Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
Crit Rev Biochem Mol Biol ; 53(1): 83-98, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29239216

RESUMEN

Protein depalmitoylation describes the removal of thioester-linked long chain fatty acids from cysteine residues in proteins. For many S-palmitoylated proteins, this process is promoted by acyl protein thioesterase enzymes, which catalyze thioester hydrolysis to solubilize and displace substrate proteins from membranes. The closely related enzymes acyl protein thioesterase 1 (APT1; LYPLA1) and acyl protein thioesterase 2 (APT2; LYPLA2) were initially identified from biochemical assays as G protein depalmitoylases, yet later were shown to accept a number of S-palmitoylated protein and phospholipid substrates. Leveraging the development of isoform-selective APT inhibitors, several studies report distinct roles for APT enzymes in growth factor and hormonal signaling. Recent crystal structures of APT1 and APT2 reveal convergent acyl binding channels, suggesting additional factors beyond acyl chain recognition mediate substrate selection. In addition to APT enzymes, the ABHD17 family of hydrolases contributes to the depalmitoylation of Ras-family GTPases and synaptic proteins. Overall, enzymatic depalmitoylation ensures efficient membrane targeting by balancing the palmitoylation cycle, and may play additional roles in signaling, growth, and cell organization. In this review, we provide a perspective on the biochemical, structural, and cellular analysis of protein depalmitoylases, and outline opportunities for future studies of systems-wide analysis of protein depalmitoylation.


Asunto(s)
Lipoilación , Monoacilglicerol Lipasas/metabolismo , Procesamiento Proteico-Postraduccional , Serina Proteasas/metabolismo , Tioléster Hidrolasas/metabolismo , Animales , Humanos , Modelos Moleculares , Monoacilglicerol Lipasas/química , Proteínas/química , Proteínas/metabolismo , Serina Proteasas/química , Tioléster Hidrolasas/química
2.
J Am Chem Soc ; 142(4): 1801-1810, 2020 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-31881155

RESUMEN

Heteroaromatic sulfones react with cysteine via nucleophilic aromatic substitution, providing a mechanistically selective and irreversible scaffold for cysteine conjugation. Here we evaluate a library of heteroaromatic sulfides with different oxidation states, heteroatom substitutions, and a series of electron-donating and electron-withdrawing substituents. Select substitutions profoundly influence reactivity and stability compared to conventional cysteine conjugation reagents, increasing the reaction rate by >3 orders of magnitude. The findings establish a series of synthetically accessible electrophilic scaffolds tunable across multiple centers. New electrophiles and their corresponding alkyne conjugates were profiled directly in cultured cells, achieving thiol saturation in a few minutes at submillimolar concentrations. Direct addition of desthiobiotin-functionalized probes to cultured cells simplified enrichment and elution to enable the mass spectrometry discovery of >3000 reactive and/or accessible thiols labeled in their native cellular environments in a fraction of the standard analysis time. Surprisingly, only half of the annotated cysteines were identified by both iodoacetamide-desthiobiotin and methylsulfonylbenzothiazole-desthiobiotin in replicate experiments, demonstrating complementary detection by mass spectrometry analysis. These probes offer advantages over existing cysteine alkylation reagents, including accelerated reaction rates, improved stability, and robust ionization for mass spectrometry applications. Overall, heteroaromatic sulfones provide modular tunability, shifted chromatographic elution times, and superior in-cell cysteine profiling for in-depth proteome-wide analysis and covalent ligand discovery.


Asunto(s)
Cisteína/química , Sulfonas/química , Alquinos/química , Indicadores y Reactivos/química , Sondas Moleculares/química , Oxidación-Reducción , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
3.
J Biol Chem ; 292(12): 4766-4769, 2017 03 24.
Artículo en Inglés | MEDLINE | ID: mdl-28188288

RESUMEN

Itaconic acid is an important metabolite produced by macrophages after stimulation with LPS. The role of itaconate in the inflammatory cascade is unclear. Here we used [13C]itaconate and dimethyl [13C]itaconate (DMI) to probe itaconate metabolism, and find that [13C]DMI is not metabolized to itaconate. [13C]Itaconate in the cell culture medium leads to elevated intracellular levels of unlabeled succinate, with no evidence of intracellular uptake. The goal of this study is to encourage the development of effective pro-drug strategies to increase the intracellular levels of itaconate, which will enable more conclusive analysis of its action on macrophages and other cell and tissue types.


Asunto(s)
Inflamación/metabolismo , Macrófagos/metabolismo , Metaboloma , Succinatos/metabolismo , Animales , Células Cultivadas , Lipopolisacáridos/metabolismo , Metabolómica , Ratones , Ratones Endogámicos C57BL , Células RAW 264.7 , Ácido Succínico/metabolismo
4.
Anal Chem ; 90(15): 8722-8726, 2018 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-29989796

RESUMEN

Quantitative mass spectrometry-based protein profiling is widely used to measure protein levels across different treatments or disease states, yet current mass spectrometry acquisition methods present distinct limitations. While data-independent acquisition (DIA) bypasses the stochastic nature of data-dependent acquisition (DDA), fragment spectra derived from DIA are often complex and challenging to deconvolve. In-line ion mobility separation (IMS) adds an additional dimension to increase peak capacity for more efficient product ion assignment. As a similar strategy to sequential window acquisition methods (SWATH), IMS-enabled DIA methods rival DDA methods for protein annotation. Here we evaluate IMS-DIA quantitative accuracy using stable isotope labeling by amino acids in cell culture (SILAC). Since SILAC analysis doubles the sample complexity, we find that IMS-DIA analysis is not sufficiently accurate for sensitive quantitation. However, SILAC precursor pairs share common retention and drift times, and both species cofragment to yield multiple quantifiable isotopic y-ion peak pairs. Since y-ion SILAC ratios are intrinsic for each quantified precursor, combined MS1 and y-ion ratio analysis significantly increases the total number of measurements. With increased sampling, we present DIA-SIFT ( SILAC Intrinsic Filtering Tool), a simple statistical algorithm to identify and eliminate poorly quantified MS1 and/or MS2 events. DIA-SIFT combines both MS1 and y-ion ratios, removes outliers, and provides more accurate and precise quantitation (<15% CV) without removing any proteins from the final analysis. Overall, pooled MS1 and MS2 quantitation increases sampling in IMS-DIA SILAC analyses for accurate and precise quantitation.


Asunto(s)
Aminoácidos/análisis , Espectrometría de Masas/métodos , Proteoma/análisis , Proteómica/métodos , Técnicas de Cultivo de Célula/métodos , Células HEK293 , Humanos , Marcaje Isotópico/métodos , Programas Informáticos
5.
Anal Chem ; 90(16): 9682-9686, 2018 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-30063332

RESUMEN

Tandem mass spectrometry (MS/MS) is the primary method for discovering, identifying, and localizing post-translational modifications (PTMs) in proteins. However, conventional positive ion mode collision induced dissociation (CID)-based MS/MS often fails to yield site-specific information for labile and acidic modifications due to low ionization efficiency in positive ion mode and/or preferential PTM loss. While a number of alternative methods have been developed to address this issue, most require specialized instrumentation or indirect detection. In this work, we present an amine-reactive TEMPO-based free radical initiated peptide sequencing (FRIPS) approach for negative ion mode analysis of phosphorylated and sulfated peptides. FRIPS-based fragmentation generates sequence informative ions for both phosphorylated and sulfated peptides with no significant PTM loss. Furthermore, FRIPS is compared to positive ion mode CID, electron transfer dissociation (ETD), as well as negative ion mode electron capture dissociation (niECD) and CID, both in terms of sequence coverage and fragmentation efficiency for phospho- and sulfo-peptides. Because FRIPS-based fragmentation has no particular instrumentation requirements and shows limited PTM loss, we propose this approach as a promising alternative to current techniques for analysis of labile and acidic PTMs.


Asunto(s)
Radicales Libres/química , Oligopéptidos/análisis , Fosfopéptidos/análisis , Colecistoquinina/análisis , Colecistoquinina/química , Hirudinas/análisis , Hirudinas/química , Oligopéptidos/química , Fosfopéptidos/química , Fosforilación , Procesamiento Proteico-Postraduccional , Análisis de Secuencia de Proteína , Espectrometría de Masas en Tándem/métodos
6.
J Biol Chem ; 291(39): 20295-20302, 2016 09 23.
Artículo en Inglés | MEDLINE | ID: mdl-27528603

RESUMEN

Heterotrimeric G proteins are localized to the plasma membrane where they transduce extracellular signals to intracellular effectors. G proteins also act at intracellular locations, and can translocate between cellular compartments. For example, Gαs can leave the plasma membrane and move to the cell interior after activation. However, the mechanism of Gαs translocation and its intracellular destination are not known. Here we use bioluminescence resonance energy transfer (BRET) to show that after activation, Gαs rapidly associates with the endoplasmic reticulum, mitochondria, and endosomes, consistent with indiscriminate sampling of intracellular membranes from the cytosol rather than transport via a specific vesicular pathway. The primary source of Gαs for endosomal compartments is constitutive endocytosis rather than activity-dependent internalization. Recycling of Gαs to the plasma membrane is complete 25 min after stimulation is discontinued. We also show that an acylation-deacylation cycle is important for the steady-state localization of Gαs at the plasma membrane, but our results do not support a role for deacylation in activity-dependent Gαs internalization.


Asunto(s)
Cromograninas/metabolismo , Endocitosis/fisiología , Retículo Endoplásmico/enzimología , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Membranas Intracelulares/enzimología , Acilación , Transferencia de Energía por Resonancia de Bioluminiscencia/métodos , Cromograninas/genética , Retículo Endoplásmico/genética , Activación Enzimática/fisiología , Subunidades alfa de la Proteína de Unión al GTP Gs/genética , Células HEK293 , Humanos , Transporte de Proteínas/fisiología
7.
J Am Chem Soc ; 139(45): 16222-16227, 2017 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-29035536

RESUMEN

The histone deacetylase family comprises 18 enzymes that catalyze deacetylation of acetylated lysine residues; however, the specificity and substrate profile of each isozyme remains largely unknown. Due to transient enzyme-substrate interactions, conventional co-immunoprecipitation methods frequently fail to identify enzyme-specific substrates. Additionally, compensatory mechanisms often limit the ability of knockdown or chemical inhibition studies to achieve significant fold changes observed by acetylation proteomics methods. Furthermore, measured alterations do not guarantee a direct link between enzyme and substrate. Here we present a chemical crosslinking strategy that incorporates a photoreactive, non-natural amino acid, p-benzoyl-l-phenylalanine, into various positions of the structurally characterized isozyme histone deacetylase 8 (HDAC8). After covalent capture, co-immunoprecipitation, and mass spectrometric analysis, we identified a subset of HDAC8 substrates from human cell lysates, which were further validated for catalytic turnover. Overall, this chemical crosslinking approach identified novel HDAC8-specific substrates with high catalytic efficiency, thus presenting a general strategy for unbiased deacetylase substrate discovery.


Asunto(s)
Dominio Catalítico/genética , Dominio Catalítico/efectos de la radiación , Reactivos de Enlaces Cruzados/efectos de la radiación , Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , Procesos Fotoquímicos , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Acetilación , Benzofenonas/metabolismo , Extractos Celulares , Histona Desacetilasas/química , Humanos , Lisina/química , Lisina/metabolismo , Fenilalanina/análogos & derivados , Fenilalanina/metabolismo , Proteómica , Proteínas Represoras/química , Reproducibilidad de los Resultados , Especificidad por Sustrato
8.
Anal Chem ; 89(16): 8304-8310, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28708386

RESUMEN

Protein S-sulfinylation (R-SO2-) and S-sulfonylation (R-SO3-) are irreversible oxidative post-translational modifications of cysteine residues. Greater than 5% of cysteines are reported to occupy these higher oxidation states, which effectively inactivate the corresponding thiols and alter the electronic and physical properties of modified proteins. Such higher oxidation states are reached after excessive exposure to cellular oxidants, and accumulate across different disease states. Despite widespread and functionally relevant cysteine oxidation across the proteome, there are currently no robust methods to profile higher order cysteine oxidation. Traditional data-dependent liquid chromatography/tandem mass spectrometry (LC/MS/MS) methods generally miss low-occupancy modifications in complex analyses. Here, we present a data-independent acquisition (DIA) LC/MS-based approach, leveraging the high IR absorbance of sulfoxides at 10.6 µm, for selective dissociation and discovery of S-sulfonated peptides. Across peptide standards and protein digests, we demonstrate selective infrared multiphoton dissociation (IRMPD) of S-sulfonated peptides in the background of unmodified peptides. This selective DIA IRMPD LC/MS-based approach allows identification and annotation of S-sulfonated peptides across complex mixtures while providing sufficient sequence information to localize the modification site.


Asunto(s)
Cisteína/análogos & derivados , Péptidos/química , Cisteína/química , Cisteína/efectos de la radiación , Rayos Infrarrojos , Espectrometría de Masas/métodos , Oxidación-Reducción , Péptidos/metabolismo , Péptidos/efectos de la radiación , Procesamiento Proteico-Postraduccional/efectos de la radiación
9.
Anal Chem ; 89(11): 5669-5672, 2017 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-28471653

RESUMEN

High mass accuracy, data-dependent acquisition is the current standard method in mass spectrometry-based peptide annotation and quantification. In high complexity samples, limited instrument scan speeds often result in under-sampling. In contrast, all-ion data-independent acquisition methods bypass precursor selection, alternating high and low collision energies to analyze product and precursor ions across wide mass ranges. Despite capturing data for all events, peptide annotation is limited by inadequate alignment algorithms or overlapping ions. Ion mobility separation can add an orthogonal analytical dimension, reducing ion interference to improve reproducibility, peak capacity, and peptide identifications to rival modern hybrid quadrupole orbitrap systems. Despite the advantages of ion mobility separation in complex proteomics analyses, there has been no quantitative measure of ion mobility resolution in a complex proteomic sample. Here, we present TWIMExtract, a data extraction tool to export defined slices of liquid chromatography/ion mobility/mass spectrometry (LC-IM-MS) data, providing a route to quantify ion mobility resolution from a commercial traveling-wave ion mobility time-of-flight mass spectrometer. Using standard traveling-wave ion mobility parameters (600 m/s, 40 V), 90% of the annotated peptides occupied just 23% of the ion mobility drift space, yet inclusion of ion mobility nearly doubled the overall peak capacity. Relative to fixed velocity traveling-wave ion mobility settings, ramping the traveling-wave velocity increased drift space occupancy, amplifying resolution by 16%, peak capacity by nearly 50%, and peptide/protein identifications by 40%. Overall, variable-velocity traveling-wave ion mobility-mass spectrometry significantly enhances proteomics analysis in all-ion fragmentation acquisition.


Asunto(s)
Espectrometría de Movilidad Iónica/métodos , Proteómica/métodos , Cromatografía Liquida , Células HeLa , Humanos , Péptidos/análisis , Proteínas/análisis , Reproducibilidad de los Resultados , Espectrometría de Masas en Tándem
10.
Chembiochem ; 18(20): 2028-2032, 2017 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-28809078

RESUMEN

Cysteine residues are susceptible to oxidation to form S-sulfinyl (R-SO2 H) and S-sulfonyl (R-SO3 H) post-translational modifications. Here we present a simple bioconjugation strategy to label S-sulfinated proteins by using reporter-linked maleimides. After alkylation of free thiols with iodoacetamide, S-sulfinated cysteines react with maleimide to form a sulfone Michael adduct that remains stable under acidic conditions. Using this sequential alkylation strategy, we demonstrate differential S-sulfination across mouse tissue homogenates, as well as enhanced S-sulfination following pharmacological induction of endoplasmic reticulum stress, lipopolysaccharide stimulation, and inhibitors of the electron transport chain. Overall, this study reveals a broadened profile of maleimide reactivity across cysteine modifications, and outlines a simple method for profiling the physiological role of cysteine S-sulfination in disease.


Asunto(s)
Maleimidas/química , Sondas Moleculares/química , Proteínas/química , Proteínas/metabolismo , Ácidos Sulfínicos/metabolismo , Azufre/metabolismo , Células HEK293 , Humanos , Modelos Moleculares , Conformación Proteica
11.
J Am Chem Soc ; 138(6): 1852-9, 2016 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-26780921

RESUMEN

Cysteine S-nitrosation and S-sulfination are naturally occurring post-translational modifications (PTMs) on proteins induced by physiological signals and redox stress. Here we demonstrate that sulfinic acids and nitrosothiols react to form a stable thiosulfonate bond, and leverage this reactivity using sulfinate-linked probes to enrich and annotate hundreds of endogenous S-nitrosated proteins. In physiological buffers, sulfinic acids do not react with iodoacetamide or disulfides, enabling selective alkylation of free thiols and site-specific analysis of S-nitrosation. In parallel, S-nitrosothiol-linked probes enable enrichment and detection of endogenous S-sulfinated proteins, confirming that a single sulfinic acid can react with a nitrosothiol to form a thiosulfonate linkage. Using this approach, we find that hydrogen peroxide addition increases S-sulfination of human DJ-1 (PARK7) at Cys106, whereas Cys46 and Cys53 are fully oxidized to sulfonic acids. Comparative gel-based analysis of different mouse tissues reveals distinct profiles for both S-nitrosation and S-sulfination. Quantitative proteomic analysis demonstrates that both S-nitrosation and S-sulfination are widespread, yet exhibit enhanced occupancy on select proteins, including thioredoxin, peroxiredoxins, and other validated redox active proteins. Overall, we present a direct, bidirectional method to profile select redox cysteine modifications based on the unique nucleophilicity of sulfinic acids.


Asunto(s)
Cisteína/química , Reacciones Cruzadas , Humanos , Péptidos y Proteínas de Señalización Intracelular/química , Compuestos Nitrosos/química , Proteínas Oncogénicas/química , Oxidación-Reducción , Proteína Desglicasa DJ-1 , Compuestos de Sulfhidrilo/química , Ácidos Sulfínicos/química
12.
J Biol Chem ; 289(9): 6249-57, 2014 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-24385443

RESUMEN

Reversible attachment and removal of palmitate or other long-chain fatty acids on proteins has been hypothesized, like phosphorylation, to control diverse biological processes. Indeed, palmitate turnover regulates Ras trafficking and signaling. Beyond this example, however, the functions of palmitate turnover on specific proteins remain poorly understood. Here, we show that a mechanism regulating G protein-coupled receptor signaling in neuronal cells requires palmitate turnover. We used hexadecyl fluorophosphonate or palmostatin B to inhibit enzymes in the serine hydrolase family that depalmitoylate proteins, and we studied R7 regulator of G protein signaling (RGS)-binding protein (R7BP), a palmitoylated allosteric modulator of R7 RGS proteins that accelerate deactivation of Gi/o class G proteins. Depalmitoylation inhibition caused R7BP to redistribute from the plasma membrane to endomembrane compartments, dissociated R7BP-bound R7 RGS complexes from Gi/o-gated G protein-regulated inwardly rectifying K(+) (GIRK) channels and delayed GIRK channel closure. In contrast, targeting R7BP to the plasma membrane with a polybasic domain and an irreversibly attached lipid instead of palmitate rendered GIRK channel closure insensitive to depalmitoylation inhibitors. Palmitate turnover therefore is required for localizing R7BP to the plasma membrane and facilitating Gi/o deactivation by R7 RGS proteins on GIRK channels. Our findings broaden the scope of biological processes regulated by palmitate turnover on specific target proteins. Inhibiting R7BP depalmitoylation may provide a means of enhancing GIRK activity in neurological disorders.


Asunto(s)
Proteínas Portadoras/metabolismo , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/metabolismo , Lipoilación/fisiología , Procesamiento Proteico-Postraduccional/fisiología , Proteínas RGS/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Proteínas Portadoras/genética , Línea Celular Tumoral , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/genética , Humanos , Péptidos y Proteínas de Señalización Intracelular , Lipoilación/efectos de los fármacos , Ratones , Propiolactona/análogos & derivados , Propiolactona/farmacología , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Proteínas RGS/genética , Receptores Acoplados a Proteínas G/genética
13.
Mol Pharm ; 12(9): 3399-407, 2015 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-26262434

RESUMEN

Understanding the mechanistic basis of prodrug delivery and activation is critical for establishing species-specific prodrug sensitivities necessary for evaluating preclinical animal models and potential drug-drug interactions. Despite significant adoption of prodrug methodologies for enhanced pharmacokinetics, functional annotation of prodrug activating enzymes is laborious and often unaddressed. Activity-based protein profiling (ABPP) describes an emerging chemoproteomic approach to assay active site occupancy within a mechanistically similar enzyme class in native proteomes. The serine hydrolase enzyme family is broadly reactive with reporter-linked fluorophosphonates, which have shown to provide a mechanism-based covalent labeling strategy to assay the activation state and active site occupancy of cellular serine amidases, esterases, and thioesterases. Here we describe a modified ABPP approach using direct substrate competition to identify activating enzymes for an ethyl ester prodrug, the influenza neuraminidase inhibitor oseltamivir. Substrate-competitive ABPP analysis identified carboxylesterase 1 (CES1) as an oseltamivir-activating enzyme in intestinal cell homogenates. Saturating concentrations of oseltamivir lead to a four-fold reduction in the observed rate constant for CES1 inactivation by fluorophosphonates. WWL50, a reported carbamate inhibitor of mouse CES1, blocked oseltamivir hydrolysis activity in human cell homogenates, confirming CES1 is the primary prodrug activating enzyme for oseltamivir in human liver and intestinal cell lines. The related carbamate inhibitor WWL79 inhibited mouse but not human CES1, providing a series of probes for analyzing prodrug activation mechanisms in different preclinical models. Overall, we present a substrate-competitive activity-based profiling approach for broadly surveying candidate prodrug hydrolyzing enzymes and outline the kinetic parameters for activating enzyme discovery, ester prodrug design, and preclinical development of ester prodrugs.


Asunto(s)
Hidrolasas de Éster Carboxílico/metabolismo , Inhibidores Enzimáticos/farmacología , Ésteres/farmacología , Oseltamivir/farmacología , Profármacos/farmacología , Animales , Línea Celular , Humanos , Hidrólisis , Mucosa Intestinal/metabolismo , Intestinos/efectos de los fármacos , Cinética , Hígado/efectos de los fármacos , Hígado/metabolismo , Ratones , Especificidad por Sustrato
15.
Nat Methods ; 9(1): 84-9, 2011 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-22056678

RESUMEN

The reversible thioester linkage of palmitic acid on cysteines, known as protein S-palmitoylation, facilitates the membrane association and proper subcellular localization of proteins. Here we report the metabolic incorporation of the palmitic acid analog 17-octadecynoic acid (17-ODYA) in combination with stable-isotope labeling with amino acids in cell culture (SILAC) and pulse-chase methods to generate a global quantitative map of dynamic protein palmitoylation events in cells. We distinguished stably palmitoylated proteins from those that turn over rapidly. Treatment with a serine lipase-selective inhibitor identified a pool of dynamically palmitoylated proteins regulated by palmitoyl-protein thioesterases. This subset was enriched in oncoproteins and other proteins linked to aberrant cell growth, migration and cancer. Our method provides a straightforward way to characterize global palmitoylation dynamics in cells and confirms enzyme-mediated depalmitoylation as a critical regulatory mechanism for a specific subset of rapidly cycling palmitoylated proteins.


Asunto(s)
Cisteína/metabolismo , Ácidos Grasos Insaturados/metabolismo , Lipoilación , Ácido Palmítico/metabolismo , Animales , Lipasa/antagonistas & inhibidores , Lipoilación/efectos de los fármacos , Espectrometría de Masas , Ratones , Organofosfonatos/farmacología , Procesamiento Proteico-Postraduccional , Serina Endopeptidasas/metabolismo , Tioléster Hidrolasas/metabolismo
16.
Biopolymers ; 101(2): 173-9, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23828013

RESUMEN

Cysteine is a uniquely reactive amino acid, capable of undergoing both nucleophlilic and oxidative post-translational modifications. One such oxidation reaction involves the covalent modification of cysteine via the gaseous second messenger nitric oxide (NO), termed S-nitrosylation (SNO). This dynamic post-translational modification is involved in the redox regulation of proteins across all phylogenic kingdoms. In mammals, calcium-dependent activation of NO synthase triggers the local release of NO, which activates nearby guanylyl cyclases and cGMP-dependent pathways. In parallel, diffusible NO can locally modify redox active cellular thiols, functionally modulating many redox sensitive enzymes. Aberrant SNO is implicated in the pathology of many diseases, including neurodegeneration, inflammation, and stroke. In this review, we discuss current methods to label sites of SNO for biochemical analysis. The most popular method involves a series of biochemical steps to mask free thiols followed by selective nitrosothiol reduction and capture. Other emerging methods include mechanism-based phosphine probes and mercury enrichment chemistry. By bridging new enrichment approaches with high-resolution mass spectrometry, large-scale analysis of protein nitrosylation has highlighted new pathways of oxidative regulation.


Asunto(s)
Bioquímica/métodos , Animales , Ácido Ascórbico/metabolismo , Humanos , Óxido Nítrico/metabolismo , Donantes de Óxido Nítrico/metabolismo , Nitrosación , Compuestos de Sulfhidrilo/metabolismo
17.
J Biol Chem ; 287(1): 523-530, 2012 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-22081607

RESUMEN

Post-translational palmitoylation of intracellular proteins is mediated by protein palmitoyltransferases belonging to the DHHC family, which share a common catalytic Asp-His-His-Cys (DHHC) motif. Several members have been implicated in neuronal development, neurotransmission, and synaptic plasticity. We previously observed that mice homozygous for a hypomorphic allele of the ZDHHC5 gene are impaired in context-dependent learning and memory. To identify potentially relevant protein substrates of DHHC5, we performed a quantitative proteomic analysis of stable isotope-labeled neuronal stem cell cultures from forebrains of normal and DHHC5-GT (gene-trapped) mice using the bioorthogonal palmitate analog 17-octadecynoic acid. We identified ∼300 17-octadecynoic acid-modified and hydroxylamine-sensitive proteins, of which a subset was decreased in abundance in DHHC5-GT cells. Palmitoylation and oligomerization of one of these proteins (flotillin-2) was abolished in DHHC5-GT neuronal stem cells. In COS-1 cells, overexpression of DHHC5 markedly stimulated the palmitoylation of flotillin-2, strongly suggesting a direct enzyme-substrate relationship. Serendipitously, we found that down-regulation of DHHC5 was triggered within minutes following growth factor withdrawal from normal neural stem cells, a maneuver that is used to induce neural differentiation in culture. The effect was reversible for up to 4 h, and degradation was partially prevented by inhibitors of ubiquitin-mediated proteolysis. These findings suggest that protein palmitoylation can be regulated through changes in DHHC PAT levels in response to differentiation signals.


Asunto(s)
Diferenciación Celular , Lipoilación , Proteínas de la Membrana/metabolismo , Neuronas/citología , Neuronas/metabolismo , Proteolisis , Aciltransferasas , Animales , Células Cultivadas , Humanos , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Ratones , Mutagénesis Sitio-Dirigida , Mutación , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Multimerización de Proteína , Estructura Cuaternaria de Proteína
18.
Biochem Soc Trans ; 41(1): 43-9, 2013 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23356256

RESUMEN

Protein palmitoylation is a critical post-translational modification important for membrane compartmentalization, trafficking and regulation of many key signalling proteins. Recent non-radioactive chemo-proteomic labelling methods have enabled a new focus on this emerging regulatory modification. Palmitoylated proteins can now be profiled in complex biological systems by MS for direct annotation and quantification. Based on these analyses, palmitoylation is clearly widespread and broadly influences the function of many cellular pathways. The recent introduction of selective chemical labelling approaches has opened new opportunities to revisit long-held questions about the enzymatic regulation of this widespread post-translational modification. In the present review, we discuss the impact of new chemical labelling approaches and future challenges for the dynamic global analysis of protein palmitoylation.


Asunto(s)
Lipoilación , Ácido Palmítico/metabolismo , Proteínas/metabolismo , Aciltransferasas/metabolismo , Espectrometría de Masas , Proteínas/química , Proteómica , Especificidad por Sustrato
19.
Nat Chem Biol ; 7(7): 469-78, 2011 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-21572424

RESUMEN

Serine hydrolases are a diverse enzyme class representing ∼1% of all human proteins. The biological functions of most serine hydrolases remain poorly characterized owing to a lack of selective inhibitors to probe their activity in living systems. Here we show that a substantial number of serine hydrolases can be irreversibly inactivated by 1,2,3-triazole ureas, which show negligible cross-reactivity with other protein classes. Rapid lead optimization by click chemistry-enabled synthesis and competitive activity-based profiling identified 1,2,3-triazole ureas that selectively inhibit enzymes from diverse branches of the serine hydrolase class, including peptidases (acyl-peptide hydrolase, or APEH), lipases (platelet-activating factor acetylhydrolase-2, or PAFAH2) and uncharacterized hydrolases (α,ß-hydrolase-11, or ABHD11), with exceptional potency in cells (sub-nanomolar) and mice (<1 mg kg(-1)). We show that APEH inhibition leads to accumulation of N-acetylated proteins and promotes proliferation in T cells. These data indicate 1,2,3-triazole ureas are a pharmacologically privileged chemotype for serine hydrolase inhibition, combining broad activity across the serine hydrolase class with tunable selectivity for individual enzymes.


Asunto(s)
Química Clic/métodos , Serina Proteasas/metabolismo , Inhibidores de Serina Proteinasa/síntesis química , Triazoles/síntesis química , Urea/análogos & derivados , Animales , Encéfalo/enzimología , Línea Celular Tumoral , Humanos , Ratones , Estructura Molecular , Miocardio/enzimología , Serina Proteasas/genética , Inhibidores de Serina Proteinasa/química , Inhibidores de Serina Proteinasa/farmacología , Linfocitos T/efectos de los fármacos , Linfocitos T/enzimología , Transfección , Triazoles/química , Triazoles/farmacología , Urea/farmacología
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA