Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Nat Chem Biol ; 20(3): 365-372, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37828400

RESUMEN

Stimulator of interferon genes (STING) is a dimeric transmembrane adapter protein that plays a key role in the human innate immune response to infection and has been therapeutically exploited for its antitumor activity. The activation of STING requires its high-order oligomerization, which could be induced by binding of the endogenous ligand, cGAMP, to the cytosolic ligand-binding domain. Here we report the discovery through functional screens of a class of compounds, named NVS-STGs, that activate human STING. Our cryo-EM structures show that NVS-STG2 induces the high-order oligomerization of human STING by binding to a pocket between the transmembrane domains of the neighboring STING dimers, effectively acting as a molecular glue. Our functional assays showed that NVS-STG2 could elicit potent STING-mediated immune responses in cells and antitumor activities in animal models.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Proteínas de la Membrana , Animales , Humanos , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Bioensayo , Citosol , Inmunidad Innata , Ligandos , Proteínas de la Membrana/metabolismo
2.
Cell Chem Biol ; 28(10): 1407-1419.e6, 2021 10 21.
Artículo en Inglés | MEDLINE | ID: mdl-33794192

RESUMEN

Three limonoid natural products with selective anti-proliferative activity against BRAF(V600E) and NRAS(Q61K)-mutation-dependent melanoma cell lines were identified. Differential transcriptome analysis revealed dependency of compound activity on expression of the mitochondrial cytochrome P450 oxidase CYP27A1, a transcriptional target of melanogenesis-associated transcription factor (MITF). We determined that CYP27A1 activity is necessary for the generation of a reactive metabolite that proceeds to inhibit cellular proliferation. A genome-wide small interfering RNA screen in combination with chemical proteomics experiments revealed gene-drug functional epistasis, suggesting that these compounds target mitochondrial biogenesis and inhibit tumor bioenergetics through a covalent mechanism. Our work suggests a strategy for melanoma-specific targeting by exploiting the expression of MITF target gene CYP27A1 and inhibiting mitochondrial oxidative phosphorylation in BRAF mutant melanomas.


Asunto(s)
Colestanotriol 26-Monooxigenasa/metabolismo , Limoninas/farmacología , Mitocondrias/efectos de los fármacos , Antineoplásicos/química , Antineoplásicos/metabolismo , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Productos Biológicos/química , Productos Biológicos/metabolismo , Productos Biológicos/farmacología , Productos Biológicos/uso terapéutico , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Colestanotriol 26-Monooxigenasa/antagonistas & inhibidores , Colestanotriol 26-Monooxigenasa/genética , Humanos , Limoninas/química , Limoninas/metabolismo , Limoninas/uso terapéutico , Melanoma/tratamiento farmacológico , Melanoma/patología , Factor de Transcripción Asociado a Microftalmía/genética , Factor de Transcripción Asociado a Microftalmía/metabolismo , Mitocondrias/metabolismo , Fosforilación Oxidativa/efectos de los fármacos , Regiones Promotoras Genéticas , Unión Proteica , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas B-raf/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo
4.
SLAS Discov ; 25(4): 350-360, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31997692

RESUMEN

Protein turnover is highly regulated by the posttranslational process of ubiquitination. Deregulation of the ubiquitin proteasome system (UPS) has been implicated in cancer and neurodegenerative diseases, and modulating this system has proven to be a viable approach for therapeutic intervention. The development of novel technologies that enable high-throughput studies of substrate protein ubiquitination is key for UPS drug discovery. Conventional approaches for studying ubiquitination either have high protein requirements or rely on exogenous or modified ubiquitin moieties, thus limiting their utility. In order to circumvent these issues, we developed a high-throughput live-cell assay that combines the NanoBiT luminescence-based technology with tandem ubiquitin binding entities (TUBEs) to resolve substrate ubiquitination. To demonstrate the effectiveness and utility of this assay, we studied compound-induced ubiquitination of the G to S Phase Transition 1 (GSPT1) protein. Using this assay, we characterized compounds with varying levels of GSPT1 ubiquitination activity. This method provides a live-cell-based approach for assaying substrate ubiquitination that can be adapted to study the kinetics of ubiquitin transfer onto a substrate protein of interest. In addition, our results show that this approach is portable for studying the ubiquitination of target proteins with diverse functions.


Asunto(s)
Descubrimiento de Drogas , Ensayos Analíticos de Alto Rendimiento , Complejo de la Endopetidasa Proteasomal/genética , Ubiquitina/genética , Humanos , Luminiscencia , Unión Proteica/genética , Transporte de Proteínas/genética , Ubiquitinación/genética
5.
Nat Chem Biol ; 16(1): 50-59, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31819276

RESUMEN

The post-genomic era has seen many advances in our understanding of cancer pathways, yet resistance and tumor heterogeneity necessitate multiple approaches to target even monogenic tumors. Here, we combine phenotypic screening with chemical genetics to identify pre-messenger RNA endonuclease cleavage and polyadenylation specificity factor 3 (CPSF3) as the target of JTE-607, a small molecule with previously unknown target. We show that CPSF3 represents a synthetic lethal node in a subset of acute myeloid leukemia (AML) and Ewing's sarcoma cancer cell lines. Inhibition of CPSF3 by JTE-607 alters expression of known downstream effectors in AML and Ewing's sarcoma lines, upregulates apoptosis and causes tumor-selective stasis in mouse xenografts. Mechanistically, it prevents the release of newly synthesized pre-mRNAs, resulting in read-through transcription and the formation of DNA-RNA hybrid R-loop structures. This study implicates pre-mRNA processing, and specifically CPSF3, as a druggable target providing an avenue to therapeutic intervention in cancer.


Asunto(s)
Factor de Especificidad de Desdoblamiento y Poliadenilación/metabolismo , Leucemia Mieloide Aguda/metabolismo , Precursores del ARN/metabolismo , Sarcoma de Ewing/metabolismo , Animales , Apoptosis/efectos de los fármacos , Sitios de Unión , Hidrolasas de Éster Carboxílico/metabolismo , Línea Celular Tumoral , Supervivencia Celular , Factor de Especificidad de Desdoblamiento y Poliadenilación/genética , Células HEK293 , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Masculino , Espectrometría de Masas , Ratones , Ratones Endogámicos C57BL , Trasplante de Neoplasias , Fenotipo , Fenilalanina/análogos & derivados , Fenilalanina/farmacología , Piperazinas/farmacología , Unión Proteica , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Sarcoma de Ewing/tratamiento farmacológico
6.
ACS Chem Biol ; 13(4): 1066-1081, 2018 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-29553248

RESUMEN

We screened a library of bioactive small molecules for activators and inhibitors of innate immune signaling through IRF3 and NFkB pathways with the goals of advancing pathway understanding and discovering probes for immunology research. We used high content screening to measure the translocation from the cytoplasm to nucleus of IRF3 and NFkB in primary human macrophages; these transcription factors play a critical role in the activation of STING and other pro-inflammatory pathways. Our pathway activator screen yielded a diverse set of hits that promoted nuclear translocation of IRF3 and/or NFkB, but the majority of these compounds did not cause activation of downstream pathways. Screening for antagonists of the STING pathway yielded multiple kinase inhibitors, some of which inhibit kinases not previously known to regulate the activity of this pathway. Structure-activity relationships (SARs) and subsequent chemical proteomics experiments suggested that MAPKAPK5 (PRAK) is a kinase that regulates IRF3 translocation in human macrophages. Our work establishes a high content screening approach for measuring pro-inflammatory pathways in human macrophages and identifies novel ways to inhibit such pathways; among the targets of the screen are several molecules that may merit further development as anti-inflammatory drugs.


Asunto(s)
Factor 3 Regulador del Interferón/antagonistas & inhibidores , Macrófagos/química , Proteínas de la Membrana/antagonistas & inhibidores , FN-kappa B/metabolismo , Bibliotecas de Moléculas Pequeñas/farmacología , Transporte Activo de Núcleo Celular/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Humanos , Péptidos y Proteínas de Señalización Intracelular/fisiología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/fisiología , Transducción de Señal/efectos de los fármacos
7.
Bioorg Med Chem Lett ; 27(20): 4589-4596, 2017 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-28911816

RESUMEN

Protein degradation is critical for proteostasis, and the addition of polyubiquitin chains to a substrate is necessary for its recognition by the 26S proteasome. Therapeutic intervention in the ubiquitin proteasome system has implications ranging from cancer to neurodegeneration. Novel screening methods and chemical biology tools for targeting E1-activating, E2-conjugating and deubiquitinating enzymes will be discussed in this review. Approaches for targeting E3 ligase-substrate interactions as well as the proteasome will also be covered, with a focus on recently described approaches.


Asunto(s)
Complejo de la Endopetidasa Proteasomal/metabolismo , Ubiquitina/metabolismo , Descubrimiento de Drogas , Humanos , Factores Inmunológicos/química , Factores Inmunológicos/metabolismo , Complejo de la Endopetidasa Proteasomal/química , Bibliotecas de Moléculas Pequeñas/química , Ubiquitina/antagonistas & inhibidores , Enzimas Activadoras de Ubiquitina/antagonistas & inhibidores , Enzimas Activadoras de Ubiquitina/metabolismo , Enzimas Ubiquitina-Conjugadoras/antagonistas & inhibidores , Enzimas Ubiquitina-Conjugadoras/metabolismo
8.
Elife ; 42015 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-26651998

RESUMEN

Cyclic peptide natural products have evolved to exploit diverse protein targets, many of which control essential cellular processes. Inspired by a series of cyclic peptides with partially elucidated structures, we designed synthetic variants of ternatin, a cytotoxic and anti-adipogenic natural product whose molecular mode of action was unknown. The new ternatin variants are cytotoxic toward cancer cells, with up to 500-fold greater potency than ternatin itself. Using a ternatin photo-affinity probe, we identify the translation elongation factor-1A ternary complex (eEF1A·GTP·aminoacyl-tRNA) as a specific target and demonstrate competitive binding by the unrelated natural products, didemnin and cytotrienin. Mutations in domain III of eEF1A prevent ternatin binding and confer resistance to its cytotoxic effects, implicating the adjacent hydrophobic surface as a functional hot spot for eEF1A modulation. We conclude that the eukaryotic elongation factor-1A and its ternary complex with GTP and aminoacyl-tRNA are common targets for the evolution of cytotoxic natural products.


Asunto(s)
Antineoplásicos/farmacología , Muerte Celular , Factor 1 de Elongación Peptídica/antagonistas & inhibidores , Péptidos Cíclicos/farmacología , Antineoplásicos/síntesis química , Línea Celular Tumoral , Resistencia a Medicamentos , Guanosina Trifosfato/metabolismo , Humanos , Proteínas Mutantes/antagonistas & inhibidores , Proteínas Mutantes/genética , Mutación , Factor 1 de Elongación Peptídica/genética , Péptidos Cíclicos/síntesis química , Unión Proteica , ARN de Transferencia/metabolismo
9.
Angew Chem Int Ed Engl ; 54(35): 10149-54, 2015 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-26179970

RESUMEN

Cultivation of myxobacteria of the Nannocystis genus led to the isolation and structure elucidation of a class of novel cyclic lactone inhibitors of elongation factor 1. Whole genome sequence analysis and annotation enabled identification of the putative biosynthetic cluster and synthesis process. In biological assays the compounds displayed anti-fungal and cytotoxic activity. Combined genetic and proteomic approaches identified the eukaryotic translation elongation factor 1α (EF-1α) as the primary target for this compound class. Nannocystin A (1) displayed differential activity across various cancer cell lines and EEF1A1 expression levels appear to be the main differentiating factor. Biochemical and genetic evidence support an overlapping binding site of 1 with the anti-cancer compound didemnin B on EF-1α. This myxobacterial chemotype thus offers an interesting starting point for further investigations of the potential of therapeutics targeting elongation factor 1.


Asunto(s)
Antineoplásicos/química , Proliferación Celular/efectos de los fármacos , Compuestos Macrocíclicos/farmacología , Myxococcales/fisiología , Neoplasias/patología , Factor 1 de Elongación Peptídica/antagonistas & inhibidores , Antifúngicos/química , Antifúngicos/farmacología , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Candida albicans/efectos de los fármacos , Genómica/métodos , Humanos , Compuestos Macrocíclicos/química , Estructura Molecular , Neoplasias/tratamiento farmacológico , Factor 1 de Elongación Peptídica/genética , Factor 1 de Elongación Peptídica/metabolismo , Proteómica/métodos , Relación Estructura-Actividad , Células Tumorales Cultivadas
10.
PLoS One ; 10(6): e0127498, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26098886

RESUMEN

Englerin A is a structurally unique natural product reported to selectively inhibit growth of renal cell carcinoma cell lines. A large scale phenotypic cell profiling experiment (CLiP) of englerin A on ¬over 500 well characterized cancer cell lines showed that englerin A inhibits growth of a subset of tumor cell lines from many lineages, not just renal cell carcinomas. Expression of the TRPC4 cation channel was the cell line feature that best correlated with sensitivity to englerin A, suggesting the hypothesis that TRPC4 is the efficacy target for englerin A. Genetic experiments demonstrate that TRPC4 expression is both necessary and sufficient for englerin A induced growth inhibition. Englerin A induces calcium influx and membrane depolarization in cells expressing high levels of TRPC4 or its close ortholog TRPC5. Electrophysiology experiments confirmed that englerin A is a TRPC4 agonist. Both the englerin A induced current and the englerin A induced growth inhibition can be blocked by the TRPC4/C5 inhibitor ML204. These experiments confirm that activation of TRPC4/C5 channels inhibits tumor cell line proliferation and confirms the TRPC4 target hypothesis generated by the cell line profiling. In selectivity assays englerin A weakly inhibits TRPA1, TRPV3/V4, and TRPM8 which suggests that englerin A may bind a common feature of TRP ion channels. In vivo experiments show that englerin A is lethal in rodents near doses needed to activate the TRPC4 channel. This toxicity suggests that englerin A itself is probably unsuitable for further drug development. However, since englerin A can be synthesized in the laboratory, it may be a useful chemical starting point to identify novel modulators of other TRP family channels.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Sesquiterpenos de Guayano/farmacología , Canales Catiónicos TRPC/agonistas , Animales , Antineoplásicos/farmacología , Carcinoma de Células Renales/tratamiento farmacológico , Línea Celular Tumoral , Células HEK293 , Humanos , Indoles/farmacología , Neoplasias Renales/tratamiento farmacológico , Ratones , Ratones Desnudos , Piperidinas/farmacología , Interferencia de ARN , ARN Interferente Pequeño , Ratas , Canales Catiónicos TRPC/antagonistas & inhibidores , Canales Catiónicos TRPC/genética , Transfección
11.
Front Pharmacol ; 5: 121, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24904423

RESUMEN

The drug discovery process classically revolves around a set of biochemical and cellular assays to drive potency optimization and structural-activity relationship models. Layered on top of these concepts is the inclusion of molecular features that affect final drug use, things like: bioavailability, toxicity, stability, solubility, formulation, route of administration, etc. Paradoxically, most drugs entering clinical trials are only tested in a handful of human genetic backgrounds before they are given to people. Here we review efforts and opine on the use of large scale in vitro cellular and in vivo models that attempt to model human disease and include diversity found in the human genetic population. Because hundreds to thousands of individual assays are needed to scratch the surface of human genetic diversity, sophisticated high throughput automation technologies or pooling and deconvolution strategies are required. Characterization of each model needs to be extensive to enable non-biased informatics based modeling. Such approaches will enable deep understanding of genetic to pharmacological response and result in new methods for patient stratification in the clinic. Oncology medicines and cancer genetics have been paving the way for these approaches and we expect to see continued expansion to other fields such as immunology and neuroscience.

12.
Nat Chem Biol ; 9(8): 514-20, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23728495

RESUMEN

Cell-based therapies hold the potential to alleviate the growing burden of liver diseases. Such therapies require human hepatocytes, which, within the stromal context of the liver, are capable of many rounds of replication. However, this ability is lost ex vivo, and human hepatocyte sourcing has limited many fields of research for decades. Here we developed a high-throughput screening platform for primary human hepatocytes to identify small molecules in two different classes that can be used to generate renewable sources of functional human hepatocytes. The first class induced functional proliferation of primary human hepatocytes in vitro. The second class enhanced hepatocyte functions and promoted the differentiation of induced pluripotent stem cell-derived hepatocytes toward a more mature phenotype than what was previously obtainable. The identification of these small molecules can help address a major challenge affecting many facets of liver research and may lead to the development of new therapeutics for liver diseases.


Asunto(s)
Hepatocitos/citología , Hepatocitos/efectos de los fármacos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología , Diferenciación Celular/efectos de los fármacos , Proliferación Celular , Relación Dosis-Respuesta a Droga , Ensayos Analíticos de Alto Rendimiento , Humanos , Estructura Molecular , Bibliotecas de Moléculas Pequeñas/química , Relación Estructura-Actividad
13.
ACS Med Chem Lett ; 3(12): 1034-1038, 2012 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-23256033

RESUMEN

The KRAS oncogene is found in up to 30% of all human tumors. In 2009, RNAi experiments revealed that lowering mRNA levels of a transcript encoding the serine/threonine kinase STK33 was selectively toxic to KRAS-dependent cancer cell lines, suggesting that small-molecule inhibitors of STK33 might selectively target KRAS-dependent cancers. To test this hypothesis, we initiated a high-throughput screen using compounds in the Molecular Libraries Small Molecule Repository (MLSMR). Several hits were identified, and one of these, a quinoxalinone derivative, was optimized. Extensive SAR studies were performed and led to the chemical probe ML281 that showed low nanomolar inhibition of purified recombinant STK33 and a distinct selectivity profile as compared to other STK33 inhibitors that were reported in the course of these studies. Even at the highest concentration tested (10 µM), ML281 had no effect on the viability of KRAS-dependent cancer cells. These results are consistent with other recent reports using small-molecule STK33 inhibitors. Small molecules having different chemical structures and kinase-selectivity profiles are needed to fully understand the role of STK33 in KRAS-dependent cancers. In this regard, ML281 is a valuable addition to small-molecule probes of STK33.

14.
J Clin Invest ; 122(4): 1377-92, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22406537

RESUMEN

In Alzheimer disease (AD), amyloid ß peptide (Aß) accumulates in plaques in the brain. Receptor for advanced glycation end products (RAGE) mediates Aß-induced perturbations in cerebral vessels, neurons, and microglia in AD. Here, we identified a high-affinity RAGE-specific inhibitor (FPS-ZM1) that blocked Aß binding to the V domain of RAGE and inhibited Aß40- and Aß42-induced cellular stress in RAGE-expressing cells in vitro and in the mouse brain in vivo. FPS-ZM1 was nontoxic to mice and readily crossed the blood-brain barrier (BBB). In aged APPsw/0 mice overexpressing human Aß-precursor protein, a transgenic mouse model of AD with established Aß pathology, FPS-ZM1 inhibited RAGE-mediated influx of circulating Aß40 and Aß42 into the brain. In brain, FPS-ZM1 bound exclusively to RAGE, which inhibited ß-secretase activity and Aß production and suppressed microglia activation and the neuroinflammatory response. Blockade of RAGE actions at the BBB and in the brain reduced Aß40 and Aß42 levels in brain markedly and normalized cognitive performance and cerebral blood flow responses in aged APPsw/0 mice. Our data suggest that FPS-ZM1 is a potent multimodal RAGE blocker that effectively controls progression of Aß-mediated brain disorder and that it may have the potential to be a disease-modifying agent for AD.


Asunto(s)
Enfermedad de Alzheimer/prevención & control , Péptidos beta-Amiloides/metabolismo , Benzamidas/uso terapéutico , Modelos Animales de Enfermedad , Fármacos Neuroprotectores/uso terapéutico , Fragmentos de Péptidos/metabolismo , Receptores Inmunológicos/antagonistas & inhibidores , Envejecimiento/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Péptidos beta-Amiloides/genética , Animales , Benzamidas/farmacología , Benzamidas/toxicidad , Barrera Hematoencefálica/efectos de los fármacos , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Células CHO/efectos de los fármacos , Circulación Cerebrovascular/efectos de los fármacos , Cricetinae , Cricetulus , Evaluación Preclínica de Medicamentos , Humanos , Masculino , Ratones , Ratones Transgénicos , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/toxicidad , Fragmentos de Péptidos/genética , Desempeño Psicomotor/efectos de los fármacos , Receptor para Productos Finales de Glicación Avanzada , Proteínas Recombinantes de Fusión/metabolismo , Bibliotecas de Moléculas Pequeñas
15.
Proc Natl Acad Sci U S A ; 109(8): 2860-5, 2012 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-22323609

RESUMEN

Approximately 30% of human cancers harbor oncogenic gain-of-function mutations in KRAS. Despite interest in KRAS as a therapeutic target, direct blockade of KRAS function with small molecules has yet to be demonstrated. Based on experiments that lower mRNA levels of protein kinases, KRAS-dependent cancer cells were proposed to have a unique requirement for the serine/threonine kinase STK33. Thus, it was suggested that small-molecule inhibitors of STK33 might have therapeutic benefit in these cancers. Here, we describe the development of selective, low nanomolar inhibitors of STK33's kinase activity. The most potent and selective of these, BRD8899, failed to kill KRAS-dependent cells. While several explanations for this result exist, our data are most consistent with the view that inhibition of STK33's kinase activity does not represent a promising anti-KRAS therapeutic strategy.


Asunto(s)
Isoquinolinas/química , Neoplasias/enzimología , Neoplasias/patología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/metabolismo , Proteínas ras/metabolismo , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/análogos & derivados , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/farmacología , Bioensayo , Biomarcadores de Tumor/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Ensayos Analíticos de Alto Rendimiento , Humanos , Isoquinolinas/farmacología , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/química , Proteínas Serina-Treonina Quinasas/metabolismo , Proteómica , Proteínas Proto-Oncogénicas p21(ras) , Bibliotecas de Moléculas Pequeñas/farmacología
16.
J Am Chem Soc ; 132(47): 16962-76, 2010 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-21067169

RESUMEN

An aldol-based build/couple/pair (B/C/P) strategy was applied to generate a collection of stereochemically and skeletally diverse small molecules. In the build phase, a series of asymmetric syn- and anti-aldol reactions were performed to produce four stereoisomers of a Boc-protected γ-amino acid. In addition, both stereoisomers of O-PMB-protected alaninol were generated to provide a chiral amine coupling partner. In the couple step, eight stereoisomeric amides were synthesized by coupling the chiral acid and amine building blocks. The amides were subsequently reduced to generate the corresponding secondary amines. In the pair phase, three different reactions were employed to enable intramolecular ring-forming processes: nucleophilic aromatic substitution (S(N)Ar), Huisgen [3+2] cycloaddition, and ring-closing metathesis (RCM). Despite some stereochemical dependencies, the ring-forming reactions were optimized to proceed with good to excellent yields, providing a variety of skeletons ranging in size from 8- to 14-membered rings. Scaffolds resulting from the RCM pairing reaction were diversified on the solid phase to yield a 14 400-membered library of macrolactams. Screening of this library led to the discovery of a novel class of histone deacetylase inhibitors, which display mixed enzyme inhibition, and led to increased levels of acetylation in a primary mouse neuron culture. The development of stereo-structure/activity relationships was made possible by screening all 16 stereoisomers of the macrolactams produced through the aldol-based B/C/P strategy.


Asunto(s)
Aldehídos/química , Descubrimiento de Drogas/métodos , Inhibidores de Histona Desacetilasas/síntesis química , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/metabolismo , Compuestos Macrocíclicos/síntesis química , Compuestos Macrocíclicos/farmacología , Animales , Productos Biológicos/síntesis química , Productos Biológicos/química , Productos Biológicos/farmacología , Evaluación Preclínica de Medicamentos , Inhibidores de Histona Desacetilasas/química , Compuestos Macrocíclicos/química , Ratones , Modelos Moleculares , Conformación Molecular , Estereoisomerismo , Especificidad por Sustrato
17.
J Am Chem Soc ; 132(19): 6749-54, 2010 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-20415446

RESUMEN

Artificially controlled cell recognition has potentially far-reaching applications in both the understanding and altering of biological function. The event of recognition often involves a multimeric protein binding a cellular membrane. While such an interaction is energetically favorable, it has been surprisingly underexploited in artificial control of recognition. Herein we describe how changing properties of substrate (phosphocholine, PC) self-assembly can affect both binding behavior and substrate affinity to a pentameric recognition protein (C-reactive protein, CRP). PC was modified with a short, self-assembling DNA strand to make the substrate self-assembly sensitive and responsive to ionic environment. A significant shift in CRP binding affinity was observed when substrates were assembled in the presence of Cs(+) rather than K(+). Furthermore, alteration of the linker length tethering PC to DNA showed trends similar to other multivalent systems. In optimizing these linker lengths, positive cooperativity increased and K(d) of the substrate assembly to CRP improved roughly 1000-fold. Such experiments both inform our understanding of biological, multivalent interactions in self-assembling systems and present a potential method to exogenously control events in cell recognition.


Asunto(s)
Multimerización de Proteína , Proteína C-Reactiva/química , Proteína C-Reactiva/metabolismo , Metales Alcalinos/metabolismo , Oligodesoxirribonucleótidos/metabolismo , Fosforilcolina/metabolismo , Unión Proteica , Estructura Cuaternaria de Proteína , Termodinámica
18.
Philos Trans A Math Phys Eng Sci ; 368(1914): 989-1008, 2010 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-20123744

RESUMEN

Proteins modulate the majority of all biological functions and are primarily composed of highly organized secondary structural elements such as helices, turns and sheets. Many of these functions are affected by a small number of key protein-protein contacts, often involving one or more of these well-defined structural elements. Given the ubiquitous nature of these protein recognition domains, their mimicry by peptidic and non-peptidic scaffolds has become a major focus of contemporary research. This review examines several key advances in secondary structure mimicry over the past several years, particularly focusing upon scaffolds that show not only promising projection of functional groups, but also a proven effect in biological systems.


Asunto(s)
Materiales Biomiméticos/química , Materiales Biomiméticos/síntesis química , Biomimética/métodos , Proteínas/química , Secuencia de Aminoácidos , Materiales Biomiméticos/metabolismo , Humanos , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Proteínas/metabolismo
20.
ChemMedChem ; 4(4): 649-56, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19330783

RESUMEN

The Bcl-x(L)/Bak protein-protein interaction has emerged as an important target for cancer therapy due to its role in apoptosis. Inhibition of this interaction by small-molecule antagonists induces apoptosis in unhealthy cells. Bak, a pro-apoptotic Bcl-2 protein, projects four hydrophobic side chains (V74, L78, I81, and I85), corresponding to the i, i+4, i+7, and i+11 positions of an alpha-helix, into a hydrophobic cleft on Bcl-x(L). Herein, we present a novel family of rationally designed alpha-helix mimetics with improved solubility and synthetic feasibility based on a benzoylurea scaffold. These benzoylurea derivatives favor a linear conformation stabilized by an intramolecular hydrogen bond, and are able to mimic the spatial projection of the i, i+4, and i+7 residues of an alpha-helix. The binding of the benzoylurea derivatives to Bcl-x(L) was assessed using fluorescence polarization competition assays, isothermal titration calorimetry, and (15)N-HSQC experiments. These experiments showed that these agents bind to and disrupt Bcl-x(L) with low micromolar inhibition and dissociation constants, with (15)N-HSQC experiments confirming binding to the hydrophobic pocket of Bcl-x(L) normally occupied by the Bak helix.


Asunto(s)
Benceno/química , Materiales Biomiméticos/química , Urea/química , Proteína Destructora del Antagonista Homólogo bcl-2/química , Proteína Destructora del Antagonista Homólogo bcl-2/metabolismo , Proteína bcl-X/química , Proteína bcl-X/metabolismo , Calorimetría , Cristalografía por Rayos X , Modelos Moleculares , Estructura Molecular , Resonancia Magnética Nuclear Biomolecular , Unión Proteica , Relación Estructura-Actividad , Termodinámica , Volumetría
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA