Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Biochimie ; 133: 103-111, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28034716

RESUMEN

Excessive proliferation of vascular smooth muscle cells (SMC) is an important contributor to the progression of atherosclerosis. Inhibition of proliferation can be achieved by endogenously produced and exogenously supplied nitrogen monoxide, commonly known as nitric oxide (NO). We report herein the dichotomous effects of two isomeric families of secondary amines, precursors to the N-nitrosated NO-donors, on HASMC proliferation. The syntheses of these two families were carried out using two equivalents of homologous, aliphatic monoamines and 2,6-difluoro-3-nitrobenzonitrile (2,6-DFNBN, O family) or 2,4-difluoro-5-nitrobenzonitrile (2,4-DFNBN, P family). The secondary amines belonging to the P family inhibited HASMC proliferation at all concentrations, whereas the O family induced HASMC proliferation at low concentrations, and exhibited inhibitory properties at high concentrations. A probable explanation of these behaviors is proposed herein. l-homocysteine (HCY) is known to induce HASMC proliferation at low concentrations (<1 mM) and inhibit HASMC proliferation at higher concentrations (>2.5 mM). Our findings suggest that these two families of amines inhibit cystathionine-γ-lyase (CSE) to varying extents, which directly results in altered levels of intracellular HCY and consequent changes in HASMC proliferation.


Asunto(s)
Aminas/química , Proliferación Celular/efectos de los fármacos , Cistationina gamma-Liasa/antagonistas & inhibidores , Óxido Nítrico/biosíntesis , Aminas/administración & dosificación , Aorta/citología , Aorta/efectos de los fármacos , Línea Celular , Humanos , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/enzimología , Óxido Nítrico/química , Donantes de Óxido Nítrico/química , Nitrilos/química
2.
Biochem Biophys Res Commun ; 450(1): 208-12, 2014 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-24878532

RESUMEN

Atherosclerosis is the leading cause of cerebral and myocardial infarction. It is believed that neointimal growth common in the later stages of atherosclerosis is a result of vascular smooth muscle cell (SMC) de-differentiation in response to endothelial injury. However, the claims of the SMC de-differentiation theory have not been substantiated by monitoring the fate of mature SMCs in response to such injuries. A recent study suggests that atherosclerosis is a consequence of multipotent vascular stem cell (MVSC) differentiation. Nitric oxide (NO) is a well-known mediator against atherosclerosis, in part because of its inhibitory effect on SMC proliferation. Using three different NO-donors, we have investigated the effects of NO on MVSC proliferation. Results indicate that NO inhibits MVSC proliferation in a concentration dependent manner. A slow and sustained delivery of NO proved to inhibit proliferation without causing cell death. On the other hand, larger, single-burst NO concentrations, inhibits proliferation, with concurrent significant cell death. Furthermore, our results indicate that endogenously produced NO inhibits MVSC differentiation to mesenchymal-like stem cells (MSCs) and subsequently to SMC as well.


Asunto(s)
Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/fisiología , Células Madre Multipotentes/citología , Células Madre Multipotentes/fisiología , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/fisiología , Donantes de Óxido Nítrico/administración & dosificación , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Preparaciones de Acción Retardada/administración & dosificación , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Multipotentes/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Ratas
3.
Bioorg Med Chem ; 21(5): 1123-35, 2013 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-23375096

RESUMEN

Atherosclerosis, a leading cause of death worldwide, is associated with the excessive proliferation of vascular smooth muscle cells. Nitrogen monoxide, more commonly known as nitric oxide, inhibits this uncontrolled proliferation. Herein we report the preparation of two families of nitric oxide donors; beginning with the syntheses of secondary amine precursors, obtained through the reaction between 2 equiv of various monoamines with 2,4 or 2,6-difluoronitrobenzene. The purified secondary amines were nitrosated then subjected to a Griess reagent test to examine the slow and sustained nitric oxide release rate for each compound in both the absence and presence of reduced glutathione. The release rate profiles of these two isomeric families of NO-donors were strongly dependent on the number of side chain methylene units and the relative orientations of the nitro groups with respect to the N-nitroso moieties. The nitrosated compounds were then added to human aortic smooth muscle cell cultures, individually and in tandem with S-2-amino-6-boronic acid (ABH), a potent arginase inhibitor. Cell viability studies indicated a lack of toxicity of the amine precursors, in addition to anti-proliferative effects exhibited by the nitrosated compounds, which were enhanced in the presence of ABH.


Asunto(s)
Aminas/química , Arginasa/antagonistas & inhibidores , Inhibidores Enzimáticos/química , Músculo Liso Vascular/efectos de los fármacos , Óxido Nítrico/metabolismo , Arginasa/metabolismo , Ácidos Borónicos/química , Ácidos Borónicos/toxicidad , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Sinergismo Farmacológico , Inhibidores Enzimáticos/toxicidad , Humanos , Isomerismo , Músculo Liso Vascular/citología , Donantes de Óxido Nítrico/química , Donantes de Óxido Nítrico/toxicidad , Nitrosación
4.
Arch Biochem Biophys ; 504(2): 190-6, 2010 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-20850413

RESUMEN

Peptidylarginine deiminase (PAD), which catalyzes the deimination of the guanidino group from peptidylarginine residues, belongs to a superfamily of guanidino group modifying enzymes that have been shown to produce an S-alkylthiouronium ion intermediate during catalysis. Thiol-directed reagents iodoacetamide and iodoacetate inactivate recombinant PAD, and substrate protects the enzyme from inactivation. Activity measurements together with peptide mapping by mass spectrometry of PAD modified in the absence and presence of substrate demonstrated that cysteine-351 is modified by iodoacetamide. The pK(a) value of the cysteine residue, 7.7±0.2 as determined by iodoacetamide modification, agrees well with a critical pK value identified in pH rate studies. The role of cysteine-351 in catalysis was tested by site-directed mutagenesis in which the cysteine was replaced with serine to eliminate the proposed nucleophilic interaction. Binding studies carried out using fluorescence spectrometry established the structural integrity of the C351S PAD. However, the C351S PAD variant was catalytically inactive, exhibiting <0.01% wild-type activity. These results indicate that Cys 351 is a nucleophile that initiates the enzymatic reaction.


Asunto(s)
Cisteína/química , Hidrolasas/química , Porphyromonas gingivalis/enzimología , Catálisis , Concentración de Iones de Hidrógeno , Hidrolasas/antagonistas & inhibidores , Hidrolasas/genética , Yodoacetamida/química , Ácido Yodoacético/química , Ligandos , Espectrometría de Masas , Mutagénesis Sitio-Dirigida , Mapeo Peptídico , Unión Proteica , Desiminasas de la Arginina Proteica , Proteínas Recombinantes/antagonistas & inhibidores , Proteínas Recombinantes/química
5.
Arch Biochem Biophys ; 488(1): 14-22, 2009 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-19545534

RESUMEN

Porphyromonas gingivalis peptidylarginine deiminase (PAD) catalyzes the deimination of peptidylarginine residues of various peptides to produce peptidylcitrulline and ammonia. P. gingivalis is associated with adult-onset periodontitis and cardiovascular disease, and its proliferation depends on secretion of PAD. We have expressed two recombinant forms of the P. gingivalis PAD in Escherichia coli, a truncated form with a 43-amino acid N-terminal deletion and the full-length form of PAD as predicted from the DNA sequence. Both forms contain a poly-His tag and Xpress epitope at the N-terminus to aid in detection and purification. The activities and stabilities of these two forms have been evaluated. PAD is cold sensitive; it aggregates within 30 min at 4 degrees C, and optimal storage conditions are at 25 degrees C in the presence of a reducing agent. PAD is not a metalloenzyme and does not need a cofactor for catalysis or stability. Multiple l-arginine analogs, various arginine-containing peptides, and free l-arginine were used to evaluate substrate specificity and determine kinetic parameters.


Asunto(s)
Escherichia coli/genética , Hidrolasas/aislamiento & purificación , Hidrolasas/metabolismo , Porphyromonas gingivalis/enzimología , Secuencia de Aminoácidos , Biocatálisis , Coenzimas/metabolismo , Estabilidad de Enzimas , Eliminación de Gen , Expresión Génica , Concentración de Iones de Hidrógeno , Hidrolasas/biosíntesis , Hidrolasas/química , Iminas/metabolismo , Metales/metabolismo , Datos de Secuencia Molecular , Peso Molecular , Porphyromonas gingivalis/genética , Desiminasas de la Arginina Proteica , Análisis de Secuencia de ADN , Especificidad por Sustrato
6.
Biochemistry ; 48(1): 121-31, 2009 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-19093830

RESUMEN

Arginase is a binuclear manganese metalloenzyme that serves as a therapeutic target for the treatment of asthma, erectile dysfunction, and atherosclerosis. In order to better understand the molecular basis of inhibitor affinity, we have employed site-directed mutagenesis, enzyme kinetics, and X-ray crystallography to probe the molecular recognition of the amino acid moiety (i.e., the alpha-amino and alpha-carboxylate groups) of substrate l-arginine and inhibitors in the active site of arginase I. Specifically, we focus on (1) a water-mediated hydrogen bond between the substrate alpha-carboxylate and T135, (2) a direct hydrogen bond between the substrate alpha-carboxylate and N130, and (3) a direct charged hydrogen bond between the substrate alpha-amino group and D183. Amino acid substitutions for T135, N130, and D183 generally compromise substrate affinity as reflected by increased K(M) values but have less pronounced effects on catalytic function as reflected by minimal variations of k(cat). As with substrate K(M) values, inhibitor K(d) values increase for binding to enzyme mutants and suggest that the relative contribution of intermolecular interactions to amino acid affinity in the arginase active site is water-mediated hydrogen bond < direct hydrogen bond < direct charged hydrogen bond. Structural comparisons of arginase with the related binuclear manganese metalloenzymes agmatinase and proclavaminic acid amidinohydrolase suggest that the evolution of substrate recognition in the arginase fold occurs by mutation of residues contained in specificity loops flanking the mouth of the active site (especially loops 4 and 5), thereby allowing diverse guanidinium substrates to be accommodated for catalysis.


Asunto(s)
Aminoácidos/química , Arginasa/química , Sustitución de Aminoácidos , Animales , Arginasa/genética , Catálisis , Dominio Catalítico , Cristalografía por Rayos X , Inhibidores Enzimáticos/química , Guanidinas/química , Humanos , Enlace de Hidrógeno , Cinética , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Unión Proteica , Conformación Proteica , Ratas , Ureohidrolasas/química
7.
Methods Enzymol ; 440: 221-30, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18423220

RESUMEN

Of all arginine catabolic enzymes, the arginases and nitric oxide (NO) synthases are the ones that are of greatest interest to many investigators. Mammalian arginases catalyze the hydrolysis of arginine to ornithine and urea and are composed of two distinct isozymes: arginase I, located within the cytosol, and arginase II, located within mitochondria. The arginases not only can inhibit NO synthesis by reducing arginine availability, but also can promote the synthesis of polyamines or proline via production of the common precursor ornithine. Because of their inducibility in many cell types and to their potential impact on multiple biochemical pathways in health and disease, there is growing interest in assays of arginase activity. Although arginase activity may be determined by either spectrophotometric or radiochemical assays, radiochemical assays afford greater sensitivity and do not require correction for any ornithine or urea that may be present in the samples. Part of the arginase assay protocol described in this chapter also can be used for radiochemical assays of enzymes that catalyze decarboxylation reactions. No activity assay currently available is capable of distinguishing the arginase isozymes.


Asunto(s)
Arginasa/análisis , Arginina/metabolismo , Mamíferos/metabolismo , Animales , Arginasa/fisiología , Activación Enzimática/fisiología , Humanos , Especificidad por Sustrato/fisiología
8.
Arch Biochem Biophys ; 444(1): 15-26, 2005 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-16266687

RESUMEN

Rat liver arginase (arginase I) is potently inactivated by diethyl pyrocarbonate, with a second-order rate constant of 113M(-1)s(-1) for the inactivation process at pH 7.0, 25 degrees C. Partial protection from inactivation is provided by the product of the reaction, l-ornithine, while nearly complete protection is afforded by the inhibitor pair, l-ornithine and borate. The role of H141 has been probed by mutagenesis, chemical modulation, and X-ray diffraction. The hyper-reactivity of H141 towards diethyl pyrocarbonate can be explained by its proximity to E277. A proton shuttling role for H141 is supported by its conformational mobility observed among the known arginase structures. H141 is proposed to serve as an acid/base catalyst, deprotonating the metal-bridging water molecule to generate the metal-bridging hydroxide nucleophile, and by protonating the amino group of the product to facilitate its departure.


Asunto(s)
Arginasa/química , Histidina/química , Animales , Arginasa/antagonistas & inhibidores , Boratos/química , Cristalografía por Rayos X , Dietil Pirocarbonato/química , Cinética , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Ornitina/química , Conformación Proteica , Ratas
9.
J Nutr ; 134(10 Suppl): 2760S-2764S; discussion 2765S-2767S, 2004 10.
Artículo en Inglés | MEDLINE | ID: mdl-15465781

RESUMEN

The arginases catalyze the divalent cation dependent hydrolysis of L-arginine to produce L-ornithine and urea. Although traditionally considered in terms of its role as the final enzyme of the urea cycle, the enzyme is found in a variety of nonhepatic tissues. These findings suggest that the enzyme may have other functions in addition to its role in nitrogen metabolism. High-resolution crystal structures have been determined for recombinant rat liver (type I) arginase and for recombinant human kidney (type II) arginase, their variants, and complexes with products and inhibitors. Each identical subunit of the trimeric enzyme contains an active site that lies at the bottom of a 15 A deep cleft. The 2 essential Mn(II) ions are located at the bottom of this cleft, separated by approximately 3.3 A and bridged by oxygens derived from 2 aspartic acid residues and a solvent-derived hydroxide. This metal bridging hydroxide is proposed to be the nucleophile that attacks the guanidinium carbon of substrate arginine. On the basis of this proposed mechanism, boronic acid inhibitors of the enzyme have been synthesized and characterized kinetically and structurally. These inhibitors display slow-onset inhibition at the pH optimum of the enzyme, and are found as tetrahedral species at the active site, as determined by X-ray diffraction. The potent inhibition of arginases I and II by these compounds has not only delineated key enzyme-substrate interactions, but has also led to a greater understanding of the role of arginase in nonhepatic tissues.


Asunto(s)
Arginasa/química , Arginasa/metabolismo , Arginina/análogos & derivados , Animales , Arginasa/antagonistas & inhibidores , Arginina/farmacología , Ácidos Borónicos/farmacología , Catálisis , Inhibidores Enzimáticos/farmacología , Humanos , Cinética , Metales/metabolismo , Relación Estructura-Actividad
10.
Biochemistry ; 43(28): 8987-99, 2004 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-15248756

RESUMEN

Arginase is a manganese metalloenzyme that catalyzes the hydrolysis of L-arginine to form L-ornithine and urea. The structure and stability of the binuclear manganese cluster are critical for catalytic activity as it activates the catalytic nucleophile, metal-bridging hydroxide ion, and stabilizes the tetrahedral intermediate and its flanking states. Here, we report X-ray structures of a series of inhibitors bound to the active site of arginase, and each inhibitor exploits a different mode of coordination with the Mn(2+)(2) cluster. Specifically, we have studied the binding of fluoride ion (F(-); an uncompetitive inhibitor) and L-arginine, L-valine, dinor-N(omega)-hydroxy-L-arginine, descarboxy-nor-N(omega)-hydroxy-L-arginine, and dehydro-2(S)-amino-6-boronohexanoic acid. Some inhibitors, such as fluoride ion, dinor-N(omega)-hydroxy-L-arginine, and dehydro-2(S)-amino-6-boronohexanoic acid, cause the net addition of one ligand to the Mn(2+)(2) cluster. Other inhibitors, such as descarboxy-nor-N(omega)-hydroxy-L-arginine, simply displace the metal-bridging hydroxide ion of the native enzyme and do not cause any net change in the metal coordination polyhedra. The highest affinity inhibitors displace the metal-bridging hydroxide ion (and sometimes occupy a Mn(2+)(A) site found vacant in the native enzyme) and maintain a conserved array of hydrogen bonds with their alpha-amino and -carboxylate groups.


Asunto(s)
Arginasa/química , Inhibidores Enzimáticos/química , Manganeso , Animales , Arginasa/antagonistas & inhibidores , Arginina/análogos & derivados , Arginina/química , Sitios de Unión , Cristalografía por Rayos X , Flúor/química , Enlace de Hidrógeno , Metaloproteínas/antagonistas & inhibidores , Metaloproteínas/química , Estructura Molecular , Unión Proteica , Ratas , Proteínas Recombinantes , Valina/química
11.
Biochemistry ; 42(28): 8445-51, 2003 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-12859189

RESUMEN

Arginase is a binuclear manganese metalloenzyme that catalyzes the hydrolysis of l-arginine to form l-ornithine and urea. The X-ray crystal structure of a fully active, truncated form of human arginase II complexed with a boronic acid transition state analogue inhibitor has been determined at 2.7 A resolution. This structure is consistent with the hydrolysis of l-arginine through a metal-activated hydroxide mechanism. Given that human arginase II appears to play a role in regulating l-arginine bioavailability to NO synthase in human penile corpus cavernosum smooth muscle, the inhibition of human arginase II is a potential new strategy for the treatment of erectile dysfunction [Kim, N. N., Cox, J. D., Baggio, R. F., Emig, F. A., Mistry, S., Harper, S. L., Speicher, D. W., Morris, S. M., Ash, D. E., Traish, A. M., and Christianson, D. W. (2001) Biochemistry 40, 2678-2688]. Since NO synthase is found in human clitoral corpus cavernosum and vagina, we hypothesized that human arginase II is similarly present in these tissues and functions to regulate l-arginine bioavailability to NO synthase. Accordingly, hemodynamic studies conducted with a boronic acid arginase inhibitor in vivo are summarized, suggesting that the extrahepatic arginase plays a role in both male and female sexual arousal. Therefore, arginase II is a potential target for the treatment of male and female sexual arousal disorders.


Asunto(s)
Arginasa/química , Nivel de Alerta/fisiología , Hemodinámica/fisiología , Sexualidad/fisiología , Secuencia de Aminoácidos , Animales , Arginasa/antagonistas & inhibidores , Arginasa/genética , Secuencia de Bases , Sitios de Unión , Cristalografía por Rayos X/métodos , Cartilla de ADN , Inhibidores Enzimáticos/farmacología , Femenino , Variación Genética , Humanos , Isoenzimas , Masculino , Modelos Moleculares , Reacción en Cadena de la Polimerasa , Estructura Secundaria de Proteína , Conejos , Proteínas Recombinantes/antagonistas & inhibidores , Proteínas Recombinantes/química , Eliminación de Secuencia
12.
Biochemistry ; 42(25): 7748-58, 2003 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-12820884

RESUMEN

Arginase is a binuclear manganese metalloenzyme that hydrolyzes l-arginine to form l-ornithine and urea. The three-dimensional structures of D128E, D128N, D232A, D232C, D234E, H101N, and H101E arginases I have been determined by X-ray crystallographic methods to elucidate the roles of the first-shell metal ligands in the stability and catalytic activity of the enzyme. This work represents the first structure-based dissection of the binuclear manganese cluster using site-directed mutagenesis and X-ray crystallography. Substitution of the metal ligands compromises the catalytic activity of the enzyme, either by the loss or disruption of the metal cluster or the nucleophilic metal-bridging hydroxide ion. However, the substitution of the metal ligands or the reduction of Mn(2+)(A) or Mn(2+)(B) occupancy does not compromise enzyme-substrate affinity as reflected by K(M), which remains relatively invariant across this series of arginase variants. This implicates a nonmetal binding site for substrate l-arginine in the precatalytic Michaelis complex, as proposed based on analysis of the native enzyme structure (Kanyo, Z. F., Scolnick, L. R., Ash, D. E., and Christianson, D. W. (1996) Nature 383, 554-557).


Asunto(s)
Arginasa/metabolismo , Manganeso/metabolismo , Sustitución de Aminoácidos , Animales , Arginasa/genética , Cristalografía por Rayos X , Ligandos , Ratas , Relación Estructura-Actividad
13.
Arch Biochem Biophys ; 399(1): 49-55, 2002 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-11883902

RESUMEN

Hyperargininemia is a rare autosomal disorder that results from a deficiency in hepatic type I arginase. This deficiency is the consequence of random point mutations that occur throughout the gene. The G235R patient mutation has been proposed to affect the catalytic activity and structural integrity of the protein [D. E. Ash, L. R. Scolnick, Z. F. Kanyo, J. G. Vockley, S. D. Cederbaum, and D. W. Christianson (1998) Mol. Genet. Metab. 64, 243-249]. The G235R (patient) and G235A (control) arginase mutants of rat liver arginase have been generated to probe the effects of these point mutations on the structure and function of hepatic type I arginase. Both mutant arginases were trimeric by gel filtration, but the control G235A mutant had 56% of wild-type activity and the G235R mutant had less than 0.03% activity compared to the wild-type enzyme. The G235R mutant contained undetectable levels of tightly bound manganese as determined by electron paramagnetic resonance, while the G235A mutant had a Mn(II) stoichiometry of 2 Mn/subunit. Molecular modeling indicates that the introduction of an arginine residue at position 235 results in a major rearrangement of the metal ligands that compromise Mn(II) binding.


Asunto(s)
Arginasa/genética , Arginasa/fisiología , Hígado/metabolismo , Mutación Puntual , Animales , Arginasa/química , Sitios de Unión , Cromatografía en Gel , Espectroscopía de Resonancia por Spin del Electrón , Estabilidad de Enzimas , Hiperargininemia/enzimología , Hiperargininemia/etiología , Cinética , Manganeso/química , Metales/metabolismo , Modelos Químicos , Modelos Moleculares , Peso Molecular , Mutagénesis Sitio-Dirigida , Ratas , Relación Estructura-Actividad , Temperatura
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...