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












Base de datos
Intervalo de año de publicación
1.
Biochim Biophys Acta Gen Subj ; 1862(9): 1948-1955, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29908816

RESUMEN

Human porphobilinogen deaminase (PBGD), the third enzyme in the heme pathway, catalyzes four times a single reaction to convert porphobilinogen into hydroxymethylbilane. Remarkably, PBGD employs a single active site during the process, with a distinct yet chemically equivalent bond formed each time. The four intermediate complexes of the enzyme have been biochemically validated and they can be isolated but they have never been structurally characterized other than the apo- and holo-enzyme bound to the cofactor. We present crystal structures for two human PBGD intermediates: PBGD loaded with the cofactor and with the reaction intermediate containing two additional substrate pyrrole rings. These results, combined with SAXS and NMR experiments, allow us to propose a mechanism for the reaction progression that requires less structural rearrangements than previously suggested: the enzyme slides a flexible loop over the growing-product active site cavity. The structures and the mechanism proposed for this essential reaction explain how a set of missense mutations result in acute intermittent porphyria.


Asunto(s)
Hidroximetilbilano Sintasa/química , Hidroximetilbilano Sintasa/metabolismo , Pirroles/química , Pirroles/metabolismo , Catálisis , Dominio Catalítico , Cristalografía por Rayos X , Humanos , Polimerizacion , Porfobilinógeno/química , Porfobilinógeno/metabolismo , Conformación Proteica , Uroporfirinógenos/química , Uroporfirinógenos/metabolismo
2.
Structure ; 26(4): 565-571.e3, 2018 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-29551288

RESUMEN

There are numerous applications that use the structures of protein-ligand complexes from the PDB, such as 3D pharmacophore identification, virtual screening, and fragment-based drug design. The structures underlying these applications are potentially much more informative if they contain biologically relevant bound ligands, with high similarity to the cognate ligands. We present a study of ligand-enzyme complexes that compares the similarity of bound and cognate ligands, enabling the best matches to be identified. We calculate the molecular similarity scores using a method called PARITY (proportion of atoms residing in identical topology), which can conveniently be combined to give a similarity score for all cognate reactants or products in the reaction. Thus, we generate a rank-ordered list of related PDB structures, according to the biological similarity of the ligands bound in the structures.


Asunto(s)
Acetilcolina/química , Acetilcolinesterasa/química , Biosimilares Farmacéuticos/química , Uroporfirinógeno III Sintetasa/química , Uroporfirinógenos/química , Acetilcolina/metabolismo , Acetilcolinesterasa/metabolismo , Sitios de Unión , Biosimilares Farmacéuticos/metabolismo , Humanos , Ligandos , Simulación del Acoplamiento Molecular , Unión Proteica , Especificidad por Sustrato , Uroporfirinógeno III Sintetasa/metabolismo , Uroporfirinógenos/metabolismo
3.
Proteins ; 85(1): 46-53, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27756106

RESUMEN

In an earlier study, we showed that two-domain segment-swapped proteins can evolve by domain swapping and fusion, resulting in a protein with two linkers connecting its domains. We proposed that a potential evolutionary advantage of this topology may be the restriction of interdomain motions, which may facilitate domain closure by a hinge-like movement, crucial for the function of many enzymes. Here, we test this hypothesis computationally on uroporphyrinogen III synthase, a two-domain segment-swapped enzyme essential in porphyrin metabolism. To compare the interdomain flexibility between the wild-type, segment-swapped enzyme (having two interdomain linkers) and circular permutants of the same enzyme having only one interdomain linker, we performed geometric and molecular dynamics simulations for these species in their ligand-free and ligand-bound forms. We find that in the ligand-free form, interdomain motions in the wild-type enzyme are significantly more restricted than they would be with only one interdomain linker, while the flexibility difference is negligible in the ligand-bound form. We also estimated the entropy costs of ligand binding associated with the interdomain motions, and find that the change in domain connectivity due to segment swapping results in a reduction of this entropy cost, corresponding to ∼20% of the total ligand binding free energy. In addition, the restriction of interdomain motions may also help the functional domain-closure motion required for catalysis. This suggests that the evolution of the segment-swapped topology facilitated the evolution of enzyme function for this protein by influencing its dynamic properties. Proteins 2016; 85:46-53. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Proteínas Bacterianas/química , Thermus thermophilus/química , Uroporfirinógeno III Sintetasa/química , Uroporfirinógenos/química , Biocatálisis , Entropía , Evolución Molecular , Ligandos , Simulación de Dinámica Molecular , Movimiento (Física) , Unión Proteica , Dominios Proteicos , Estructura Secundaria de Proteína , Thermus thermophilus/enzimología
4.
Cell Mol Life Sci ; 71(15): 2837-63, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24515122

RESUMEN

Hemes (a, b, c, and o) and heme d 1 belong to the group of modified tetrapyrroles, which also includes chlorophylls, cobalamins, coenzyme F430, and siroheme. These compounds are found throughout all domains of life and are involved in a variety of essential biological processes ranging from photosynthesis to methanogenesis. The biosynthesis of heme b has been well studied in many organisms, but in sulfate-reducing bacteria and archaea, the pathway has remained a mystery, as many of the enzymes involved in these characterized steps are absent. The heme pathway in most organisms proceeds from the cyclic precursor of all modified tetrapyrroles uroporphyrinogen III, to coproporphyrinogen III, which is followed by oxidation of the ring and finally iron insertion. Sulfate-reducing bacteria and some archaea lack the genetic information necessary to convert uroporphyrinogen III to heme along the "classical" route and instead use an "alternative" pathway. Biosynthesis of the isobacteriochlorin heme d 1, a cofactor of the dissimilatory nitrite reductase cytochrome cd 1, has also been a subject of much research, although the biosynthetic pathway and its intermediates have evaded discovery for quite some time. This review focuses on the recent advances in the understanding of these two pathways and their surprisingly close relationship via the unlikely intermediate siroheme, which is also a cofactor of sulfite and nitrite reductases in many organisms. The evolutionary questions raised by this discovery will also be discussed along with the potential regulation required by organisms with overlapping tetrapyrrole biosynthesis pathways.


Asunto(s)
Vías Biosintéticas , Hemo/análogos & derivados , Tetrapirroles/metabolismo , Animales , Hemo/química , Hemo/metabolismo , Humanos , Modelos Moleculares , Tetrapirroles/química , Uroporfirinógenos/química , Uroporfirinógenos/metabolismo
5.
J Biol Chem ; 286(30): 26754-67, 2011 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-21632530

RESUMEN

During the biosynthesis of heme d(1), the essential cofactor of cytochrome cd(1) nitrite reductase, the NirE protein catalyzes the methylation of uroporphyrinogen III to precorrin-2 using S-adenosyl-L-methionine (SAM) as the methyl group donor. The crystal structure of Pseudomonas aeruginosa NirE in complex with its substrate uroporphyrinogen III and the reaction by-product S-adenosyl-L-homocysteine (SAH) was solved to 2.0 Å resolution. This represents the first enzyme-substrate complex structure for a SAM-dependent uroporphyrinogen III methyltransferase. The large substrate binds on top of the SAH in a "puckered" conformation in which the two pyrrole rings facing each other point into the same direction either upward or downward. Three arginine residues, a histidine, and a methionine are involved in the coordination of uroporphyrinogen III. Through site-directed mutagenesis of the nirE gene and biochemical characterization of the corresponding NirE variants the amino acid residues Arg-111, Glu-114, and Arg-149 were identified to be involved in NirE catalysis. Based on our structural and biochemical findings, we propose a potential catalytic mechanism for NirE in which the methyl transfer reaction is initiated by an arginine catalyzed proton abstraction from the C-20 position of the substrate.


Asunto(s)
Proteínas Bacterianas/química , Metiltransferasas/química , Pseudomonas aeruginosa/enzimología , Uroporfirinógenos/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Catálisis , Cristalografía por Rayos X , Hemo/análogos & derivados , Hemo/biosíntesis , Hemo/química , Hemo/metabolismo , Metiltransferasas/genética , Metiltransferasas/metabolismo , Mutagénesis Sitio-Dirigida , Mutación Missense , Estructura Terciaria de Proteína , Pseudomonas aeruginosa/genética , Uroporfirinógenos/genética , Uroporfirinógenos/metabolismo
6.
Biometals ; 22(2): 345-51, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18956144

RESUMEN

Accumulating evidence, including experiments using cytochrome P450 1a2 (Cyp1a2) gene knock-out mice (Cyp1a2(-/-)), indicates that the development of chemically induced porphyria requires the expression of CYP1A2. It has also been demonstrated that iron enhances and expedites the development of experimental uroporphyria, but that iron alone without CYP1A2 expression, as in Cyp1a2(-/-) mice, does not cause uroporphyria. The role of iron in the development of porphyria has not been elucidated. We examined the in vivo effect of iron deficiency on hepatic URO accumulation in experimental porphyria. Mice were fed diets containing low (iron-deficient diet (IDD), 8.5 mg iron/kg) or normal (normal diet (ND), 213.7 mg iron/kg) levels of iron. They were treated with 3-methylcholanthrene (MC), an archetypal inducer of CYP1A, and 5-aminolevulinate (ALA), precursors of porphyrin and heme. We found that uroporphyrin (URO) levels and uroporphyrinogen oxidation (UROX) activity were markedly increased in ND mice treated with MC and ALA, while the levels were not raised in IDD mice with the same treatments. CYP1A2 levels and methoxyresorufin O-demethylase (MROD) activities, the CYP1A2-mediated reaction, were markedly induced in the livers of both ND and IDD mice treated with MC and ALA. UROX activity, supposedly a CYP1A2-dependent activity, was not enhanced in iron-deficient mice in spite of the fact of induction of CYP1A2. We showed that a sufficient level of iron is essential for the development of porphyria and UROX activity.


Asunto(s)
Ácido Aminolevulínico/farmacología , Hierro/metabolismo , Metilcolantreno/farmacología , Porfirias/inducido químicamente , Uroporfirinas/metabolismo , Animales , Citocromo P-450 CYP1A2/genética , Hierro/química , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oxígeno/química , Porfirias/metabolismo , Factores de Tiempo , Uroporfirinógenos/química , Uroporfirinógenos/metabolismo , Uroporfirinas/química
7.
Biochemistry ; 47(33): 8648-55, 2008 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-18651750

RESUMEN

Uroporphyrinogen III synthase (U3S) catalyzes the asymmetrical cyclization of a linear tetrapyrrole to form the physiologically relevant uroporphyrinogen III (uro'gen III) isomer during heme biosynthesis. Here, we report four apoenzyme and one product complex crystal structures of the Thermus thermophilus (HB27) U3S protein. The overlay of eight crystallographically unique U3S molecules reveals a huge range of conformational flexibility, including a "closed" product complex. The product, uro'gen III, binds between the two domains and is held in place by a network of hydrogen bonds between the product's side chain carboxylates and the protein's main chain amides. Interactions of the product A and B ring carboxylate side chains with both structural domains of U3S appear to dictate the relative orientation of the domains in the closed enzyme conformation and likely remain intact during catalysis. The product C and D rings are less constrained in the structure, consistent with the conformational changes required for the catalytic cyclization with inversion of D ring orientation. A conserved tyrosine residue is potentially positioned to facilitate loss of a hydroxyl from the substrate to initiate the catalytic reaction.


Asunto(s)
Uroporfirinógeno III Sintetasa/química , Uroporfirinógeno III Sintetasa/metabolismo , Uroporfirinógenos/química , Uroporfirinógenos/metabolismo , Cristalización , Modelos Moleculares , Estructura Molecular , Thermus thermophilus/enzimología
8.
Proc Natl Acad Sci U S A ; 104(12): 5079-84, 2007 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-17360334

RESUMEN

Porphyria cutanea tarda (PCT), the most common form of porphyria in humans, is due to reduced activity of uroporphyrinogen decarboxylase (URO-D) in the liver. Previous studies have demonstrated that protein levels of URO-D do not change when catalytic activity is reduced, suggesting that an inhibitor of URO-D is generated in hepatocytes. Here, we describe the identification and characterization of an inhibitor of URO-D in liver cytosolic extracts from two murine models of PCT: wild-type mice treated with iron, delta-aminolevulinic acid, and polychlorinated biphenyls; and mice with one null allele of Uro-d and two null alleles of the hemochromatosis gene (Uro-d(+/-), Hfe(-/-)) that develop PCT with no treatments. In both models, we identified an inhibitor of recombinant human URO-D (rhURO-D). The inhibitor was characterized by solid-phase extraction, chromatography, UV-visible spectroscopy, and mass spectroscopy and proved to be uroporphomethene, a compound in which one bridge carbon in the uroporphyrinogen macrocycle is oxidized. We synthesized uroporphomethene by photooxidation of enzymatically generated uroporphyrinogen I or III. Both uroporphomethenes inhibited rhURO-D, but the III isomer porphomethene was a more potent inhibitor. Finally, we detected an inhibitor of rhURO-D in cytosolic extracts of liver biopsy samples of patients with PCT. These studies define the mechanism underlying clinical expression of the PCT phenotype, namely oxidation of uroporphyrinogen to uroporphomethene, a competitive inhibitor of URO-D. The oxidation reaction is iron-dependent.


Asunto(s)
Porfiria Cutánea Tardía/etiología , Porfirinas/farmacología , Uroporfirinógeno Descarboxilasa/antagonistas & inhibidores , Animales , Cromatografía Líquida de Alta Presión , Citosol/efectos de los fármacos , Citosol/enzimología , Humanos , Hígado/efectos de los fármacos , Hígado/enzimología , Extractos Hepáticos , Espectrometría de Masas , Ratones , Ratones Endogámicos C57BL , Porfiria Cutánea Tardía/inducido químicamente , Porfirinas/análisis , Porfirinas/química , Proteínas Recombinantes/metabolismo , Uroporfirinógenos/química
9.
Transl Res ; 149(2): 85-91, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17240319

RESUMEN

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.


Asunto(s)
Codón sin Sentido , Mutación Missense , Porfiria Hepatoeritropoyética/genética , Uroporfirinógeno Descarboxilasa/genética , Adulto , Línea Celular Transformada , Preescolar , Coproporfirinógenos/biosíntesis , Coproporfirinógenos/química , Cristalografía , Activación Enzimática/genética , Femenino , Genotipo , Heterocigoto , Homocigoto , Humanos , Masculino , Fenotipo , Porfiria Hepatoeritropoyética/metabolismo , Embarazo , Diagnóstico Prenatal , Estructura Terciaria de Proteína , Uroporfirinógeno Descarboxilasa/química , Uroporfirinógeno Descarboxilasa/metabolismo , Uroporfirinógenos/química , Uroporfirinógenos/metabolismo
10.
Nat Prod Rep ; 19(4): 390-412, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12195810

RESUMEN

Vitamin B12, or cobalamin, is one of the most structurally complex small molecules made in Nature. Major progress has been made over the past decade in understanding how this synthesis is accomplished. This review covers some of the most important findings that have been made and provides the reader with a complete description of the transformation of uroporphyrinogen III into adenosylcobalamin (AdoCbl). 183 references are cited.


Asunto(s)
Bacterias/metabolismo , Uroporfirinógenos/biosíntesis , Uroporfirinógenos/química , Vitamina B 12 , Catálisis , Cobalto/metabolismo , Cobamidas/metabolismo , Ciclización , Genes Bacterianos , Metiltransferasas/química , Metiltransferasas/metabolismo , Estructura Molecular , Nucleotidiltransferasas/metabolismo , Conformación Proteica , Uroporfirinas/biosíntesis , Uroporfirinas/química , Vitamina B 12/análogos & derivados , Vitamina B 12/biosíntesis , Vitamina B 12/química
11.
Biochem Pharmacol ; 63(4): 615-24, 2002 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-11992629

RESUMEN

In previous work it was shown that hepatic microsomes from rats treated with 3-methylcholanthrene and similar inducers had increased bilirubin-degrading activity. The activity was further stimulated by addition of 3,4-tetrachlorobiphenyl (TCB), a response specifically dependent on CYP1A1. Here, we compared the effect of adding PCBs of either planar or non-planar configuration on rate of bilirubin degradation, monooxygenase activity and NADPH/O(2) consumption by liver microsomes from animals treated with either phenobarbital or 3-methylcholanthrene/beta-naphthoflavone. We also examined the oxidation of uroporphyrinogen (hexahydro-uroporphyrin) (URO'gen) under these conditions. Polychlorinated biphenyl (PCBs) stimulated the rate of bilirubin and URO'gen oxidation with microsomes expressing high levels of either CYP2B or CYP1A, inhibiting at the same time their monooxygenase activities (PROD and EROD, respectively); however, non-planar di-ortho-substituted PCBs were preferentially active with phenobarbitone-induced microsomes, in contrast to those active with 3-methylcholanthrene/beta-naphthoflavone microsomes, where a planar configuration was required for activity. An antibody raised against CYP2B1 markedly inhibited the PCB-dependent bilirubin degradation and PROD activities of phenobarbital-induced microsomes with similar dose-response curves for the two effects. Increased microsomal utilizations of NADPH and O(2) were also caused by PCBs with both types of induced microsomes and here again PCBs of different configuration were preferentially active. It is concluded that PCBs of the appropriate configuration may interact with either CYP1A1 or CYP2B1, increase production of oxidative species by an uncoupling mechanism, and lead to oxidation of target molecules in the cell, among these uroporphyrinogen and bilirubin.


Asunto(s)
Hidrocarburo de Aril Hidroxilasas , Bilirrubina/metabolismo , Sistema Enzimático del Citocromo P-450/metabolismo , Microsomas Hepáticos/metabolismo , Oxidorreductasas N-Desmetilantes/metabolismo , Bifenilos Policlorados/metabolismo , Uroporfirinógenos/metabolismo , Animales , Anticuerpos/inmunología , Bilirrubina/química , Citocromo P-450 CYP2B6 , Sistema Enzimático del Citocromo P-450/biosíntesis , Masculino , Microsomas Hepáticos/enzimología , Conformación Molecular , NADP/metabolismo , Oxidación-Reducción , Oxígeno/metabolismo , Fenobarbital/farmacología , Ratas , Ratas Wistar , Uroporfirinógenos/química
12.
Proc Natl Acad Sci U S A ; 98(1): 259-64, 2001 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-11134514

RESUMEN

Approximately one-third of patients with porphyria cutanea tarda (PCT), the most common porphyria in humans, inherit a single mutant allele of the uroporphyrinogen decarboxylase (URO-D) gene. PCT associated with URO-D mutations is designated familial PCT. The phenotype is characterized by a photosensitive dermatosis with hepatic accumulation and urinary excretion of uroporphyrin and hepta-carboxylic porphyrins. Most heterozygotes for URO-D mutations do not express a porphyric phenotype unless hepatic siderosis is present. Hemochromatosis gene (HFE) mutations are frequently found when the phenotype is expressed. We used homologous recombination to disrupt one allele of murine URO-D. URO-D(+/-) mice had half-wild type (wt) URO-D protein and enzymatic activity in all tissues but did not accumulate hepatic porphyrins, indicating that half-normal URO-D activity is not rate limiting. When URO-D(+/-) mice were injected with iron-dextran and given drinking water containing delta-aminolevulinic acid for 21 days, hepatic porphyrins accumulated, and hepatic URO-D activity was reduced to 20% of wt. We bred mice homozygous for an HFE gene disruption (HFE(-/-)) to URO-D(+/-) mice, generating mice with the URO-D(+/-)/HFE(-/-) genotype. These animals developed a porphyric phenotype by 14 weeks of age without ALA supplementation, and URO-D activity was reduced to 14% of wt. These data indicate that iron overload alone is sufficient to reduce URO-D activity to rate-limiting levels in URO-D(+/-) mice. The URO-D(+/-) mouse serves as an excellent model of familial PCT and affords the opportunity to define the mechanism by which iron influences URO-D activity.


Asunto(s)
Modelos Animales de Enfermedad , Hemocromatosis/genética , Porfiria Cutánea Tardía/genética , Uroporfirinógeno Descarboxilasa/genética , Ácido Aminolevulínico/farmacología , Animales , Clonación Molecular , Coproporfirinógenos/química , Coproporfirinógenos/metabolismo , Inhibidores Enzimáticos/farmacología , Eliminación de Gen , Marcación de Gen , Genotipo , Humanos , Hierro/análisis , Complejo Hierro-Dextran/administración & dosificación , Complejo Hierro-Dextran/farmacología , Hígado/química , Hígado/metabolismo , Ratones , Ratones Noqueados , Fenotipo , Porfiria Cutánea Tardía/inducido químicamente , Porfiria Cutánea Tardía/enzimología , Porfiria Cutánea Tardía/metabolismo , Porfirinas/análisis , Porfirinas/orina , Células Madre/metabolismo , Uroporfirinógeno Descarboxilasa/análisis , Uroporfirinógeno Descarboxilasa/antagonistas & inhibidores , Uroporfirinógeno Descarboxilasa/metabolismo , Uroporfirinógenos/química , Uroporfirinógenos/metabolismo
13.
Nihon Rinsho ; 53(6): 1319-28, 1995 Jun.
Artículo en Japonés | MEDLINE | ID: mdl-7616643

RESUMEN

There are two ways for numbering the positions of C and N atoms in the porphyrin ring. The Fisher numeration is relatively old styled, but at present, is most widely employed among chemists because of its simpler and easier technique than that of the IUPAC numeration which is more effective in numbering the complicated structures of recent synthetic chemical including recent porphyrin derivatives. Uroporphyrinogen III, Coproporphyrinogen III and Protoporphynogen IX have been described as the porphyrin intermediates in heme biosynthesis. Uroporphyrins, Coproporphyrins and Protoporphyrin IX which are found in blood, urine, feces of either normal or porphyric subjects are their auto-oxidized products and can not be true intermediates for heme biosynthesis except for Protoporphyrin. Delta aminolevulinic acid (ALA), Porphobilinogen (PBG) and Hydroxymethylbilane have been described as the precursors to porphyrin biosynthesis. Besides the recent advance of porphyrin research within the limit of medical fields the pure organic chemistry of porphyrins symbolized by its large electron conjugated system have been unexpectedly developed for the last 20 years, suggesting that its application will quickly be extended to the new fields of macromolecular engineering and polymer electronics.


Asunto(s)
Hemo/biosíntesis , Porfirinas/química , Ácido Aminolevulínico/química , Humanos , Porfobilinógeno/química , Uroporfirinógenos/química
14.
Bioorg Med Chem ; 2(4): 267-77, 1994 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-7922138

RESUMEN

A novel hydroxymethylbilane analog, 19-Br-HMB (11), has been synthesized. Its activity with the enzyme Uro'gen III synthase shows competitive inhibition.


Asunto(s)
Uroporfirinógeno III Sintetasa/antagonistas & inhibidores , Uroporfirinógenos/farmacología , Unión Competitiva , Técnicas In Vitro , Espectroscopía de Resonancia Magnética , Uroporfirinógenos/biosíntesis , Uroporfirinógenos/síntesis química , Uroporfirinógenos/química
15.
Bioorg Med Chem ; 2(3): 153-68, 1994 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-7922129

RESUMEN

Evidence for an azafulvene intermediate in the enzymatic formation of Uroporphyrinogen III has been obtained. Using conditions to slow down the enzyme activity (high pH, low temperature), the transient species was trapped with ammonium ions as aminomethylbilane and with sodium borohydride as methylbilane, and observed by 13C-NMR.


Asunto(s)
Uroporfirinógenos/biosíntesis , Escherichia coli/enzimología , Escherichia coli/genética , Genes Bacterianos , Concentración de Iones de Hidrógeno , Técnicas In Vitro , Espectroscopía de Resonancia Magnética , Estructura Molecular , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Temperatura , Uroporfirinógeno III Sintetasa/metabolismo , Uroporfirinógenos/química , Uroporfirinógenos/genética , Uroporfirinógenos/metabolismo
16.
Ciba Found Symp ; 180: 111-23; discussion 124-30, 1994.
Artículo en Inglés | MEDLINE | ID: mdl-7842849

RESUMEN

In the course of the cyclization of the linear tetrapyrrole hydroxymethylbilane to uroporphyrinogen III, catalysed by uroporphyrinogen III synthase (cosynthase), ring D of the bilane becomes inverted. Many different mechanisms have been proposed for this transformation but the most economical is one involving a spirocyclic pyrrolenine. Synthesis of a spirolactam, and other compounds closely related to the spirocyclic pyrrolenine, has shown that such compounds are not impossibly strained. The spirolactam is a powerful inhibitor of the enzyme, which suggests it does resemble an intermediate in the enzymic process. In the synthetic procedure to make an ester of the spirolactam the two products obtained were initially thought to be conformational isomers. However, molecular mechanics calculations on a model of the spirolactam predicted that several low energy conformations should exist and that the energy barriers for their interconversion are all lower than 32 kJ/mol. Reinvestigation revealed that one of the two products is in fact a macrocyclic dimer with a 28-membered ring. On the basis of the predicted preferred conformations of the spirolactam and of uroporphyrinogen III, a detailed three-dimensional mechanism is proposed, along with a rationalization of how the rearrangement of ring D may be directed by the enzyme.


Asunto(s)
Compuestos de Espiro/química , Uroporfirinógeno III Sintetasa/metabolismo , Uroporfirinógenos/biosíntesis , Estructura Molecular , Conformación Proteica , Uroporfirinógenos/química
17.
Enzyme Protein ; 48(5-6): 275-81, 1994.
Artículo en Inglés | MEDLINE | ID: mdl-8792872

RESUMEN

The enzyme porphobilinogen deaminase (PBG deaminase, EC 4.3.1.8) catalyzes the condensation of four molecules of PBG to give the linear tetrapyrrol, hydroxymethylbilane. It has been shown that this enzyme forms stable mono-, di-, tri- and tetrapyrrole-enzyme covalent complexes. When the enzyme, partially purified in the absence or presence of phenylmethylsulfonyl fluoride (PMSF) and preincubated with PBG, was applied on DEAE-cellulose columns, three peaks with PBG deaminase activity were detected. Using Ehrlich's reagent, it was found that the active peaks corresponded to mono-, di- and tri-pyrrylmethane-enzyme complexes. Therefore, the mechanism of action of PBG deaminase from Saccharomyces cerevisiae also involves the sequential addition of four PBG units, leading to the formation of the enzyme-substrate intermediate complexes, as has already been described for the same enzyme from other sources.


Asunto(s)
Hidroximetilbilano Sintasa/análisis , Hidroximetilbilano Sintasa/metabolismo , Pirroles/metabolismo , Saccharomyces cerevisiae/enzimología , Cromatografía DEAE-Celulosa/métodos , Indicadores y Reactivos , Fluoruro de Fenilmetilsulfonilo , Porfobilinógeno/química , Porfobilinógeno/metabolismo , Inhibidores de Proteasas , Uroporfirinógenos/química
19.
Trends Biochem Sci ; 15(12): 486-91, 1990 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-2077690

RESUMEN

Data obtained using a combination of molecular biology and NMR spectroscopy has transformed our thinking about the evolution of the biochemical machinery required for the synthesis of the vital metallopigments: haem, chlorophyll, vitamin B12 and factor F430. One of the most recent advances is the discovery of a unique dipyrromethane cofactor that is bound covalently at the active site of porphobillinogen deaminase, the key enzyme of tetrapyrrole assembly. We will also discuss how the oxidation level and chromophoric arrangement of the uroporphinoid ring, rather than its substitution pattern, provides the necessary molecular recognition for some of the later enzymes, whose function is to decorate the template by C-methylation on the way to the biologically active cofactors.


Asunto(s)
Pirroles/química , Uroporfirinógenos/química , Sitios de Unión , Hidroximetilbilano Sintasa/química , Ligandos , Metilación , Porfobilinógeno/química , Tetrapirroles , Uroporfirinógenos/metabolismo
20.
Photochem Photobiol ; 42(3): 239-44, 1985.
Artículo en Inglés | MEDLINE | ID: mdl-11539651

RESUMEN

The photochemical ramifications of the high ultraviolet flux on the primordial earth prior to the formation of the ozone layer have been considered in a study of the ultraviolet photochemistry of uroporphyrinogen (urohexahydroporphyrin), a colorless compound which absorbs strongly at wavelengths less than 220 nanometers. Urohexahydroporphyrin was investigated since it is the first macrocycle formed on the biosynthetic pathway of chlorophyll and can be used to test the hypothesis that the biosynthetic pathway to chlorophyll recapitulates the evolutionary history of photosynthesis. When urohexahydroporphyrin is illuminated in aqueous anaerobic solution, hydrogen gas is produced. More hydrogen gas is produced in the presence of a colloidal platinum catalyst. The products of the photooxidation of urohexahydroporphyrin are urotetrahydroporphyrin (uroporphomethene) and uroporphyrin. This research shows how the oxidation of uroporphyrinogen to uroporphyrin, the first biogenetic porphyrin, could have occurred anaerobically and abiotically on the primordial earth.


Asunto(s)
Planeta Tierra , Evolución Química , Evolución Planetaria , Fotosíntesis , Rayos Ultravioleta , Uroporfirinas/química , Atmósfera , Clorofila/química , Hidrógeno/química , Oxígeno/química , Fotoquímica , Uroporfirinógenos/química , Uroporfirinógenos/efectos de la radiación , Uroporfirinas/efectos de la radiación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...