Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Mol Cancer Ther ; 18(10): 1765-1774, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31341033

RESUMEN

The preclinical antitumor agent RITA (2,5-bis[5-hydroxymethyl-2-thienyl] furan, NSC 652287), an analog of the natural product α-terthiophene, failed during the development phase due to acute pulmonary toxicity in animal models. A series of synthetic modifications to RITA's heterocyclic scaffold resulted in activity ranging from broadly cytotoxic to highly selective. In the NCI 60-cell line screen, these "hyperselective" agents (e.g., imatinib) are rare. A selectivity index (SI) was developed to quantify this desirable feature, which is 20 for imatinib, whereas RITA's SI is only 0.10. One of the described hyperselective RITA analogs (SI = 7.9) completely lost activity in the presence of a known SULT1A1 inhibitor. These results, coupled with previous evidence that RITA is a SULT1A1 substrate, suggest that carbinol modification by a sulfate leaving group and subsequent formation of a reactive carbocation may explain RITA's broad cytotoxicity. Although SULT1A1 expression is required for susceptibility, hyperselective analogs exhibited reduced association of activity with SULT1A1 mRNA expression compared with RITA, apparently requiring some additional target(s). In support of this hypothesis, there is a strong correlation (P < 0.01, r = 0.95) between quantum mechanically calculated energy barriers for carbocation formation from sulfonated analogs and SI, indicating that hyperselective RITA analogs generate reactive carbocations less readily after sulfate activation. Importantly, narrowing the cytotoxicity profile of RITA did not eliminate its analogs' in vivo antitumor activity, as several new hyperselective agents, NSC 773097 (1), 773392 (2), and 782846 (6), displayed impressive activity against A498 xenografts in mice.


Asunto(s)
Antineoplásicos/farmacología , Furanos/farmacología , Animales , Antineoplásicos/química , Arilsulfotransferasa/genética , Arilsulfotransferasa/metabolismo , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Femenino , Furanos/química , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Ratones Endogámicos BALB C , Ratones Desnudos
2.
Eur J Med Chem ; 159: 74-89, 2018 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-30268825

RESUMEN

Small molecules that target microtubules (MTs) represent promising therapeutics to treat certain types of cancer, including glioblastoma multiform (GBM). We synthesized modified carbazoles and evaluated their antitumor activity in GBM cells in culture. Modified carbazoles with an ethyl moiety linked to the nitrogen of the carbazole and a carbonyl moiety linked to distinct biaromatic rings exhibited remarkably different killing activities in human GBM cell lines and patient-derived GBM cells, with IC50 values from 67 to >10,000 nM. Measures of the activity of modified carbazoles with tubulin and microtubules coupled to molecular docking studies show that these compounds bind to the colchicine site of tubulin in a unique low interaction space that inhibits tubulin assembly. The modified carbazoles reported here represent novel chemical tools to better understand how small molecules disrupt MT functions and kill devastating cancers such as GBM.


Asunto(s)
Antineoplásicos/farmacología , Carbazoles/farmacología , Glioblastoma/tratamiento farmacológico , Microtúbulos/efectos de los fármacos , Antineoplásicos/síntesis química , Antineoplásicos/química , Carbazoles/síntesis química , Carbazoles/química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Glioblastoma/patología , Humanos , Simulación del Acoplamiento Molecular , Estructura Molecular , Relación Estructura-Actividad
3.
Org Biomol Chem ; 15(19): 4096-4114, 2017 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-28352916

RESUMEN

The turnstile motion of two neighboring threonines sets up a dynamic side chain interplay that can accommodate both polar and apolar ligands in a small molecule allosteric protein binding site. A computational model based on SAR data and both X-ray and cryo-EM structures of the AAA ATPase p97 was used to analyze the effects of paired threonines at the inhibitor site. Specifically, the Thr side chain hydroxyl groups form a hydrogen bonding network that readily accommodates small, highly polar ligand substituents. Conversely, diametric rotation of the χ1 torsion by 150-180° orients the side chain ß-methyl groups into the binding cleft, creating a hydrophobic pocket that can accommodate small, apolar substituents. This motif was found to be critical for rationalizing the affinities of a structurally focused set of inhibitors of p97 covering a > 2000-fold variation in potencies, with a preference for either small-highly polar or small-apolar groups. The threonine turnstile motif was further validated by a PDB search that identified analogous binding modes in ligand interactions in PKB, as well as by an analysis of NMR structures demonstrating additional gear-like interactions between adjacent Thr pairs. Combined, these data suggest that the threonine turnstile motif may be a general feature of interest in protein binding pockets.


Asunto(s)
Adenosina Trifosfatasas/química , Adenosina Trifosfatasas/metabolismo , Sitio Alostérico , Interacciones Hidrofóbicas e Hidrofílicas , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Treonina , Secuencias de Aminoácidos , Ligandos , Modelos Moleculares , Unión Proteica
4.
Org Lett ; 16(7): 2034-7, 2014 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-24641272

RESUMEN

Two complementary approaches for the preparation of linked 5-membered heterocycles were developed. The Pd-catalyzed Suzuki-Miyaura cross-coupling with halogenated furan, thiophene, and selenophene led to higher overall yields, but C,H-bond activation was a more efficient strategy for the coupling at C(2) of oxazoles. Potency and selectivity of the final hydroxymethyl products in renal (A498), lung (NCI-H226), kidney (CAKI-1), and breast (MDA-MB-468, MCF7) carcinoma cell lines were determined.


Asunto(s)
Antineoplásicos/síntesis química , Compuestos Heterocíclicos/química , Compuestos Heterocíclicos/síntesis química , Paladio/química , Antineoplásicos/química , Antineoplásicos/toxicidad , Ácidos Borónicos/química , Catálisis , Química Orgánica/métodos , Ensayos de Selección de Medicamentos Antitumorales , Furanos/química , Compuestos Heterocíclicos/toxicidad , Humanos , Estructura Molecular , Oxazoles/química , Relación Estructura-Actividad
5.
Biochem Pharmacol ; 84(4): 444-50, 2012 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-22634405

RESUMEN

We purified pseudolaric acid B (PAB) from the root and stem bark of Pseudolarix kaempferi (Lindl.) Gorden. Confirming previous findings, we found that the compound had high nanomolar IC50 antiproliferative effects in several cultured cell lines, causing mitotic arrest and the disappearance of intracellular microtubules. PAB strongly inhibited tubulin assembly (IC50, 1.1 µM) but weakly inhibited the binding of colchicine to tubulin, as demonstrated by fluorescence and with [³H]colchicine. Kinetic analysis demonstrated that the mechanism of inhibition was competitive, with an apparent K(i) of 12-15 µM. Indirect studies demonstrated that PAB bound rapidly to tubulin and dissociated more rapidly from tubulin than the colchicine analog 2-methoxy-5-(2',3',4'-trimethoxyphenyl)tropone, whose complex with tubulin is known to have a half-life of 17s at 37 °C. We modeled PAB into the colchicine site of tubulin, using the crystal structure 1SA0 that contains two αß-tubulin heterodimers, both bound to a colchicinoid and to a stathmin fragment. The binding model of PAB revealed common pharmacophoric features between PAB and colchicinoids, not readily apparent from their chemical structures.


Asunto(s)
Colchicina/metabolismo , Diterpenos/farmacología , Pinaceae/química , Moduladores de Tubulina/farmacología , Tubulina (Proteína)/metabolismo , Sitios de Unión , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Colchicina/química , Diterpenos/química , Diterpenos/aislamiento & purificación , Fluorescencia , Humanos , Cinética , Mitosis/efectos de los fármacos , Modelos Moleculares , Unión Proteica , Estatmina/química , Tubulina (Proteína)/química , Moduladores de Tubulina/química , Moduladores de Tubulina/aislamiento & purificación
6.
Mol Cancer Ther ; 11(5): 1103-11, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22442310

RESUMEN

Tumor resistance to antitubulin drugs resulting from P-glycoprotein (Pgp) drug-efflux activity, increased expression of the ßIII tubulin isotype, and alterations in the drug-binding sites are major obstacles in cancer therapy. Consequently, novel antitubulin drugs that overcome these challenges are of substantial interest. Here, we study a novel chemotype named furan metotica that localizes to the colchicine-binding site in ß-tubulin, inhibits tubulin polymerization, and is not antagonized by Pgp. To elucidate the structure-activity properties of this chiral chemotype, the enantiomers of its most potent member were separated and their absolute configurations determined by X-ray crystallography. Both isomers were active and inhibited all 60 primary cancer cell lines tested at the U.S. National Cancer Institute. They also efficiently killed drug-resistant cancer cells that overexpressed the Pgp drug-efflux pump 10(6)-fold. In vitro, the R-isomer inhibited tubulin polymerization at least 4-fold more potently than the S-isomer, whereas in human cells the difference was 30-fold. Molecular modeling showed that the two isomers bind to ß-tubulin in distinct manners: the R-isomer binds in a colchicine-like mode and the S-isomer in a podophyllotoxin-like fashion. In addition, the dynamic binding trajectory and occupancy state of the R-isomer were energetically more favorable then those of the S-isomer, explaining the observed differences in biologic activities. The ability of a racemic drug to assume the binding modes of two prototypical colchicine-site binders represents a novel mechanistic basis for antitubulin activity and paves the way toward a comprehensive design of novel anticancer agents.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Antineoplásicos/química , Antineoplásicos/farmacología , Furanos/química , Furanos/farmacología , Indoles/química , Indoles/farmacología , Moduladores de Tubulina/química , Moduladores de Tubulina/farmacología , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/antagonistas & inhibidores , Línea Celular Tumoral , Resistencia a Antineoplásicos , Humanos , Neoplasias/metabolismo , Estereoisomerismo
7.
J Chem Inf Model ; 51(6): 1393-404, 2011 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-21539396

RESUMEN

Compounds that modulate microtubule dynamics include highly effective anticancer drugs, leading to continuing efforts to identify new agents and improve the activity of established ones. Here, we demonstrate that [(3)H]-labeled halichondrin B (HB), a complex, sponge-derived natural product, is bound to and dissociated from tubulin rapidly at one binding site per αß-heterodimer, with an apparent K(d) of 0.31 µM. We found no HB-induced aggregation of tubulin by high-performance liquid chromatography, even following column equilibration with HB. Binding of [(3)H]HB was competitively inhibited by a newly approved clinical agent, the truncated HB analogue eribulin (apparent K(i), 0.80 µM) and noncompetitively by dolastatin 10 and vincristine (apparent K(i)'s, 0.35 and 5.4 µM, respectively). Our earlier studies demonstrated that HB inhibits nucleotide exchange on ß-tubulin, and this, together with the results presented here, indicated the HB site is located on ß-tubulin. Using molecular dynamics simulations, we determined complementary conformations of HB and ß-tubulin that delineated in atomic detail binding interactions of HB with only ß-tubulin, with no involvement of the α-subunit in the binding interaction. Moreover, the HB model served as a template for an eribulin binding model that furthered our understanding of the properties of eribulin as a drug. Overall, these results established a mechanistic basis for the antimitotic activity of the halichondrin class of compounds.


Asunto(s)
Antimitóticos/metabolismo , Éteres Cíclicos/metabolismo , Furanos/metabolismo , Cetonas/metabolismo , Modelos Moleculares , Tubulina (Proteína)/metabolismo , Animales , Sitios de Unión , Bovinos , Macrólidos , Simulación de Dinámica Molecular , Poríferos , Unión Proteica , Multimerización de Proteína , Estructura Cuaternaria de Proteína , Tubulina (Proteína)/química
8.
PLoS One ; 5(6): e11378, 2010 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-20614028

RESUMEN

The botulinum neurotoxin serotype A light chain (BoNT/A LC) protease is the catalytic component responsible for the neuroparalysis that is characteristic of the disease state botulism. Three related peptide-like molecules (PLMs) were designed using previous information from co-crystal structures, synthesized, and assayed for in vitro inhibition against BoNT/A LC. Our results indicate these PLMS are competitive inhibitors of the BoNT/A LC protease and their K(i) values are in the nM-range. A co-crystal structure for one of these inhibitors was determined and reveals that the PLM, in accord with the goals of our design strategy, simultaneously involves both ionic interactions via its P1 residue and hydrophobic contacts by means of an aromatic group in the P2' position. The PLM adopts a helical conformation similar to previously determined co-crystal structures of PLMs, although there are also major differences to these other structures such as contacts with specific BoNT/A LC residues. Our structure further demonstrates the remarkable plasticity of the substrate binding cleft of the BoNT/A LC protease and provides a paradigm for iterative structure-based design and development of BoNT/A LC inhibitors.


Asunto(s)
Toxinas Botulínicas Tipo A/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Péptidos/farmacología , Catálisis , Cristalización , Cristalografía por Rayos X , Inhibidores Enzimáticos/química , Modelos Moleculares , Péptidos/química , Conformación Proteica , Especificidad por Sustrato
9.
PLoS One ; 5(7): e11603, 2010 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-20657644

RESUMEN

BACKGROUND: Protein assemblies named kinetochores bind sister chromatids to the mitotic spindle and orchestrate sister chromatid segregation. Interference with kinetochore activity triggers a spindle checkpoint mediated arrest in mitosis, which frequently ends in cell death. We set out to identify small compounds that inhibit kinetochore-microtubule binding for use in kinetochore-spindle interaction studies and to develop them into novel anticancer drugs. METHODOLOGY/PRINCIPAL FINDINGS: A fluorescence microscopy-based in vitro assay was developed to screen compound libraries for molecules that prevented the binding of a recombinant human Ndc80 kinetochore complex to taxol-stabilized microtubules. An active compound was identified that acted at the microtubule level. More specifically, by localizing to the colchicine-binding site in alphabeta-tubulin the hit compound prevented the Ndc80 complex from binding to the microtubule surface. Next, structure-activity analyses distinguished active regions in the compound and led to the identification of highly potent analogs that killed cancer cells with an efficacy equaling that of established spindle drugs. CONCLUSIONS/SIGNIFICANCE: The compound identified in our screen and its subsequently identified analogs represent new antitubulin chemotypes that can be synthetically developed into a novel class of antimitotic spindle drugs. In addition, they are stereochemically unique as their R- and S-isomers mimic binding of colchicine and podophyllotoxin, respectively, two antitubulin drugs that interact differently with the tubulin interface. Model-driven manipulation of our compounds promises to advance insight into how antitubulin drugs act upon tubulin. These advances in turn may lead to tailor-made colchicine site agents which would be valuable new assets to fight a variety of tumors, including those that have become resistant to the (antispindle) drugs used today.


Asunto(s)
Cinetocoros/metabolismo , Microtúbulos/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Recombinantes/metabolismo , Antimitóticos/farmacología , Antineoplásicos/farmacología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Proteínas del Citoesqueleto , Células HeLa , Humanos , Microscopía Fluorescente , Proteínas Nucleares/genética , Unión Proteica/efectos de los fármacos , Proteínas Recombinantes/genética , Tubulina (Proteína)/metabolismo
10.
J Chem Pharm Res ; 2(5): 587-598, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-21593990

RESUMEN

Due to its overexpression and activation in human cancer cells and tissues, an emerging molecular target in cancer therapeutics is p90 ribosomal s6 kinase 2 (RSK2). While a growing number of RSK2 inhibitors have been reported in the literature, only the crystal structure of RSK2 in complex with an AMP analogue provides a structural basis for understanding RSK2 inhibition. To remedy this, we used our fluorescence polarization assay to determine the RSK2 activity for a set of structurally diverse compounds, and followed this by modeling their binding modes in an all-atom, energy refined crystal structure of RSK2. These binding models reveal that Val131 and Leu147 are key interaction sites for potent RSK2 inhibition. This structure-based pharmacophore is an important tool for new lead discovery and refinement.

11.
Antimicrob Agents Chemother ; 53(10): 4283-91, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19635954

RESUMEN

Given the limited number of structural classes of clinically available antimicrobial drugs, the discovery of antibacterials with novel chemical scaffolds is an important strategy in the development of effective therapeutics for both naturally occurring and engineered resistant strains of pathogenic bacteria. In this study, several diarylamidine derivatives were evaluated for their ability to protect macrophages from cell death following infection with Bacillus anthracis, a gram-positive spore-forming bacterium. Four bis-(imidazolinylindole) compounds were identified with potent antibacterial activity as measured by the protection of macrophages and by the inhibition of bacterial growth in vitro. These compounds were effective against a broad range of gram-positive and gram-negative bacterial species, including several antibiotic-resistant strains. Minor structural variations among the four compounds correlated with differences in their effects on bacterial macromolecular synthesis and mechanisms of resistance. In vivo studies revealed protection by two of the compounds of mice lethally infected with B. anthracis, Staphylococcus aureus, or Yersinia pestis. Taken together, these results indicate that the bis-(imidazolinylindole) compounds represent a new chemotype for the development of therapeutics for both gram-positive and gram-negative bacterial species as well as against antibiotic-resistant infections.


Asunto(s)
Antibacterianos/farmacología , Bacillus anthracis/efectos de los fármacos , Animales , Antibacterianos/química , Muerte Celular/efectos de los fármacos , Línea Celular , Bacterias Gramnegativas/efectos de los fármacos , Bacterias Grampositivas/efectos de los fármacos , Macrófagos/efectos de los fármacos , Macrófagos/microbiología , Ratones , Staphylococcus aureus/efectos de los fármacos , Yersinia pestis/efectos de los fármacos
12.
J Biol Chem ; 284(19): 12874-85, 2009 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-19269962

RESUMEN

The modulation of cellular processes by small molecule inhibitors, gene inactivation, or targeted knockdown strategies combined with phenotypic screens are powerful approaches to delineate complex cellular pathways and to identify key players involved in disease pathogenesis. Using chemical genetic screening, we tested a library of known phosphatase inhibitors and identified several compounds that protected Bacillus anthracis infected macrophages from cell death. The most potent compound was assayed against a panel of sixteen different phosphatases of which CD45 was found to be most sensitive to inhibition. Testing of a known CD45 inhibitor and antisense phosphorodiamidate morpholino oligomers targeting CD45 also protected B. anthracis-infected macrophages from cell death. However, reduced CD45 expression did not protect anthrax lethal toxin (LT) treated macrophages, suggesting that the pathogen and independently added LT may signal through distinct pathways. Subsequent, in vivo studies with both gene-targeted knockdown of CD45 and genetically engineered mice expressing reduced levels of CD45 resulted in protection of mice after infection with the virulent Ames B. anthracis. Intermediate levels of CD45 expression were critical for the protection, as mice expressing normal levels of CD45 or disrupted CD45 phosphatase activity or no CD45 all succumbed to this pathogen. Mechanism-based studies suggest that the protection provided by reduced CD45 levels results from regulated immune cell homeostasis that may diminish the impact of apoptosis during the infection. To date, this is the first report demonstrating that reduced levels of host phosphatase CD45 modulate anthrax pathogenesis.


Asunto(s)
Carbunco/enzimología , Carbunco/prevención & control , Bacillus anthracis/patogenicidad , Antígenos Comunes de Leucocito/metabolismo , Animales , Antígenos Bacterianos/metabolismo , Antígenos Bacterianos/toxicidad , Apoptosis , Bacillus anthracis/fisiología , Toxinas Bacterianas/antagonistas & inhibidores , Toxinas Bacterianas/metabolismo , Toxinas Bacterianas/toxicidad , Supervivencia Celular , Femenino , Citometría de Flujo , Técnicas de Silenciamiento del Gen , Pruebas Genéticas , Immunoblotting , Técnicas para Inmunoenzimas , Antígenos Comunes de Leucocito/antagonistas & inhibidores , Macrófagos/microbiología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Morfolinas/farmacología , Morfolinos , Fagocitosis , Monoéster Fosfórico Hidrolasas/metabolismo , Esporas Bacterianas/crecimiento & desarrollo , Esporas Bacterianas/patogenicidad
13.
ChemMedChem ; 3(12): 1905-12, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19006141

RESUMEN

A search query consisting of two aromatic centers and two cationic centers was defined based on previously identified small molecule inhibitors of the botulinum neurotoxin serotype A light chain (BoNT/A LC) and used to mine the National Cancer Institute Open Repository. Ten small molecule hits were identified, and upon testing, three demonstrated inhibitory activity. Of these, one was structurally unique, possessing a rigid diazachrysene scaffold. The steric limitations of the diazachrysene imposed a separation between the overlaps of previously identified inhibitors, revealing an extended binding mode. As a result, the pharmacophore for BoNT/A LC inhibition has been modified to encompass three zones. To demonstrate the utility of this model, a novel three-zone inhibitor was mined and its activity was confirmed.


Asunto(s)
Toxinas Botulínicas Tipo A/antagonistas & inhibidores , Crisenos/química , Modelos Moleculares , Toxinas Botulínicas Tipo A/farmacología , Crisenos/farmacología , Simulación por Computador , Bases de Datos Factuales , Diseño de Fármacos , Imagenología Tridimensional , Relación Estructura-Actividad
14.
Biophys J ; 95(3): 1157-64, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18645196

RESUMEN

Nonelectrolyte polymers of poly(ethylene glycol) (PEG) were used to estimate the diameter of the ion channel formed by the Bacillus anthracis protective antigen 63 (PA(63)). Based on the ability of different molecular weight PEGs to partition into the pore and reduce channel conductance, the pore appears to be narrower than the one formed by Staphylococcus aureus alpha-hemolysin. Numerical integration of the PEG sample mass spectra and the channel conductance data were used to refine the estimate of the pore's PEG molecular mass cutoff (approximately 1400 g/mol). The results suggest that the limiting diameter of the PA(63) pore is <2 nm, which is consistent with an all-atom model of the PA(63) channel and previous experiments using large ions.


Asunto(s)
Antígenos Bacterianos/química , Antígenos Bacterianos/ultraestructura , Bacillus anthracis/química , Toxinas Bacterianas/química , Modelos Químicos , Modelos Moleculares , Polietilenglicoles/química , Simulación por Computador , Electrólitos/química , Porosidad , Conformación Proteica
15.
Chem Biol ; 14(3): 245-55, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17379140

RESUMEN

Anthrax lethal toxin (LT)-induced cell death via mitogen-activated protein kinase kinase (MAPKK) cleavage remains questionable. Here, a chemical genetics approach was used to investigate what pathways mediate LT-induced cell death. Several small molecules were found to protect macrophages from anthrax LT cytotoxicity and MAPKK from cleavage by lethal factor (LF), without inhibiting LF enzymatic activity or cellular proteasome activity. Interestingly, the compounds activated MAPK-signaling molecules, induced proinflammatory cytokine production, and inhibited LT-induced macrophage apoptosis in a concentration-dependent manner. We propose that induction of antiapoptotic responses by MAPK-dependent or -independent pathways and activation of host innate responses may protect macrophages from anthrax LT-induced cell death. Altering host responses through a chemical genetics approach can help identify critical cellular pathways involved in the pathogenesis of anthrax and can be exploited to further explore host-pathogen interactions.


Asunto(s)
Antígenos Bacterianos/toxicidad , Toxinas Bacterianas/toxicidad , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Animales , Apoptosis/efectos de los fármacos , Proteínas de Ciclo Celular/antagonistas & inhibidores , Muerte Celular/efectos de los fármacos , Línea Celular , Citocinas/metabolismo , Pruebas Genéticas/métodos , Macrófagos/citología , Macrófagos/efectos de los fármacos , Macrófagos/fisiología , Ratones , Microscopía Confocal , Necrosis , Fosfatasas cdc25/antagonistas & inhibidores
16.
J Biol Chem ; 282(7): 5004-5014, 2007 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-17092934

RESUMEN

An efficient research strategy integrating empirically guided, structure-based modeling and chemoinformatics was used to discover potent small molecule inhibitors of the botulinum neurotoxin serotype A light chain. First, a modeled binding mode for inhibitor 2-mercapto-3-phenylpropionyl-RATKML (K(i) = 330 nM) was generated, and required the use of a molecular dynamic conformer of the enzyme displaying the reorientation of surface loops bordering the substrate binding cleft. These flexible loops are conformationally variable in x-ray crystal structures, and the model predicted that they were pivotal for providing complementary binding surfaces and solvent shielding for the pseudo-peptide. The docked conformation of 2-mercapto-3-phenylpropionyl-RATKML was then used to refine our pharmacophore for botulinum serotype A light chain inhibition. Data base search queries derived from the pharmacophore were employed to mine small molecule (non-peptidic) inhibitors from the National Cancer Institute's Open Repository. Four of the inhibitors possess K(i) values ranging from 3.0 to 10.0 microM. Of these, NSC 240898 is a promising lead for therapeutic development, as it readily enters neurons, exhibits no neuronal toxicity, and elicits dose-dependent protection of synaptosomal-associated protein (of 25 kDa) in a primary culture of embryonic chicken neurons. Isothermal titration calorimetry showed that the interaction between NSC 240898 and the botulinum A light chain is largely entropy-driven, and occurs with a 1:1 stoichiometry and a dissociation constant of 4.6 microM.


Asunto(s)
Toxinas Botulínicas Tipo A/química , Metaloproteasas/química , Modelos Moleculares , Neuronas/química , Inhibidores de Proteasas/química , Animales , Toxinas Botulínicas Tipo A/metabolismo , Botulismo/tratamiento farmacológico , Botulismo/enzimología , Células Cultivadas , Embrión de Pollo , Metaloproteasas/metabolismo , Neuronas/enzimología , Inhibidores de Proteasas/metabolismo , Inhibidores de Proteasas/uso terapéutico
17.
Mol Pharmacol ; 70(6): 1866-75, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16940412

RESUMEN

The complex marine natural product halichondrin B was compared with NSC 707389 (E7389), a structurally simplified, synthetic macrocyclic ketone analog, which has been selected for clinical trials in human patients. NSC 707389 was invariably more potent than halichondrin B in its interactions with tubulin. Both compounds inhibited tubulin assembly, inhibited nucleotide exchange on beta-tubulin, and were noncompetitive inhibitors of the binding of radiolabeled vinblastine and dolastatin 10 to tubulin. Neither compound seemed to induce an aberrant tubulin assembly reaction, as occurs with vinblastine (tight spirals) or dolastatin 10 (aggregated rings and spirals). We modeled the two compounds into a shared binding site on tubulin consistent with their biochemical properties. Of the two tubulin structures available, we selected for modeling the complex of a stathmin fragment with two tubulin heterodimers with two bound colchicinoid molecules and a single bound vinblastine between the two heterodimers (Nature (Lond) 435:519-522, 2005). Halichondrin B and NSC 707389 fit snugly between the two heterodimers adjacent to the exchangeable site nucleotide. Fitting the compounds into this site, which was also close to the vinblastine site, resulted in enough movement of amino acid residues at the vinblastine site to cause the latter compound to bind less well to tubulin. The model suggests that halichondrin B and NSC 707389 most likely form highly unstable, small aberrant tubulin polymers rather than the massive stable structures observed with vinca alkaloids and antimitotic peptides.


Asunto(s)
Éteres Cíclicos/farmacología , Furanos/farmacología , Cetonas/farmacología , Tubulina (Proteína)/metabolismo , Sitios de Unión , División Celular , Línea Celular Tumoral , Cristalografía por Rayos X , Éteres Cíclicos/química , Éteres Cíclicos/metabolismo , Furanos/química , Furanos/metabolismo , Humanos , Cetonas/química , Cetonas/metabolismo , Macrólidos , Modelos Moleculares , Estructura Molecular
18.
Bioorg Med Chem ; 14(17): 6097-105, 2006 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-16723234

RESUMEN

Ribosomal S6 kinase 2 (RSK2) is a serine/threonine kinase that plays a role in human cancer and Coffin-Lowry syndrome and is comprised of two nonidentical kinase domains, each domain with its own ATP-binding site. RSK2 can be inactivated by different types of small organic molecules. Potent RSK2 inhibitors include the two classic bisindole maleimide PKC inhibitors, Ro31-8220 and GF109203X, and the natural product SL0101 that was shown to bind specifically to the ATP pocket of the N-terminal domain (NTD). In this paper, we present an atomic model of the RSK2 NTD (residues 68-323), which was built to simultaneously bind the distinctive molecular scaffolds of SL0101, Ro31-8220, and GF109203X. The RSK2 NTD model was used to identify two novel RSK2 inhibitors from the National Cancer Institute open chemical repository and to develop a preliminary structure-based pharmacophore model.


Asunto(s)
Proteínas Quinasas S6 Ribosómicas 90-kDa/antagonistas & inhibidores , Proteínas Quinasas S6 Ribosómicas 90-kDa/química , Benzopiranos/química , Sitios de Unión , Indoles/química , Quempferoles/química , Modelos Moleculares , Monosacáridos/química , Naftalenos/química , Conformación Proteica , Estructura Terciaria de Proteína , Pirimidinas/química , Proteínas Quinasas S6 Ribosómicas 90-kDa/metabolismo , Relación Estructura-Actividad
19.
J Med Chem ; 48(19): 6107-16, 2005 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-16162011

RESUMEN

Modulating the structure and function of tubulin and microtubules is an important route to anticancer therapeutics, and therefore, small molecules that bind to tubulin and cause mitotic arrest are of immense interest. A large number of synthetic and natural compounds with diverse structures have been shown to bind at the colchicine site, one of the major binding sites on tubulin, and inhibit tubulin assembly. Using the recently determined X-ray structure of the tubulin:colchicinoid complex as the template, we employed docking studies to determine the binding modes of a set of structurally diverse colchicine site inhibitors. These binding models were subsequently used to construct a comprehensive, structure-based pharmacophore that in combination with molecular dynamics simulations confirms and extends our understanding of binding interactions at the colchicine site.


Asunto(s)
Colchicina/química , Modelos Moleculares , Moduladores de Tubulina , Tubulina (Proteína)/química , 2-Metoxiestradiol , 4-Butirolactona/análogos & derivados , 4-Butirolactona/química , Aminofenoles/química , Sitios de Unión , Chalcona/química , Ciclopropanos/química , Estradiol/análogos & derivados , Estradiol/química , Indanos/química , Lignanos/química , Estructura Molecular , Nocodazol/química , Podofilotoxina/química , Unión Proteica , Estilbenos/química , Relación Estructura-Actividad , Sulfonamidas/química , Tiazoles/química
20.
Biochemistry ; 44(34): 11525-38, 2005 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-16114889

RESUMEN

Cyclostreptin (FR182877), a bacterial natural product, was reported to have weak paclitaxel-like activity with tubulin but antitumor activity in vivo. We used synthetic cyclostreptin in studies of its mechanism of action. Although less potent than paclitaxel in several human cancer cell lines, cyclostreptin was active against cells resistant to paclitaxel and epothilone A. At equitoxic concentrations with paclitaxel, cyclostreptin was more effective in arresting MCF-7 cells in mitosis and equivalent in bundling microtubules in PtK(2) cells. Tubulin assembly with paclitaxel occurs at low temperatures and in the absence of GTP or microtubule-associated proteins (MAPs). Brisk assembly with cyclostreptin required MAPs, GTP, and higher reaction temperatures. On the basis of turbidimetry, cyclostreptin-induced microtubules were more stable in the cold than the paclitaxel-induced polymer. Moreover, at 37 degrees C cyclostreptin was a strong competitive inhibitor of the binding of radiolabeled paclitaxel to tubulin polymer, with an apparent K(i) value of 88 nM. Competition studies versus a fluorescent taxoid across a temperature range, in comparison with paclitaxel and docetaxel, showed that only the binding of cyclostreptin to microtubules was markedly reduced at 4 degrees C versus temperatures over 30 degrees C. The binding of cyclostreptin to microtubules was characterized by a relatively greater endothermic and entropic profile as compared with those of the taxoid binding reactions, which are characterized more by exothermic and enthalpic interactions. Molecular modeling showed that cyclostreptin formed a pharmacophore with taxoids but formed hydrogen bonds only with the S9-S10 and M loops in the taxoid site. Initial studies also indicate that, relative to paclitaxel, cyclostreptin is more deficient in nucleation than elongation of polymer.


Asunto(s)
Antineoplásicos/farmacología , Compuestos Policíclicos/farmacología , Taxoides/metabolismo , Tubulina (Proteína)/química , Tubulina (Proteína)/metabolismo , Animales , Sitios de Unión , Bovinos , Línea Celular Tumoral , Femenino , Humanos , Cinética , Ligandos , Microtúbulos/efectos de los fármacos , Microtúbulos/ultraestructura , Modelos Moleculares , Neoplasias Ováricas , Paclitaxel/farmacología , Conformación Proteica , Taxoides/química , Termodinámica , Tubulina (Proteína)/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA