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










Base de dados
Intervalo de ano de publicação
1.
Mol Cancer Ther ; 18(10): 1765-1774, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31341033

RESUMO

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.


Assuntos
Antineoplásicos/farmacologia , Furanos/farmacologia , Animais , Antineoplásicos/química , Arilsulfotransferase/genética , Arilsulfotransferase/metabolismo , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Feminino , Furanos/química , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Camundongos Endogâmicos BALB C , Camundongos Nus
2.
Eur J Med Chem ; 159: 74-89, 2018 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-30268825

RESUMO

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.


Assuntos
Antineoplásicos/farmacologia , Carbazóis/farmacologia , Glioblastoma/tratamento farmacológico , Microtúbulos/efeitos dos fármacos , Antineoplásicos/síntese química , Antineoplásicos/química , Carbazóis/síntese química , Carbazóis/química , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Glioblastoma/patologia , Humanos , Simulação de Acoplamento Molecular , Estrutura Molecular , Relação Estrutura-Atividade
3.
Org Biomol Chem ; 15(19): 4096-4114, 2017 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-28352916

RESUMO

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.


Assuntos
Adenosina Trifosfatases/química , Adenosina Trifosfatases/metabolismo , Sítio Alostérico , Interações Hidrofóbicas e Hidrofílicas , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Treonina , Motivos de Aminoácidos , Ligantes , Modelos Moleculares , Ligação Proteica
4.
PLoS One ; 10(6): e0129264, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26061731

RESUMO

There is an urgent need to develop novel treatments to counter Botulinum neurotoxin (BoNT) poisoning. Currently, the majority of BoNT drug development efforts focus on directly inhibiting the proteolytic components of BoNT, i.e. light chains (LC). Although this is a rational approach, previous research has shown that LCs are extremely difficult drug targets and that inhibiting multi-serotype BoNTs with a single LC inhibitor may not be feasible. An alternative approach would target neuronal pathways involved in intoxication/recovery, rather than the LC itself. Phosphorylation-related mechanisms have been implicated in the intoxication pathway(s) of BoNTs. However, the effects of phosphatase inhibitors upon BoNT activity in the physiological target of BoNTs, i.e. motor neurons, have not been investigated. In this study, a small library of phosphatase inhibitors was screened for BoNT antagonism in the context of mouse embryonic stem cell-derived motor neurons (ES-MNs). Four inhibitors were found to function as BoNT/A antagonists. Subsequently, we confirmed that these inhibitors protect against BoNT/A in a dose-dependent manner in human ES-MNs. Additionally, these compounds provide protection when administered in post-intoxication scenario. Importantly, the inhibitors were also effective against BoNT serotypes B and E. To the best of our knowledge, this is the first study showing phosphatase inhibitors as broad-spectrum BoNT antagonists.


Assuntos
Toxinas Botulínicas/toxicidade , Células-Tronco Embrionárias/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Neurônios Motores/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/farmacologia , Animais , Toxinas Botulínicas/antagonistas & inibidores , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Células-Tronco Embrionárias/metabolismo , Humanos , Camundongos , Neurônios Motores/metabolismo , Monoéster Fosfórico Hidrolases/antagonistas & inibidores , Proteínas SNARE/metabolismo
5.
Neurotox Res ; 27(4): 384-98, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25782580

RESUMO

Botulinum neurotoxins (BoNTs), the causative agents of botulism, are potent inhibitors of neurotransmitter release from motor neurons. There are currently no drugs to treat BoNT intoxication after the onset of the disease symptoms. In this study, we explored how modulation of key host pathways affects the process of BoNT intoxication in human motor neurons, focusing on Src family kinase (SFK) signaling. Motor neurons derived from human embryonic stem (hES) cells were treated with a panel of SFK inhibitors and intoxicated with BoNT serotypes A, B, or E (which are responsible for >95 % of human botulism cases). Subsequently, it was found that bosutinib, dasatinib, KX2-391, PP1, PP2, Src inhibitor-1, and SU6656 significantly antagonized all three of the serotypes. Furthermore, the data indicated that the treatment of hES-derived motor neurons with multiple SFK inhibitors increased the antagonistic effect synergistically. Mechanistically, the small molecules appear to inhibit BoNTs by targeting host pathways necessary for intoxication and not by directly inhibiting the toxins' proteolytic activity. Importantly, the identified inhibitors are all well-studied with some in clinical trials while others are FDA-approved drugs. Overall, this study emphasizes the importance of targeting host neuronal pathways, rather than the toxin's enzymatic components, to antagonize multiple BoNT serotypes in motor neurons.


Assuntos
Toxinas Botulínicas/toxicidade , Neurônios Motores/efeitos dos fármacos , Neurônios Motores/metabolismo , Transdução de Sinais/efeitos dos fármacos , Quinases da Família src/antagonistas & inibidores , Quinases da Família src/metabolismo , Células-Tronco Embrionárias/citologia , Humanos , Proteólise/efeitos dos fármacos , Sorogrupo
6.
Org Lett ; 16(7): 2034-7, 2014 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-24641272

RESUMO

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.


Assuntos
Antineoplásicos/síntese química , Compostos Heterocíclicos/química , Compostos Heterocíclicos/síntese química , Paládio/química , Antineoplásicos/química , Antineoplásicos/toxicidade , Ácidos Borônicos/química , Catálise , Química Orgânica/métodos , Ensaios de Seleção de Medicamentos Antitumorais , Furanos/química , Compostos Heterocíclicos/toxicidade , Humanos , Estrutura Molecular , Oxazóis/química , Relação Estrutura-Atividade
7.
J Chem Phys ; 139(6): 065101, 2013 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-23947891

RESUMO

We demonstrate experimentally that anthrax toxin complexes rupture artificial lipid bilayer membranes when isolated from the blood of infected animals. When the solution pH is temporally acidified to mimic that process in endosomes, recombinant anthrax toxin forms an irreversibly bound complex, which also destabilizes membranes. The results suggest an alternative mechanism for the translocation of anthrax toxin into the cytoplasm.


Assuntos
Antígenos de Bactérias/toxicidade , Toxinas Bacterianas/toxicidade , Membrana Celular/efeitos dos fármacos , Bicamadas Lipídicas/química , Animais , Antígenos de Bactérias/genética , Toxinas Bacterianas/genética , Células Sanguíneas/efeitos dos fármacos , Endossomos/efeitos dos fármacos , Cobaias , Haplorrinos , Humanos , Membranas Artificiais , Coelhos
8.
Curr Pharm Des ; 19(23): 4310-5, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23170887

RESUMO

In this review, a summary of methodologies is covered to enable medicinal chemists to access an overview of pK(a) estimation devices. In order to stave overutilization of costly synthetic resources, the chemist requires an accurate and computationally tractable solution for estimating a pK(a) of a candidate molecule. We focus on the cationic moieties, since they are so fundamentally important in the chemistry of drugs, and possess unique requirements to obtain a reasonably reliable pK(a) estimation.


Assuntos
Preparações Farmacêuticas/química , Cátions , Termodinâmica
9.
Biochem Pharmacol ; 84(4): 444-50, 2012 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-22634405

RESUMO

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.


Assuntos
Colchicina/metabolismo , Diterpenos/farmacologia , Pinaceae/química , Moduladores de Tubulina/farmacologia , Tubulina (Proteína)/metabolismo , Sítios de Ligação , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Colchicina/química , Diterpenos/química , Diterpenos/isolamento & purificação , Fluorescência , Humanos , Cinética , Mitose/efeitos dos fármacos , Modelos Moleculares , Ligação Proteica , Estatmina/química , Tubulina (Proteína)/química , Moduladores de Tubulina/química , Moduladores de Tubulina/isolamento & purificação
10.
Eur J Med Chem ; 53: 374-9, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22516424

RESUMO

Botulinum neurotoxins (BoNTs), composed of a family of seven serotypes (categorized A-G), are the deadliest of known biological toxins. The activity of the metalloprotease, light chain (LC) component of the toxins is responsible for causing the life-threatening paralysis associated with the disease botulism. Herein we report significantly more potent analogs of novel, lead BoNT serotype A LC inhibitor 2,5-bis(4-amidinophenyl)thiophene (K(i) = 10.88 µM ± 0.90 µM). Specifically, synthetic modifications involved simultaneously replacing the lead inhibitor's terminal bis-amidines with secondary amines and the systematic tethering of 4-amino-7-chloroquinoline substituents to provide derivatives with K(i) values ranging from 0.302 µM (± 0.03 µM) to 0.889µM (± 0.11 µM).


Assuntos
Toxinas Botulínicas Tipo A/antagonistas & inibidores , Inibidores de Proteases/síntese química , Inibidores de Proteases/farmacologia , Tiofenos/síntese química , Tiofenos/farmacologia , Toxinas Botulínicas Tipo A/química , Técnicas de Química Sintética , Relação Dose-Resposta a Droga , Inibidores de Proteases/química , Tiofenos/química
11.
Mol Cancer Ther ; 11(5): 1103-11, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22442310

RESUMO

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.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Antineoplásicos/química , Antineoplásicos/farmacologia , Furanos/química , Furanos/farmacologia , Indóis/química , Indóis/farmacologia , Moduladores de Tubulina/química , Moduladores de Tubulina/farmacologia , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/antagonistas & inibidores , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos , Humanos , Neoplasias/metabolismo , Estereoisomerismo
12.
J Chem Inf Model ; 51(6): 1393-404, 2011 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-21539396

RESUMO

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.


Assuntos
Antimitóticos/metabolismo , Éteres Cíclicos/metabolismo , Furanos/metabolismo , Cetonas/metabolismo , Modelos Moleculares , Tubulina (Proteína)/metabolismo , Animais , Sítios de Ligação , Bovinos , Macrolídeos , Simulação de Dinâmica Molecular , Poríferos , Ligação Proteica , Multimerização Proteica , Estrutura Quaternária de Proteína , Tubulina (Proteína)/química
13.
Stem Cell Res ; 6(3): 195-205, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21353660

RESUMO

Botulinum neurotoxins (BoNTs) inhibit cholinergic synaptic transmission by specifically cleaving proteins that are crucial for neurotransmitter exocytosis. Due to the lethality of these toxins, there are elevated concerns regarding their possible use as bioterrorism agents. Moreover, their widespread use for cosmetic purposes, and as medical treatments, has increased the potential risk of accidental overdosing and environmental exposure. Hence, there is an urgent need to develop novel modalities to counter BoNT intoxication. Mammalian motoneurons are the main target of BoNTs; however, due to the difficulty and poor efficiency of the procedures required to isolate the cells, they are not suitable for high-throughput drug screening assays. Here, we explored the suitability of embryonic stem (ES) cell-derived motoneurons as a renewable, reproducible, and physiologically relevant system for BoNT studies. We found that the sensitivity of ES-derived motoneurons to BoNT/A intoxication is comparable to that of primary mouse spinal motoneurons. Additionally, we demonstrated that several BoNT/A inhibitors protected SNAP-25, the BoNT/A substrate, in the ES-derived motoneuron system. Furthermore, this system is compatible with immunofluorescence-based high-throughput studies. These data suggest that ES-derived motoneurons provide a highly sensitive system that is amenable to large-scale screenings to rapidly identify and evaluate the biological efficacies of novel therapeutics.


Assuntos
Antitoxina Botulínica/farmacologia , Toxinas Botulínicas/antagonistas & inibidores , Descoberta de Drogas , Avaliação Pré-Clínica de Medicamentos/métodos , Células-Tronco Embrionárias/efeitos dos fármacos , Ensaios de Triagem em Larga Escala/métodos , Neurônios Motores/efeitos dos fármacos , Animais , Toxinas Botulínicas/toxicidade , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Avaliação Pré-Clínica de Medicamentos/instrumentação , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Ensaios de Triagem em Larga Escala/instrumentação , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Neurônios Motores/citologia , Neurônios Motores/metabolismo , Proteína 25 Associada a Sinaptossoma/metabolismo
14.
J Med Chem ; 54(5): 1157-69, 2011 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-21265542

RESUMO

A 1,7-bis(alkylamino)diazachrysene-based small molecule was previously identified as an inhibitor of the botulinum neurotoxin serotype A light chain metalloprotease. Subsequently, a variety of derivatives of this chemotype were synthesized to develop structure-activity relationships, and all are inhibitors of the BoNT/A LC. Three-dimensional analyses indicated that half of the originally discovered 1,7-DAAC structure superimposed well with 4-amino-7-chloroquinoline-based antimalarial agents. This observation led to the discovery that several of the 1,7-DAAC derivatives are potent in vitro inhibitors of Plasmodium falciparum and, in general, are more efficacious against CQ-resistant strains than against CQ-susceptible strains. In addition, by inhibiting ß-hematin formation, the most efficacious 1,7-DAAC-based antimalarials employ a mechanism of action analogous to that of 4,7-ACQ-based antimalarials and are well tolerated by normal cells. One candidate was also effective when administered orally in a rodent-based malaria model. Finally, the 1,7-DAAC-based derivatives were examined for Ebola filovirus inhibition in an assay employing Vero76 cells, and three provided promising antiviral activities and acceptably low toxicities.


Assuntos
Antibacterianos/síntese química , Antimaláricos/síntese química , Antivirais/síntese química , Toxinas Botulínicas Tipo A/antagonistas & inibidores , Crisenos/síntese química , Ebolavirus/efeitos dos fármacos , Plasmodium falciparum/efeitos dos fármacos , Quinolinas/síntese química , Animais , Anopheles/parasitologia , Antibacterianos/química , Antibacterianos/farmacologia , Antimaláricos/química , Antimaláricos/farmacologia , Antivirais/química , Antivirais/farmacologia , Linhagem Celular , Chlorocebus aethiops , Crisenos/química , Crisenos/farmacologia , Hemeproteínas/antagonistas & inibidores , Malária/tratamento farmacológico , Camundongos , Modelos Moleculares , Plasmodium berghei , Quinolinas/química , Quinolinas/farmacologia , Ratos , Estereoisomerismo , Relação Estrutura-Atividade
15.
ACS Med Chem Lett ; 1(7): 301-305, 2010 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-21116458

RESUMO

Botulinum neurotoxins (BoNTs) are the deadliest of microbial toxins. The enzymes' Zinc(II) metalloprotease, referred to as the light chain (LC) component, inhibits acetylcholine release into neuromuscular junctions, resulting in the disease botulism. Currently, no therapies counter BoNT poisoning post-neuronal intoxication; however, it is hypothesized that small molecules may be used to inhibit BoNT LC activity in the neuronal cytosol. Herein, we describe the pharmacophore-based design and chemical synthesis of potent (non-Zinc(II) chelating) small molecule (non-peptidic) inhibitors (SMNPIs) of the BoNT serotype A LC (the most toxic of the BoNT serotype LCs). Specifically, the three-dimensional superimpositions of 2-[4-(4-amidinephenoxy)-phenyl]-indole-6-amidine-based SMNPI regioisomers (K(i) = 0.600 µM (± 0.100 µM)), with a novel lead bis-[3-amide-5-(imidazolino)-phenyl]-terephthalamide (BAIPT)-based SMNPI (K(i) = 8.52 µM (± 0.53 µM)), resulted in a refined 4-zone pharmacophore. The refined model guided the design of BAIPT-based SMNPIs possessing K(i) values = 0.572 µM (± 0.041 µM) and 0.900 µM (± 0.078 µM).

16.
J Chem Inf Model ; 50(11): 2019-28, 2010 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-21028850

RESUMO

We used synthetic peloruside A for the commercial preparation of [³H]peloruside A. The radiolabeled compound bound to preformed tubulin polymer in amounts stoichiometric with the polymer's tubulin content, with an apparent K(d) value of 0.35 µM. A less active peloruside A analogue, (11-R)-peloruside A and laulimalide acted as competitive inhibitors of the binding of the [³H]peloruside A, with apparent K(i) values of 9.3 and 0.25 µM, respectively. Paclitaxel, epothilone B, and discodermolide had essentially no ability to inhibit [³H]peloruside A binding, confirming that these compounds bind to a different site on tubulin polymer. We modeled both laulimalide and peloruside A into the binding site on ß-tubulin that was identified by Huzil et al. (J. Mol. Biol. 2008, 378, 1016-1030), but our model provides a more reasonable structural basis for the protein-ligand interaction. There is a more complete desolvation of the peloruside A ligand and a greater array of favorable hydrophobic and electrostatic interactions exhibited by peloruside A at its ß-tubulin binding site. In addition, the protein architecture in our peloruside A binding model was suitable for binding laulimalide. With the generation of both laulimalide and peloruside A binding models, it was possible to delineate the structural basis for the greater activity of laulimalide relative to peloruside A and to rationalize the known structure-activity relationship data for both compounds.


Assuntos
Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Lactonas/farmacologia , Macrolídeos/farmacologia , Modelos Moleculares , Multimerização Proteica/efeitos dos fármacos , Trítio/química , Tubulina (Proteína)/química , Tubulina (Proteína)/metabolismo , Animais , Sítios de Ligação , Compostos Bicíclicos Heterocíclicos com Pontes/química , Compostos Bicíclicos Heterocíclicos com Pontes/metabolismo , Bovinos , Lactonas/química , Lactonas/metabolismo , Macrolídeos/química , Macrolídeos/metabolismo , Estrutura Quaternária de Proteína , Estereoisomerismo
17.
PLoS One ; 5(7): e11603, 2010 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-20657644

RESUMO

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.


Assuntos
Cinetocoros/metabolismo , Microtúbulos/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Recombinantes/metabolismo , Antimitóticos/farmacologia , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Proteínas do Citoesqueleto , Células HeLa , Humanos , Microscopia de Fluorescência , Proteínas Nucleares/genética , Ligação Proteica/efeitos dos fármacos , Proteínas Recombinantes/genética , Tubulina (Proteína)/metabolismo
18.
PLoS One ; 5(6): e11378, 2010 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-20614028

RESUMO

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.


Assuntos
Toxinas Botulínicas Tipo A/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Peptídeos/farmacologia , Catálise , Cristalização , Cristalografia por Raios X , Inibidores Enzimáticos/química , Modelos Moleculares , Peptídeos/química , Conformação Proteica , Especificidade por Substrato
19.
J Chem Pharm Res ; 2(5): 587-598, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-21593990

RESUMO

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.

20.
Antimicrob Agents Chemother ; 53(10): 4283-91, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19635954

RESUMO

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.


Assuntos
Antibacterianos/farmacologia , Bacillus anthracis/efeitos dos fármacos , Animais , Antibacterianos/química , Morte Celular/efeitos dos fármacos , Linhagem Celular , Bactérias Gram-Negativas/efeitos dos fármacos , Bactérias Gram-Positivas/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Macrófagos/microbiologia , Camundongos , Staphylococcus aureus/efeitos dos fármacos , Yersinia pestis/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...