Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros








Intervalo de ano de publicação
1.
Int J Mol Sci ; 23(12)2022 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-35742891

RESUMO

Arginase catalyzes the hydrolysis of L-arginine into L-ornithine and urea. This enzyme has several analogies with agmatinase, which catalyzes the hydrolysis of agmatine into putrescine and urea. However, this contrasts with the highlighted specificity that each one presents for their respective substrate. A comparison of available crystal structures for arginases reveals an important difference in the extension of two loops located in the entrance of the active site. The first, denominated loop A (I129-L140) contains the residues that interact with the alpha carboxyl group or arginine of arginase, and the loop B (D181-P184) contains the residues that interact with the alpha amino group of arginine. In this work, to determine the importance of these loops in the specificity of arginase, single, double, and triple arginase mutants in these loops were constructed, as well as chimeras between type I human arginase and E. coli agmatinase. In previous studies, the substitution of N130D in arginase (in loop A) generated a species capable of hydrolyzing arginine and agmatine. Now, the specificity of arginase is completely altered, generating a chimeric species that is only active with agmatine as a substrate, by substituting I129T, N130Y, and T131A together with the elimination of residues P132, L133, and T134. In addition, Quantum Mechanic/Molecular Mechanic (QM/MM) calculations were carried out to study the accommodation of the substrates in in the active site of this chimera. With these results it is concluded that this loop is decisive to discriminate the type of substrate susceptible to be hydrolyzed by arginase. Evidence was also obtained to define the loop B as a structural determinant for substrate affinity. Concretely, the double mutation D181T and V182E generate an enzyme with an essentially unaltered kcat value, but with a significantly increased Km value for arginine and a significant decrease in affinity for its product ornithine.


Assuntos
Agmatina , Arginase , Arginase/metabolismo , Arginina/química , Escherichia coli/genética , Escherichia coli/metabolismo , Humanos , Ornitina , Especificidade por Substrato , Ureia
2.
J Struct Biol ; 211(2): 107533, 2020 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-32450233

RESUMO

Arginase (EC 3.5.3.1) catalyzes the hydrolysis of L-arginine to L-ornithine and urea, and requires a bivalent cation, especially Mn2+ for its catalytic activity. It is a component of the urea cycle and regulates the intracellular levels of l-arginine, which makes the arginase a target for treatment of vascular diseases and asthma. Mammalian arginases contain an unusual S-shaped motif located at the intermonomeric interface. Until now, the studies were limited to structural role of the motif. Then, our interest was focused on functional aspects and our hypothesis has been that the motif is essential for maintain the oligomeric state, having Arg308 as a central axis. Previously, we have shown that the R308A mutant is monomeric and re-associates to the trimeric-cooperative state in the presence of low concentrations of guanidine chloride. We have now mutated Asp204 that interacts with Arg308 in the neighbor subunit, and also we mutated Glu256, proposed as important for oligomerization. Concretely, the human arginase I mutants D204A, D204E, E256A, E256Q and E256D were generated and examined. No differences were observed in the kinetic parameters at pH 9.5 or in tryptophan fluorescence. However, the D204A and E256Q variants were monomeric. On the other hand, D204E and E256D proved to be trimeric and kinetically cooperative at pH 7.5, whereas hyperbolic kinetics was exhibited by E256A, also trimeric. The results obtained strongly support the importance of the interaction between Arg255 and Glu256 in the cooperative properties of arginase, and Asp204 would be relevant to maintain the oligomeric state through salt bridges with Arg255 and Arg308.


Assuntos
Arginase/ultraestrutura , Arginina/genética , Ácido Aspártico/genética , Conformação Proteica , Arginase/química , Arginase/genética , Arginina/química , Ácido Aspártico/química , Ácido Glutâmico/química , Ácido Glutâmico/genética , Humanos , Cinética , Substâncias Macromoleculares , Modelos Moleculares , Mutação/genética , Multimerização Proteica/genética
3.
J Inorg Biochem ; 202: 110812, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31731096

RESUMO

Ureohydrolases form a conserved family of enzymes with a strict requirement for divalent metal ions for catalytic activity. They catalyze the hydrolysis of the guanidino group and produce urea. In their active sites six highly conserved amino acid residues form a binding pocket for two catalytically essential metal ions that are needed to activate a water molecule to initiate the hydrolysis of the guanidino group in a nucleophilic attack. Focus in this review is on two members of the ureohydrolase family, the Mn2+-dependent arginase and agmatinase, which play important roles in functions related to replication and cell survival. We will focus in particular on Mn2+ binding interactions, and on how this metal ion contributes to the reaction catalyzed by these enzymes. We also include the agmatinase-like protein (ALP) because it is functionally closely related to agmatinase, also requires at least one Mn2+ ion for catalytic activity, but may possess an active site that differs significantly from all other known ureohydrolases.


Assuntos
Arginase , Manganês , Ureo-Hidrolases , Arginase/química , Arginase/metabolismo , Catálise , Manganês/química , Manganês/metabolismo , Ureo-Hidrolases/química , Ureo-Hidrolases/metabolismo
4.
Metabolism ; 81: 35-44, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29162499

RESUMO

Agmatine (1-amino-4-guanidinobutane), a precursor for polyamine biosynthesis, has been identified as an important neuromodulator with anticonvulsant, antineurotoxic and antidepressant actions in the brain. In this context it has emerged as an important mediator of addiction/satiety pathways associated with alcohol misuse. Consequently, the regulation of the activity of key enzymes in agmatine metabolism is an attractive strategy to combat alcoholism and related addiction disorders. Agmatine results from the decarboxylation of L-arginine in a reaction catalyzed by arginine decarboxylase (ADC), and can be converted to either guanidine butyraldehyde by diamine oxidase (DAO) or putrescine and urea by the enzyme agmatinase (AGM) or the more recently identified AGM-like protein (ALP). In rat brain, agmatine, AGM and ALP are predominantly localised in areas associated with roles in appetitive and craving (drug-reinstatement) behaviors. Thus, inhibitors of AGM or ALP are promising agents for the treatment of addictions. In this review, the properties of DAO, AGM and ALP are discussed with a view to their role in the agmatine metabolism in mammals.


Assuntos
Agmatina/metabolismo , Neurotransmissores/metabolismo , Amina Oxidase (contendo Cobre)/fisiologia , Animais , Carboxiliases/fisiologia , Humanos , Ureo-Hidrolases/fisiologia
5.
J Inorg Biochem ; 166: 122-125, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27846445

RESUMO

Agmatine (1-amino-4-guanidinobutane) plays an important role in a range of metabolic functions, in particular in the brain. Agmatinases (AGMs) are enzymes capable of converting agmatine to the polyamine putrescine and urea. AGMs belong to the family of Mn2+-dependent ureahydrolases. However, no AGM from a mammalian source has yet been extracted in catalytically active form. While in human AGM the six amino acid ligands that coordinate the two Mn2+ ions in the active site are conserved, four mutations are observed in the murine enzyme. Here, we demonstrate that similar to its human counterpart murine AGM does not appear to have in vitro catalytic activity, independent of the presence of Mn2+. However, in presence of agmatine both enzymes are very efficient in promoting cell growth of a yeast strain that is deficient in polyamine biosynthesis (Saccharomyces cerevisiae strain TRY104Δspe1). Furthermore, mutations among the putative Mn2+ binding residues had no effect on the ability of murine AGM to promote growth of the yeast culture. It thus appears that mammalian AGMs form a distinct group within the family of ureahydrolases that (i) either fold in a manner distinct from other members in this family, or (ii) require accessory proteins to bind Mn2+ in a mechanism related to that observed for the Ni2+-dependent urease.


Assuntos
Agmatina/metabolismo , Manganês/metabolismo , Ureo-Hidrolases/metabolismo , Agmatina/química , Animais , Sítios de Ligação , Manganês/química , Camundongos , Proteínas Recombinantes , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Ureo-Hidrolases/química , Ureo-Hidrolases/genética
6.
Histochem Cell Biol ; 145(3): 305-13, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26678503

