Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Protein Sci ; 29(3): 789-802, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31930600

RESUMO

Acinetobacter baumannii is well known for causing hospital-associated infections due in part to its intrinsic antibiotic resistance as well as its ability to remain viable on surfaces and resist cleaning agents. In a previous publication, A. baumannii strain AB5075 was studied by transposon mutagenesis and 438 essential gene candidates for growth on rich-medium were identified. The Seattle Structural Genomics Center for Infectious Disease entered 342 of these candidate essential genes into our pipeline for structure determination, in which 306 were successfully cloned into expression vectors, 192 were detectably expressed, 165 screened as soluble, 121 were purified, 52 crystalized, 30 provided diffraction data, and 29 structures were deposited in the Protein Data Bank. Here, we report these structures, compare them with human orthologs where applicable, and discuss their potential as drug targets for antibiotic development against A. baumannii.


Assuntos
Acinetobacter baumannii/química , Acinetobacter baumannii/efeitos dos fármacos , Antibacterianos/farmacologia , Proteínas de Bactérias/antagonistas & inibidores , Genoma Bacteriano/efeitos dos fármacos , Genoma Bacteriano/genética , Acinetobacter baumannii/genética , Proteínas de Bactérias/genética , Coproporfirinogênio Oxidase/química , Coproporfirinogênio Oxidase/metabolismo , Farmacorresistência Bacteriana/efeitos dos fármacos , Humanos , Metionina tRNA Ligase/química , Metionina tRNA Ligase/metabolismo , Modelos Moleculares , Conformação Proteica , Uroporfirinogênio Descarboxilase/química , Uroporfirinogênio Descarboxilase/metabolismo
2.
Adv Protein Chem Struct Biol ; 100: 153-85, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26415844

RESUMO

In this chapter, we discuss the influence of an anisotropic protein environment on the reaction mechanisms of saccharopine reductase and uroporphyrinogen decarboxylase, respectively, via the use of a quantum mechanical and molecular mechanical (QM/MM) approach. In addition, we discuss the importance of selecting a suitable DFT functional to be used in a QM/MM study of a key intermediate in the mechanism of 8R-lipoxygenase, a nonheme iron enzyme. In the case of saccharopine reductase, while the enzyme utilizes a substrate-assisted catalytic pathway, it was found that only through treating the polarizing effect of the active site, via the use of an electronic embedding formalism, was agreement with experimental kinetic data obtained. Similarly, in the case of uroporphyrinogen decarboxylase, the effect of the protein environment on the catalytic mechanism was found to be such that the calculated rate-limiting barrier is in good agreement with related experimentally determined values for the first decarboxylation of the substrate. For 8R-lipoxygenase, it was found that the geometries and energies of the multicentered open-shell intermediate complexes formed during the mechanism are quite sensitive to the choice of the density functional theory method. Thus, while density functional theory has become the method of choice in QM/MM studies, care must be taken in the selection of a particular high-level method.


Assuntos
Araquidonato Lipoxigenases/química , Simulação de Dinâmica Molecular , Sacaropina Desidrogenases/química , Uroporfirinogênio Descarboxilase/química , Animais , Anisotropia , Antozoários/química , Antozoários/enzimologia , Domínio Catalítico , Humanos , Cinética , Teoria Quântica , Especificidade por Substrato , Termodinâmica
3.
Folia Biol (Praha) ; 61(6): 219-26, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26789143

RESUMO

Porphyrias are metabolic disorders resulting from mutations in haem biosynthetic pathway genes. Hepatoerythropoietic porphyria (HEP) is a rare type of porphyria caused by the deficiency of the fifth enzyme (uroporphyrinogen decarboxylase, UROD) in this pathway. The defect in the enzymatic activity is due to biallelic mutations in the UROD gene. Currently, 109 UROD mutations are known. The human disease has an early onset, manifesting in infancy or early childhood with red urine, skin photosensitivity in sun-exposed areas, and hypertrichosis. Similar defects and links to photosensitivity and hepatopathy exist in several animal models, including zebrafish and mice. In the present study, we report a new mutation in the UROD gene in Egyptian patients with HEP. We show that the homozygous c.T163A missense mutation leads to a substitution of a conserved phenylalanine (amino acid 55) for isoleucine in the enzyme active site, causing a dramatic decrease in the enzyme activity (19 % of activity of wild-type enzyme). Inspection of the UROD crystal structure shows that Phe-55 contacts the substrate and is located in the loop that connects helices 2 and 3. Phe-55 is strictly conserved in both prokaryotic and eukaryotic UROD. The F55I substitution likely interferes with the enzyme-substrate interaction.


Assuntos
Alelos , Predisposição Genética para Doença , Mutação/genética , Porfiria Hepatoeritropoética/enzimologia , Porfiria Hepatoeritropoética/genética , Uroporfirinogênio Descarboxilase/genética , Adolescente , Sequência de Aminoácidos , Sequência de Bases , Criança , Cicatriz/complicações , Análise Mutacional de DNA , Egito , Família , Feminino , Humanos , Hipertricose/complicações , Masculino , Modelos Moleculares , Dados de Sequência Molecular , Taxa de Mutação , Linhagem , Porfiria Hepatoeritropoética/complicações , Proteínas Recombinantes de Fusão/metabolismo , Alinhamento de Sequência , Uroporfirinogênio Descarboxilase/química
4.
Cell Biochem Biophys ; 70(2): 735-46, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24777812

RESUMO

Uroporphyrinogen decarboxylase is a cytosolic enzyme involved in the biosynthetic pathway of heme production. Decreased activity of this enzyme results in porphyria cutanea tarda and hepato erythropoietic porphyria. Nonsynonymous single nucleotide polymorphisms (nsSNPs) alter protein sequence and can cause disease. Identifying the deleterious nsSNPs that contribute to disease is an important task. We used five different in silico tools namely SIFT, PANTHER, PolyPhen2, SNPs&GO, and I-mutant3 to identify deleterious nsSNPs in UROD gene. Further, we used molecular dynamic (MD) approach to evaluate the impact of deleterious mutations on UROD protein structure. By comparing the results of all the five prediction results, we screened 35 (51.47 %) nsSNPs as highly deleterious. MD analysis results show that all the three L161Q, L282R, and I334T deleterious variants were affecting the UROD protein structural stability and flexibility. Our findings provide strong evidence on the effect of deleterious nsSNPs in UROD gene. A detailed MD study provides a new insight in the conformational changes occurred in the mutant structures of UROD protein.


Assuntos
Doença/genética , Simulação de Dinâmica Molecular , Polimorfismo de Nucleotídeo Único , Uroporfirinogênio Descarboxilase/química , Uroporfirinogênio Descarboxilase/genética , Estabilidade Enzimática , Humanos , Ligação de Hidrogênio , Mutação , Estrutura Secundária de Proteína , Eletricidade Estática
5.
PLoS One ; 9(2): e89889, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24587102

RESUMO

Uroporphyrinogen decarboxylase (UROD) catalyzes the conversion of uroporphyrinogen to coproporphyrinogen during heme biosynthesis. This enzyme was recently identified as a potential anticancer target; its inhibition leads to an increase in reactive oxygen species, likely mediated by the Fenton reaction, thereby decreasing cancer cell viability and working in cooperation with radiation and/or cisplatin. Because there is no known chemical UROD inhibitor suitable for use in translational studies, we aimed to design, synthesize, and characterize such a compound. Initial in silico-based design and docking analyses identified a potential porphyrin analogue that was subsequently synthesized. This species, a porphodimethene (named PI-16), was found to inhibit UROD in an enzymatic assay (IC50 = 9.9 µM), but did not affect porphobilinogen deaminase (at 62.5 µM), thereby exhibiting specificity. In cellular assays, PI-16 reduced the viability of FaDu and ME-180 cancer cells with half maximal effective concentrations of 22.7 µM and 26.9 µM, respectively, and only minimally affected normal oral epithelial (NOE) cells. PI-16 also combined effectively with radiation and cisplatin, with potent synergy being observed in the case of cisplatin in FaDu cells (Chou-Talalay combination index <1). This work presents the first known synthetic UROD inhibitor, and sets the foundation for the design, synthesis, and characterization of higher affinity and more effective UROD inhibitors.


Assuntos
Modelos Moleculares , Porfirinas/síntese química , Proteínas Recombinantes/metabolismo , Uroporfirinogênio Descarboxilase/antagonistas & inibidores , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Humanos , Concentração Inibidora 50 , Estrutura Molecular , Porfirinas/química , Porfirinas/farmacologia , Proteínas Recombinantes/química , Especificidade por Substrato , Uroporfirinogênio Descarboxilase/química
6.
J Biomol Struct Dyn ; 31(12): 1358-69, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23140436

RESUMO

Overexpression of epidermal growth factor receptor (EGFR), Her2, and uroporphyrinogen decarboxylase (UROD) occurs in a variety of malignant tumor tissues. UROD has potential to modulate tumor response of radiotherapy for head and neck cancer, and EGFR and Her2 are common drug targets for the treatment of head and neck cancer. This study attempts to find a possible lead compound backbone from TCM Database@Taiwan ( http://tcm.cmu.edu.tw/ ) for EGFR, Her2, and UROD proteins against head and neck cancer using computational techniques. Possible traditional Chinese medicine (TCM) lead compounds had potential binding affinities with EGFR, Her2, and UROD proteins. The candidates formed stable interactions with residues Arg803, Thr854 in EGFR, residues Thr862, Asp863 in Her2 protein, and residues Arg37, Arg41 in UROD protein, which are key residues in the binding or catalytic domain of EGFR, Her2, and UROD proteins. Thus, the TCM candidates indicated a possible molecule backbone for evolving potential inhibitors for three drug target proteins against head and neck cancer.


Assuntos
Inibidores Enzimáticos/uso terapêutico , Receptores ErbB/antagonistas & inibidores , Neoplasias de Cabeça e Pescoço/prevenção & controle , Receptor ErbB-2/antagonistas & inibidores , Uroporfirinogênio Descarboxilase/antagonistas & inibidores , Antineoplásicos/química , Antineoplásicos/metabolismo , Antineoplásicos/uso terapêutico , Simulação por Computador , Bases de Dados Factuais , Inibidores Enzimáticos/química , Inibidores Enzimáticos/metabolismo , Receptores ErbB/química , Receptores ErbB/metabolismo , Neoplasias de Cabeça e Pescoço/metabolismo , Humanos , Medicina Tradicional Chinesa/métodos , Modelos Moleculares , Simulação de Dinâmica Molecular , Estrutura Molecular , Ligação Proteica , Estrutura Terciária de Proteína , Receptor ErbB-2/química , Receptor ErbB-2/metabolismo , Taiwan , Uroporfirinogênio Descarboxilase/química , Uroporfirinogênio Descarboxilase/metabolismo
7.
PLoS One ; 7(11): e50087, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23209648

RESUMO

Uroporphyrinogen decarboxylase (UROD) has been suggested as a protectant against radiation for head and neck cancer (HNC). In this study, we employed traditional Chinese medicine (TCM) compounds from TCM Database@Taiwan (http://tcm.cmu.edu.tw/) to screen for drug-like candidates with potential UROD inhibition characteristics using virtual screening techniques. Isopraeroside IV, scopolin, and nodakenin exhibited the highest Dock Scores, and were predicted to have good Absorption, Distribution, Metabolism, Excretion, and Toxicity (ADMET) properties. Two common moieties, 2H-chromen-2-one and glucoside, were observed among the top TCM candidates. Cross comparison of the docking poses indicated that candidates formed stable interactions with key binding and catalytic residues of UROD through these two moieties. The 2H-chromen-2-one moiety enabled pi-cation interactions with Arg37 and H-bonds with Tyr164. The glucoside moiety was involved in forming H-bonds with Arg37 and Asp86. From our computational results, we propose isopraeroside IV, scopolin, and nodakenin as ligands that might exhibit drug-like inhibitory effects on UROD. The glucoside and 2H-chromen-2-one moieties may potentially be used for designing inhibitors of UROD.


Assuntos
Medicina Tradicional Chinesa/métodos , Uroporfirinogênio Descarboxilase/metabolismo , Algoritmos , Sítios de Ligação , Simulação por Computador , Cristalografia por Raios X/métodos , Desenho de Fármacos , Glucosídeos/metabolismo , Humanos , Ligação de Hidrogênio , Concentração de Íons de Hidrogênio , Ligantes , Modelos Químicos , Simulação de Dinâmica Molecular , Ligação Proteica , Software , Tirosina/genética , Uroporfirinogênio Descarboxilase/química
8.
J Comput Chem ; 32(5): 822-34, 2011 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-20941734

RESUMO

In humans, uroporphyrinogen decarboxylase is intimately involved in the synthesis of heme, where the decarboxylation of the uroporphyrinogen-III occurs in a single catalytic site. Several variants of the mechanistic proposal exist; however, the exact mechanism is still debated. Thus, using an ONIOM quantum mechanical/molecular mechanical approach, the mechanism by which uroporphyrinogen decarboxylase decarboxylates ring D of uroporphyrinogen-III has been investigated. From the study performed, it was found that both Arg37 and Arg50 are essential in the decarboxylation of ring D, where experimentally both have been shown to be critical to the catalytic behavior of the enzyme. Overall, the reaction was found to have a barrier of 10.3 kcal mol(-1) at 298.15 K. The rate-limiting step was found to be the initial proton transfer from Arg37 to the substrate before the decarboxylation. In addition, it has been found that several key interactions exist between the substrate carboxylate groups and backbone amides of various active site residues as well as several other functional groups.


Assuntos
Simulação de Dinâmica Molecular , Porfirinas/biossíntese , Teoria Quântica , Uroporfirinogênio Descarboxilase/química , Sítios de Ligação , Biocatálise , Biologia Computacional , Descarboxilação , Humanos , Modelos Moleculares , Conformação Molecular , Uroporfirinogênio Descarboxilase/metabolismo
9.
J Phys Chem B ; 114(27): 8994-9001, 2010 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-20553007

RESUMO

Uroporphyrinogen III decarboxylase catalyzes the fifth step in heme biosynthesis, the elimination of carboxyl groups from the four acetate side chains of uroporphyrinogen III to yield coproporphyrinogen III. We have previously found that the rate-limiting step of uroporphyrinogen III decarboxylase is substrate protonation rather than the decarboxylation reaction. This protonation can be effected by an arginine residue (Arg37) in close proximity to the substrate. In this report, we present evidence for the function of this arginine residue as a general acid catalyst. Although substrate protonation by H(3)O(+) is both exergonic and very fast, our density functional calculations show that in the presence of a protonated Arg37 substrate, decarboxylation becomes rate-limiting, and the substrate spontaneously breaks upon protonation. These results suggest that the active site must be shielded from solvent protons. Consequently, H(3)O(+) can be excluded from a role in both protonations proposed for the enzyme mechanism. In agreement with these conclusions, a second arginine residue (Arg41) is uniquely positioned to act as donor of the second proton, with an activation barrier below 2 kcal mol(-1). Generated mutant uroporphyrinogen III decarboxylase variants carrying amino acid exchanges in the position of both arginine residues (R41A, R41K, R37A, and R37K) failed to produce coproporphyrinogen III. The proposed unusual use of two basic residues as general acids in two different proton donation steps by uroporphyrinogen III decarboxylase provides an elegant solution to the problem of simultaneously binding the very negative uroporphyrinogen (which requires a positively charged active site), and selectively protonating it while preventing excessive carboxylate stabilization by positive charges.


Assuntos
Arginina , Oniocompostos , Uroporfirinogênio Descarboxilase/química , Uroporfirinogênio Descarboxilase/metabolismo , Domínio Catalítico , Humanos , Simulação de Dinâmica Molecular , Mutagênese Sítio-Dirigida , Prótons , Uroporfirinogênio Descarboxilase/genética , Uroporfirinogênios/metabolismo
10.
Cell Mol Biol (Noisy-le-grand) ; 55(2): 40-5, 2009 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-19656450

RESUMO

Porphyria cutanea tarda (PCT) is caused by inhibition of uroporphyrinogen decarboxylase (URO-D) activity in hepatocytes. Subnormal URO-D activity results in accumulation and urinary excretion of uroporphyrin and heptacarboxyl porphyrin. Heterozygosity for mutations in the URO-D gene is found in the familial form of PCT (F-PCT). Over 70 mutations of URO-D have been described but very few have been characterized structurally. Here we characterize 3 mutations in the URO-D gene found in patients with F-PCT, G318R, K297N, and D306Y. Expression of the D306Y mutation results in an insoluble recombinant protein. G318R and K297N have little effect on the structure or activity of recombinant URO-D, but the proteins display reduced stability in vitro.


Assuntos
Uroporfirinogênio Descarboxilase/metabolismo , Adulto , Idoso , Domínio Catalítico , Cristalografia por Raios X , Feminino , Genótipo , Heterozigoto , Humanos , Cinética , Masculino , Pessoa de Meia-Idade , Polimorfismo de Nucleotídeo Único , Porfiria Cutânea Tardia/etiologia , Porfiria Cutânea Tardia/genética , Estabilidade Proteica , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Uroporfirinogênio Descarboxilase/química , Uroporfirinogênio Descarboxilase/genética
11.
J Mol Biol ; 389(2): 306-14, 2009 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-19362562

RESUMO

Uroporphyrinogen decarboxylase (URO-D; EC 4.1.1.37), the fifth enzyme of the heme biosynthetic pathway, is required for the production of heme, vitamin B12, siroheme, and chlorophyll precursors. URO-D catalyzes the sequential decarboxylation of four acetate side chains in the pyrrole groups of uroporphyrinogen to produce coproporphyrinogen. URO-D is a stable homodimer, with the active-site clefts of the two subunits adjacent to each other. It has been hypothesized that the two catalytic centers interact functionally, perhaps by shuttling of reaction intermediates between subunits. We tested this hypothesis by construction of a single-chain protein (single-chain URO-D) in which the two subunits were connected by a flexible linker. The crystal structure of this protein was shown to be superimposable with wild-type activity and to have comparable catalytic activity. Mutations that impaired one or the other of the two active sites of single-chain URO-D resulted in approximately half of wild-type activity. The distributions of reaction intermediates were the same for mutant and wild-type sequences and were unaltered in a competition experiment using I and III isomer substrates. These observations indicate that communication between active sites is not required for enzyme function and suggest that the dimeric structure of URO-D is required to achieve conformational stability and to create a large active-site cleft.


Assuntos
Coproporfirinogênios/biossíntese , Uroporfirinogênio Descarboxilase/metabolismo , Domínio Catalítico , Cristalografia por Raios X , Conformação Proteica , Multimerização Proteica , Subunidades Proteicas , Uroporfirinogênio Descarboxilase/química , Uroporfirinogênios/metabolismo
12.
Transl Res ; 149(2): 85-91, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17240319

RESUMO

Hepatoerythropoietic porphyria (HEP) is a rare form of porphyria in humans. The disorder is caused by homozygosity or compound heterozygosity for mutations of the uroporphyrinogen decarboxylase (URO-D) gene. Subnormal URO-D activity results in accumulation of uroporphyrin in the liver, which ultimately mediates the photosensitivity that clinically characterizes HEP. Two previously undescribed URO-D mutations found in a 2-year-old Caucasian boy with HEP, a maternal nonsense mutation (Gln71Stop), and a paternal missense mutation (Gly168Arg) are reported here. Recombinant Gly168Arg URO-D retained 65% of wild-type URO-D activity and studies in Epstein-Barr Virus (EBV)-transformed lymphoblasts indicated that protein levels are reduced, suggesting that the mutant protein might be subjected to accelerated turnover. The crystal structure of Gly168Arg was determined both as the apo-enzyme and with the reaction product bound. These studies revealed little distortion of the active site, but a loop containing residues 167-172 was displaced, possibly indicating small changes in the catalytic geometry or in substrate binding or increased accessibility to a cellular proteolytic pathway. A second pregnancy occurred in this family, and in utero genotyping revealed a fetus heterozygous for the maternal nonsense mutation (URO-D genotype WT/Gln71Stop). A healthy infant was born with no clinical evidence of porphyria.


Assuntos
Códon sem Sentido , Mutação de Sentido Incorreto , Porfiria Hepatoeritropoética/genética , Uroporfirinogênio Descarboxilase/genética , Adulto , Linhagem Celular Transformada , Pré-Escolar , Coproporfirinogênios/biossíntese , Coproporfirinogênios/química , Cristalografia , Ativação Enzimática/genética , Feminino , Genótipo , Heterozigoto , Homozigoto , Humanos , Masculino , Fenótipo , Porfiria Hepatoeritropoética/metabolismo , Gravidez , Diagnóstico Pré-Natal , Estrutura Terciária de Proteína , Uroporfirinogênio Descarboxilase/química , Uroporfirinogênio Descarboxilase/metabolismo , Uroporfirinogênios/química , Uroporfirinogênios/metabolismo
13.
J Bacteriol ; 189(9): 3573-80, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17122346

RESUMO

Uroporphyrinogen decarboxylase (UROD) is a branch point enzyme in the biosynthesis of the tetrapyrroles. It catalyzes the decarboxylation of four acetate groups of uroporphyrinogen III to yield coproporphyrinogen III, leading to heme and chlorophyll biosynthesis. UROD is a special type of nonoxidative decarboxylase, since no cofactor is essential for catalysis. In this work, the first crystal structure of a bacterial UROD, Bacillus subtilis UROD (UROD(Bs)), has been determined at a 2.3 A resolution. The biological unit of UROD(Bs) was determined by dynamic light scattering measurements to be a homodimer in solution. There are four molecules in the crystallographic asymmetric unit, corresponding to two homodimers. Structural comparison of UROD(Bs) with eukaryotic URODs reveals a variation of two loops, which possibly affect the binding of substrates and release of products. Structural comparison with the human UROD-coproporphyrinogen III complex discloses a similar active cleft, with five invariant polar residues (Arg29, Arg33, Asp78, Tyr154, and His322) and three invariant hydrophobic residues (Ile79, Phe144, and Phe207), in UROD(Bs). Among them, Asp78 may interact with the pyrrole NH groups of the substrate, and Arg29 is a candidate for positioning the acetate groups of the substrate. Both residues may also play catalytic roles.


Assuntos
Bacillus subtilis/enzimologia , Uroporfirinogênio Descarboxilase/química , Sequência de Aminoácidos , Domínio Catalítico , Sequência Conservada , Cristalografia por Raios X , Dimerização , Modelos Moleculares , Dados de Sequência Molecular , Estrutura Quaternária de Proteína , Alinhamento de Sequência
14.
J Proteome Res ; 5(7): 1751-62, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16823983

RESUMO

The Harderian gland is an orbital gland located behind the ocular bulb in most terrestrial vertebrates probably functioning for production of lipid secretion to protect the eye. We herein present a protein reference database of the rat Harderian gland that may serve as analytical tool for future proteomic work, report lipid and porphyrin handling cascades, address sequence conflicts and report structures that have not been so far described by proteomics methods.


Assuntos
Glândula de Harder/metabolismo , Proteoma/análise , Ratos/anatomia & histologia , Sequência de Aminoácidos , Animais , Bases de Dados de Proteínas , Imuno-Histoquímica , Metabolismo dos Lipídeos , Lipoproteínas/química , Masculino , Dados de Sequência Molecular , Mapeamento de Peptídeos , Porfirinas/química , Porfirinas/metabolismo , Proteômica/métodos , Ratos/metabolismo , Ratos Sprague-Dawley , Homologia de Sequência de Aminoácidos , Uroporfirinogênio Descarboxilase/química
15.
Scand J Clin Lab Invest ; 65(3): 227-35, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16095052

RESUMO

The functional consequence of six uroporphyrinogen decarboxylase (UROD) gene mutations found in Danish patients with familial porphyria cutanea tarda was investigated. Wild-type UROD and the 6 mutants (3 missense, 1 nonsense and 2 frameshift mutants) were cloned and expressed using the prokaryotic gGEX-6P system, in which the protein is produced in fusion with glutathione S-transferase (GST). Enzymatic activity of the purified recombinant mutant fusion proteins ranged from undetectable to less than 12% of the recombinant wild-type protein. Mutant proteins cleaved from the GST part did not retain any catalytic activity. These observations can be ascribed to the structure/function relationships of the enzyme, and the fact that the enzyme is a dimer in its active form. Although the clinical manifestation of familial porphyria cutanea tarda is complex, the findings support the notion that different mutations may affect individuals differently.


Assuntos
Mutação/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Uroporfirinogênio Descarboxilase/genética , Uroporfirinogênio Descarboxilase/metabolismo , Expressão Gênica , Humanos , Cinética , Proteínas Recombinantes/química , Proteínas Recombinantes/isolamento & purificação , Uroporfirinogênio Descarboxilase/química , Uroporfirinogênio Descarboxilase/isolamento & purificação
16.
J Phys Chem B ; 109(38): 18195-200, 2005 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-16853337

RESUMO

Uroporphyrinogen III decarboxylase catalyzes the fifth step in heme biosynthesis: the elimination of carboxyl groups from the four acetate side chains of uroporphyrinogen III to yield coproporphyrinogen III. The enzyme acts by successively protonating each of the four pyrrole rings present in the substrate, thereby allowing decarboxylation of their side chains, but the identity of the proton donors has not been established yet. Tyr164 has been suggested as a proton donor, and Asp86 has been proposed to act either as a proton donor or as an intermediate-stabilizing residue. We have performed density-functional calculations to study this reaction mechanism, and found that the rate-limiting step is substrate protonation, rather than decarboxylation. Surprisingly, whereas Tyr164 is unable to protonate the substrate, this protonation can be effected by a nearby arginine residue (Arg37), with a free energy barrier of 21.4 kcal.mol(-1), in remarkable agreement with the experimental value of 19.5 kcal.mol(-1). The central positioning of this residue in close proximity to all four pyrrole rings in the substrate may play a key role in the sequential activation of each of these moieties.


Assuntos
Uroporfirinogênio Descarboxilase/química , Uroporfirinogênio Descarboxilase/metabolismo , Coproporfirinogênios/metabolismo , Cinética , Modelos Moleculares , Conformação Molecular , Uroporfirinogênios/metabolismo
17.
Bioorg Med Chem Lett ; 14(22): 5559-64, 2004 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-15482924

RESUMO

Of the heme biosynthetic pathway enzymes, coproporphyrinogen oxidase is one of the least understood. Substrate recognition studies [Prepr. Biochem. Biotech.1997, 27, 47, J. Org. Chem.1999, 64, 464] have been done using chicken blood hemolysates (CBH) as the source of this enzyme. However, the enzyme uroporphyrinogen decarboxylase is also present in these preparations and separation of these two enzymes from CBH had not yet been achieved. Thus, a substrate ligand column was developed by covalently linking coproporphyrin-III to a sepharose resin following a similar procedure previously used for the purification of uroporphyrinogen decarboxylase [Int. J. Biochem.1992, 24, 105]. The ligand-resin chromatography step rapidly separates coproporphyrinogen oxidase from uroporphyrinogen decarboxylase as well as the majority of the hemoglobin.


Assuntos
Coproporfirinogênio Oxidase/química , Eritrócitos/enzimologia , Heme/biossíntese , Uroporfirinogênio Descarboxilase/química , Animais , Galinhas , Cromatografia Líquida de Alta Pressão/métodos , Coproporfirinogênio Oxidase/isolamento & purificação , Ativação Enzimática , Estrutura Molecular , Relação Estrutura-Atividade , Uroporfirinogênio Descarboxilase/isolamento & purificação
18.
EMBO J ; 22(23): 6225-33, 2003 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-14633982

RESUMO

Uroporphyrinogen decarboxylase (URO-D), an essential enzyme that functions in the heme biosynthetic pathway, catalyzes decarboxylation of all four acetate groups of uroporphyrinogen to form coproporphyrinogen. Here we report crystal structures of URO-D in complex with the I and III isomer coproporphyrinogen products. Crystallization required use of a novel enzymatic approach to generate the highly oxygen-sensitive porphyrinogen substrate in situ. The tetrapyrrole product adopts a domed conformation that lies against a collar of conserved hydrophobic residues and allows formation of hydrogen bonding interactions between a carboxylate oxygen atom of the invariant Asp86 residue and the pyrrole NH groups. Structural and biochemical analyses of URO-D proteins mutated at Asp86 support the conclusion that this residue makes important contributions to binding and likely promotes catalysis by stabilizing a positive charge on a reaction intermediate. The central coordination geometry of Asp86 allows the initial substrates and the various partially decarboxylated intermediates to be bound with equivalent activating interactions, and thereby explains how all four of the substrate acetate groups can be decarboxylated at the same catalytic center.


Assuntos
Tetrapirróis/química , Uroporfirinogênio Descarboxilase/química , Sequência de Aminoácidos , Animais , Ácido Aspártico , Sítios de Ligação , Cristalografia por Raios X , Ligação de Hidrogênio , Processamento de Imagem Assistida por Computador , Dados de Sequência Molecular , Conformação Proteica , Estrutura Secundária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo
19.
J Biochem Biophys Methods ; 55(3): 241-9, 2003 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-12706908

RESUMO

Uroporphyrinogen decarboxylase (UROD) and coproporphyrinogen oxidase (copro'gen oxidase) are two of the least well understood enzymes in the heme biosynthetic pathway. In the fifth step of the pathway, UROD converts uroporphyrinogen III to coproporphyrinogen III by the decarboxylation of the four acetic acid side chains. Copro'gen oxidase then converts coproporphyrinogen III to protoporphyrinogen IX via two sequential oxidative decarboxylations. Studies of these two enzymes are important to increase our understanding of their mechanisms. Assay comparisons of UROD and copro'gen oxidase from chicken blood hemolysates (CBH), using a newly developed micro-assay, showed that the specific activity of both enzymes is increased in the micro-assay relative to the large-scale assay. The micro-assay has distinct advantages in terms of cost, labor intensity, amount of enzyme required, and sensitivity.


Assuntos
Cromatografia Líquida de Alta Pressão/métodos , Coproporfirinogênio Oxidase/análise , Coproporfirinogênio Oxidase/química , Uroporfirinogênio Descarboxilase/análise , Uroporfirinogênio Descarboxilase/química , Animais , Galinhas , Coproporfirinogênio Oxidase/sangue , Ativação Enzimática , Eritrócitos/enzimologia , Uroporfirinogênio Descarboxilase/sangue
20.
Artigo em Inglês | MEDLINE | ID: mdl-12381387

RESUMO

We characterized Uroporphyrinogen decarboxylase (UroD) (E.C. 4.1.1.37) in hepatopancreas of the crab Chasmagnathus granulatus as a first step to establish this enzyme as a possible biomarker for environmental contamination. We performed a comparative study of crab UroD with the enzyme UroD present in Wistar rat liver, which is known as a useful indicator of intoxication by polyhalogenated aromatic hydrocarbons (PAHs). The final products were the same in crab and rat UroD: the remaining substrate (8-carboxyl-porphyrinogen), the final product Coproporphyrinogen (4-COOH) and intermediate compounds with 7-, 6- and 5-COOH. The elimination of the second carboxyl group seems to be the rate-limiting step in this multiple decarboxylation, because large amounts of 7-COOH porphyrinogen are accumulated. The V(max)/K(m) ratio was 100-fold higher for rat liver UroD than for crab hepatopancreas UroD, suggesting a higher efficiency of the rat enzyme. Optimum pH for enzyme activity was 7.2 and 6.8 for crab and rat, respectively. Although both systems showed the same optimum temperature (47 degrees C), the activation energy was clearly different, 51.5 kJ/mol for C. granulatus and 5.4 kJ/mol for Rattus norvegicus (Wistar strain). Superdex 75 gel chromatography yielded a single symmetrical peak with an apparent molecular mass of 48+/-3 kDa for crab hepatopancreas UroD, suggesting the existence of only one enzymatic species in C. granulatus.


Assuntos
Decápodes/enzimologia , Sistema Digestório/enzimologia , Uroporfirinogênio Descarboxilase/metabolismo , Animais , Descarboxilação , Poluentes Ambientais/metabolismo , Hidrocarbonetos Halogenados/metabolismo , Concentração de Íons de Hidrogênio , Cinética , Fígado/enzimologia , Porfirinogênios/metabolismo , Ratos , Ratos Wistar , Temperatura , Uroporfirinogênio Descarboxilase/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA