxílico/análogos & derivados , Inhibidores de la Transcriptasa Inversa/química , Fármacos Anti-VIH/farmacología , Ácido Aurintricarboxílico/química , Ácido Aurintricarboxílico/farmacología , Antígenos CD4/química , Antígenos CD4/metabolismo , Proteína gp120 de Envoltorio del VIH/química , Proteína gp120 de Envoltorio del VIH/metabolismo , Humanos , Inhibidores de la Transcriptasa Inversa/farmacología , Relación Estructura-Actividad
15.
Spectrochim Acta A Mol Biomol Spectrosc ; 74(5): 1145-51, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19836295

RESUMEN

In the investigation of interaction of aurintricarboxylic acid (ATA) with four biologically important proteins we observed inhibition of enzymatic activity of DNase I, RNase A, M-MLV reverse transcriptase and Taq polymerase by ATA in vitro assay. As the telomerase reverse transcriptase (TERT) is the main catalytic subunit of telomerase holoenzyme, we also monitored effect of ATA on telomerase activity in vivo and observed dose-dependent inhibition of telomerase activity in Chinese hamster V79 cells treated with ATA. Direct association of ATA with DNase I (K(d)=9.019 microM)), RNase A (K(d)=2.33 microM) reverse transcriptase (K(d)=0.255 microM) and Taq polymerase (K(d)=81.97 microM) was further shown by tryptophan fluorescence quenching studies. Such association altered the three-dimensional conformation of DNase I, RNase A and Taq polymerase as detected by circular dichroism. We propose ATA inhibits enzymatic activity of the four proteins through interfering with DNA or RNA binding to the respective proteins either competitively or allosterically, i.e. by perturbing three-dimensional structure of enzymes.


Asunto(s)
Ácido Aurintricarboxílico/metabolismo , Desoxirribonucleasa I/metabolismo , ADN Polimerasa Dirigida por ARN/metabolismo , Ribonucleasa Pancreática/metabolismo , Polimerasa Taq/metabolismo , Animales , Ácido Aurintricarboxílico/química , Ácido Aurintricarboxílico/farmacología , Línea Celular , Dicroismo Circular , Desoxirribonucleasa I/antagonistas & inhibidores , Cabras , Cinética , Reacción en Cadena de la Polimerasa , ARN/metabolismo , Espectrometría de Fluorescencia , Telomerasa/antagonistas & inhibidores , Temperatura
16.
Pharm Dev Technol ; 14(4): 341-2, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19235046

RESUMEN

Factors associated with the intravaginal release of an anti-HIV agent from an alginate complex were considered. Among these is citrate associated with prostatic fluid. This study demonstrates that citrate, at a physiologically appropriate concentration, facilitates the release of an anti-HIV polymer from a calcium alginate complex. The release of the agent can be modified by the concentration of the calcium and alginate in the complex. These results suggest that seminal and prostatic fluid can be considered in the design of an intravaginal system for HIV chemoprophylaxis.


Asunto(s)
Alginatos/química , Fármacos Anti-VIH/química , Ácido Aurintricarboxílico/química , Citratos/química , Administración Intravaginal , Fármacos Anti-VIH/administración & dosificación , Ácido Aurintricarboxílico/administración & dosificación , Química Farmacéutica , Portadores de Fármacos/química , Ácido Glucurónico/química , Infecciones por VIH/prevención & control , Ácidos Hexurónicos/química , Citrato de Sodio , Solubilidad
17.
Genome Biol ; 9(3): R47, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18312689

RESUMEN

Here we describe a proof-of-concept experiment designed to explore the possibility of using gene expression-based high-throughput screening (GE-HTS) to find inhibitors of a signaling cascade, using platelet derived growth factor receptor (PDGFR) signaling as the example. The previously unrecognized ability of aurintricarboxylic acid to inhibit PDGFR signaling, discovered through a screen of 1,739 compounds, demonstrates the feasibility and generalizability of GE-HTS for the discovery of small molecule modulators of any signaling pathway of interest.


Asunto(s)
Ácido Aurintricarboxílico/farmacología , Expresión Génica/efectos de los fármacos , Inhibidores de Proteínas Quinasas/aislamiento & purificación , Inhibidores de Proteínas Quinasas/farmacología , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Ácido Aurintricarboxílico/química , Línea Celular Tumoral , Evaluación Preclínica de Medicamentos/métodos , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Inhibidores de Proteínas Quinasas/química , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/agonistas , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/agonistas , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Transducción de Señal/efectos de los fármacos , Relación Estructura-Actividad
18.
Mol Biol Cell ; 17(11): 4632-44, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16928960

RESUMEN

A cornerstone of the antiviral interferon response is phosphorylation of eukaryotic initiation factor (eIF)2alpha. This limits the availability of eIF2.GTP.Met-tRNA(i)(Met) ternary complexes, reduces formation of 43S preinitiation complexes, and blocks viral (and most cellular) mRNA translation. However, many viruses have developed counterstrategies that circumvent this cellular response. Herein, we characterize a novel class of translation initiation inhibitors that block ternary complex formation and prevent the assembly of 43S preinitiation complexes. We find that translation driven by the HCV IRES is refractory to inhibition by these compounds at concentrations that effectively block cap-dependent translation in vitro and in vivo. Analysis of initiation complexes formed on the HCV IRES in the presence of inhibitor indicates that eIF2alpha and Met-tRNA(i)(Met) are present, defining a tactic used by HCV to evade part of the antiviral interferon response.


Asunto(s)
Factor 2 Eucariótico de Iniciación/metabolismo , Guanosina Trifosfato/metabolismo , Hepacivirus/genética , Biosíntesis de Proteínas/genética , ARN de Transferencia de Metionina/metabolismo , Animales , Ácido Aurintricarboxílico/química , Ácido Aurintricarboxílico/farmacología , Hepacivirus/efectos de los fármacos , Ratones , Modelos Genéticos , Biosíntesis de Proteínas/efectos de los fármacos , Inhibidores de la Síntesis de la Proteína/química , Inhibidores de la Síntesis de la Proteína/farmacología , Secuencias Reguladoras de Ácidos Nucleicos/genética , Ribosomas/efectos de los fármacos , Ribosomas/metabolismo
19.
J Med Chem ; 49(12): 3595-601, 2006 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-16759101

RESUMEN

Previously, we have reported that aurintricarboxylic acid (ATA) is one of the most potent inhibitors of the DNA binding of transcription factor NF-kappaB. We now report the NF-kappaB-DNA binding inhibitory activity of ATA analogues. An electrophoretic mobility shift assay has shown that bromopyrogallol red (BPR) is the most effective inhibitor of NF-kappaB-DNA binding among the studied analogues. The molecular modeling studies showed that BPR makes a strong network of hydrogen bonds with the DNA-binding region of the p50 subunit of NF-kappaB and has electronegative potential on its peripheral surface. Because zinc has been reported to influence the DNA binding of NF-kappaB, the interaction of these analogues with zinc was studied. Chemical speciation and formation-constant studies showed that BPR forms the most stable 1:1 complex with zinc. BPR has also been found to be the most potent antioxidant among the studied analogues.


Asunto(s)
Antivirales/química , Ácido Aurintricarboxílico/análogos & derivados , Ácido Aurintricarboxílico/química , Quelantes/química , ADN/metabolismo , Modelos Moleculares , FN-kappa B/metabolismo , Compuestos Organometálicos/química , Zinc , ADN/química , Electricidad , Ensayo de Cambio de Movilidad Electroforética , Enlace de Hidrógeno , FN-kappa B/química , Subunidad p50 de NF-kappa B/química , Unión Proteica , Pirogalol/química , Relación Estructura-Actividad , Zinc/química
20.
Biopolymers ; 83(4): 327-36, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16609969

RESUMEN

The gastrointestinal tract poses a variety of morphological and physiological barriers to the expression of target genes. The aim of this study was to evaluate the stability of cationic polymer/pDNA nanoparticles toward salts and enzymes of the intestinal fluid. Within this study, a chitosan-enzyme inhibitor conjugate has been generated and characterized. Based on this conjugate, nanoparticles with pDNA were generated to enhance transfection rate in oral gene delivery. The enzyme inhibitor aurintricarboxylic acid (ATA) was covalently bound to chitosan to improve the enzymatic stability of nanoparticles formed with this polymer and pDNA. Chitosan-ATA/pDNA nanoparticles showed a size of 98.5 +/- 26 nm and a zeta potential of -13.26 +/- 0.24 mV (n = 3-4). Stability studies with salt solution, lysozyme, DNase, and freshly collected porcine intestinal fluid showed that chitosan-ATA/pDNA nanoparticles are significantly (p < 0.05) more stable than unmodified chitosan/pDNA nanoparticles. Apart from improved stability, chitosan-ATA/pDNA nanoparticles showed a 2.6-fold higher transfection rate than chitosan/pDNA nanoparticles in the Caco-2 cell line, thus creating a promising carrier for orally administered therapeutic genes.


Asunto(s)
Ácido Aurintricarboxílico/química , Quitosano/química , ADN/administración & dosificación , Enfermedades Intestinales/terapia , Plásmidos/administración & dosificación , Administración Oral , Células CACO-2 , ADN/química , ADN/farmacocinética , Desoxirribonucleasa I/metabolismo , Sistemas de Liberación de Medicamentos/métodos , Electroforesis en Gel de Agar/métodos , Expresión Génica/genética , Terapia Genética/métodos , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Absorción Intestinal , Enfermedades Intestinales/genética , Enfermedades Intestinales/metabolismo , Microscopía Electrónica de Transmisión/métodos , Estructura Molecular , Muramidasa/metabolismo , Nanoestructuras/química , Nanoestructuras/ultraestructura , Tamaño de la Partícula , Plásmidos/química , Plásmidos/farmacocinética , Transfección/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...