Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biochemistry ; 40(31): 9356-62, 2001 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-11478904

RESUMO

Arginases catalyze the hydrolysis of L-arginine to yield L-ornithine and urea. Recent studies indicate that arginases, both the type I and type II isozymes, participate in the regulation of nitric oxide production by modulating the availability of arginine for nitric oxide synthase. Due to the reciprocal regulation between arginase and nitric oxide synthase, arginase inhibitors have therapeutic potential in treating nitric oxide-dependent smooth muscle disorders, such as erectile dysfunction. We demonstrate the competitive inhibition of the mitochondrial human type II arginase by N(omega)-hydroxy-L-arginine, the intermediate in the reaction catalyzed by nitric oxide synthase, and its analogue N(omega)-hydroxy-nor-L-arginine, with K(i) values of 1.6 microM and 51 nM at pH 7.5, respectively. We also demonstrate the inhibition of human type II arginase by the boronic acid-based transition-state analogues 2(S)-amino-6-boronohexanoic acid (ABH) and S-(2-boronoethyl)-L-cysteine (BEC), which are known inhibitors of type I arginase. At pH 7.5, both ABH and BEC are classical, competitive inhibitors of human type II arginase with K(i) values of 0.25 and 0.31 microM, respectively. However, at pH 9.5, ABH and BEC are slow-binding inhibitors of the enzyme with K(i) values of 8.5 and 30 nM, respectively. The findings presented here indicate that the design of arginine analogues with uncharged, tetrahedral functional groups will lead to the development of more potent inhibitors of arginases at physiological pH.


Assuntos
Arginase/antagonistas & inibidores , Arginase/metabolismo , Arginina/análogos & derivados , Inibidores Enzimáticos/metabolismo , Aminocaproatos/metabolismo , Arginina/metabolismo , Ligação Competitiva , Boro , Compostos de Boro/metabolismo , Ácidos Borônicos/química , Ácidos Borônicos/metabolismo , Inibidores Enzimáticos/classificação , Guanidinas/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Isoenzimas/antagonistas & inibidores , Isoenzimas/metabolismo , Cinética , Ressonância Magnética Nuclear Biomolecular , Especificidade por Substrato , Fatores de Tempo
2.
Arch Biochem Biophys ; 389(1): 135-43, 2001 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-11370664

RESUMO

Human type II arginase, which is extrahepatic and mitochondrial in location, catalyzes the hydrolysis of arginine to form ornithine and urea. While type I arginases function in the net production of urea for excretion of excess nitrogen, type II arginases are believed to function primarily in the net production of ornithine, a precursor of polyamines, glutamate, and proline. Type II arginases may also regulate nitric oxide biosynthesis by modulating arginine availability for nitric oxide synthase. Recombinant human type II arginase was expressed in Escherichia coli and purified to apparent homogeneity. The Km of arginine for type II arginase is approximately 4.8 mM at physiological pH. Type II arginase exists primarily as a trimer, although higher order oligomers were observed. Borate is a noncompetitive inhibitor of the enzyme, with a Kis of 0.32 mM and a Kii of 0.3 mM. Ornithine, a product of the reaction catalyzed by arginase and a potent inhibitor of type I arginase, is a poor inhibitor of the type II isozyme. The findings presented here indicate that isozyme-selectivity exists between type I and type II arginases for binding of substrate and products, as well as inhibitors. Therefore, inhibitors with greater isozyme-selectivity for type II arginase may be identified and utilized for the therapeutic treatment of smooth muscle disorders, such as erectile dysfunction.


Assuntos
Arginase/química , Arginase/antagonistas & inibidores , Arginase/genética , Arginina/metabolismo , Arginina/farmacologia , Ligação Competitiva/efeitos dos fármacos , Catálise/efeitos dos fármacos , Cátions Bivalentes/farmacologia , Cromatografia em Gel , Cromatografia por Troca Iônica , DNA Complementar/genética , Espectroscopia de Ressonância de Spin Eletrônica , Eletroforese em Gel de Poliacrilamida , Inibidores Enzimáticos/farmacologia , Estabilidade Enzimática/efeitos dos fármacos , Expressão Gênica , Humanos , Concentração de Íons de Hidrogênio , Isoenzimas/antagonistas & inibidores , Isoenzimas/química , Isoenzimas/genética , Mitocôndrias/enzimologia , Ornitina/biossíntese , Ornitina/farmacologia , Reação em Cadeia da Polimerase , Proteínas Recombinantes/antagonistas & inibidores , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Análise de Sequência de Proteína , Ureia/metabolismo
3.
J Biol Chem ; 276(17): 14242-8, 2001 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-11278703

RESUMO

The structure of the trimeric, manganese metalloenzyme, rat liver arginase, has been previously determined at 2.1-A resolution (Kanyo, Z. F., Scolnick, L. R., Ash, D. E., and Christianson, D. W., (1996) Nature 383, 554-557). A key feature of this structure is a novel S-shaped oligomerization motif at the carboxyl terminus of the protein that mediates approximately 54% of the intermonomer contacts. Arg-308, located within this oligomerization motif, nucleates a series of intramonomer and intermonomer salt links. In contrast to the trimeric wild-type enzyme, the R308A, R308E, and R308K variants of arginase exist as monomeric species, as determined by gel filtration and analytical ultracentrifugation, indicating that mutation of Arg-308 shifts the equilibrium for trimer dissociation by at least a factor of 10(5). These monomeric arginase variants are catalytically active, with k(cat)/K(m) values that are 13-17% of the value for wild-type enzyme. The arginase variants are characterized by decreased temperature stability relative to the wild-type enzyme. Differential scanning calorimetry shows that the midpoint temperature for unfolding of the Arg-308 variants is in the range of 63.6-65.5 degrees C, while the corresponding value for the wild-type enzyme is 70 degrees C. The three-dimensional structure of the R308K variant has been determined at 3-A resolution. At the high protein concentrations utilized in the crystallizations, this variant exists as a trimer, but weakened salt link interactions are observed for Lys-308.


Assuntos
Arginase/química , Arginase/genética , Mutação , Sequência de Aminoácidos , Aminoácidos/química , Animais , Arginina/química , Varredura Diferencial de Calorimetria , Catálise , Cromatografia em Gel , Dicroísmo Circular , Cristalografia por Raios X , Dimerização , Espectroscopia de Ressonância de Spin Eletrônica , Elétrons , Cinética , Fígado/enzimologia , Manganês/química , Camundongos , Modelos Químicos , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Ligação Proteica , Conformação Proteica , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína , Ratos , Temperatura , Ultracentrifugação , Xenopus
4.
Biochemistry ; 40(9): 2678-88, 2001 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-11258879

RESUMO

The boronic acid-based arginine analogue S-(2-boronoethyl)-L-cysteine (BEC) has been synthesized and assayed as a slow-binding competitive inhibitor of the binuclear manganese metalloenzyme arginase. Kinetic measurements indicate a K(I) value of 0.4-0.6 microM, which is in reasonable agreement with the dissociation constant of 2.22 microM measured by isothermal titration calorimetry. The X-ray crystal structure of the arginase-BEC complex has been determined at 2.3 A resolution from crystals perfectly twinned by hemihedry. The structure of the complex reveals that the boronic acid moiety undergoes nucleophilic attack by metal-bridging hydroxide ion to yield a tetrahedral boronate anion that bridges the binuclear manganese cluster, thereby mimicking the tetrahedral intermediate (and its flanking transition states) in the arginine hydrolysis reaction. Accordingly, the binding mode of BEC is consistent with the structure-based mechanism proposed for arginase as outlined in Cox et al. [Cox, J. D., Cama, E., Colleluori D. M., Pethe, S., Boucher, J. S., Mansuy, D., Ash, D. E., and Christianson, D. W. (2001) Biochemistry 40, 2689-2701.]. Since BEC does not inhibit nitric oxide synthase, BEC serves as a valuable reagent to probe the physiological relationship between arginase and nitric oxide (NO) synthase in regulating the NO-dependent smooth muscle relaxation in human penile corpus cavernosum tissue that is required for erection. Consequently, we demonstrate that arginase is present in human penile corpus cavernosum tissue, and that the arginase inhibitor BEC causes significant enhancement of NO-dependent smooth muscle relaxation in this tissue. Therefore, human penile arginase is a potential target for the treatment of sexual dysfunction in the male.


Assuntos
Arginase/antagonistas & inibidores , Arginina/análogos & derivados , Arginina/metabolismo , Ácidos Borônicos/metabolismo , Inibidores Enzimáticos/metabolismo , Relaxamento Muscular/efeitos dos fármacos , Músculo Liso Vascular/enzimologia , Ereção Peniana/fisiologia , Animais , Arginase/biossíntese , Arginase/genética , Arginase/metabolismo , Arginina/farmacologia , Ligação Competitiva , Ácidos Borônicos/síntese química , Ácidos Borônicos/farmacologia , Calorimetria , Cristalografia por Raios X , Inibidores Enzimáticos/farmacologia , Humanos , Técnicas In Vitro , Cinética , Substâncias Macromoleculares , Masculino , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Ereção Peniana/efeitos dos fármacos , Pênis/irrigação sanguínea , Pênis/enzimologia , Pênis/inervação , RNA Mensageiro/biossíntese , Coelhos , Ratos , Termodinâmica
5.
Biochemistry ; 40(9): 2689-701, 2001 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-11258880

RESUMO

Arginase is a binuclear Mn(2+) metalloenzyme that catalyzes the hydrolysis of L-arginine to L-ornithine and urea. X-ray crystal structures of arginase complexed to substrate analogues N(omega)-hydroxy-L-arginine and N(omega)-hydroxy-nor-L-arginine, as well as the products L-ornithine and urea, complete a set of structural "snapshots" along the reaction coordinate of arginase catalysis when interpreted along with the X-ray crystal structure of the arginase-transition-state analogue complex described in Kim et al. [Kim, N. N., Cox, J. D., Baggio, R. F., Emig, F. A., Mistry, S., Harper, S. L., Speicher, D. W., Morris, Jr., S. M., Ash, D. E., Traish, A. M., and Christianson, D. W. (2001) Biochemistry 40, 2678-2688]. Taken together, these structures render important insight on the structural determinants of tight binding inhibitors. Furthermore, we demonstrate for the first time the structural mechanistic link between arginase and NO synthase through their respective complexes with N(omega)-hydroxy-L-arginine. That N(omega)-hydroxy-L-arginine is a catalytic intermediate for NO synthase and an inhibitor of arginase reflects the reciprocal metabolic relationship between these two critical enzymes of L-arginine catabolism.


Assuntos
Arginase/química , Arginase/metabolismo , Arginina/análogos & derivados , Substituição de Aminoácidos/genética , Animais , Arginase/antagonistas & inibidores , Arginase/genética , Arginina/química , Arginina/metabolismo , Ligação Competitiva/genética , Catálise , Cristalografia por Raios X , Cisteína/genética , Inibidores Enzimáticos/química , Inibidores Enzimáticos/metabolismo , Histidina/genética , Substâncias Macromoleculares , Modelos Moleculares , Mutagênese Sítio-Dirigida , Ornitina/química , Ornitina/metabolismo , Ratos , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Relação Estrutura-Atividade , Especificidade por Substrato/genética , Ureia/química , Ureia/metabolismo
7.
J Pharmacol Exp Ther ; 290(3): 1409-16, 1999 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10454520

RESUMO

An increase in arginase activity has been associated with the pathophysiology of a number of conditions, including an impairment in nonadrenergic and noncholinergic (NANC) nerve-mediated relaxation of the gastrointestinal smooth muscle. An arginase inhibitor may rectify this condition. We compared the effects of a newly designed arginase inhibitor, 2(S)-amino-6-boronohexanoic acid (ABH), with the currently available N(omega)-hydroxy-L-arginine (L-HO-Arg), on the NANC nerve-mediated internal anal sphincter (IAS) smooth-muscle relaxation and the arginase activity in the IAS and other tissues. Arginase caused an attenuation of the IAS smooth-muscle relaxations by NANC nerve stimulation that was restored by the arginase inhibitors. L-HO-Arg but not ABH caused dose-dependent and complete reversal of N(omega)-nitro-L-arginine-suppressed IAS relaxation that was similar to that seen with L-arginine. Both ABH and L-HO-Arg caused an augmentation of NANC nerve-mediated relaxation of the IAS. In the IAS, ABH was found to be approximately 250 times more potent than L-HO-Arg in inhibiting the arginase activity. L-HO-Arg was found to be 10 to 18 times more potent in inhibiting the arginase activity in the liver than in nonhepatic tissues. We conclude that arginase plays a significant role in the regulation of nitric oxide synthase-mediated NANC relaxation in the IAS. The advent of new and selective arginase inhibitors may play a significant role in the discrimination of arginase isozymes and have important pathophysiological and therapeutic implications in gastrointestinal motility disorders.


Assuntos
Aminoácidos/farmacologia , Aminocaproatos , Arginase/antagonistas & inibidores , Compostos de Boro/farmacologia , Inibidores Enzimáticos/farmacologia , Canal Anal/enzimologia , Canal Anal/inervação , Canal Anal/fisiologia , Animais , Arginase/metabolismo , Arginina/análogos & derivados , Arginina/farmacologia , Encéfalo/enzimologia , Feminino , Técnicas In Vitro , Isoenzimas/antagonistas & inibidores , Isoenzimas/metabolismo , Fígado/enzimologia , Masculino , Relaxamento Muscular/efeitos dos fármacos , Relaxamento Muscular/fisiologia , Músculo Liso/efeitos dos fármacos , Músculo Liso/inervação , Músculo Liso/fisiologia , Óxido Nítrico Sintase/antagonistas & inibidores , Nitroarginina/farmacologia , Gambás , Reto/enzimologia , Especificidade por Substrato
8.
Mol Genet Metab ; 64(4): 243-9, 1998 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9758714

RESUMO

Hyperargininemia is a rare autosomal recessive disorder that results from a deficiency of hepatic type I arginase. At the genetic level, this deficiency in arginase activity is a consequence of random point mutations throughout the gene that lead to premature termination of the protein or to substitution mutations. Given the high degree of sequence homology between human liver and rat liver enzymes, we have mapped both patient and nonpatient mutations of the human enzyme onto the structure of the rat liver enzyme to rationalize the molecular basis for the low activities of these mutant arginases. Mutations identified in hyperargininemia patients affect the structure and function of the enzyme by compromising active-site residues, packing interactions in the protein scaffolding, and/or quaternary structure by destabilizing the assembly of the arginase trimer.


Assuntos
Erros Inatos do Metabolismo dos Aminoácidos/genética , Arginase/genética , Arginina/sangue , Fígado/enzimologia , Mutação , Erros Inatos do Metabolismo dos Aminoácidos/enzimologia , Animais , Arginase/química , Sítios de Ligação , Biopolímeros , Humanos , Estrutura Molecular , Ratos
9.
J Neurosci ; 18(11): 4083-95, 1998 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-9592089

RESUMO

Catalase is an antioxidant enzyme that has been shown to inhibit apoptotic or necrotic neuronal death induced by hydrogen peroxide. We report the purification of a contaminating antiapoptotic activity from a commercial bovine liver catalase preparation by following its ability to inhibit apoptosis when applied extracellularly in multiple death paradigms. The antiapoptotic activity was identified by protein microsequencing as arginase, a urea cycle and nitric oxide synthase-regulating enzyme, and confirmed by demonstrating the presence of antiapoptotic activity in a >97% pure preparation of recombinant arginase. The pluripotency of recombinant arginase was demonstrated by its ability to inhibit apoptosis in multiple paradigms including rat cortical neurons induced to die by glutathione depletion and oxidative stress, by 100 nM staurosporine treatment, or by Sindbis virus infection. The protective effects of arginase in these apoptotic paradigms, in contrast to previous studies on excitotoxic neuronal necrosis, are independent of nitric oxide synthase inhibition. Rather, arginase-induced depletion of arginine leads to inhibition of protein synthesis, resulting in cell survival. Because inhibitors of nitric oxide synthesis and of protein synthesis have been shown to decrease necrotic and apoptotic death, respectively, in animal models of stroke and spinal cord injury, arginine-depleting enzymes, capable of simultaneously inhibiting protein synthesis and nitric oxide generation, may be propitious therapeutic agents for acute neurological diseases. Furthermore, our results suggest caution in attributing the cytoprotective effects of some catalase preparations to catalase.


Assuntos
Apoptose/fisiologia , Arginase/metabolismo , Fígado/enzimologia , Neurônios/citologia , Óxido Nítrico/metabolismo , Sequência de Aminoácidos , Aminoácidos/metabolismo , Animais , Apoptose/efeitos dos fármacos , Arginase/genética , Arginase/farmacologia , Catalase/genética , Catalase/metabolismo , Catalase/farmacologia , Bovinos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Córtex Cerebral/citologia , Inibidores Enzimáticos/farmacologia , Feto/citologia , Glutationa/metabolismo , Dados de Sequência Molecular , Fármacos Neuroprotetores/farmacologia , Estresse Oxidativo/fisiologia , Ratos , Ratos Sprague-Dawley , Estaurosporina/farmacologia
10.
Biochemistry ; 37(23): 8539-50, 1998 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-9622506

RESUMO

Rat liver arginase contains a dimanganese(II,II) center per subunit that is required for catalytic hydrolysis of l-arginine to form urea and l-ornithine. A recent crystallographic study has shown that the Mn2 center consists of two coordinatively inequivalent manganese(II) ions, MnA and MnB, bridged by a water (hydroxide) molecule and two aspartate residues [Kanyo et al. (1996) Nature 383, 554-557]. A conserved residue, His141, is located near the proposed substrate binding region at 4.2 A from the bridging solvent molecule. The present EPR studies reveal that there is no essential alteration of the Mn2 site upon mutation of His141 to an Asn residue, which lacks a potential acid/base residue, while the catalytic activity of the mutant enzyme is 10 times lower vs wild-type enzyme. The binding affinity of l-lysine, l-arginine (substrate), and Nomega-OH-l-arginine (type 2 binders) increases inversely with the pKa of the side chain. Binding of l-lysine is more than 10 times weaker, and the substrate Michaelis constant (Km) is >6-fold greater (weaker binding) in the His141Asn mutant than in wild-type arginase. L-Lysine and Nomega-OH-L-arginine, type 2 binders, induce extensive loss of the EPR intensity, suggesting direct coordination to the Mn2 center. From these data and the pH dependence of type 2 binders, we conclude that His141 functions as the base for deprotonation of the side-chain amino group of L-lysine and the substrate guanidinium group, -NH-C(NH2)2+ and that the unprotonated side chain of these amino acids is responsible for binding to the active site. A different class of inhibitors (type 1), including L-isoleucine, L-ornithine, and L-citrulline, suppresses enzymatic activity, producing only minor change in the zero-field splitting of the Mn2 EPR signal and no change in the EPR intensity, suggestive of minimal conformational transformation. We propose that type 1 alpha-amino acid inhibitors do not bind directly to either Mn ion, but interact with the recognition site on arginase for the alpha-aminocarboxylate groups of the substrate. A new mechanism for the arginase-catalyzed hydrolysis of L-arginine is proposed which has general relevance to all binuclear hydrolases: (1) Deprotonation of substrate l-arginine(H+) by His141 permits entry of the neutral guanidinium group into the buried Mn2 region. Binding of the substrate imino group (>C=NH), most likely to MnB, is coupled to breaking of the MnB-(mu-H2O) bond, forming a terminal aquo ligand on MnA. (2) Proton transfer from the terminal MnA-aqua ligand to the substrate Ndelta-guanidino atom forms the nucleophilic hydroxide on MnA and the cationic NdeltaH2+-guanidino leaving group. Protonation of the substrate -NdeltaH2+-group is likely assisted by hydrogen bonding to the juxtaposed anionic carboxylate group of Glu277. (3) Attack of the MnA-bound hydroxide at the electrophilic guanidino C-atom forms a tetrahedral intermediate. (4) Formation of products is initiated by cleavage of the Cepsilon-NdeltaH2+ bond, yielding urea and L-ornithine(H+).


Assuntos
Arginase/metabolismo , Arginina/metabolismo , Guanidina/metabolismo , Histidina/metabolismo , Fígado/enzimologia , Manganês/metabolismo , Animais , Arginase/antagonistas & inibidores , Arginase/genética , Arginina/análogos & derivados , Arginina/farmacologia , Asparagina/genética , Sítios de Ligação , Boratos/farmacologia , Citrulina/farmacologia , Espectroscopia de Ressonância de Spin Eletrônica , Histidina/genética , Concentração de Íons de Hidrogênio , Isoleucina/farmacologia , Lisina/farmacologia , Mutagênese Sítio-Dirigida , Ornitina/farmacologia , Prótons , Ratos , Especificidade por Substrato
11.
Biochemistry ; 36(47): 14392-402, 1997 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-9398157

RESUMO

Chicken liver pyruvate carboxylase catalyzes a nonclassical ping-pong mechanism in which the carboxylation of biotin at subsite 1 of the active site is coupled to the biotin-dependent carboxylation of pyruvate at subsite 2. The functions of two divalent cation cofactors and at least one monovalent cation cofactor in catalysis are not well understood. The oxyvanadyl cation, VO2+ does not support phosphoryl transfer at the first subsite, and uncouples the decarboxylation of oxaloacetate at subsite 2 from the formation of ATP at subsite 1. Stimulation of this oxaloacetate decarboxylase activity in the presence of substrates and cofactors of the first subsite, including VO2+, VOADP-, Pi, and acetyl CoA, suggests that these cofactors and substrates induce the movement of carboxybiotin from the second subsite to the first subsite, where it is decarboxylated. VO2+ EPR has provided evidence for enzymic and nucleotide divalent cation binding sites within the first subsite. The EPR properties of enzyme bound VO2+ were altered by bicarbonate, suggesting that this substrate ligands directly to VO2+ at the enzymic metal site. Fluorescence quenching experiments suggest that a monovalent cation may interact with bicarbonate at the first subsite as well. The results of this study provide evidence that (i) the extrinsic metal ion cofactors interact with the substrates at the first subsite, and that (ii) divalent cations play a role in coupling catalysis at the two nonoverlapping subsites by inducing the decarboxylation of carboxybiotin at the first subsite.


Assuntos
Fígado/enzimologia , Oxaloacetatos/metabolismo , Piruvato Carboxilase/química , Piruvato Carboxilase/metabolismo , Vanadatos/metabolismo , Animais , Bicarbonatos/farmacologia , Cátions Bivalentes/farmacologia , Galinhas , Espectroscopia de Ressonância de Spin Eletrônica , Ativação Enzimática , Cinética , Magnésio/farmacologia , Especificidade por Substrato , Vanadatos/farmacologia
12.
Biochemistry ; 36(32): 9847-58, 1997 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-9245417

RESUMO

The solution structures of the binuclear Mn centers in arginase, Mn catalase, and the Mn-substituted forms of the Fe enzymes ribonucleotide reductase and hemerythrin have been determined using X-ray absorption spectroscopy (XAS). X-ray absorption near edge structure (XANES) spectra for these proteins were compared to those obtained for Mn(II) models. The Mn model spectra show an inverse correlation between the XANES peak maximum and the root-mean-square (RMS) deviation in metal-ligand bond lengths. For these complexes, the XANES maxima appear to be more effective than the 1s --> 3d areas as an indicator of metal-site symmetry. Arginase and Mn-substituted ribonucleotide reductase have symmetric nearest neighbor environments with low RMS deviation in bond length, while Mn catalase and Mn-substituted hemerythrin appear to have a larger RMS bond length deviation. The 1s --> 3d areas for arginase and Mn-substituted ribonucleotide reductase are consistent with six coordinate Mn, while the 1s --> 3d areas for Mn catalase and Mn-substituted hemerythrin are larger, suggesting that one or both of the Mn ions are five-coordinate in these proteins. Extended x-ray absorption fine structure (EXAFS) spectra were used to determine the Mn2 core structure for the four proteins. In order to quantitate the number of histidine residues bound to the Mn2 centers, EXAFS data for the crystallographically characterized model hexakis-imidazole Mn(II) dichloride tetrahydrate were used to calibrate the Mn-imidazole multiple scattering interactions. These calibrated parameters allowed the outer shell EXAFS to be fit to give a lower limit on the number of bound histidine residues. The EXAFS spectra for Mn-substituted ribonucleotide reductase and arginase are nearly identical, with symmetric Mn-nearest neighbor environments and outer shell scattering consistent with a lower limit of one histidine per Mn2 core. In contrast, the EXAFS data for Mn catalase and Mn-substituted hemerythrin show two distinct Mn-nearest neighbor shells, modeled as Mn-O at ca. 2.1 A and Mn-N at ca. 2.3 A, and outer shell carbon scattering consistent with a lower limit of ca. 2-3 His residues per Mn2 core. Only Mn catalase shows clear evidence for Mn...Mn scattering. The observed Mn...Mn distance is 3.53 A, which is significantly longer than the approximately 3.3 A distances that are typically observed for Mn(II)2 cores with two single atom bridges, but which is typical of the distances seen in Mn(II)2 cores having one single atom bridge (e.g., aqua or hydroxo) together with one or two carboxylate bridges. The absence of EXAFS-detectable Mn...Mn interactions for the other three proteins suggests either that there are no single atom bridges in these cases or that the Mn...Mn interactions are more disordered.


Assuntos
Arginase/química , Catalase/química , Hemeritrina/química , Manganês/química , Ribonucleotídeo Redutases/química , Animais , Proteínas de Bactérias/química , Fígado/química , Fígado/enzimologia , Modelos Químicos , Ratos , Espectrometria por Raios X
13.
Biochemistry ; 36(34): 10558-65, 1997 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-9265637

RESUMO

Arginase is a thermostable (Tm = 75 degrees C) binuclear manganese metalloenzyme which hydrolyzes l-arginine to form l-ornithine and urea. The three-dimensional structures of native metal-depleted arginase, metal-loaded H101N arginase, and metal-depleted H101N arginase have been determined by X-ray crystallographic methods to probe the roles of the manganese ion in site A (Mn2+A) and its ligand H101 in catalysis and thermostability. We correlate these structures with thermal stability and catalytic activity measurements reported here and elsewhere [Cavalli, R. C., Burke, C. J., Kawamoto, S., Soprano, D. R., and Ash, D. E. (1994) Biochemistry 33, 10652-10657]. We conclude that the substitution of a wild-type histidine ligand to Mn2+A compromises metal binding, which in turn compromises protein thermostability and catalytic function. Therefore, a fully occupied binuclear manganese metal cluster is required for optimal catalysis and thermostability.


Assuntos
Arginase/química , Arginase/metabolismo , Manganês/metabolismo , Animais , Sítios de Ligação , Catálise , Dicroísmo Circular , Cristalografia por Raios X , Ativação Enzimática , Estabilidade Enzimática , Escherichia coli/genética , Fígado/enzimologia , Manganês/química , Manganês/farmacologia , Metaloproteínas/química , Metaloproteínas/metabolismo , Modelos Moleculares , Estrutura Molecular , Conformação Proteica , Dobramento de Proteína , Ratos , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Temperatura
14.
Biochemistry ; 36(51): 16239-46, 1997 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-9405058

RESUMO

Bifunctional peptidylglycine alpha-amidating enzyme (alpha-AE) catalyzes the two-step conversion of C-terminal glycine-extended peptides to C-terminal alpha-amidated peptides and glyoxylate. The first step is the ascorbate-, O2-, and copper-dependent hydroxylation of the alpha-carbon of the glycyl residue, producing an alpha-hydroxyglycine-extended peptide. The second step is the ascorbate-, O2-, and copper-independent dealkylation of the carbinolamide intermediate. We show that alpha-AE requires 1.1 +/- 0. 2 mol of zinc/mol of enzyme for maximal (S)-N-dansyl-Tyr-Val-alpha-hydroxyglycine dealkylation activity. Treatment of the enzyme with EDTA abolishes both the peptide hydroxylation and the carbinolamide dealkylation activities. Addition of Zn(II), Co(II), Cd(II), and Mn(II) partially restores carbinolamide dealkylation activity to the EDTA-treated enzyme. Addition of Co(II) produces the greatest restoration of dealkylation activity, 32% relative to a control not treated with EDTA, while Mn(II) addition results in the smallest restoration of dealkylation activity, only 3% relative to an untreated control. The structure and coordination of the zinc center has been investigated by X-ray absorption spectroscopy. EXAFS data are best interpreted by an average coordination of 2-3 histidine ligands and 1-2 non-histidine O/N ligands. Since catalytic zinc centers in other zinc metalloenzymes generally exhibit only O/N ligands to the zinc atom, a zinc-bound water or hydroxide may serve as a general base for the abstraction of the hydroxyl proton from the carbinolamide intermediate. Alternatively, the zinc may function in a structural role.


Assuntos
Oxigenases de Função Mista/química , Oxigenases de Função Mista/metabolismo , Complexos Multienzimáticos/química , Oligopeptídeos/metabolismo , Zinco/metabolismo , Amidas/metabolismo , Animais , Apoproteínas/isolamento & purificação , Apoproteínas/metabolismo , Células CHO , Cobre/metabolismo , Cricetinae , Ácido Edético/farmacologia , Cinética , Complexos Multienzimáticos/metabolismo , Ratos , Análise Espectral , Raios X , Zinco/análise , Zinco/química
15.
Nature ; 383(6600): 554-7, 1996 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-8849731

RESUMO

Each individual excretes roughly 10 kg of urea per year, as a result of the hydrolysis of arginine in the final cytosolic step of the urea cycle. This reaction allows the disposal of nitrogenous waste from protein catabolism, and is catalysed by the liver arginase enzyme. In other tissues that lack a complete urea cycle, arginase regulates cellular arginine and ornithine concentrations for biosynthetic reactions, including nitric oxide synthesis: in the macrophage, arginase activity is reciprocally coordinated with that of NO synthase to modulate NO-dependent cytotoxicity. The bioinorganic chemistry of arginase is particularly rich because this enzyme is one of very few that specifically requires a spin-coupled Mn2+-Mn2+ cluster for catalytic activity in vitro and in vivo. The 2.1 angstrom-resolution crystal structure of trimeric rat liver arginase reveals that this unique metal cluster resides at the bottom of an active-site cleft that is 15 angstroms deep. Analysis of the structure indicates that arginine hydrolysis is achieved by a metal-activated solvent molecule which symmetrically bridges the two Mn2+ ions.


Assuntos
Arginase/química , Manganês/química , Animais , Arginase/metabolismo , Arginina/metabolismo , Sítios de Ligação , Cristalografia por Raios X , Fígado/enzimologia , Metaloproteínas/química , Modelos Moleculares , Conformação Proteica , Estrutura Secundária de Proteína , Ratos
16.
Arch Biochem Biophys ; 327(1): 107-12, 1996 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-8615679

RESUMO

Treatment of rat liver arginase with N-bromosuccinimide results in modification of six tryptophan residues per enzyme molecule and is accompanied by loss of catalytic activity (E. Ber and G. Muzynska (1979) Acta Biochim. Pol. 26, 103-114). In order to probe the chemistry of N-bromosuccinimide inactivation and the role of tryptophan residues in catalysis, the two tryptophan residues of rat liver arginase, Trp122 and Trp164, have been separately mutated to phenylalanine using site-directed mutagenesis of the protein expressed in Escherichia coli. Both single Trp -> Phe mutant enzymes have kinetic parameters nearly identical to those for the wild-type enzyme. Treatment of native, wild-type, and each of the Trp -> Phe mutant enzymes with N-bromosuccinimide results in loss of absorbance at 280 nm and is accompanied by a loss of catalytic activity. However, treatment of the wild-type enzyme with N-bromosuccinimide in the presence of the arginase inhibitors NG-hydroxy-L-arginine or the combination of L-ornithine and borate protects against inactivation, even though tryptophan residues are modified. Treatment of the H101N and H126N mutant arginases with N-bromosuccinimide also results in loss of catalytic activity and modification of tryptophan residues. In contrast, the H141N mutant arginase is not inactivated by N-bromosuccinimide, indicating that His141 is the critical target for the N-bromosuccinimide inactivation of the enzyme.


Assuntos
Arginase/química , Arginase/metabolismo , Bromosuccinimida/farmacologia , Histidina , Fígado/enzimologia , Sequência de Aminoácidos , Animais , Arginase/antagonistas & inibidores , Sequência de Bases , Primers do DNA , Inibidores Enzimáticos/farmacologia , Humanos , Cinética , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Neurospora crassa/enzimologia , Fenilalanina , Mutação Puntual , Reação em Cadeia da Polimerase , Ratos , Proteínas Recombinantes/antagonistas & inibidores , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Rhizobium/enzimologia , Saccharomyces cerevisiae/enzimologia , Homologia de Sequência de Aminoácidos , Espectrofotometria Ultravioleta , Triptofano , Xenopus
17.
FEBS Lett ; 366(2-3): 165-9, 1995 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-7540562

RESUMO

Peptidylglycine alpha-amidating enzyme (alpha-AE) and dopamine beta-monooxygenase (D beta M), two copper-dependent monooxygenases that have catalytic and structural similarities, are irreversibly inactivated by sodium sulfite in a time- and concentration-dependent manner. Studies with alpha-AE show that the sulfite-mediated inactivation is dependent on the presence of redox active transition metals free in solution, with Cu(II) being the most effective in supporting the inactivation reaction. Sulfite inactivation of alpha-AE is specific for the monooxygenase reaction of this bifunctional enzyme and amidated peptides provide protection against the inactivation. Consequently, the sulfite-mediated inactivation of alpha-AE and D beta M most likely results from the transition metal-catalyzed oxidation of sulfite to the sulfite radical, SO3-.


Assuntos
Cobre/farmacologia , Dopamina beta-Hidroxilase/antagonistas & inibidores , Oxigenases de Função Mista/antagonistas & inibidores , Complexos Multienzimáticos , Sulfitos/farmacologia , Sequência de Aminoácidos , Animais , Células CHO , Cricetinae , Ativação Enzimática/efeitos dos fármacos , Radicais Livres , Dados de Sequência Molecular , Proteínas de Neoplasias/antagonistas & inibidores , Oxirredução , Ratos , Proteínas Recombinantes de Fusão/antagonistas & inibidores , Substância P/farmacologia , Neoplasias da Glândula Tireoide/enzimologia
18.
Arch Biochem Biophys ; 317(1): 93-102, 1995 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-7872809

RESUMO

Peptidylglycine alpha-amidating enzyme catalyzes the two-step conversion of C-terminal glycine-extended peptides to C-terminal alpha-amidated peptides and glyoxylate in a reaction that requires O2, ascorbate and 2 mol of copper per mole of enzyme [Kulathila et al. (1994) Arch. Biochem. Biophys. 311, 191-195]. Peptides with a C-terminal alpha-hydroxyglycine residue are intermediates in the amidation reaction. Benzylhydrazine inactivates the enzymatic conversion of dansyl-Tyr-Val-Gly to dansyl-Tyr-Val-NH2 in a time- and concentration-dependent manner. In contrast, the enzymatic conversion of dansyl-Tyr-Val-alpha-hydroxyglycine to dansyl-Tyr-Val-NH2 is unaffected by benzylhydrazine. The plot of 1/(inactivation rate) vs 1/[benzylhydrazine] is parabolic, indicating that the inactivation results from the interaction of 2 mol of benzylhydrazine per mole of enzyme. EPR spectra obtained from benzylhydrazine inactivation reactions carried out in the presence of a radical trap, alpha-(4-pyridyl-1-oxide)-N-tert-butylnitrone, show the formation of a carbon-centered benzyl radical. The benzyl radical most likely results from redox chemistry between benzylhydrazine and the enzyme-bound Cu(II) ions because EPR studies show that enzyme-bound Cu(II) is reduced to Cu(I) in the presence of benzylhydrazine. The kinetic constants for benzylhydrazine as a reductant in the amidation reaction were determined at benzylhydrazine concentrations too low to cause significant enzyme inactivation. Mimosine exhibits mixed inhibition vs benzylhydrazine; however, previous results have shown that benzylhydrazine is competitive vs ascorbate [Miller et al. (1992) Arch. Biochem. Biophys. 298, 380-388]. This change in kinetic mechanism coupled with the nonlinear inactivation kinetics have lead to a proposal that the two enzyme-bound Cu(II) atoms are nonequivalent with respect to their reduction by benzylhydrazine.


Assuntos
Cobre/química , Hidrazinas/farmacologia , Oxigenases de Função Mista/antagonistas & inibidores , Complexos Multienzimáticos , Animais , Sítios de Ligação , Células CHO , Cricetinae , Ativação Enzimática/efeitos dos fármacos , Hidrazinas/química , Cinética , Mimosina/farmacologia , Oxigenases de Função Mista/química
19.
Biochemistry ; 34(6): 2015-25, 1995 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-7849059

RESUMO

The dimanganese (II,II) catalase from Thermus thermophilus, MnCat(II,II), arginase from rat liver, Arg(II,II), and several dimanganese(II,II) compounds, LMn2XY2, which are functional catalase mimics, all possess a pair of coupled Mn(II) ions in their catalytic sites. For each of these, we have measured by EPR spectroscopy the relative energies separating the three lowest electronic states (singlet, triplet, and quintet), described a general method for extracting the individual spectra for these states by multicomponent analysis, and determined the Mn-Mn separation. The triplet-singlet and quintet-singlet energy gaps were modeled well by fitting the temperature dependence of the EPR intensities to a Boltzmann expression for a pair of Mn(II) ions coupled by isotropic Heisenberg spin exchange (-2JS1S2). This dependence indicates diamagnetic ground states with delta E10 (cm-1) = magnitude of 2J = 4 and 11.2 cm-1 for Arg-(II,II)(+borate) and MnCat(II,II)(phosphate), respectively. This large difference in magnitude of 2J reflects either a difference in the bridging ligands or, possibly, a weaker ligand field (larger ionization potential) for the Mn(II) ions in arginase. In n-butanol/CH2Cl2 the triplet-singlet energy gaps for [LMn2(CH3CO2)](C1O4)2 (1), [LMn2(CH3CO2)3] (2), and [LMn2Cl3] (3), where HL = N,N,N',N'-tetrakis(2-methylenebenzimidazole)-1,3-diaminopropan+ ++-2-ol, are 23-24 cm-1. Comparison of the Heisenberg exchange interaction constants for more than 30 dimanganese(II,II) complexes suggests a possible bridging structure of (mu-OH)(mu-carboxylate)1-2 for MnCat(II,II), while the 3-fold weaker coupling in Arg(II,II) suggests mu-aqua in place of mu-hydroxide. EPR spectra of both the triplet and quintet electronic states were extracted and found to exhibit zero-field splittings (ZFS) and resolved 55Mn hyperfine splittings indicating spin-coupled Mn2-(II,II) species. The major ZFS interaction could be attributed to the magnetic dipole-dipole interaction between the Mn(II) ions. A linear correlation is observed between the crystallographically determined Mn-Mn distance and the ZFS of the quintet state (D2) for five dimanganese pairs for which both data sets are available. Using this correlation, the Mn-Mn distance in Arg(II,II) is predicted to be 3.36-3.57 A for the native enzyme (multiple forms) and 3.59 A for MnCat(II,II)(phosphate). Addition of the inhibitor borate to Arg(II,II) simplifies the ZFS, indicative of conversion to a single species with mean Mn-Mn separation of 3.50 A. The second metal ion in dinuclear complexes possessing a shared bridging ligand has been shown to attenuate the strength of the mu-ligand field potential, as monitored by the strength of the single ion ZFS.(ABSTRACT TRUNCATED AT 250 WORDS)


Assuntos
Arginase/química , Catalase/química , Fígado/enzimologia , Manganês/química , Animais , Sítios de Ligação , Cloretos/química , Espectroscopia de Ressonância de Spin Eletrônica , Fosfatos/química , Ratos , Temperatura , Termodinâmica , Thermus thermophilus/enzimologia
20.
Biochemistry ; 33(35): 10652-7, 1994 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-8075066

RESUMO

Rat liver arginase has been overexpressed in Escherichia coli using a T7-based expression system. The kinetic properties of the recombinant wild-type protein are essentially identical to those of the native rat liver enzyme. The recombinant wild-type protein contains six Mn(II) ions per trimer, in good agreement with results obtained with the fully active native enzyme. However, in contrast to the native enzyme which loses three Mn(II) per trimer upon extended dialysis, the recombinant protein binds Mn(II) tenaciously, and retains six Mn(II) per trimer even after extensive dialysis. Three histidine residues, corresponding to His101, His126, and His141 in the rat liver enzyme, are highly conserved in arginases from evolutionarily divergent species. The replacement of His101 and His126 with Asn by site-directed mutagenesis produced only modest effects on enzymatic activity when measured in the presence of Mn(II) ions. However, EDTA treatment of these mutant enzymes reduced activity to < 0.2% of that for the wild-type enzyme. The activity of wild-type enzyme and the His141 Asn mutant was unaffected by treatment with EDTA. Thus, His101 and His126 are proposed to be ligands to the binuclear Mn(II) center of the enzyme. The His141 Asn mutation produced an enzyme which, in contrast to the native, wild-type, His101 Asn, and His126 Asn arginases, was not inactivated by diethyl pyrocarbonate. These results suggest a catalytic role for His141.


Assuntos
Arginase/metabolismo , Animais , Arginase/genética , Dicroísmo Circular , Dietil Pirocarbonato/química , Ácido Edético/química , Escherichia coli , Histidina/química , Humanos , Cinética , Fígado/enzimologia , Manganês/química , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Oligodesoxirribonucleotídeos/química , Estrutura Secundária de Proteína , Ratos , Proteínas Recombinantes , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Relação Estrutura-Atividade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...