RESUMO

Agmatine, a precursor for polyamine biosynthesis, is also associated with neurotransmitter, anticonvulsant, antineurotoxic and antidepressant actions in the brain. This molecule results from the decarboxylation of L-arginine by arginine decarboxylase, and it is hydrolyzed to urea and putrescine by agmatinase. Recently, we have described a new protein that also hydrolyzes agmatine, agmatinase-like protein (ALP), which was identified through immunohistochemical analysis in the hypothalamus and hippocampus of rats. However, its sequence differs greatly from all known agmatinases and does not contain the typical Mn(2+) ligands associated with the urea hydrolase family of proteins. ALP has a LIM-like domain close to its carboxyl terminus, and the removal of which results in a truncated variant with a tenfold increased k cat value and a threefold decreased K m value for agmatine. Analysis of the gene database revealed several transcripts, denominated LIMCH1 isoforms, with extreme 3' sequences identical to ALP. Limch1 gene products have been described as members of a multi-domain family of proteins with the biggest isoform containing a calponin homology (CH) domain at its N-terminus. Here, we cloned two LIMCH1 transcripts, one of 3177 bp and the other of 2709 bp (ALP contains 1569 bp) and analyzed LIMCH1 expression and distribution in rat brain using RT-PCR, Western blot and immunohistochemical analyses. LIMCH1 was detected mainly in the hypothalamic and hippocampal regions, which is similar to the distribution of ALP and agmatine in brain. In addition, we cloned and expressed both isoforms in E. coli and confirmed that they were catalytically active on agmatine with kinetic parameters similar to ALP. LIM domain-truncated variants of both isoforms moderately increased the k cat and catalytic efficiency. Thus, we propose that LIMCH1 is useful to regulate the intracellular concentrations of the neurotransmitter/neuromodulator, agmatine.


Assuntos
Encéfalo/metabolismo , Ureo-Hidrolases/genética , Ureo-Hidrolases/metabolismo , Animais , Linhagem Celular , Clonagem Molecular , Masculino , Isoformas de Proteínas/análise , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Ratos , Ratos Sprague-Dawley , Ureo-Hidrolases/análise
7.
J Inorg Biochem ; 145: 65-9, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25635913

RESUMO

Agmatinase is an enzyme that catalyzes the hydrolysis of agmatine, a compound that is associated with numerous functions in the brain of mammalian organisms such as neurotransmitter, anticonvulsant, antinociceptive, anxiolytic and antidepressant-like actions. To date the only characterized agmatinases with significant enzymatic activity were extracted from bacterial organisms. These agmatinases are closely related to another ureahydrolase, arginase; both have binuclear Mn(2+) centers in their active sites. An agmatinase-like protein (ALP) from rat brain was identified that bears no sequence homology to known agmatinases (E. Uribe, M. Salas, S. Enriquez, M.S. Orellana, N. Carvajal, Arch. Biochem. Biophys. 461(2007) 146-150). Since all known ureahydrolases contain histidines in their binuclear Mn(2+) site each of the five histidine residues in ALP was individually replaced by alanines to identify those that may be involved in metal ion binding. Reactivation assays and thermal stability measurements indicated that His206 is likely to interact with a Mn(2+) bound to a high affinity site. In contrast, His65 and possibly His435 are important for binding of a second Mn(2+) to a lower affinity site. Metal ion binding to that site is not only leading to an increase in reactivity but also enzyme stability. Thus, similar to bacterial agmatinases and some of the antibiotic-degrading, Zn(2+)-dependent metallo-ß-lactamases ALP appears to be active in the mono and binuclear form, with binding of the second metal ion increasing both reactivity and stability.


Assuntos
Manganês/química , Ureo-Hidrolases/química , Cátions Bivalentes , Ligação Proteica
8.
Biochimie ; 108: 8-12, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25447142

RESUMO

To substantiate the functionality of a crystallographically evidenced allosteric site in Bacillus caldovelox arginase (Bewley et al., 1999), we have examined the kinetic consequences of the single mutations of Asp199 and Glu256, which interact with l-arginine in this site. The introduced mutations (Asp199 → Asn, Asp199 → Ala, Glu256 → Gln and Glu256 → Ala) had no effect on the hexameric structure of the enzyme (mol. wt. 195 ± 10 kDa). However, in contrast with the Michaelis-Menten kinetics exhibited by the wild-type species, the D199A, D199N, E256A and E256Q variants exhibited positive cooperativity with respect to l-arginine. The Glu278 → Ala mutation, which compromise interactions at the trimer-trimer interface, yielded trimeric species (mol. wt. 100 ± 5 kDa) exhibiting hyperbolic kinetics that changed to sigmoidal by the additional Glu256 → Ala mutation. In addition to demonstrating the total functionality of the trimer, our results also suggest that B. caldovelox is kinetically cooperative and that the commonly detected hyperbolic behavior results from binding of l-arginine as a typical allosteric activator.


Assuntos
Sítio Alostérico/genética , Arginase/química , Arginase/metabolismo , Arginina/farmacologia , Bacillus/enzimologia , Mutagênese Sítio-Dirigida , Arginase/genética , Ativação Enzimática/efeitos dos fármacos , Cinética , Modelos Moleculares , Mutação , Multimerização Proteica , Estrutura Quaternária de Proteína
9.
J Inorg Biochem ; 132: 92-5, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24411595

RESUMO

Agmatine is a precursor for polyamine biosynthesis also associated to neurotransmitter, anticonvulsant, antineurotoxic and antidepressant actions in the brain. It results from decarboxylation of l-arginine by arginine decarboxylase and it is hydrolyzed to urea and putrescine by agmatinase. Recently, we have described a new protein which also hydrolyzes agmatine although its sequence greatly differs from all known agmatinases. This agmatinase-like protein (ALP) contains a LIM-like double Zn-finger domain close to its carboxyl terminus, whose removal results in a truncated variant with a 10-fold increased kcat, and a 3-fold decreased Km value for agmatine. Our proposal was that the LIM-domain functions as an autoinhibitory, regulatory entity for ALP. Results in this report provide additional support for the postulated inhibitory effect. The purified isolated LIM domain was shown to be competitively inhibitory to a truncated variant ALP (lacking the LIM-domain), but not to the wild-type species. The C453A variant was shown to be a Zn(2+)-free enzyme with kinetic parameters similar to those of the truncated-ALP. A molecular dynamic simulation of a modeled LIM-domain 3D structure showed that, as a consequence of C453A mutation, the coordination of the zinc ion is broken and the structure of the zinc finger is melted. The inhibitory action of the LIM/double Zinc-finger motif was associated to a significant conformational change, as detected by tryptophan fluorescence studies, but was not related to changes in the association of the enzyme with the catalytically essential Mn(2+).


Assuntos
Proteínas Correpressoras/química , Proteínas com Domínio LIM/química , Modelos Moleculares , Ureo-Hidrolases/química , Dedos de Zinco , Sequência de Aminoácidos , Variação Genética , Humanos , Mutação , Dobramento de Proteína , Ureo-Hidrolases/genética , Ureo-Hidrolases/metabolismo
10.
Arch Biochem Biophys ; 512(1): 107-10, 2011 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-21601560

RESUMO

We recently cloned a rat brain agmatinase-like protein (ALP) whose amino acid sequence greatly differs from other agmatinases and exhibits a LIM-like domain close to its carboxyl terminus. The protein was immunohistochemically detected in the hypothalamic region and hippocampal astrocytes and neurons. We now show that truncated species, lacking the LIM-type domain, retains the dimeric structure of the wild-type protein but exhibits a 10-fold increased k(cat), a 3-fold decreased K(m) value for agmatine and altered intrinsic tryptophan fluorescent properties. As expected for a LIM protein, zinc was detected only in the wild-type ALP (∼2 Zn(2+)/monomer). Our proposal is that the LIM domain functions as an autoinhibitory entity and that inhibition is reversed by interaction of the domain with some yet undefined brain protein.


Assuntos
Encéfalo/enzimologia , Ureo-Hidrolases/química , Ureo-Hidrolases/metabolismo , Agmatina/metabolismo , Animais , Sequência Consenso , Cinética , Estrutura Terciária de Proteína , Ratos , Deleção de Sequência , Ureo-Hidrolases/genética , Dedos de Zinco
11.
Histochem Cell Biol ; 134(2): 137-44, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20607275

RESUMO

Agmatinase catalyzes the hydrolysis of agmatine into putrescine and urea, and agmatine (decarboxylated L: -arginine) plays several roles in mammalian tissues, including neurotransmitter/neuromodulatory actions in the brain. Injection of agmatine in animals produces anticonvulsant, antineurotoxic and antidepressant-like actions. Information regarding the enzymatic aspects of agmatine metabolism in mammals, especially related to its degradation, is relatively scarce. The explanation for this is the lack of enzymatically active preparations of mammalian agmatinase. Recently, we have cloned a protein from a cDNA rat brain library having agmatinase activity although its amino acid sequence greatly differs from all known agmatinases, we called agmatinase-like protein. In this work, we analyzed the expression of this enzyme in the rat brain by means of RT-PCR and immunohistochemical analysis using a polyclonal antibody generated against the recombinant agmatinase-like protein. The agmatinase-like protein was detected in the hypothalamus in glial cells and arcuate nucleus neurons, and in hippocampus astrocytes and neurons, but not in brain cortex. In general, detected localization of agmatinase-like protein coincides with that described for its substrate agmatine and our results help to explain several reported effects of agmatine in the brain. Concretely, a role in the regulation of intracellular concentrations of the neurotransmitter/neuromodulator agmatine is suggested for the brain agmatinase-like protein.


Assuntos
Encéfalo/enzimologia , Ureo-Hidrolases/análise , Agmatina/metabolismo , Animais , Química Encefálica , Hipotálamo/enzimologia , Neuroglia/enzimologia , Ratos , Proteínas Recombinantes/análise , Distribuição Tecidual
12.
High Alt Med Biol ; 10(4): 373-81, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-20039818

RESUMO

The aim of this study was to analyze the activity and expression levels of arginase I and II and to monitor the cardiovascular and hematological responses in tolerant and intolerant rats exposed to chronic intermittent hypobaric hypoxia (CIHH). Male Wistar rats (age: 3.0 +/- 0.4 months, weight: 250 +/- 25 g; n = 30) were randomly divided into two groups: CIHH2 x 2 (2 days hypoxia, 2 days normoxia, n = 20) and NX (normoxia, n = 10). The hypoxia was simulated in a hypobaric chamber at 428 torr. Tolerance was determined according to a previous protocol. Arginase activity was measured in lung and heart tissues, and the expression levels were determined by a (RT-PCR) assay in lung tissue. Results showed that the intolerants rats had lower body weight, higher hematocrit (Hct) (74 +/- 4% vs. 61 +/- 2%, p < 0.05), higher values of systolic blood pressure (SBP) (183 +/- 3.7 mmHg vs. 147 +/- 5.4 mmHg, p < 0.05), and higher arginase activity. In addition, RT-PCR analysis from lung tissue showed an overexpression of arginase II in the intolerant group (p < 0.01). However, tolerants had similar values as the NX group (p = ns). Further, a correlation was found between arginase activity and SBP in the heart (r(2) = 0.596, p < 0.001). An upregulation of arginase type II could be pivotal in understanding the pathogenesis of systolic hypertension and probably other phenomena associated with intermittent hypobaric hypoxia. A schematic explanation of these relations is proposed.


Assuntos
Aclimatação , Altitude , Arginase/metabolismo , Hipertensão/metabolismo , Hipóxia/metabolismo , Regulação para Cima , Animais , Pressão Atmosférica , Pressão Sanguínea , Peso Corporal , Frequência Cardíaca , Hematócrito , Hipertensão/complicações , Hipertensão/fisiopatologia , Hipóxia/complicações , Pulmão/metabolismo , Masculino , Miocárdio/metabolismo , Nitratos/sangue , Nitritos/sangue , Ratos , Ratos Wistar
13.
Sao Paulo Med J ; 127(3): 140-4, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19820874

RESUMO

CONTEXT AND OBJECTIVE: Epidemiological data provide useful information for clinical practice and investigations. This study aimed to determine glomerular disease frequencies in a region of Colombia and it represents the basis for future studies. DESIGN AND SETTING: Single-center retrospective analysis at the University of Antioquia, Colombia. METHODS: All native renal biopsies (July 1998 to December 2007) were reviewed, but only glomerular diseases were analyzed. The diagnosis of each case was based on histological, immunopathological and clinical features. RESULTS: A total of 1,040 biopsies were included. In 302 cases (29.0%), the patient's age was

Assuntos
Glomerulosclerose Segmentar e Focal/epidemiologia , Rim/patologia , Adulto , Distribuição por Idade , Idoso , Biópsia , Colômbia/epidemiologia , Bases de Dados Factuais , Feminino , Glomerulonefrite por IGA/epidemiologia , Glomerulosclerose Segmentar e Focal/patologia , Humanos , Lactente , Recém-Nascido , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos , Adulto Jovem
14.
Arch Biochem Biophys ; 481(1): 16-20, 2009 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-18957279

RESUMO

The functional significance of a C-terminal S-shaped motif (residues 304-322) in human arginase I was explored by examining the kinetic properties of the R308A mutant and truncated species terminating in either Arg-308 or Ala-308. Replacement of Arg-308 with alanine, with or without truncation, yielded monomeric species. All mutants were kinetically indistinguishable from the wild-type enzyme at the optimum pH of 9.5. At the more physiological, pH 7.5, hyperbolic kinetics was observed for all the mutants, in contrast with the cooperative behavior exhibited by the wild-type species. In the presence of 2mM guanidinium chloride (Gdn(+)), the single mutant R308A changed to a trimeric and kinetically cooperative form, whereas the other enzyme variants were not altered. The S-shaped motif is suggested as essential for the cooperative response of the enzyme to l-arginine at pH 7.5. Gdn(+) is suggested to mimic the guanidine group of Arg-308 at the monomer-monomer interface.


Assuntos
Arginase/química , Motivos de Aminoácidos , Arginase/genética , Arginina/química , Guanidina , Humanos , Concentração de Íons de Hidrogênio , Mutação , Proteínas Recombinantes/química , Proteínas Recombinantes/genética
15.
São Paulo med. j ; 127(3): 140-144, 2009. graf, tab
Artigo em Inglês | LILACS | ID: lil-528109

RESUMO

CONTEXT AND OBJECTIVE: Epidemiological data provide useful information for clinical practice and investigations. This study aimed to determine glomerular disease frequencies in a region of Colombia and it represents the basis for future studies. DESIGN AND SETTING: Single-center retrospective analysis at the University of Antioquia, Colombia. METHODS: All native renal biopsies (July 1998 to December 2007) were reviewed, but only glomerular diseases were analyzed. The diagnosis of each case was based on histological, immunopathological and clinical features. RESULTS: A total of 1,040 biopsies were included. In 302 cases (29.0 percent), the patient's age was < 15 years. Primary glomerular diseases were diagnosed in 828 biopsies (79.6 percent) and secondary in 212 (20.4 percent). The most common primary diseases were focal and segmental glomerulosclerosis (FSGS) (34.8 percent), immunoglobulin A (IgA) nephropathy (IgAN) (11.8 percent), membranous glomerulonephritis (MGN) (10.6 percent), minimal change disease (MCD) (10.6 percent), crescentic glomerulonephritis (GN) (5.6 percent), and non-IgA mesangial proliferative GN (5.6 percent). Postinfectious GN represented 10.7 percent of the diagnoses if included as primary GN. Lupus nephritis corresponded to 17.8 percent of the entire series. In adults, the order of the most frequent primary diseases was: FSGS, IgAN, MGN, crescentic GN and MCD. In children (< 15 years), the most frequent were: FSGS, postinfectious GN, MCD, non-IgA mesangial proliferative GN, endocapillary diffuse GN and IgAN. CONCLUSIONS: As among Afro-Americans, FSGS is the most frequent type of glomerulopathy in our population, but in our group, there are more cases of IgAN. The reasons for these findings are unclear. This information is an important contribution towards understanding the prevalence of renal diseases in Latin America.


CONTEXTO Y OBJETIVO: Los datos epidemiológicos dan información útil en clínica e investigación. Nuestro objetivo fue determinar frecuencias de enfermedad glomerular en una región de Colombia y representa la base para trabajos futuros. DISEÑO Y UBICACIÓN: Análisis retrospectivo en un único centro: Universidad de Antioquia, Colombia. MÉTODOS: Todas las biopsias de riñón nativo fueron revisadas (1998 - 2007), pero solo analizamos enfermedades glomerulares. El diagnóstico en cada caso estuvo basado en histología, inmunopatología y características clínicas. RESULTS: 1.040 biopsias fueron incluidas. En 302 casos (29,0 por ciento) la edad del paciente fue < 15 años. El diagnóstico fue enfermedad primaria en 828 biopsias (79,6 por ciento) y secundaria en 212 (20,4 por ciento). Las enfermedades primarias más frecuentes fueron glomeruloesclerosis focal y segmentaria (GEFyS) (34,8 por ciento), nefropatía IgA (NIgA) (11,8 por ciento), glomerulonefritis membranosa (GNM) (10,6 por ciento), enfermedad de cambios mínimos (ECM) (10,6 por ciento), glomerulonefritis (GN) extracapilar (5,6 por ciento) y GN proliferativa mesangial no-IgA (5,6 por ciento). La GN postinfecciosa representa el 10,7 por ciento de glomerulopatías primarias. La nefritis lúpica corresponde al 17,8 por ciento de todos los casos. En adultos el orden de frecuencia de enfermedades primarias es: GEFyS, NIgA, GNM, GN extracapilar y ECM. En niños (< 15 años) las más frecuentes fueron: GEFyS, GN postinfecciosa, ECM, GN mesangial no-IgA, GN endocapilar difusa y NIgA. CONCLUSIONES: Al igual que en afro-americanos, en nuestra población la GEFyS es la glomerulopatía más frecuente, pero en nuestro grupo hay más NIgA. Las razones para estos hallazgos no se conocen. La información presentada aquí es una contribución importante para el entendimiento de la prevalencia de enfermedades renales en Latinoamérica.


Assuntos
Adulto , Idoso , Feminino , Humanos , Lactente , Recém-Nascido , Masculino , Pessoa de Meia-Idade , Adulto Jovem , Glomerulosclerose Segmentar e Focal/epidemiologia , Rim/patologia , Distribuição por Idade , Biópsia , Colômbia/epidemiologia , Bases de Dados Factuais , Glomerulonefrite por IGA/epidemiologia , Glomerulosclerose Segmentar e Focal/patologia , Estudos Retrospectivos , Adulto Jovem
16.
Arch Biochem Biophys ; 461(1): 146-50, 2007 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-17291445

RESUMO

A rat brain cDNA encoding for a novel protein with agmatinase activity was cloned and functionally expressed. The protein was expressed as a histidine-tagged fusion product with a molecular weight of about 63 kDa. Agmatine hydrolysis was strictly dependent on Mn(2+); K(m) and k(cat) values were 2.5+/-0.2 mM and 0.8+/-0.2 s(-1), respectively. The product putrescine was a linear competitive inhibitor (K(i)=5+/-0.5 mM). The substrate specificity, metal ion requirement and pH optimum (9.5) coincide with those reported for Escherichia coli agmatinase, the best characterized of the agmatinases. However, as indicated by the k(cat)/K(m) (320 M(-1)s(-1)), the recombinant protein was about 290-fold less efficient than the bacterial enzyme. The deduced amino sequence revealed great differences with all known agmatinases, thus excluding the protein from the arginase family. It was, however, highly identical (>85%) to the predicted sequences for fragments of hypothetical or unnamed LIM domain-containing proteins. As a suggestion, the agmatinase activity is adscribed to a protein with an active site that promiscuously catalyze a reaction other than the one it evolved to catalyze.


Assuntos
Arginase/química , Arginase/genética , Encéfalo/enzimologia , Clonagem Molecular , DNA Complementar/genética , Família Multigênica , Ureo-Hidrolases/química , Ureo-Hidrolases/genética , Sequência de Aminoácidos , Animais , Arginase/fisiologia , Sítios de Ligação/genética , Catálise , DNA Complementar/biossíntese , DNA Complementar/fisiologia , Humanos , Dados de Sequência Molecular , Ratos , Ureo-Hidrolases/biossíntese
17.
FEBS J ; 273(24): 5625-31, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17212779

RESUMO

Upon mutation of Asn130 to aspartate, the catalytic activity of human arginase I was reduced to approximately 17% of wild-type activity, the Km value for arginine was increased approximately 9-fold, and the kcat/Km value was reduced approximately 50-fold. The kinetic properties were much less affected by replacement of Asn130 with glutamine. In contrast with the wild-type and N130Q enzymes, the N130D variant was active not only on arginine but also on its decarboxylated derivative, agmatine. Moreover, it exhibited no preferential substrate specificity for arginine over agmatine (kcat/Km values of 2.48 x 10(3) M(-1) x s(-1) and 2.14 x 10(3) M(-1) x s(-1), respectively). After dialysis against EDTA and assay in the absence of added Mn2+, the N130D mutant enzyme was inactive, whereas about 50% full activity was expressed by the wild-type and N130Q variants. Mutations were not accompanied by changes in the tryptophan fluorescence properties, thermal stability or chromatographic behavior of the enzyme. An active site conformational change is proposed as an explanation for the altered substrate specificity and low catalytic efficiency of the N130D variant.


Assuntos
Arginase/genética , Arginase/metabolismo , Mutagênese Sítio-Dirigida , Ureo-Hidrolases/genética , Ureo-Hidrolases/metabolismo , Substituição de Aminoácidos , Western Blotting , Catálise , Ativação Enzimática/genética , Estabilidade Enzimática , Fluorescência , Humanos , Cinética , Especificidade por Substrato/genética , Temperatura
18.
FEBS J ; 272(17): 4540-8, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16128822

RESUMO

To examine the interaction of human arginase II (EC 3.5.3.1) with substrate and manganese ions, the His120Asn, His145Asn and Asn149Asp mutations were introduced separately. About 53% and 95% of wild-type arginase activity were expressed by fully manganese activated species of the His120Asn and His145Asn variants, respectively. The K(m) for arginine (1.4-1.6 mM) was not altered and the wild-type and mutant enzymes were essentially inactive on agmatine. In contrast, the Asn149Asp mutant expressed almost undetectable activity on arginine, but significant activity on agmatine. The agmatinase activity of Asn149Asp (K(m) = 2.5 +/- 0.2 mM) was markedly resistant to inhibition by arginine. After dialysis against EDTA, the His120Asn variant was totally inactive in the absence of added Mn(2+) and contained < 0.1 Mn(2+).subunit(-1), whereas wild-type and His145Asn enzymes were half active and contained 1.1 +/- 0.1 Mn(2+).subunit(-1) and 1.3 +/- 0.1 Mn(2+).subunit(-1), respectively. Manganese reactivation of metal-free to half active species followed hyperbolic kinetics with K(d) of 1.8 +/- 0.2 x 10(-8) M for the wild-type and His145Asn enzymes and 16.2 +/- 0.5 x 10(-8) m for the His120Asn variant. Upon mutation, the chromatographic behavior, tryptophan fluorescence properties (lambda(max) = 338-339 nm) and sensitivity to thermal inactivation were not altered. The Asn149-->Asp mutation is proposed to generate a conformational change responsible for the altered substrate specificity of arginase II. We also conclude that, in contrast with arginase I, Mn(2+) (A) is the more tightly bound metal ion in arginase II.


Assuntos
Arginase/genética , Arginase/metabolismo , Substituição de Aminoácidos , Sequência de Bases , DNA/genética , Humanos , Técnicas In Vitro , Cinética , Manganês/metabolismo , Mutagênese Sítio-Dirigida , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Especificidade por Substrato
19.
Arch Biochem Biophys ; 430(2): 185-90, 2004 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-15369817

RESUMO

The interaction of Escherichia coli agmatinase (EC 3.5.3.11) with the substrate guanidinium group was investigated by kinetic and site-directed mutagenesis studies. Putrescine and guanidinium ions (Gdn+) were slope-linear, competitive inhibitors with respect to agmatine and their bindings to the enzyme were not mutually exclusive. By site-directed mutagenesis, the E274A variant exhibiting about 1-2% of wild-type activity was obtained. Mutation produced a moderate, but significant, increase in the Km value for agmatine (from 1.1 +/- 0.2 mM to 6.3 +/- 0.3 mM) and the Ki value for competitive inhibition by Gdn+ (from 15.0 +/- 0.1 mM to 44.2 +/- 2.1 mM), but the Ki value for putrescine inhibition (2.8 +/- 0.2 mM) was not altered. The tryptophan fluorescence properties (lambdamax = 342 nm) and circular dichroism spectra were not significantly altered by the Glu274 --> Ala mutation. The dimeric structure of the enzyme was also maintained. We conclude that Glu274 is involved in binding and positioning of the guanidinium moiety of the substrate for efficient catalysis. A kinetic mechanism involving rapid equilibrium random release of products is proposed for E. coli agmatinase.


Assuntos
Escherichia coli/enzimologia , Glutamina/metabolismo , Guanidina/metabolismo , Sítios de Ligação , Catálise , Escherichia coli/genética , Variação Genética , Glutamina/química , Glutamina/genética , Cinética , Modelos Moleculares , Mutagênese Sítio-Dirigida , Mutação , Relação Estrutura-Atividade , Especificidade por Substrato , Ureo-Hidrolases
20.
J Inorg Biochem ; 98(8): 1465-9, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15271525

RESUMO

Recombinant wild-type human liver arginase (EC 3.5.3.1) expressed in Escherichia coli was markedly resistant to inhibition by ethylene diamine tetraacetic acid (EDTA). In contrast, half and fully activated species of the H101N variant were totally inactive in the presence of approximately 1 mM EDTA. Dilution of inhibited species in metal-free buffer lead to a time dependent recovery of activity, even when measured in the absence of added Mn2+. The inhibition was mixed type, with predominance of a competitive component (Kii=0.31 mM; Kis=0.022 mM). The structurally related N,N,N',N'-tetramethylethylenediamine was not inhibitory, indicating the importance of the carboxyl groups in EDTA inhibition. We conclude that EDTA inhibition of H101N arginase is not due to interaction with a weakly bound Mn2+ or chelation of essential metal ions.


Assuntos
Arginase/antagonistas & inibidores , Ácido Edético/farmacologia , Isoenzimas/antagonistas & inibidores , Fígado/enzimologia , Arginase/genética , Arginase/metabolismo , Quelantes/metabolismo , Quelantes/farmacologia , Ácido Edético/metabolismo , Humanos , Íons/metabolismo , Isoenzimas/genética , Isoenzimas/metabolismo , Manganês/metabolismo , Metais/metabolismo , Proteínas Recombinantes/antagonistas & inibidores , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA