Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Bioorg Med Chem ; 66: 116816, 2022 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-35598478

RESUMO

Dimethylarginine dimethylaminohydrolase (DDAH, EC 3.5.3.18) catalyzes the hydrolysis of asymmetric Nω,Nω-dimethyl-l-arginine (ADMA), an endogenous inhibitor of human nitric oxide synthases. The active-site cysteine residue has been proposed to serve as the catalytic nucleophile, forming an S-alkylthiourea reaction intermediate, and serving as a target for covalent inhibitors. Inhibition can lead to ADMA accumulation and downstream inhibition of nitric oxide production. Prior studies have provided experimental evidence for formation of this covalent adduct but have not characterized it kinetically. Here, rapid quench-flow is used with ADMA and the DDAH from Pseudomonas aeruginosa to determine the rate constants for formation (k2 = 17 ± 2 s-1) and decay (k3 = 1.5 ± 0.1 s-1) of the covalent S-alkylthiourea adduct. A minimal kinetic mechanism for DDAH is proposed that supports the kinetic competence of this species as a covalent reaction intermediate and assigns the rate-limiting step in substrate turnover as hydrolysis of this intermediate. This work helps elucidate the different reactivities of S-alkylthiourea intermediates found among the mechanistically diverse pentein superfamily of guanidine-modifying enzymes and provides information useful for inhibitor development.


Assuntos
Amidoidrolases , Óxido Nítrico , Amidoidrolases/química , Amidoidrolases/metabolismo , Arginina/farmacologia , Humanos , Cinética , Óxido Nítrico/química , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase/química , Óxido Nítrico Sintase/metabolismo
2.
Chem Sci ; 10(34): 8025-8034, 2019 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-31853358

RESUMO

Mutations in the GTPase enzyme K-Ras, specifically at codon G12, remain the most common genetic alterations in human cancers. The mechanisms governing activation of downstream signaling pathways and how they relate back to the identity of the mutation have yet to be completely defined. Here we use native mass spectrometry (MS) combined with ultraviolet photodissociation (UVPD) to investigate the impact of three G12X mutations (G12C, G12V, G12S) on the homodimerization of K-Ras as well as heterodimerization with a downstream effector protein, Raf. Electrospray ionization (ESI) was used to transfer complexes of WT or G12X K-Ras bound to guanosine 5'-diphosphate (GDP) or GppNHp (non-hydrolyzable analogue of GTP) into the gas phase. Relative abundances of homo- or hetero-dimer complexes were estimated from ESI-MS spectra. K-Ras + Raf heterocomplexes were activated with UVPD to probe structural changes responsible for observed differences in the amount of heterocomplex formed for each variant. Holo (ligand-bound) fragment ions resulting from photodissociation suggest the G12X mutants bind Raf along the expected effector binding region (ß-interface) but may interact with Raf via an alternative α-interface as well. Variations in backbone cleavage efficiencies during UV photoactivation of each variant were used to relate mutation identity to structural changes that might impact downstream signaling. Specifically, oncogenic upregulation for hydrogen-bonding amino acid substitutions (G12C, G12S) is achieved by stabilizing ß-interface interactions with Raf, while a bulkier, hydrophobic G12V substitution leads to destabilization of this interface and instead increases the proximity of residues along the α-helical bundles. This study deciphers new pieces of the complex puzzle of how different K-Ras mutations exert influence in downstream signaling.

3.
ACS Chem Biol ; 14(4): 613-618, 2019 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-30933557

RESUMO

Nicotinamide N-methyltransferase (NNMT) catalyzes the S-adenosyl-l-methionine-dependent methylation of nicotinamide to form N-methylnicotinamide. This enzyme detoxifies xenobiotics and regulates NAD+ biosynthesis. Additionally, NNMT is overexpressed in various cancers. Herein, we describe the first NNMT-targeted suicide substrates. These compounds, which include 4-chloropyridine and 4-chloronicotinamide, exploit the broad substrate scope of NNMT; methylation of the pyridine nitrogen enhances the electrophilicity of the C4 position, thereby promoting an aromatic nucleophilic substitution by C159, a noncatalytic cysteine. On the basis of this activity, we developed a suicide inhibition-based protein labeling strategy using an alkyne-substituted 4-chloropyridine that selectively labels NNMT in vitro and in cells. In total, this study describes the first NNMT-directed activity-based probes.


Assuntos
Nicotinamida N-Metiltransferase/metabolismo , Proteínas/metabolismo , Catálise , Células HEK293 , Humanos , Cinética , Metilação , Nicotinamida N-Metiltransferase/antagonistas & inibidores
4.
Biochemistry ; 57(24): 3326-3337, 2018 06 19.
Artigo em Inglês | MEDLINE | ID: mdl-29689165

RESUMO

Covalent enzyme inhibitors are widely applied as biochemical tools and therapeutic agents. As a complement to categorization of these inhibitors by reactive group or modification site, we present a categorization by mechanism, which highlights common advantages and disadvantages inherent to each approach. Established categories for reversible and irreversible covalent inhibition are reviewed with representative examples given for each class, including covalent reversible inhibitors, slow substrates, residue-specific reagents, affinity labels (classical, quiescent, and photoaffinity), and mechanism-based inactivators. The relationships of these categories to proteomic profiling probes (activity-based and reactivity-based) as well as complementary approaches such as prodrug and soft drug design are also discussed. A wide variety of strategies are used to balance reactivity and selectivity in the design of covalent enzyme inhibitors. Use of a shared terminology is encouraged to clearly convey these mechanisms, to relate them to prior use of covalent inhibitors in enzymology, and to facilitate the development of more effective covalent inhibitors.


Assuntos
Pró-Fármacos/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Proteínas Quinases S6 Ribossômicas 90-kDa/antagonistas & inibidores , Humanos , Estrutura Molecular , Pró-Fármacos/síntese química , Pró-Fármacos/química , Inibidores de Proteínas Quinases/síntese química , Inibidores de Proteínas Quinases/química , Proteínas Quinases S6 Ribossômicas 90-kDa/metabolismo
5.
Chembiochem ; 18(15): 1551-1556, 2017 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-28470883

RESUMO

We have investigated 4-halopyridines as selective, tunable, and switchable covalent protein modifiers for use in the development of chemical probes. Nonenzymatic reactivity of 4-chloropyridine with amino acids and thiols was ranked with respect to common covalent protein-modifying reagents and found to have reactivity similar to that of acrylamide, but could be switched to a reactivity similar to that of iodoacetamide upon stabilization of the positively charged pyridinium. Diverse, fragment-sized 4-halopyridines inactivated human dimethylarginine dimethylaminohydrolase-1 (DDAH1) through covalent modification of the active site cysteine, acting as quiescent affinity labels that required off-pathway catalysis through stabilization of the protonated pyridinium by a neighboring aspartate residue. A series of 2-fluoromethyl-substituted 4-chloropyridines demonstrated that the pKa and kinact /KI values could be predictably varied over several orders of magnitude. Covalent labeling of proteins in an Escherichia coli lysate was shown to require folded proteins, indicating that alternative proteins can be targeted, and modification is likely to be catalysisdependent. 4-Halopyridines, and quiescent affinity labels in general, represent an attractive strategy to develop reagents with switchable electrophilicity as selective covalent protein modifiers.


Assuntos
Amidoidrolases/química , Piridinas/química , Acrilamida/química , Marcadores de Afinidade/química , Cisteína/química , Escherichia coli/metabolismo , Glutationa/química , Humanos , Iodoacetamida/química , Fenóis/química , Proteoma/química , Proteoma/metabolismo , Compostos de Piridínio/química , Compostos de Sulfidrila/química
6.
J Am Chem Soc ; 138(40): 13187-13196, 2016 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-27665622

RESUMO

Single-residue mutations at Gly12 (G12X) in the GTP-ase protein K-Ras can lead to activation of different downstream signaling pathways, depending on the identity of the mutation, through a poorly defined mechanism. Herein, native mass spectrometry combined with top-down ultraviolet photodissociation (UVPD) was employed to investigate the structural changes occurring from G12X mutations of K-Ras. Complexes between K-Ras or the G12X mutants and guanosine 5'-diphosphate (GDP) or GDPnP (a stable GTP analogue) were transferred to the gas phase by nano-electrospray ionization and characterized using UVPD. Variations in the efficiencies of backbone cleavages were observed upon substitution of GDPnP for GDP as well as for the G12X mutants relative to wild-type K-Ras. An increase in the fragmentation efficiency in the segment containing the first 50 residues was observed for the K-Ras/GDPnP complexes relative to the K-Ras/GDP complexes, whereas a decrease in fragmentation efficiency occurred in the segment containing the last 100 residues. Within these general regions, the specific residues at which changes in fragmentation efficiency occurred correspond to the phosphate and guanine binding regions, respectively, and are indicative of a change in the binding motif upon replacement of the ligand (GDP versus GDPnP). Notably, unique changes in UVPD were observed for each G12X mutant with the cysteine and serine mutations exhibiting similar UVPD changes whereas the valine mutation was significantly different. These findings suggest a mechanism that links the identity of the G12X substitution to different downstream effects through long-range conformational or dynamic effects as detected by variations in UVPD fragmentation.

7.
Curr Drug Targets ; 17(9): 1029-50, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26424398

RESUMO

Metallo-ß-Lactamases (MBLs) are class Bß-lactamases that hydrolyze almost all clinically-availableß-lactam antibiotics. MBLs feature the distinctive αß/ßα sandwich fold of the metallo-hydrolase/oxidoreductase superfamily and possess a shallow active-site groove containing one or two divalent zinc ions, flanked by flexible loops. According to sequence identity and zinc ion dependence, MBLs are classified into three subclasses (B1, B2 and B3), of which the B1 subclass enzymes have emerged as the most clinically significant. Differences among the active site architectures, the nature of zinc ligands, and the catalytic mechanisms have limited the development of a common inhibitor. In this review, we will describe the molecular epidemiology and structural studies of the most prominent representatives of class B1 MBLs (NDM-1, IMP-1 and VIM-2) and describe the implications for inhibitor design to counter this growing clinical threat.


Assuntos
Antineoplásicos/química , Bactérias Gram-Negativas/enzimologia , Zinco/metabolismo , beta-Lactamases/química , beta-Lactamases/metabolismo , Antineoplásicos/farmacologia , Sítios de Ligação , Domínio Catalítico , Cristalografia por Raios X , Desenho de Fármacos , Bactérias Gram-Negativas/química , Bactérias Gram-Negativas/efeitos dos fármacos , Humanos , Hidrólise , Estrutura Molecular , Estrutura Secundária de Proteína , Relação Estrutura-Atividade , Especificidade por Substrato , beta-Lactamases/classificação
8.
ChemMedChem ; 9(4): 792-7, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24574257

RESUMO

Inhibitors of the human enzyme dimethylarginine dimethylaminohydrolase-1 (DDAH-1) can raise endogenous levels of asymmetric dimethylarginine (ADMA) and lead to a subsequent inhibition of nitric oxide synthesis. In this study, N(5) -(1-imino-2-chloroethyl)-L-ornithine (Cl-NIO) is shown to be a potent time- and concentration-dependent inhibitor of purified human DDAH-1 (KI =1.3±0.6 µM; kinact =0.34±0.07 min(-1) ), with >500-fold selectivity against two arginine-handling enzymes in the same pathway. An activity probe is used to measure the "in cell" IC50 value (6.6±0.2 µM) for Cl-NIO inhibition of DDAH-1 artificially expressed within cultured HEK293T cells. A screen of diverse melanoma cell lines reveals that a striking 50/64 (78 %) of melanoma lines tested showed increased levels of DDAH-1 relative to normal melanocyte control lines. Treatment of the melanoma A375 cell line with Cl-NIO shows a subsequent decrease in cellular nitric oxide production. Cl-NIO is a promising tool for the study of methylarginine-mediated nitric oxide control and a potential therapeutic lead compound for other indications with elevated nitric oxide production, such as septic shock and idiopathic pulmonary fibrosis.


Assuntos
Amidoidrolases/antagonistas & inibidores , Desenho de Fármacos , Inibidores Enzimáticos/farmacologia , Melanoma/enzimologia , Ornitina/análogos & derivados , Amidoidrolases/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Células HEK293 , Humanos , Melanoma/metabolismo , Conformação Molecular , Óxido Nítrico/antagonistas & inibidores , Óxido Nítrico/biossíntese , Ornitina/síntese química , Ornitina/química , Ornitina/farmacologia , Relação Estrutura-Atividade , Regulação para Cima/efeitos dos fármacos
9.
Bioorg Med Chem ; 21(11): 3138-46, 2013 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-23591260

RESUMO

Due to the global threat of antibiotic resistance mediated by New Delhi metallo-beta-lactamase-1 (NDM-1) and the lack of structurally diverse inhibitors reported for this enzyme, we developed screening and counter-screening assays for manual and automated formats. The manual assay is a trans-well absorbance-based endpoint assay in 96-well plates and has a Z' factor of 0.8. The automated assay is an epi-absorbance endpoint assay in 384-well plates, has a Z' factor of ≥0.8, good signal/baseline ratios (>3.8), and is likely scalable for high-throughput screening (HTS). A TEM-1-based counter-screen is also presented to eliminate false positives due to assay interference or off-target activities. A pilot screen of a pharmacologically characterized compound library identified two thiol-modifying compounds as authentic NDM-1 inhibitors: p-chloromecuribenzoate (p-CMB) and nitroprusside. Recombinant NDM-1 has one Cys residue that serves as a conserved active-site primary zinc ligand and is selectively modified by p-CMB as confirmed by LC-MS/MS. However a C208D mutation results in an enzyme that maintains almost full lactamase activity, yet is completely resistant to the inhibitor. These results predict that covalent targeting of the conserved active-site Cys residue may have drawbacks as a drug design strategy.


Assuntos
Antibacterianos/química , Nitroprussiato/química , Zinco/química , Inibidores de beta-Lactamases , Ácido p-Cloromercurobenzoico/química , Domínio Catalítico , Cisteína/química , Cisteína/genética , Enterobacteriaceae/química , Enterobacteriaceae/enzimologia , Enterobacteriaceae/genética , Ensaios de Triagem em Larga Escala , Testes de Sensibilidade Microbiana , Mutação , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Espectrometria de Massas em Tandem , Resistência beta-Lactâmica , beta-Lactamases/química , beta-Lactamases/genética
10.
Biochemistry ; 52(9): 1603-10, 2013 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-23387521

RESUMO

Autoinducer inactivator A (AiiA) is a metal-dependent N-acyl homoserine lactone hydrolase that displays broad substrate specificity but shows a preference for substrates with long N-acyl substitutions. Previously, crystal structures of AiiA in complex with the ring-opened product N-hexanoyl-l-homoserine revealed binding interactions near the metal center but did not identify a binding pocket for the N-acyl chains of longer substrates. Here we report the crystal structure of an AiiA mutant, F107W, determined in the presence and absence of N-decanoyl-l-homoserine. F107 is located in a hydrophobic cavity adjacent to the previously identified ligand binding pocket, and the F107W mutation results in the formation of an unexpected interaction with the ring-opened product. Notably, the structure reveals a previously unidentified hydrophobic binding pocket for the substrate's N-acyl chain. Two aromatic residues, F64 and F68, form a hydrophobic clamp, centered around the seventh carbon in the product-bound structure's decanoyl chain, making an interaction that would also be available for longer substrates, but not for shorter substrates. Steady-state kinetics using substrates of various lengths with AiiA bearing mutations at the hydrophobic clamp, including insertion of a redox-sensitive cysteine pair, confirms the importance of this hydrophobic feature for substrate preference. Identifying the specificity determinants of AiiA will aid the development of more selective quorum-quenching enzymes as tools and as potential therapeutics.


Assuntos
Amidoidrolases/química , Amidoidrolases/metabolismo , Bacillus thuringiensis/enzimologia , Homosserina/análogos & derivados , Amidoidrolases/genética , Bacillus thuringiensis/química , Bacillus thuringiensis/genética , Bacillus thuringiensis/metabolismo , Cristalografia por Raios X , Homosserina/metabolismo , Cinética , Modelos Moleculares , Mutagênese Sítio-Dirigida , Fenilalanina/química , Fenilalanina/genética , Fenilalanina/metabolismo , Mutação Puntual , Conformação Proteica , Percepção de Quorum , Especificidade por Substrato
11.
Bioorg Med Chem ; 20(18): 5550-8, 2012 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-22921743

RESUMO

Potent and selective inhibitors of the enzyme dimethylarginine dimethylaminohydrolase (DDAH) are useful as molecular probes to better understand cellular regulation of nitric oxide. Inhibitors are also potential therapeutic agents for treatment of pathological states associated with the inappropriate overproduction of nitric oxide, such as septic shock, selected types of cancer, and other conditions. Inhibitors with structures dissimilar to substrate may overcome limitations inherent to substrate analogs. Therefore, to identify structurally-diverse inhibitor scaffolds, high-throughput screening (HTS) of a 4000-member library of fragment-sized molecules was completed using the Pseudomonas aeruginosa DDAH and human DDAH-1 isoforms. Use of a substrate concentration equal to its K(M) value during the primary screen allowed for the detection of inhibitors with different modes of inhibition. A series of validation tests were designed and implemented in the identification of four inhibitors of human DDAH-1 that were unknown prior to the screen. Two inhibitors share a 4-halopyridine scaffold and act as quiescent affinity labels that selectively and covalently modify the active-site Cys residue. Two inhibitors are benzimidazole-like compounds that reversibly and competitively inhibit human DDAH-1 with Ligand Efficiency values ≥0.3 kcal/mol/heavy (non-hydrogen) atom, indicating their suitability for further development. Both inhibitor scaffolds have available sites to derivatize for further optimization. Therefore, use of this fragment-based HTS approach is demonstrated to successfully identify two novel scaffolds for development of DDAH-1 inhibitors.


Assuntos
Amidoidrolases/antagonistas & inibidores , Descoberta de Drogas , Inibidores Enzimáticos/farmacologia , Ensaios de Triagem em Larga Escala , Bibliotecas de Moléculas Pequenas/farmacologia , Amidoidrolases/isolamento & purificação , Amidoidrolases/metabolismo , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/química , Humanos , Isoenzimas/antagonistas & inibidores , Modelos Moleculares , Pseudomonas aeruginosa/enzimologia , Bibliotecas de Moléculas Pequenas/química , Relação Estrutura-Atividade
12.
Mol Biosyst ; 7(7): 2286-95, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21584310

RESUMO

Peptidylarginine deiminase (PAD) catalyzes the posttranslational citrullination of selected proteins in a calcium dependent manner. The PAD4 isoform has been implicated in multiple sclerosis, rheumatoid arthritis, some types of cancer, and plays a role in gene regulation. However, the substrate selectivity of PAD4 is not well defined, nor is the impact of citrullination on many other pathways. Here, a high-density protein array is used as a primary screen to identify 40 previously unreported PAD4 substrates, 10 of which are selected and verified in a cell lysate-based secondary assay. One of the most prominent hits, human 40S ribosomal protein S2 (RPS2), is characterized in detail. PAD4 citrullinates the Arg-Gly repeat region of RPS2, which is also an established site for Arg methylation by protein arginine methyltransferase 3 (PRMT3). As in other systems, crosstalk is observed; citrullination and methylation modifications are found to be antagonistic to each other, suggesting a conserved posttranslational regulatory strategy. Both PAD4 and PRMT3 are found to co-sediment with the free 40S ribosomal subunit fraction from cell extracts. These findings are consistent with participation of citrullination in the regulation of RPS2 and ribosome assembly. This application of protein arrays to reveal new PAD4 substrates suggests a role for citrullination in a number of different cellular pathways.


Assuntos
Citrulina/metabolismo , Hidrolases/metabolismo , Análise Serial de Proteínas/métodos , Proteínas Ribossômicas/metabolismo , Cloreto de Cálcio/farmacologia , Células HEK293 , Humanos , Metilação , Ligação Proteica/efeitos dos fármacos , Mapeamento de Interação de Proteínas , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Proteína-Arginina Desiminase do Tipo 4 , Desiminases de Arginina em Proteínas , Especificidade por Substrato/efeitos dos fármacos
13.
J Biol Chem ; 286(19): 17069-78, 2011 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-21454715

RESUMO

Gene expression is regulated by a number of interrelated posttranslational modifications of histones, including citrullination. For example, peptidylarginine deminase 4 (PAD4) converts peptidyl arginine to citrulline in histone H3 and can repress gene expression. However, regulation of gene expression through citrullination of non-histone proteins is less well defined. Herein, we identify a tumor suppressor protein, inhibitor of growth 4 (ING4), as a novel non-histone substrate of PAD4. ING4 is known to bind p53 via its nuclear localization signal (NLS) region and to enhance transcriptional activity of p53. We show that PAD4 preferentially citrullinates ING4 in the same NLS region and thereby disrupts the interaction between ING4 and p53. A citrulline-mimicking Arg-NLS-Gln ING4 mutant, which has all Arg residues in the NLS mutated to Gln, loses its affinity for p53, can no longer promote p53 acetylation, and results in repression of downstream p21 expression. In addition, we found that citrullination leads to increased susceptibility of ING4 to degradation, likely impacting p53-independent pathways as well. These findings elucidate an interaction between posttranslational citrullination, acetylation, and methylation and highlight an unusual mechanism whereby citrullination of a non-histone protein impacts gene regulation.


Assuntos
Proteínas de Ciclo Celular/química , Citrulina/química , Regulação da Expressão Gênica , Proteínas de Homeodomínio/química , Hidrolases/química , Mutação , Proteína Supressora de Tumor p53/química , Proteínas Supressoras de Tumor/química , Encéfalo/metabolismo , Proteínas de Ciclo Celular/metabolismo , Metilação de DNA , Glutamina/química , Glutationa Transferase/metabolismo , Proteínas de Homeodomínio/metabolismo , Humanos , Hidrolases/metabolismo , Modelos Biológicos , Ligação Proteica , Processamento de Proteína Pós-Traducional , Proteína-Arginina Desiminase do Tipo 4 , Desiminases de Arginina em Proteínas , Proteínas Recombinantes/química , Proteína Supressora de Tumor p53/metabolismo , Proteínas Supressoras de Tumor/metabolismo
14.
ChemMedChem ; 6(1): 81-8, 2011 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-20979083

RESUMO

C-Alkyl amidine analogues of asymmetric N(ω),N(ω)-dimethyl-L-arginine are dual-targeted inhibitors of both human DDAH-1 and nitric oxide (NO) synthase, and provide a promising scaffold for the development of therapeutics to control NO overproduction in a variety of pathologies including septic shock and some cancers. Using a two-part click-chemistry-mediated activity probe, a homologated series of C-alkyl amidines were ranked for their ability to inhibit DDAH-1 within cultured HEK 293T cells. N5-(1-Iminopentyl)-L-ornithine was determined to be the most potent compound in vitro (K(d)=7 µM) as well as in cultured cells, and the binding conformation and covalent reversible mode of inhibition was investigated by comparison of interactions made with DDAH-1 and a catalytically inactive C274S variant, as gauged by X-ray crystallography and isothermal titration calorimetry. By interrupting the ability of the inhibitor to form a covalent bond, the contribution of this interaction could be estimated. These results suggest that further stabilization of the covalent adduct is a promising strategy for lead optimization in the design of effective reagents to block NO synthesis.


Assuntos
Amidinas , Aminoidrolases/antagonistas & inibidores , Arginina/análogos & derivados , Inibidores Enzimáticos , Óxido Nítrico Sintase/antagonistas & inibidores , Amidinas/farmacologia , Aminoidrolases/genética , Aminoidrolases/metabolismo , Arginina/metabolismo , Disponibilidade Biológica , Varredura Diferencial de Calorimetria , Linhagem Celular , Cristalografia por Raios X , Inibidores Enzimáticos/farmacologia , Humanos , Cinética , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase/metabolismo
15.
Biochemistry ; 48(36): 8624-35, 2009 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-19663506

RESUMO

Molecules that block nitric oxide's (NO) biosynthesis are of significant interest. For example, nitric oxide synthase (NOS) inhibitors have been suggested as antitumor therapeutics, as have inhibitors of dimethylarginine dimethylaminohydrolase (DDAH), an enzyme that catabolizes endogenous NOS inhibitors. Dual-targeted inhibitors hold promise as more effective reagents to block NO biosynthesis than single-targeted compounds. In this study, a small set of known NOS inhibitors are surveyed as inhibitors of recombinant human DDAH-1. From these, an alkylamidine scaffold is selected for homologation. Stepwise lengthening of one substituent converts an NOS-selective inhibitor into a dual-targeted NOS/DDAH-1 inhibitor and then into a DDAH-1 selective inhibitor, as seen in the inhibition constants of N5-(1-iminoethyl)-, N5-(1-iminopropyl)-, N5-(1-iminopentyl)- and N(5)-(1-iminohexyl)-l-ornithine for neuronal NOS (1.7, 3, 20, >1,900 microM, respectively) and DDAH-1 (990, 52, 7.5, 110 microM, respectively). A 1.9 A X-ray crystal structure of the N5-(1-iminopropyl)-L-ornithine:DDAH-1 complex indicates covalent bond formation between the inhibitor's amidino carbon and the active-site Cys274, and solution studies show reversible competitive inhibition, consistent with a reversible covalent mode of DDAH inhibition by alkylamidine inhibitors. These represent a versatile scaffold for the development of a targeted polypharmacological approach to control NO biosynthesis.


Assuntos
Amidoidrolases/antagonistas & inibidores , Sistemas de Liberação de Medicamentos , Inibidores Enzimáticos/síntese química , Óxido Nítrico Sintase Tipo I/antagonistas & inibidores , Óxido Nítrico/antagonistas & inibidores , Amidinas/metabolismo , Amidinas/farmacologia , Amidoidrolases/metabolismo , Cristalografia por Raios X , Sistemas de Liberação de Medicamentos/métodos , Inibidores Enzimáticos/metabolismo , Inibidores Enzimáticos/farmacologia , Humanos , Óxido Nítrico/biossíntese , Óxido Nítrico Sintase Tipo I/metabolismo , Ornitina/análogos & derivados , Ornitina/metabolismo , Ornitina/farmacologia , Proteínas Recombinantes/antagonistas & inibidores , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Especificidade por Substrato , Tioureia/metabolismo , Tioureia/farmacologia
16.
Protein Sci ; 18(5): 881-92, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19384989

RESUMO

It is generally accepted that naturally existing functional domains can serve as building blocks for complex protein structures, and that novel functions can arise from assembly of different combinations of these functional domains. To inform our understanding of protein evolution and explore the modular nature of protein structure, two model enzymes were chosen for study, purT-encoded glycinamide ribonucleotide formyltransferase (PurT) and purK-encoded N(5)-carboxylaminoimidazole ribonucleotide synthetase (PurK). Both enzymes are found in the de novo purine biosynthetic pathway of Escherichia coli. In spite of their low sequence identity, PurT and PurK share significant similarity in terms of tertiary structure, active site organization, and reaction mechanism. Their characteristic three domain structures categorize both PurT and PurK as members of the ATP-grasp protein superfamily. In this study, we investigate the exchangeability of individual protein domains between these two enzymes and the in vivo and in vitro functional properties of the resulting hybrids. Six domain-swapped hybrids were unable to catalyze full wild-type reactions, but each hybrid protein could catalyze partial reactions. Notably, an additional loop replacement in one of the domain-swapped hybrid proteins was able to restore near wild-type PurK activity. Therefore, in this model system, domain-swapped proteins retained the ability to catalyze partial reactions, but further modifications were required to efficiently couple the reaction intermediates and achieve catalysis of the full reaction. Implications for understanding the role of domain swapping in protein evolution are discussed.


Assuntos
Carboxiliases/química , Proteínas de Escherichia coli/química , Hidroximetil e Formil Transferases/química , Redes e Vias Metabólicas , Purinas/metabolismo , Proteínas Recombinantes de Fusão/química , Trifosfato de Adenosina/metabolismo , Sequência de Aminoácidos , Carboxiliases/genética , Carboxiliases/metabolismo , Domínio Catalítico/genética , Domínio Catalítico/fisiologia , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Hidroximetil e Formil Transferases/genética , Hidroximetil e Formil Transferases/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Organofosfatos/metabolismo , Conformação Proteica , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
17.
J Biol Chem ; 282(48): 34684-92, 2007 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-17895252

RESUMO

The plasma concentrations of two cardiovascular risk factors, total homocysteine (tHcy) and asymmetric dimethylarginine (ADMA), correlate with decreased levels of endothelium-derived nitric oxide and subsequent endothelial dysfunction. Homocysteine has been proposed to inhibit the catabolic enzyme of ADMA, dimethylarginine dimethylaminohydrolase (DDAH), but the mechanism of this inhibition has not been fully elucidated. Here, the human DDAH isoform-1 (DDAH-1) is heterologously expressed and purified. Cys(274) and His(173) are identified as active site residues and the pH rate dependence is described. Because oxidation of the active site Cys has been suggested as an inhibitory mechanism in patients with hyperhomocysteinemia, the sensitivity of DDAH-1 to inhibition by L-homocysteine, H(2)O(2), and S-nitroso-L-homocysteine is quantified. DDAH-1 is surprisingly insensitive to inactivation by the powerful oxidant, H(2)O(2) (0.088 M(-1) s(-1)), possibly because of a substrate-assisted mechanism that allows the active site cysteine to remain predominantly protonated and less reactive in the resting enzyme. In contrast, DDAH-1 is sensitive to inactivation by S-nitroso-L-homocysteine (3.79 M(-1) s(-1)). This work illustrates how a particular catalytic mechanism can result in selective redox regulation and has possible implications for hyperhomocysteinemia.


Assuntos
Amidoidrolases/antagonistas & inibidores , Homocisteína/análogos & derivados , Peróxido de Hidrogênio/farmacologia , Hiper-Homocisteinemia/tratamento farmacológico , Hiper-Homocisteinemia/enzimologia , Amidoidrolases/química , Amidoidrolases/genética , Sequência de Aminoácidos , Animais , Sítios de Ligação , Clonagem Molecular , Cisteína/química , Relação Dose-Resposta a Droga , Desenho de Fármacos , Homocisteína/farmacologia , Humanos , Cinética , Dados de Sequência Molecular , Oxirredução , Homologia de Sequência de Aminoácidos , Especificidade por Substrato
18.
Biochemistry ; 45(17): 5618-30, 2006 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-16634643

RESUMO

The enzyme dimethylargininase (also known as dimethylarginine dimethylaminohydrolase or DDAH; EC 3.5.3.18) catalyzes the hydrolysis of endogenous nitric oxide synthase inhibitors, N(omega)-methyl-l-arginine and N(omega),N(omega)-dimethyl-l-arginine. Understanding the mechanism and regulation of DDAH activity is important for developing ways to control nitric oxide production during angiogenesis and in many cases of vascular endothelial pathobiology. Several possible physiological regulation mechanisms of DDAH depend upon the presence of an active-site cysteine residue, Cys249 in Pseudomonas aeruginosa (Pa) DDAH, which is proposed to serve as a nucleophile in the catalytic mechanism. Through the use of pH-dependent ultraviolet and visible (UV-vis) difference spectroscopy and inactivation kinetics, the pK(a) of the active-site Cys249 in the resting enzyme was found to be unperturbed from pK(a) values of typical noncatalytic cysteine residues. In contrast, the pH dependence of k(cat) values indicates a much lower apparent pK(a) value. UV-vis difference spectroscopy between wild-type and C249S DDAH shows absorbance changes consistent with Cys249 deprotonation to the anionic thiolate upon binding positively charged ligands. The proton from Cys249 is lost either to the solvent or to an unidentified general base. A mutation of the active-site histidine residue, H162G, does not eliminate cysteine nucleophilicity, further arguing against a pre-formed ion pair with Cys249. Finally, UV-vis and X-ray absorption spectroscopy revealed that inhibitory metal ions can bind at these two active-site residues, Cys249 and His162, and also stabilize the anionic form of Cys249. These results support a proposed substrate-assisted mechanism for Pa DDAH in which ligand binding modulates the reactivity of the active-site cysteine.


Assuntos
Amidoidrolases/metabolismo , Cisteína/metabolismo , Pseudomonas aeruginosa/enzimologia , Amidoidrolases/antagonistas & inibidores , Sítios de Ligação , Citrulina/análogos & derivados , Citrulina/metabolismo , Cobalto/metabolismo , Concentração de Íons de Hidrogênio , Iodoacetamida/farmacologia , Cinética , Lisina/farmacologia , Espectrometria de Massas , Modelos Químicos , Prótons , Espectrofotometria Ultravioleta , Análise Espectral , Tioureia/análogos & derivados , Tioureia/metabolismo , Raios X , Zinco/farmacologia , ômega-N-Metilarginina/metabolismo
19.
Biochemistry ; 44(42): 13744-52, 2005 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-16229464

RESUMO

The enzymes dimethylargininase [dimethylarginine dimethylaminohydrolase (DDAH); EC 3.5.3.18] and peptidylarginine deiminase (PAD; EC 3.5.3.15) catalyze hydrolysis of substituted arginines. Due to their role in normal physiology and pathophysiology, both enzymes have been identified as potential drug targets, but few useful inhibitors have been reported. Here, we find that 2-chloroacetamidine irreversibly inhibits both DDAH from Pseudomonas aeruginosa and human PAD4 in a time- and concentration-dependent manner, despite the nonoverlapping substrate specificities and low levels of amino acid identity of their catalytic domains. Substrate protection experiments indicate that inactivation occurs by modification at the active site, albeit with modest affinity. Mass spectral analysis demonstrates that irreversible inactivation of DDAH occurs through selective formation of a covalent thioether bond with the active-site Cys249 residue. The mechanism of inactivation by 2-chloroacetamidine is analogous to that of chloromethyl ketones, a set of inhibitors that have found wide application because of their specific covalent modification of active-site residues in serine and cysteine proteases. Likewise, 2-chloroacetamidine may potentially find wide applicability as a general pharmacophore useful in delineating characteristics of the amidinotransferase superfamily.


Assuntos
Amidinas/farmacologia , Amidoidrolases/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Hidrolases/antagonistas & inibidores , Amidoidrolases/genética , Amidoidrolases/metabolismo , Sequência de Aminoácidos , Sequência de Bases , Sítios de Ligação , Domínio Catalítico , Clonagem Molecular , Primers do DNA , Hidrolases/genética , Hidrolases/metabolismo , Espectrometria de Massas , Desiminases de Arginina em Proteínas
20.
Biochemistry ; 44(18): 7069-78, 2005 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-15865451

RESUMO

Dimethylarginine dimethylaminohydrolase (DDAH) regulates the concentrations of human endogenous inhibitors of nitric oxide synthase, N(omega)-methyl-l-arginine (NMMA), and asymmetric N(omega),N(omega)-dimethyl-l-arginine (ADMA). Pharmacological regulation of nitric oxide synthesis is an important goal, but the catalytic mechanism of DDAH remains largely unexplored. A DDAH from Pseudomonas aeruginosa was cloned, and asymmetrically methylated arginine analogues were shown to be the preferred substrates, with ADMA displaying a slightly higher k(cat)/K(M) value than NMMA. DDAH is similar to members of a larger superfamily of guanidino-modifying enzymes, some of which have been shown to use an S-alkylthiouronium intermediate during catalysis. No covalent intermediates were found to accumulate during steady-state turnover reactions of DDAH with NMMA or ADMA. However, identification of a new substrate with an activated leaving group, S-methyl-l-thiocitrulline (SMTC), enabled acid trapping and ESI-MS characterization of a transient covalent adduct with a mass of 158 +/- 10 Da that accumulates during steady-state turnover. Subsequent trapping, proteolysis, peptide mapping and fragmentation by mass spectrometry, and site-directed mutagenesis demonstrated that this covalent adduct was attached to an active site residue and implicates Cys249 as the catalytic nucleophile required for intermediate formation. The use of covalent catalysis clearly links DDAH to this superfamily of enzymes and suggests that an S-alkylthiouronium intermediate may be a conserved feature in their mechanisms.


Assuntos
Amidoidrolases/metabolismo , Adutos de DNA/biossíntese , Adutos de DNA/química , Pseudomonas aeruginosa/enzimologia , Ácidos/química , Amidoidrolases/química , Amidoidrolases/genética , Sequência de Aminoácidos , Catálise , Cisteína/genética , Adutos de DNA/metabolismo , Hidrólise , Cinética , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Pseudomonas aeruginosa/genética , Serina/genética , Espectrometria de Massas por Ionização por Electrospray , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Especificidade por Substrato
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA