Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Cell ; 187(10): 2465-2484.e22, 2024 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-38701782

RESUMEN

Remyelination failure in diseases like multiple sclerosis (MS) was thought to involve suppressed maturation of oligodendrocyte precursors; however, oligodendrocytes are present in MS lesions yet lack myelin production. We found that oligodendrocytes in the lesions are epigenetically silenced. Developing a transgenic reporter labeling differentiated oligodendrocytes for phenotypic screening, we identified a small-molecule epigenetic-silencing-inhibitor (ESI1) that enhances myelin production and ensheathment. ESI1 promotes remyelination in animal models of demyelination and enables de novo myelinogenesis on regenerated CNS axons. ESI1 treatment lengthened myelin sheaths in human iPSC-derived organoids and augmented (re)myelination in aged mice while reversing age-related cognitive decline. Multi-omics revealed that ESI1 induces an active chromatin landscape that activates myelinogenic pathways and reprograms metabolism. Notably, ESI1 triggered nuclear condensate formation of master lipid-metabolic regulators SREBP1/2, concentrating transcriptional co-activators to drive lipid/cholesterol biosynthesis. Our study highlights the potential of targeting epigenetic silencing to enable CNS myelin regeneration in demyelinating diseases and aging.


Asunto(s)
Epigénesis Genética , Vaina de Mielina , Oligodendroglía , Remielinización , Animales , Vaina de Mielina/metabolismo , Humanos , Ratones , Remielinización/efectos de los fármacos , Oligodendroglía/metabolismo , Sistema Nervioso Central/metabolismo , Ratones Endogámicos C57BL , Rejuvenecimiento , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Organoides/metabolismo , Organoides/efectos de los fármacos , Enfermedades Desmielinizantes/metabolismo , Enfermedades Desmielinizantes/genética , Diferenciación Celular/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología , Masculino , Regeneración/efectos de los fármacos , Esclerosis Múltiple/metabolismo , Esclerosis Múltiple/genética , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/patología
2.
J Biol Chem ; 290(20): 12879-98, 2015 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-25825487

RESUMEN

The Ras family small GTPases regulate multiple cellular processes, including cell growth, survival, movement, and gene expression, and are intimately involved in cancer pathogenesis. Activation of these small GTPases is catalyzed by a special class of enzymes, termed guanine nucleotide exchange factors (GEFs). Herein, we developed a small molecule screening platform for identifying lead hits targeting a Ras GEF enzyme, SOS1. We employed an ensemble structure-based virtual screening approach in combination with a multiple tier high throughput experimental screen utilizing two complementary fluorescent guanine nucleotide exchange assays to identify small molecule inhibitors of GEF catalytic activity toward Ras. From a library of 350,000 compounds, we selected a set of 418 candidate compounds predicted to disrupt the GEF-Ras interaction, of which dual wavelength GDP dissociation and GTP-loading experimental screening identified two chemically distinct small molecule inhibitors. Subsequent biochemical validations indicate that they are capable of dose-dependently inhibiting GEF catalytic activity, binding to SOS1 with micromolar affinity, and disrupting GEF-Ras interaction. Mutagenesis studies in conjunction with structure-activity relationship studies mapped both compounds to different sites in the catalytic pocket, and both inhibited Ras signaling in cells. The unique screening platform established here for targeting Ras GEF enzymes could be broadly useful for identifying lead inhibitors for a variety of small GTPase-activating GEF reactions.


Asunto(s)
Inhibidores Enzimáticos , Proteínas Activadoras de ras GTPasa/antagonistas & inhibidores , Factores de Intercambio de Guanina Nucleótido ras/antagonistas & inhibidores , Relación Dosis-Respuesta a Droga , Sistemas de Liberación de Medicamentos/métodos , Evaluación Preclínica de Medicamentos/métodos , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Humanos , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Proteínas Activadoras de ras GTPasa/genética , Proteínas Activadoras de ras GTPasa/metabolismo , Factores de Intercambio de Guanina Nucleótido ras/genética , Factores de Intercambio de Guanina Nucleótido ras/metabolismo
3.
Anticancer Drugs ; 26(5): 518-30, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25646742

RESUMEN

The tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a potential therapeutic agent that induces apoptosis selectively in tumor cells. However, numerous solid tumor types are resistant to TRAIL. Sensitization to TRAIL has been an area of great research interest, but has met significant challenges because of poor bioavailability, half-life, and solubility of sensitizing compounds such as curcumin. Soluble, TRAIL-sensitizing compounds were screened on the basis of similarity to the redox-active substructure of curcumin and sensitization to TRAIL-induced apoptosis. We determined the effect of the lead compound, C25, in combination with TRAIL in human cancer cell lines using MTS proliferation assays, apoptosis assays, and western blotting. Short hairpin RNA knockdown of death receptor 5 (DR5) was performed to determine whether DR5 upregulation was required for TRAIL-mediated apoptosis. In-vivo efficacy was determined using human lung tumor xenograft models. C25 helped overcome TRAIL resistance by upregulating the expression of the TRAIL receptor DR5 and apoptosis in several tumor cell lines. Blockade of DR5 expression abrogated C25 sensitization to TRAIL, demonstrating the requirement for DR5 upregulation for C25-mediated potentiation of TRAIL-mediated apoptosis. The combination of C25 and TRAIL effectively inhibited tumorigenesis in vivo. This study demonstrates the synergistic efficacy of C25 in sensitization to TRAIL-induced apoptosis in multiple tumor cell types, including highly resistant lung and ovarian tumor cell lines. Furthermore, C25 was efficacious against tumor growth in vivo. Thus, C25 may be a potential therapeutic for cancer in combination with TRAIL or DR5 agonist therapy.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Hidrazonas/farmacología , Pirazoles/farmacología , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Animales , Antineoplásicos/química , Antineoplásicos/uso terapéutico , Caspasa 3/metabolismo , Línea Celular Tumoral , Bases de Datos de Compuestos Químicos , Ensayos de Selección de Medicamentos Antitumorales , Sinergismo Farmacológico , Femenino , Xenoinjertos , Humanos , Hidrazonas/química , Hidrazonas/uso terapéutico , Ratones Desnudos , Mitocondrias/metabolismo , Trasplante de Neoplasias , Pirazoles/química , Pirazoles/uso terapéutico , Transducción de Señal , Solubilidad , Ligando Inductor de Apoptosis Relacionado con TNF/uso terapéutico , Carga Tumoral/efectos de los fármacos , Regulación hacia Arriba
4.
Neuro Oncol ; 26(4): 735-748, 2024 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-38011799

RESUMEN

BACKGROUND: Diffuse intrinsic pontine gliomas (DIPG/DMG) are devastating pediatric brain tumors with extraordinarily limited treatment options and uniformly fatal prognosis. Histone H3K27M mutation is a common recurrent alteration in DIPG and disrupts epigenetic regulation. We hypothesize that genome-wide H3K27M-induced epigenetic dysregulation makes tumors vulnerable to epigenetic targeting. METHODS: We performed a screen of compounds targeting epigenetic enzymes to identify potential inhibitors for the growth of patient-derived DIPG cells. We further carried out transcriptomic and genomic landscape profiling including RNA-seq and CUT&RUN-seq as well as shRNA-mediated knockdown to assess the effects of chaetocin and SUV39H1, a target of chaetocin, on DIPG growth. RESULTS: High-throughput small-molecule screening identified an epigenetic compound chaetocin as a potent blocker of DIPG cell growth. Chaetocin treatment selectively decreased proliferation and increased apoptosis of DIPG cells and significantly extended survival in DIPG xenograft models, while restoring H3K27me3 levels. Moreover, the loss of H3K9 methyltransferase SUV39H1 inhibited DIPG cell growth. Transcriptomic and epigenomic profiling indicated that SUV39H1 loss or inhibition led to the downregulation of stemness and oncogenic networks including growth factor receptor signaling and stemness-related programs; however, D2 dopamine receptor (DRD2) signaling adaptively underwent compensatory upregulation conferring resistance. Consistently, a combination of chaetocin treatment with a DRD2 antagonist ONC201 synergistically increased the antitumor efficacy. CONCLUSIONS: Our studies reveal a therapeutic vulnerability of DIPG cells through targeting the SUV39H1-H3K9me3 pathway and compensatory signaling loops for treating this devastating disease. Combining SUV39H1-targeting chaetocin with other agents such as ONC201 may offer a new strategy for effective DIPG treatment.


Asunto(s)
Neoplasias del Tronco Encefálico , Glioma Pontino Intrínseco Difuso , Imidazoles , Piridinas , Pirimidinas , Niño , Humanos , Epigénesis Genética , Histonas/genética , Glioma Pontino Intrínseco Difuso/genética , Neoplasias del Tronco Encefálico/tratamiento farmacológico , Neoplasias del Tronco Encefálico/genética , Neoplasias del Tronco Encefálico/patología , Metiltransferasas/genética , Metiltransferasas/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Piperazinas
5.
Mol Pharmacol ; 81(6): 811-9, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22399488

RESUMEN

Proliferating cell nuclear antigen (PCNA), a potential anticancer target, forms a homotrimer and is required for DNA replication and numerous other cellular processes. The purpose of this study was to identify novel small molecules that modulate PCNA activity to affect tumor cell proliferation. An in silico screen of a compound library against a crystal structure of PCNA and a subsequent structural similarity search of the ZINC chemical database were carried out to derive relevant docking partners. Nine compounds, termed PCNA inhibitors (PCNA-Is), were selected for further characterization. PCNA-I1 selectively bound to PCNA trimers with a dissociation constant (K(d)) of ~0.2 to 0.4 µM. PCNA-Is promoted the formation of SDS-refractory PCNA trimers. PCNA-I1 dose- and time-dependently reduced the chromatin-associated PCNA in cells. Consistent with its effects on PCNA trimer stabilization, PCNA-I1 inhibited the growth of tumor cells of various tissue types with an IC(50) of ~0.2 µM, whereas it affected the growth of nontransformed cells at significantly higher concentrations (IC(50), ~1.6 µM). Moreover, uptake of BrdU was dose-dependently reduced in cells treated with PCNA-I1. Mechanistically the PCNA-Is mimicked the effect of PCNA knockdown by siRNA, inducing cancer cell arrest at both the S and G(2)/M phases. Thus, we have identified a class of compounds that can directly bind to PCNA, stabilize PCNA trimers, reduce PCNA association with chromatin, and inhibit tumor cell growth by inducing a cell cycle arrest. They are valuable tools in studying PCNA function and may be useful for future PCNA-targeted cancer therapy.


Asunto(s)
División Celular , Cromatina/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Animales , Ciclo Celular , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Concentración 50 Inhibidora , Masculino , Ratones , Antígeno Nuclear de Célula en Proliferación/efectos de los fármacos
6.
Bioorg Med Chem Lett ; 22(13): 4458-61, 2012 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-22608389

RESUMEN

As a continuation of our efforts to discover and develop small molecules as anticancer agents, we identified GRI-394837 as an initial hit from similarity search on RGD and its analogs. Based on GRI-394837, we designed and synthesized a focused set of novel chromenes (4a-e) in a single step using microwave method. All five compounds showed activity in the nanomolar range (IC(50): 7.4-640 nM) in two melanoma, three prostate and four glioma cancer cell lines. The chromene 4e is active against all the cell lines and particularly against the A172 human glioma cell line (IC(50): 7.4 nM). Interestingly, in vitro tubulin polymerization assay shows 4e to be a weak tubulin polymerization inhibitor but it shows very strong cytotoxicity in cellular assays, therefore there must be additional unknown mechanism(s) for the anticancer activity. Additionally, the strong antiproliferative activity was verified by one of the selected chromene (4a) by the NCI 60 cell line screen. These results strongly suggest that the novel chromenes could be further developed as a potential therapeutic agent for a variety of aggressive cancers.


Asunto(s)
Antineoplásicos/química , Benzopiranos/química , Antineoplásicos/síntesis química , Antineoplásicos/toxicidad , Astrocitos/efectos de los fármacos , Benzopiranos/síntesis química , Benzopiranos/toxicidad , Sitios de Unión , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Simulación por Computador , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Oligopéptidos/química , Estructura Terciaria de Proteína , Tubulina (Proteína)/química , Tubulina (Proteína)/metabolismo , Moduladores de Tubulina/síntesis química , Moduladores de Tubulina/química , Moduladores de Tubulina/toxicidad
7.
Sci Transl Med ; 14(635): eabb7695, 2022 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-35263148

RESUMEN

Dysregulation of innate immune signaling pathways is implicated in various hematologic malignancies. However, these pathways have not been systematically examined in acute myeloid leukemia (AML). We report that AML hematopoietic stem and progenitor cells (HSPCs) exhibit a high frequency of dysregulated innate immune-related and inflammatory pathways, referred to as oncogenic immune signaling states. Through gene expression analyses and functional studies in human AML cell lines and patient-derived samples, we found that the ubiquitin-conjugating enzyme UBE2N is required for leukemic cell function in vitro and in vivo by maintaining oncogenic immune signaling states. It is known that the enzyme function of UBE2N can be inhibited by interfering with thioester formation between ubiquitin and the active site. We performed in silico structure-based and cellular-based screens and identified two related small-molecule inhibitors UC-764864/65 that targeted UBE2N at its active site. Using these small-molecule inhibitors as chemical probes, we further revealed the therapeutic efficacy of interfering with UBE2N function. This resulted in the blocking of ubiquitination of innate immune- and inflammatory-related substrates in human AML cell lines. Inhibition of UBE2N function disrupted oncogenic immune signaling by promoting cell death of leukemic HSPCs while sparing normal HSPCs in vitro. Moreover, baseline oncogenic immune signaling states in leukemic cells derived from discrete subsets of patients with AML exhibited a selective dependency on UBE2N function in vitro and in vivo. Our study reveals that interfering with UBE2N abrogates leukemic HSPC function and underscores the dependency of AML cells on UBE2N-dependent oncogenic immune signaling states.


Asunto(s)
Leucemia Mieloide Aguda , Enzimas Ubiquitina-Conjugadoras , Proliferación Celular/genética , Humanos , Leucemia Mieloide Aguda/metabolismo , Oncogenes , Transducción de Señal/genética , Enzimas Ubiquitina-Conjugadoras/antagonistas & inhibidores , Enzimas Ubiquitina-Conjugadoras/genética , Enzimas Ubiquitina-Conjugadoras/metabolismo
8.
Rapid Commun Mass Spectrom ; 23(20): 3293-300, 2009 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-19757451

RESUMEN

Mass spectrometry (MS)-based high-throughput screening (HTS) has tremendous potential as an alternative to current screening methods due to its speed, sensitivity, reproducibility and label-free readout. We recently reported that a new generation matrix-assisted laser desorption/ionization triple quadrupole (MALDI-QqQ) mass spectrometer is ideally suited for a variety of enzyme assays and screening protocols. However, all the targets measured to date had peptide substrates that were easily monitored by selected ion monitoring (SIM) without interference from the MALDI matrix. To further extend the application to enzymes with small molecule, non-peptide substrates, we evaluated this method for measuring enzyme activity and inhibition of acetylcholinesterase (AChE). Due to the potential of MALDI matrix interference, multiple reaction monitoring (MRM) was investigated for selective MS/MS transitions and to accurately measure the conversion of acetylcholine into choline. Importantly, ionization, detection and MRM transition efficiency differences between the substrate and product can be overcome by pre-balancing the MRM transitions during method development, thus allowing for a direct readout of the enzyme activity using the ratio of the substrate and product signals. Further validation of the assay showed accurate concentration-dependent inhibition measurements of AChE with several known inhibitors. Finally, a small library of 1008 drug-like compounds was screened at a single dose (10 microM) and the top 10 inhibitors from this primary screen were validated in a secondary screen to determine the rank order of inhibitory potency for each compound. Collectively, these data demonstrate that a MALDI-QqQMS-based readout platform is amenable to measuring small molecule substrates and products and offers significant advantages over current HTS methods in terms of speed, sensitivity, reproducibility and reagent costs.


Asunto(s)
Acetilcolinesterasa/química , Inhibidores de la Colinesterasa/química , Inhibidores Enzimáticos/química , Proteínas de Peces/química , Bibliotecas de Moléculas Pequeñas/química , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción/métodos , Animales , Electrophorus/metabolismo , Proteínas de Peces/antagonistas & inhibidores , Unión Proteica , Especificidad por Sustrato
9.
Proteins ; 66(3): 538-46, 2007 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-17120228

RESUMEN

In this article we describe the application of structural biology methods to the discovery of novel potent inhibitors of methionine aminopeptidases. These enzymes are employed by the cells to cleave the N-terminal methionine from nascent peptides and proteins. As this is one of the critical steps in protein maturation, it is very likely that inhibitors of these enzymes may prove useful as novel antibacterial agents. Involvement of crystallography at the very early stages of the inhibitor design process resulted in serendipitous discovery of a new inhibitor class, the pyrazole-diamines. Atomic-resolution structures of several inhibitors bound to the enzyme illuminate a new mode of inhibitor binding.


Asunto(s)
Bacterias/enzimología , Inhibidores de Proteasas/farmacología , Aminopeptidasas/química , Aminopeptidasas/aislamiento & purificación , Bacterias/efectos de los fármacos , Proteínas Bacterianas/farmacología , Cristalización , Cristalografía por Rayos X , Cinética , Espectroscopía de Resonancia Magnética , Metionil Aminopeptidasas , Modelos Moleculares , Inhibidores de Proteasas/química , Conformación Proteica , Teoría Cuántica
10.
Med Chem ; 11(4): 400-6, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25219925

RESUMEN

Glioblastoma Multiforme (GBM) is a highly fatal disease and new chemotherapeutic agents are desperately needed to treat GBM patients. With the aim of identifying new antiglioma agents we screened the UC DDC library for compounds structurally related to the antiglioma lead molecule compound 1 (SP-6-27) and clinically used compound 2 (Azixa). We identified imidazoquinoline analog 3 (S-94403) as initial hit from the first screen which included the different heterocyclic sets of 15 compounds. Based on the initial hit 3 (S-94403), a second search was performed to explore the structure activity relationship (SAR) study on imidazoquinolines. Our SAR revealed that a N-phenyl with EDGs/EWGs at C9 position, a methyl group at C7 position and an aryl or hetero-aryl groups at C2 position essential for the anticancer activity. These two consecutive screenings have identified the compounds S-94403 (IC50 = 0.625 µM) and S-98950 (IC50 = 1.04 µM) as the most potent imidazoquinoline-based antiglioma agents.


Asunto(s)
Antineoplásicos/química , Descubrimiento de Drogas , Imidazoles/química , Quinolinas/química , Antineoplásicos/farmacología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Ensayos Analíticos de Alto Rendimiento , Humanos , Imidazoles/farmacología , Neuroglía/efectos de los fármacos , Neuroglía/patología , Quinolinas/farmacología , Bibliotecas de Moléculas Pequeñas , Relación Estructura-Actividad
11.
Pharmacol Res Perspect ; 3(2): e00115, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25729582

RESUMEN

Proliferating cell nuclear antigen (PCNA) plays an essential role in DNA replication and repair. Tumor cells express high levels of PCNA, identifying it as a potentially ideal target for cancer therapy. Previously, we identified nine compounds termed PCNA inhibitors (PCNA-Is) that bind directly to PCNA, stabilize PCNA trimer structure, reduce chromatin-associated PCNA, and selectively inhibit tumor cell growth. Of these compounds, PCNA-I1 was most potent. The purpose of this study is to further establish targeting of PCNA by PCNA-I1 and to identify PCNA-I1 analogs with superior potencies. We found that PCNA-I1 does not affect the level of chromatin-associated PCNA harboring point mutations at the predicted binding site of PCNA-I1. Forty-six PCNA-I1 analogs with structures of 1-hydrazonomethyl-2-hydroxy (scaffold A), 2-hydrazonomethyl-1-hydroxy (scaffold B), 2-hydrazonomethyl-3-hydroxy (scaffold C), and 4-pyridyl hydrazine (scaffold D) were analyzed for their effects on cell growth in four tumor cell lines and PCNA trimer stabilization. Compounds in scaffold group A and group B showed the highest trimer stabilization and the most potent cell growth inhibitory activities with a significant potency advantage observed in the Z isomers of scaffold A. The absence of trimer stabilization and growth inhibitory effects in compounds of scaffold group D confirms the essentiality of the hydroxynaphthyl substructure. Compounds structure-activity relationship (SAR)-6 and SAR-24 were analyzed for their effects on and found to reduce chromatin-associated PCNA in tumor cells. This study led to the identification of SAR-24, a compound with superior potencies and potentially improved solubility, which will be used for future development of PCNA-targeting cancer therapies.

12.
J Med Chem ; 46(17): 3655-61, 2003 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-12904069

RESUMEN

Novel quinolone antibacterial agents bearing (3S)-amino-(4R)-ethylpiperidines were designed by using low energy conformation analysis and synthesized by applying a conventional coupling reaction of the quinolone nuclei with new piperidine side chains. These compounds were tested in MIC assays and found to be highly potent against Gram-positive and Gram-negative organisms. In particular, the new compounds exhibited high activity against the resistant pathogens Staphylococcus aureus (MRCR) and Streptococcus pneumoniae (PR). Importantly, when the (3S)-amino-(4R)-ethylpiperidinyl quinolones were compared with marketed quinolones sharing the same quinolone nuclei but different side chains at the C-7 position, the new quinolones showed superior activity against Gram-positive organisms, including resistant pathogens.


Asunto(s)
Antibacterianos/síntesis química , Bacterias/efectos de los fármacos , Farmacorresistencia Bacteriana , Piperidinas/síntesis química , Quinolonas/síntesis química , Antibacterianos/química , Antibacterianos/farmacología , Girasa de ADN/química , Girasa de ADN/efectos de los fármacos , ADN Superhelicoidal/química , ADN Superhelicoidal/efectos de los fármacos , Escherichia coli/enzimología , Bacterias Gramnegativas/efectos de los fármacos , Bacterias Grampositivas/efectos de los fármacos , Pruebas de Sensibilidad Microbiana , Modelos Moleculares , Conformación Molecular , Piperidinas/química , Piperidinas/farmacología , Quinolonas/química , Quinolonas/farmacología , Estereoisomerismo , Relación Estructura-Actividad
13.
Chem Biol ; 21(12): 1618-28, 2014 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-25455859

RESUMEN

Ras GTPases regulate intracellular signaling involved in cell proliferation. Elevated Ras signaling activity has been associated with human cancers. Ras activation is catalyzed by guanine nucleotide exchange factors (GEFs), of which SOS1 is a major member that transduces receptor tyrosine kinase signaling to Ras. We have developed a rational approach coupling virtual screening with experimental screening in identifying small-molecule inhibitors targeting the catalytic site of SOS1 and SOS1-regulated Ras activity. A lead inhibitor, NSC-658497, was found to bind to SOS1, competitively suppress SOS1-Ras interaction, and dose-dependently inhibit SOS1 GEF activity. Mutagenesis and structure-activity relationship studies map the NSC-658497 site of action to the SOS1 catalytic site, and define the chemical moieties in the inhibitor essential for the activity. NSC-658497 showed dose-dependent efficacy in inhibiting Ras, downstream signaling activities, and associated cell proliferation. These studies establish a proof of principle for rational design of small-molecule inhibitors targeting Ras GEF enzymatic activity.


Asunto(s)
Diseño de Fármacos , Proteína SOS1/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Dominio Catalítico , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Humanos , Ratones , Modelos Moleculares , Mutagénesis , Proteína SOS1/química , Proteína SOS1/genética , Proteína SOS1/metabolismo , Transducción de Señal/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/química , Relación Estructura-Actividad
14.
Biosci Rep ; 33(5)2013 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-24001138

RESUMEN

Most currently available small molecule inhibitors of DNA replication lack enzymatic specificity, resulting in deleterious side effects during use in cancer chemotherapy and limited experimental usefulness as mechanistic tools to study DNA replication. Towards development of targeted replication inhibitors, we have focused on Mcm2-7 (minichromosome maintenance protein 2-7), a highly conserved helicase and key regulatory component of eukaryotic DNA replication. Unexpectedly we found that the fluoroquinolone antibiotic ciprofloxacin preferentially inhibits Mcm2-7. Ciprofloxacin blocks the DNA helicase activity of Mcm2-7 at concentrations that have little effect on other tested helicases and prevents the proliferation of both yeast and human cells at concentrations similar to those that inhibit DNA unwinding. Moreover, a previously characterized mcm mutant (mcm4chaos3) exhibits increased ciprofloxacin resistance. To identify more potent Mcm2-7 inhibitors, we screened molecules that are structurally related to ciprofloxacin and identified several that compromise the Mcm2-7 helicase activity at lower concentrations. Our results indicate that ciprofloxacin targets Mcm2-7 in vitro, and support the feasibility of developing specific quinolone-based inhibitors of Mcm2-7 for therapeutic and experimental applications.


Asunto(s)
Antibióticos Antineoplásicos/farmacología , Ciprofloxacina/farmacología , Proteínas de Mantenimiento de Minicromosoma/antagonistas & inhibidores , Proteínas de Saccharomyces cerevisiae/antagonistas & inhibidores , Animales , Línea Celular , Resistencia a Antineoplásicos , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Concentración 50 Inhibidora , Ofloxacino/farmacología , Saccharomyces cerevisiae/efectos de los fármacos , Saccharomyces cerevisiae/enzimología , Saccharomyces cerevisiae/crecimiento & desarrollo
15.
J Biomol Screen ; 18(7): 837-44, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23543431

RESUMEN

Myostatin, a member of the transforming growth factor (TGF)-ß family of secreted ligands, is a strong negative regulator of muscle growth. As such, therapeutic inhibitors of myostatin are actively being investigated for their potential in the treatment of muscle-wasting diseases such as muscular dystrophy and sarcopenia. Here, we sought to develop a high-throughput screening (HTS) method for small-molecule inhibitors that target myostatin. We created a HEK293 stable cell line that expresses the (CAGA)12-luciferase reporter construct and robustly responds to signaling of certain classes of TGF-ß family ligands. After optimization and miniaturization of the assay to a 384-well format, we successfully screened a library of compounds for inhibition of myostatin and the closely related activin A. Selection of some of the tested compounds was directed by in silico screening against myostatin, which led to an enrichment of target hits as compared with random selection. Altogether, we present an HTS method that will be useful for screening potential inhibitors of not only myostatin but also many other ligands of the TGF-ß family.


Asunto(s)
Activinas/antagonistas & inhibidores , Sustancias de Crecimiento/farmacología , Miostatina/antagonistas & inhibidores , Simulación por Computador , Expresión Génica/efectos de los fármacos , Genes Reporteros , Células HEK293 , Ensayos Analíticos de Alto Rendimiento , Humanos , Luciferasas de Renilla/biosíntesis , Luciferasas de Renilla/genética , Bibliotecas de Moléculas Pequeñas
16.
Cancer Biol Ther ; 10(12): 1256-61, 2010 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-20935504

RESUMEN

We performed a high-throughput screen to identify compounds with a selective ability to induce apoptosis in Akt-expressing cells without disrupting Bcl-xL-dependent survival. Results showed that a screening strategy based on Alamar Blue underrepresented the viability of Bcl-xL-expressing cells relative to Akt-expressing cells, possibly due to metabolic differences between the two cell survival programs. Using an alternative screen based on plasma membrane integrity, we identified several compounds that target Akt-dependent survival without toxic effect to Bcl-xL-dependent survival. These compounds enhanced the cytotoxic potential of rapamycin, a chemotherapeutic that inhibits survival signaling downstream of Akt. The results demonstrate a screening method and the subsequent identification of two compounds with selective activity in counteracting Akt-dependent cell survival.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Ensayos Analíticos de Alto Rendimiento , Proteínas Proto-Oncogénicas c-akt/metabolismo , Sirolimus/farmacología , Proteína bcl-X/metabolismo , Animales , Caspasa 3/metabolismo , Línea Celular , Membrana Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Descubrimiento de Drogas , Ensayos de Selección de Medicamentos Antitumorales/métodos , Ratones , Oxazinas , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Xantenos , Proteína bcl-X/genética
17.
J Biomol Screen ; 15(8): 1001-7, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20228278

RESUMEN

Current methods for high-throughput screening (HTS) use a serial process to evaluate compounds as inhibitors toward a single therapeutic target, but as the demand to reduce screening time and cost continues to grow, one solution is the development of multiplex technology. In this communication, the multiplex assay capability of a mass spectrometry (MS)-based readout system is verified using a kinase and esterase reaction simultaneously. Furthermore, the MS-based readout is shown to be compatible with a typical HTS workflow by identifying and validating several new inhibitors for each enzyme from a small library of compounds. These data confirm that it is possible to monitor inhibition of multiple therapeutic targets with one pass through the compound repository, thus demonstrating the potential for MS-based methods to become a method of choice for HTS of isolated enzymes.


Asunto(s)
Pruebas de Enzimas/métodos , Inhibidores Enzimáticos/análisis , Inhibidores Enzimáticos/aislamiento & purificación , Ensayos Analíticos de Alto Rendimiento/métodos , Espectrometría de Masas/métodos , Acetilcolinesterasa/metabolismo , Calibración , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Pruebas de Enzimas/normas , Inhibidores Enzimáticos/farmacología , Proteínas Ligadas a GPI/antagonistas & inhibidores , Proteínas Ligadas a GPI/metabolismo , Ensayos Analíticos de Alto Rendimiento/normas , Humanos , Concentración 50 Inhibidora , Espectrometría de Masas/normas , Modelos Biológicos , Bibliotecas de Moléculas Pequeñas/análisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA