Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
ACS Chem Biol ; 10(4): 925-32, 2015 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-25602368

RESUMEN

Platelet-activating factor acetylhydrolases (PAFAHs) 1b2 and 1b3 are poorly characterized serine hydrolases that form a complex with a noncatalytic protein (1b1) to regulate brain development, spermatogenesis, and cancer pathogenesis. Determining physiological substrates and biochemical functions for the PAFAH1b complex would benefit from selective chemical probes that can perturb its activity in living systems. Here, we report a class of tetrahydropyridine reversible inhibitors of PAFAH1b2/3 discovered using a fluorescence polarization-activity-based protein profiling (fluopol-ABPP) screen of the NIH 300,000+ compound library. The most potent of these agents, P11, exhibited IC50 values of ∼40 and 900 nM for PAFAH1b2 and 1b3, respectively. We confirm selective inhibition of PAFAH1b2/3 in cancer cells by P11 using an ABPP protocol adapted for in situ analysis of reversible inhibitors and show that this compound impairs tumor cell survival, supporting a role for PAFAH1b2/3 in cancer.


Asunto(s)
1-Alquil-2-acetilglicerofosfocolina Esterasa/antagonistas & inhibidores , Evaluación Preclínica de Medicamentos/métodos , 1-Alquil-2-acetilglicerofosfocolina Esterasa/genética , 1-Alquil-2-acetilglicerofosfocolina Esterasa/metabolismo , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Línea Celular , Línea Celular Tumoral/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Polarización de Fluorescencia/métodos , Humanos , Concentración 50 Inhibidora , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Ratones , Proteómica/métodos , Piridinas/química , Piridinas/farmacología , Bibliotecas de Moléculas Pequeñas/farmacología , Relación Estructura-Actividad
2.
ACS Chem Biol ; 10(2): 364-71, 2015 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-25457457

RESUMEN

Mycobacterium tuberculosis (Mtb) maintains its intrabacterial pH (pHIB) near neutrality in the acidic environment of phagosomes within activated macrophages. A previously reported genetic screen revealed that Mtb loses this ability when the mycobacterial acid resistance protease (marP) gene is disrupted. In the present study, a high throughput screen (HTS) of compounds against the protease domain of MarP identified benzoxazinones as inhibitors of MarP. A potent benzoxazinone, BO43 (6-chloro-2-(2'-methylphenyl)-4H-1,3-benzoxazin-4-one), acylated MarP and lowered Mtb's pHIB and survival during incubation at pH 4.5. BO43 had similar effects on MarP-deficient Mtb, suggesting the existence of additional target(s). Reaction of an alkynyl-benzoxazinone, BO43T, with Mycobacterium bovis variant bacille Calmette-Guérin (BCG) followed by click chemistry with azido-biotin identified both the MarP homologue and the high temperature requirement A1 (HtrA1) homologue, an essential protein. Thus, the chemical probe identified through a target-based screen not only reacted with its intended target in the intact cells but also implicated an additional enzyme that had eluded a genetic screen biased against essential genes.


Asunto(s)
Homeostasis , Mycobacterium tuberculosis/enzimología , Mycobacterium tuberculosis/metabolismo , Periplasma/enzimología , Serina Proteasas/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Benzoxazinas/química , Benzoxazinas/farmacología , Regulación Bacteriana de la Expresión Génica/fisiología , Regulación Enzimológica de la Expresión Génica/fisiología , Concentración de Iones de Hidrógeno , Sondas Moleculares/química , Sondas Moleculares/metabolismo , Estructura Molecular , Mycobacterium tuberculosis/citología , Mycobacterium tuberculosis/genética , Serina Proteasas/genética , Inhibidores de Serina Proteinasa/química , Inhibidores de Serina Proteinasa/farmacología
3.
Biochemistry ; 52(51): 9187-201, 2013 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-24320749

RESUMEN

Myeloperoxidase (MPO) is a heme peroxidase that catalyzes the production of hypochlorous acid. Despite a high level of interest in MPO as a therapeutic target, there have been limited reports about MPO inhibitors that are suitable for evaluating MPO in pharmacological studies. 2-Thioxanthine, 3-(2-ethoxypropyl)-2-thioxo-2,3-dihydro-1H-purin-6(9H)-one (A), has recently been reported to inhibit MPO by covalently modifying the heme prosthetic group. Here we report a detailed mechanistic characterization demonstrating that A possesses all the distinguishing features of a mechanism-based inactivator. A is a time-dependent MPO inhibitor and displays saturable inactivation kinetics consistent with a two-step mechanism of inactivation and a potency (k(inact)/K(I) ratio) of 8450 ± 780 M⁻¹ s⁻¹. MPO inactivation by A is dependent on MPO catalysis and is protected by substrate. A reduces MPO compound I to compound II with a second-order rate constant of (0.801 ± 0.056) × 106 M⁻¹ s⁻¹, and its irreversible inactivation of MPO occurs prior to release of the activated inhibitory species. Despite its relatively high selectivity against a broad panel of more than 100 individual targets, including enzymes, receptors, transporters, and ion channels, we demonstrate that A labels multiple other protein targets in the presence of MPO. By synthesizing an alkyne analogue of A and utilizing click chemistry-activity-based protein profiling, we present that the MPO-activated inhibitory species can diffuse away to covalently modify other proteins, as reflected by the relatively high partition ratio of A, which we determined to be 15.6. This study highlights critical methods that can guide the discovery and development of next-generation MPO inhibitors.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Peroxidasa/antagonistas & inhibidores , Profármacos/farmacología , Tionas/farmacología , Xantinas/farmacología , Alquinos/síntesis química , Alquinos/química , Alquinos/farmacología , Unión Competitiva , Biocatálisis , Química Clic , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/metabolismo , Humanos , Peróxido de Hidrógeno/metabolismo , Cinética , Hígado/enzimología , Hígado/metabolismo , Oxazinas/metabolismo , Peroxidasa/química , Peroxidasa/metabolismo , Profármacos/química , Profármacos/metabolismo , Proteoma/química , Solubilidad , Tionas/síntesis química , Tionas/química , Tionas/metabolismo , Xantinas/síntesis química , Xantinas/química , Xantinas/metabolismo
4.
J Med Chem ; 56(21): 8257-69, 2013 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-24152245

RESUMEN

We have previously shown that 1,2,3-triazole ureas (1,2,3-TUs) act as versatile class of irreversible serine hydrolase inhibitors that can be tuned to create selective probes for diverse members of this large enzyme class, including diacylglycerol lipase-ß (DAGLß), a principal biosynthetic enzyme for the endocannabinoid 2-arachidonoylglycerol (2-AG). Here, we provide a detailed account of the discovery, synthesis, and structure-activity relationship (SAR) of (2-substituted)-piperidyl-1,2,3-TUs that selectively inactivate DAGLß in living systems. Key to success was the use of activity-based protein profiling (ABPP) with broad-spectrum and tailored activity-based probes to guide our medicinal chemistry efforts. We also describe an expanded repertoire of DAGL-tailored activity-based probes that includes biotinylated and alkyne agents for enzyme enrichment coupled with mass spectrometry-based proteomics and assessment of proteome-wide selectivity. Our findings highlight the broad utility of 1,2,3-TUs for serine hydrolase inhibitor development and their application to create selective probes of endocannabinoid biosynthetic pathways.


Asunto(s)
Descubrimiento de Drogas , Endocannabinoides/biosíntesis , Inhibidores Enzimáticos/farmacología , Piperidinas/farmacología , Triazoles/farmacología , Urea/farmacología , Relación Dosis-Respuesta a Droga , Endocannabinoides/química , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/metabolismo , Humanos , Lipoproteína Lipasa/antagonistas & inhibidores , Lipoproteína Lipasa/metabolismo , Estructura Molecular , Piperidinas/química , Piperidinas/metabolismo , Relación Estructura-Actividad , Triazoles/química , Triazoles/metabolismo , Urea/análogos & derivados , Urea/química
5.
J Med Chem ; 56(21): 8270-9, 2013 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-24152295

RESUMEN

α/ß-Hydrolase domain containing 6 (ABHD6) is a transmembrane serine hydrolase that hydrolyzes the endogenous cannabinoid 2-arachidonoylglycerol (2-AG) to regulate certain forms of cannabinoid receptor-dependent signaling in the nervous system. The full spectrum of ABHD6 metabolic activities and functions is currently unknown and would benefit from selective, in vivo-active inhibitors. Here, we report the development and characterization of an advanced series of irreversible (2-substituted)-piperidyl-1,2,3-triazole urea inhibitors of ABHD6, including compounds KT182 and KT203, which show exceptional potency and selectivity in cells (<5 nM) and, at equivalent doses in mice (1 mg kg(-1)), act as systemic and peripherally restricted ABHD6 inhibitors, respectively. We also describe an orally bioavailable ABHD6 inhibitor, KT185, that displays excellent selectivity against other brain and liver serine hydrolases in vivo. We thus describe several chemical probes for biological studies of ABHD6, including brain-penetrant and peripherally restricted inhibitors that should prove of value for interrogating ABHD6 function in animal models.


Asunto(s)
Descubrimiento de Drogas , Inhibidores Enzimáticos/farmacología , Monoacilglicerol Lipasas/antagonistas & inhibidores , Animales , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/metabolismo , Humanos , Ratones , Modelos Animales , Estructura Molecular , Monoacilglicerol Lipasas/metabolismo , Piperidinas/química , Piperidinas/metabolismo , Piperidinas/farmacología , Relación Estructura-Actividad , Triazoles/química , Triazoles/metabolismo , Triazoles/farmacología , Urea/análogos & derivados , Urea/química , Urea/farmacología
6.
Bioorg Med Chem Lett ; 23(3): 839-43, 2013 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23260346

RESUMEN

Lipoprotein-associated phospholipase A(2) (Lp-PLA(2) or PLA(2)G7) binds to low-density lipoprotein (LDL) particles, where it is thought to hydrolyze oxidatively truncated phospholipids. Lp-PLA(2) has also been implicated as a pro-tumorigenic enzyme in human prostate cancer. Several inhibitors of Lp-PLA(2) have been described, including darapladib, which is currently in phase 3 clinical development for the treatment of atherosclerosis. The selectivity that darapladib and other Lp-PLA(2) inhibitors display across the larger serine hydrolase family has not, however, been reported. Here, we describe the use of both general and tailored activity-based probes for profiling Lp-PLA(2) and inhibitors of this enzyme in native biological systems. We show that both darapladib and a novel class of structurally distinct carbamate inhibitors inactivate Lp-PLA(2) in mouse tissues and human cell lines with high selectivity. Our findings thus identify both inhibitors and chemoproteomic probes that are suitable for investigating Lp-PLA(2) function in biological systems.


Asunto(s)
1-Alquil-2-acetilglicerofosfocolina Esterasa/antagonistas & inhibidores , Piperazinas/química , Quinolinas/química , Animales , Benzaldehídos/farmacología , Carbamatos/síntesis química , Carbamatos/química , Carbamatos/farmacología , Línea Celular Tumoral , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Humanos , Ratones , Estructura Molecular , Oximas/farmacología , Piperazinas/farmacología , Quinolinas/farmacología
7.
Chembiochem ; 13(14): 2082-93, 2012 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-22907802

RESUMEN

The serine hydrolases constitute a large class of enzymes that play important roles in physiology. There is great interest in the development of potent and selective pharmacological inhibitors of these proteins. Traditional active-site inhibitors often have limited selectivity within this superfamily and are tedious and expensive to discover. Using the serine hydrolase RBBP9 as a model target, we designed a rapid and relatively inexpensive route to highly selective peptoid-based inhibitors that can be activated by visible light. This technology provides rapid access to photo-activated tool compounds capable of selectively blocking the function of particular serine hydrolases.


Asunto(s)
Proteínas de Ciclo Celular/antagonistas & inhibidores , Inhibidores Enzimáticos/química , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Luz , Proteínas de Neoplasias/antagonistas & inhibidores , Dominio Catalítico , Proteínas de Ciclo Celular/metabolismo , Inhibidores Enzimáticos/metabolismo , Eosina Amarillenta-(YS)/química , Fluoresceína/química , Células HeLa , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Magnetismo , Proteínas de Neoplasias/metabolismo , Peptoides/síntesis química , Peptoides/química , Unión Proteica , Proteoma/metabolismo , Rutenio/química
8.
Chem Sci ; 3(1): 77-83, 2012 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-22737400

RESUMEN

Fatty acid amide hydrolase (FAAH) is an integral membrane enzyme that degrades the endocannabinoid anandamide (AEA) and several other bioactive lipid amides. The catalytic mechanism of FAAH has been largely elucidated, and structural models of the enzyme suggest that it may recruit its hydrophobic substrates directly from the lipid bilayer of the cell. Testing this hypothesis, however, requires new tools to explore FAAH-substrate interactions in native cell membranes. Here, we have addressed this problem by creating clickable, photoreactive inhibitors that probe the microenvironment surrounding the FAAH active site. We show that these probes can be used directly in cell membranes, where distinct crosslinked adducts are observed for inhibitors that are buried within versus exposed to the external environment of the FAAH active site.

9.
J Am Chem Soc ; 134(25): 10345-8, 2012 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-22690931

RESUMEN

The development of small-molecule inhibitors for perturbing enzyme function requires assays to confirm that the inhibitors interact with their enzymatic targets in vivo. Determining target engagement in vivo can be particularly challenging for poorly characterized enzymes that lack known biomarkers (e.g., endogenous substrates and products) to report on their inhibition. Here, we describe a competitive activity-based protein profiling (ABPP) method for measuring the binding of reversible inhibitors to enzymes in animal models. Key to the success of this approach is the use of activity-based probes that show tempered rates of reactivity with enzymes, such that competition for target engagement with reversible inhibitors can be measured in vivo. We apply the competitive ABPP strategy to evaluate a newly described class of piperazine amide reversible inhibitors for the serine hydrolases LYPLA1 and LYPLA2, two enzymes for which selective, in vivo active inhibitors are lacking. Competitive ABPP identified individual piperazine amides that selectively inhibit LYPLA1 or LYPLA2 in mice. In summary, competitive ABPP adapted to operate with moderately reactive probes can assess the target engagement of reversible inhibitors in animal models to facilitate the discovery of small-molecule probes for characterizing enzyme function in vivo.


Asunto(s)
Amidas/química , Sistemas de Liberación de Medicamentos , Inhibidores Enzimáticos , Piperidinas/química , Bibliotecas de Moléculas Pequeñas/química , Animales , Unión Competitiva , Células Cultivadas , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Ratones , Estructura Molecular , Unión Proteica/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología , Relación Estructura-Actividad
10.
Biochemistry ; 50(51): 11143-61, 2011 Dec 27.
Artículo en Inglés | MEDLINE | ID: mdl-22074223

RESUMEN

Phosphatidylinositol polyphosphate lipids, such as phosphatidylinositol 3,4,5-trisphosphate [PI(3,4,5)P3], regulate critical biological processes, many of which are aberrant in disease. These lipids often act as site-specific ligands in interactions that enforce membrane association of protein binding partners. Herein, we describe the development of bifunctional activity probes corresponding to the headgroup of PI(3,4,5)P3 that are effective for identifying and characterizing protein binding partners from complex samples, namely cancer cell extracts. These probes contain both a photoaffinity tag for covalent labeling of target proteins and a secondary handle for subsequent detection or manipulation of labeled proteins. Probes bearing different secondary tags were exploited, either by direct attachment of a fluorescent dye for optical detection or by using an alkyne that can be derivatized after protein labeling via click chemistry. First, we describe the design and modular synthetic strategy used to generate multiple probes with different reporter tags of use for characterizing probe-labeled proteins. Next, we report initial labeling studies using purified protein, the PH domain of Akt, in which probes were found to label this target, as judged by in-gel detection. Furthermore, protein labeling was abrogated by controls including competition with an unlabeled PI(3,4,5)P3 headgroup analogue as well as through protein denaturation, indicating specific labeling. In addition, probes featuring linkers of different lengths between the PI(3,4,5)P3 headgroup and photoaffinity tag led to variations in protein labeling, indicating that a shorter linker was more effective in this case. Finally, proteomic labeling studies were performed using cell extracts; labeled proteins were observed by in-gel detection and characterized using postlabeling with biotin, affinity chromatography, and identification via tandem mass spectrometry. These studies yielded a total of 265 proteins, including both known and novel candidate PI(3,4,5)P3-binding proteins.


Asunto(s)
Sondas Moleculares/química , Sondas Moleculares/metabolismo , Proteínas de Neoplasias/química , Proteínas de Neoplasias/metabolismo , Fosfatos de Fosfatidilinositol/química , Etiquetas de Fotoafinidad/química , Etiquetas de Fotoafinidad/metabolismo , Alquinos/química , Línea Celular Tumoral , Química Clic , Colorantes Fluorescentes/química , Humanos , Ligandos , Melanoma/enzimología , Melanoma/metabolismo , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Sondas Moleculares/síntesis química , Concentración Osmolar , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Etiquetas de Fotoafinidad/síntesis química , Unión Proteica , Proteómica/métodos , Proteínas Proto-Oncogénicas c-akt/química , Proteínas Proto-Oncogénicas c-akt/metabolismo , Solubilidad , Espectrometría de Masas en Tándem
11.
J Am Chem Soc ; 133(41): 16605-16, 2011 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-21899313

RESUMEN

Glutathione S-transferases (GSTs) are a superfamily of enzymes that conjugate glutathione to a wide variety of both exogenous and endogenous compounds for biotransformation and/or removal. Glutathione S-tranferase omega 1 (GSTO1) is highly expressed in human cancer cells, where it has been suggested to play a role in detoxification of chemotherapeutic agents. Selective inhibitors of GSTO1 are, however, required to test the role that this enzyme plays in cancer and other (patho)physiological processes. With this goal in mind, we performed a fluorescence polarization activity-based protein profiling (fluopol-ABPP) high-throughput screen (HTS) with GSTO1 and the Molecular Libraries Small Molecule Repository (MLSMR) 300K+ compound library. This screen identified a class of selective and irreversible α-chloroacetamide inhibitors of GSTO1, which were optimized to generate an agent KT53 that inactivates GSTO1 with excellent in vitro (IC(50) = 21 nM) and in situ (IC(50) = 35 nM) potency. Cancer cells treated with KT53 show heightened sensitivity to the cytotoxic effects of cisplatin, supporting a role for GSTO1 in chemotherapy resistance.


Asunto(s)
Antineoplásicos/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Glutatión Transferasa/antagonistas & inhibidores , Antineoplásicos/química , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Inhibidores Enzimáticos/química , Humanos , Estructura Molecular , Proteínas Recombinantes/antagonistas & inhibidores , Estereoisomerismo , Relación Estructura-Actividad , Células Tumorales Cultivadas
12.
J Am Chem Soc ; 133(30): 11665-74, 2011 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-21692504

RESUMEN

Peptidases play vital roles in physiology through the biosynthesis, degradation, and regulation of peptides. Prolyl endopeptidase-like (PREPL) is a newly described member of the prolyl peptidase family, with significant homology to mammalian prolyl endopeptidase and the bacterial peptidase oligopeptidase B. The biochemistry and biology of PREPL are of fundamental interest due to this enzyme's homology to the biomedically important prolyl peptidases and its localization in the central nervous system. Furthermore, genetic studies of patients suffering from hypotonia-cystinuria syndrome (HCS) have revealed a deletion of a portion of the genome that includes the PREPL gene. HCS symptoms thought to be caused by lack of PREPL include neuromuscular and mild cognitive deficits. A number of complementary approaches, ranging from biochemistry to genetics, will be required to understand the biochemical, cellular, physiological, and pathological mechanisms regulated by PREPL. We are particularly interested in investigating physiological substrates and pathways controlled by PREPL. Here, we use a fluorescence polarization activity-based protein profiling (fluopol-ABPP) assay to discover selective small-molecule inhibitors of PREPL. Fluopol-ABPP is a substrate-free approach that is ideally suited for studying serine hydrolases for which no substrates are known, such as PREPL. After screening over 300,000 compounds using fluopol-ABPP, we employed a number of secondary assays to confirm assay hits and characterize a group of 3-oxo-1-phenyl-2,3,5,6,7,8-hexahydroisoquinoline-4-carbonitrile and 1-alkyl-3-oxo-3,5,6,7-tetrahydro-2H-cyclopenta[c]pyridine-4-carbonitrile PREPL inhibitors that are able to block PREPL activity in cells. Moreover, when administered to mice, 1-isobutyl-3-oxo-3,5,6,7-tetrahydro-2H-cyclopenta[c]pyridine-4-carbonitrile distributes to the brain, indicating that it may be useful for in vivo studies. The application of fluopol-ABPP has led to the first reported PREPL inhibitors, and these inhibitors will be of great value in studying the biochemistry of PREPL and in eventually understanding the link between PREPL and HCS.


Asunto(s)
Descubrimiento de Drogas , Inhibidores Enzimáticos/farmacología , Polarización de Fluorescencia/métodos , Ensayos Analíticos de Alto Rendimiento , Serina Endopeptidasas/metabolismo , Animales , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/química , Perfilación de la Expresión Génica , Ratones , Ratones Endogámicos C57BL , Estructura Molecular , Prolil Oligopeptidasas , Serina Endopeptidasas/genética , Serina Endopeptidasas/aislamiento & purificación , Relación Estructura-Actividad
13.
J Med Chem ; 54(14): 5229-36, 2011 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-21639134

RESUMEN

The serine hydrolase protein phosphatase methylesterase-1 (PME-1) regulates the methylesterification state of protein phosphatase 2A (PP2A) and has been implicated in cancer and Alzheimer's disease. We recently reported a fluorescence polarization-activity-based protein profiling (fluopol-ABPP) high-throughput screen for PME-1 that uncovered a remarkably potent and selective class of aza-ß-lactam (ABL) PME-1 inhibitors. Here, we describe a distinct set of sulfonyl acrylonitrile inhibitors that also emerged from this screen. The optimized compound, 28 (AMZ30), selectively inactivates PME-1 and reduces the demethylated form of PP2A in living cells. Considering that 28 is structurally unrelated to ABL inhibitors of PME-1, these agents, together, provide a valuable set of pharmacological probes to study the role of methylation in regulating PP2A function. We furthermore observed that several serine hydrolases were sensitive to analogues of 28, suggesting that more extensive structural exploration of the sulfonyl acrylonitrile chemotype may result in useful inhibitors for other members of this large enzyme class.


Asunto(s)
Acrilonitrilo/análogos & derivados , Acrilonitrilo/síntesis química , Hidrolasas de Éster Carboxílico/antagonistas & inhibidores , Sulfonamidas/síntesis química , Sulfonas/síntesis química , Acrilonitrilo/química , Acrilonitrilo/farmacología , Hidrolasas de Éster Carboxílico/metabolismo , Línea Celular , Humanos , Proteoma/metabolismo , Estereoisomerismo , Relación Estructura-Actividad , Sulfonamidas/química , Sulfonamidas/farmacología , Sulfonas/química , Sulfonas/farmacología
14.
Proc Natl Acad Sci U S A ; 108(17): 6811-6, 2011 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-21398589

RESUMEN

National Institutes of Health (NIH)-sponsored screening centers provide academic researchers with a special opportunity to pursue small-molecule probes for protein targets that are outside the current interest of, or beyond the standard technologies employed by, the pharmaceutical industry. Here, we describe the outcome of an inhibitor screen for one such target, the enzyme protein phosphatase methylesterase-1 (PME-1), which regulates the methylesterification state of protein phosphatase 2A (PP2A) and is implicated in cancer and neurodegeneration. Inhibitors of PME-1 have not yet been described, which we attribute, at least in part, to a dearth of substrate assays compatible with high-throughput screening. We show that PME-1 is assayable by fluorescence polarization-activity-based protein profiling (fluopol-ABPP) and use this platform to screen the 300,000+ member NIH small-molecule library. This screen identified an unusual class of compounds, the aza-ß-lactams (ABLs), as potent (IC(50) values of approximately 10 nM), covalent PME-1 inhibitors. Interestingly, ABLs did not derive from a commercial vendor but rather an academic contribution to the public library. We show using competitive-ABPP that ABLs are exquisitely selective for PME-1 in living cells and mice, where enzyme inactivation leads to substantial reductions in demethylated PP2A. In summary, we have combined advanced synthetic and chemoproteomic methods to discover a class of ABL inhibitors that can be used to selectively perturb PME-1 activity in diverse biological systems. More generally, these results illustrate how public screening centers can serve as hubs to create spontaneous collaborative opportunities between synthetic chemistry and chemical biology labs interested in creating first-in-class pharmacological probes for challenging protein targets.


Asunto(s)
Hidrolasas de Éster Carboxílico/antagonistas & inhibidores , Inhibidores Enzimáticos , Animales , Hidrolasas de Éster Carboxílico/genética , Hidrolasas de Éster Carboxílico/metabolismo , Evaluación Preclínica de Medicamentos , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Células HEK293 , Humanos , Ratones , Ratones Noqueados , National Institutes of Health (U.S.) , Proteína Fosfatasa 2/genética , Proteína Fosfatasa 2/metabolismo , Estados Unidos
15.
Acta Crystallogr Sect F Struct Biol Cryst Commun ; 66(Pt 10): 1306-8, 2010 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-20944226

RESUMEN

Liganded structures can be instrumental in assigning function to uncharacterized proteins by revealing active sites, conserved residues, binding motifs, and substrate specificity. This introduction provides an overview and commentary on the value of liganded structures emerging from the JCSG structural genomics initiative.


Asunto(s)
Cristalografía por Rayos X , Genómica , Ligandos , Modelos Moleculares , Estructura Terciaria de Proteína
16.
Curr Protoc Chem Biol ; 1: 29-41, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-21701697

RESUMEN

Activity-based protein profiling (ABPP) is a chemical proteomic method for functional interrogation of enzymes within complex proteomes. This Unit presents a protocol for in vitro and in vivo labeling using ABPP and Click Chemistry (CC)-ABPP probes for in-depth profiling using the Multi-dimensional Protein Identification Technology (MudPIT) analysis platform.

17.
Proteomics ; 8(19): 3956-64, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18780350

RESUMEN

Integral membrane proteins (IMPs) perform crucial cellular functions and are the primary targets for most pharmaceutical agents. However, the hydrophobic nature of their membrane-embedded domains and their intimate association with lipids make them difficult to handle. Numerous proteomic platforms that include LC separations have been reported for the high-throughput profiling of complex protein samples. However, there are still many challenges to overcome for proteomic analyses of IMPs, especially as compared to their soluble counterparts. In particular, considerations for the technical challenges associated with chromatographic separations are just beginning to be investigated. Here, we review the benefits of using elevated temperatures during LC for the proteomic analysis of complex membrane protein samples.


Asunto(s)
Cromatografía Liquida/métodos , Proteínas de la Membrana/análisis , Proteómica/métodos , Temperatura , Animales , Cromatografía Liquida/instrumentación , Proteómica/instrumentación , Reproducibilidad de los Resultados
18.
J Proteome Res ; 7(7): 3028-34, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18537282

RESUMEN

Integral membrane proteins perform crucial cellular functions and are the targets for the majority of pharmaceutical agents. However, the hydrophobic nature of their membrane-embedded domains makes them difficult to work with. Here, we describe a shotgun proteomic method for the high-throughput analysis of the membrane-embedded transmembrane domains of integral membrane proteins which extends the depth of coverage of the membrane proteome.


Asunto(s)
Proteínas de la Membrana/análisis , Péptidos/análisis , Membrana Celular/química , Endopeptidasa K/química , Células HeLa , Humanos , Proteínas de la Membrana/química , Microscopía Electrónica , Péptidos/química , Proteómica/métodos , Espectrometría de Masas en Tándem
20.
Nat Protoc ; 2(6): 1414-25, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17545978

RESUMEN

Activity-based protein profiling (ABPP) utilizes active site-directed chemical probes to monitor the functional state of enzymes directly in native biological systems. Identification of the specific sites of probe labeling on enzymes remains a major challenge in ABPP experiments. In this protocol, we describe an advanced ABPP platform that utilizes a tandem orthogonal proteolysis (TOP) strategy coupled with mass spectrometric analysis to simultaneously identify probe-labeled proteins together with their exact sites of probe modification. Elucidation of probe modification sites reveals fundamental insights into the molecular basis of specific probe-protein interactions. The TOP-ABPP method can be applied to any type of proteomic sample, including those derived from in vitro or in vivo labeling experiments, and is compatible with a variety of chemical probe structures. Completion of the entire protocol, including chemical synthesis of key reagents, requires approximately 8-10 days.


Asunto(s)
Perfilación de la Expresión Génica/métodos , Sondas Moleculares , Proteoma/análisis , Animales , Biotina , Ratones , Miocardio/metabolismo , Conformación Proteica , Proteómica/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA