Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
1.
Photodiagnosis Photodyn Ther ; 25: 309-316, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30639584

RESUMEN

BACKGROUND: Recently, 5-aminolevulinic acid (5-ALA)-induced protoporphyrin IX fluorescence was reported to be a useful tool during total surgical resection of high-grade gliomas. However, the labeling efficacy of protoporphyrin IX fluorescence is lower in metastatic brain tumors compared to that in high-grade gliomas, and the mechanism underlying protoporphyrin IX fluorescence in metastatic brain tumors remains unclear. Lung cancer, particularly non-small cell lung cancer (NSCLC), is the most common origin for metastatic brain tumor. Therefore, we investigated the mechanism of protoporphyrin IX fluorescence in NSCLC and associated metastatic brain tumors. METHODS: Western blotting and quantitative real-time polymerase chain reaction (qRT-PCR) was employed to evaluate the protein and mRNA levels of five transporters and enzymes involved in the porphyrin biosynthesis pathway: peptide transporter 1 (PEPT1), hydroxymethylbilane synthase (HMBS), ferrochelatase (FECH), ATP-binding cassette 2 (ABCG2), and heme oxygenase 1 (HO-1). The correlation between protein, mRNA, and protoporphyrin IX levels in NSCLC cells were evaluated in vitro. Immunohistochemistry was used to determine proteins that played a key role in intraoperative protoporphyrin IX fluorescence in clinical samples from patients with NSCLC and pathologically confirmed metastatic brain tumors. RESULTS: A significant correlation between PEPT1 expression and protoporphyrin IX accumulation in vitro was identified by western blotting (P = 0.003) and qRT-PCR (P = 0.04). Immunohistochemistry results indicated that there was a significant difference in PEPT1 between the intraoperative protoporphyrin IX fluorescence-positive and protoporphyrin IX fluorescence-negative groups (P = 0.009). CONCLUSION: Expression of PEPT1 was found to be positively correlated with 5-ALA-induced protoporphyrin IX accumulation detected by photodynamic reaction in metastatic brain tumors originating from NSCLC.


Asunto(s)
Ácido Aminolevulínico/farmacología , Transportador de Péptidos 1/biosíntesis , Fotoquimioterapia/métodos , Protoporfirinas/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/biosíntesis , Western Blotting , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/secundario , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Ferroquelatasa/biosíntesis , Hemo-Oxigenasa 1/biosíntesis , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Metástasis de la Neoplasia , Reacción en Cadena en Tiempo Real de la Polimerasa , Espectrometría de Fluorescencia , Uroporfirinógenos/biosíntesis
2.
Sheng Wu Gong Cheng Xue Bao ; 33(1): 55-67, 2017 Jan 25.
Artículo en Chino | MEDLINE | ID: mdl-28959863

RESUMEN

Biosynthesis of vitamin B12 (VB12) requires the methylation at positions C-2 and C-7 of the precursor uroporphyrinogen Ⅲ (urogen Ⅲ) to precorrin-2 by S-adenosyl-L-methionine uroporphyrinogen Ⅲ methyltransferase (SUMT), which is a potential bottleneck step. Most of SUMTs are inhibited by urogen Ⅲ and by-product S-adenosyl-L-homocysteine (SAH). In order to mine an SUMT that lacks such an inhibitory property to drive greater flux through the VB12 biosynthetic pathway, we cloned two SUMT genes (RCcobA1, RCcobA2) from Rhodobacter capsulatus SB1003 and expressed them in Escherichia coli BL21 (DE3). Thereafter, the two enzymes were purified and their specific activity of 27.3 U/mg, 68.9 U/mg were determined respectively. The latter was 2.4 times higher than PDcobA (27.9 U/mg) from Pseudomonas denitrifican. Additionally, RCcobA2 could tolerate over 70 µmol/L urogen Ⅲ, which has never been reported before. Hence, RCcobA2 can be used as an efficient enzyme to regulate the VB12 metabolic pathway and enhance VB12 production in industrial strains.


Asunto(s)
Metiltransferasas/aislamiento & purificación , Rhodobacter capsulatus/enzimología , Escherichia coli , Metionina , Pseudomonas , S-Adenosilmetionina , Uroporfirinógenos , Uroporfirinas
3.
Eukaryot Cell ; 10(11): 1536-44, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21908598

RESUMEN

The azaoxoaporphine alkaloid sampangine exhibits strong antiproliferation activity in various organisms. Previous studies suggested that it somehow affects heme metabolism and stimulates production of reactive oxygen species (ROS). In this study, we show that inhibition of heme biosynthesis is the primary mechanism of action by sampangine and that increases in the levels of reactive oxygen species are secondary to heme deficiency. We directly demonstrate that sampangine inhibits heme synthesis in the yeast Saccharomyces cerevisiae. It also causes accumulation of uroporphyrinogen and its decarboxylated derivatives, intermediate products of the heme biosynthesis pathway. Our results also suggest that sampangine likely works through an unusual mechanism-by hyperactivating uroporhyrinogen III synthase-to inhibit heme biosynthesis. We also show that the inhibitory effect of sampangine on heme synthesis is conserved in human cells. This study also reveals a surprising essential role for the interaction between the mitochondrial ATP synthase and the electron transport chain.


Asunto(s)
Alcaloides/farmacología , Hemo/biosíntesis , Saccharomyces cerevisiae/efectos de los fármacos , Proteasas ATP-Dependientes/genética , Proteasas ATP-Dependientes/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Humanos , Células Jurkat , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , ATPasas de Translocación de Protón Mitocondriales/genética , ATPasas de Translocación de Protón Mitocondriales/metabolismo , Naftiridinas , Extractos Vegetales/farmacología , Protoporfirinógeno-Oxidasa/genética , Protoporfirinógeno-Oxidasa/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Uroporfirinógeno III Sintetasa/biosíntesis , Uroporfirinógeno III Sintetasa/metabolismo , Uroporfirinógenos/metabolismo
4.
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
5.
FEBS J ; 276(20): 5973-82, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19754882

RESUMEN

Biosynthesis of heme d(1), the essential prosthetic group of the dissimilatory nitrite reductase cytochrome cd(1), requires the methylation of the tetrapyrrole precursor uroporphyrinogen III at positions C-2 and C-7. We produced Pseudomonas aeruginosa NirE, a putative S-adenosyl-L-methionine (SAM)-dependent uroporphyrinogen III methyltransferase, as a recombinant protein in Escherichia coli and purified it to apparent homogeneity by metal chelate and gel filtration chromatography. Analytical gel filtration of purified NirE indicated that the recombinant protein is a homodimer. NirE was shown to be a SAM-dependent uroporphyrinogen III methyltransferase that catalyzes the conversion of uroporphyrinogen III into precorrin-2 in vivo and in vitro. A specific activity of 316.8 nmol of precorrin-2 h(-1) x mg(-1) of NirE was found for the conversion of uroporphyrinogen III to precorrin-2. At high enzyme concentrations NirE catalyzed an overmethylation of uroporphyrinogen III, resulting in the formation of trimethylpyrrocorphin. Substrate inhibition was observed at uroporphyrinogen III concentrations above 17 microM. The protein did bind SAM, although not with the same avidity as reported for other SAM-dependent uroporphyrinogen III methyltransferases involved in siroheme and cobalamin biosynthesis. A P. aeruginosa nirE transposon mutant was not complemented by native cobA encoding the SAM-dependent uroporphyrinogen III methyltransferase involved in cobalamin formation. However, bacterial growth of the nirE mutant was observed when cobA was constitutively expressed by a complementing plasmid, underscoring the special requirement of NirE for heme d(1) biosynthesis.


Asunto(s)
Proteínas Bacterianas/metabolismo , Hemo/análogos & derivados , Metiltransferasas/química , Metiltransferasas/metabolismo , Pseudomonas aeruginosa/enzimología , Pseudomonas aeruginosa/genética , Secuencia de Aminoácidos , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Prueba de Complementación Genética , Hemo/biosíntesis , Metiltransferasas/genética , Datos de Secuencia Molecular , Unión Proteica , Pseudomonas aeruginosa/crecimiento & desarrollo , Pseudomonas aeruginosa/metabolismo , S-Adenosilmetionina/metabolismo , Homología de Secuencia de Aminoácido , Especificidad por Sustrato , Uroporfirinógenos/metabolismo , Uroporfirinas/metabolismo
6.
Res Microbiol ; 160(2): 125-33, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19154787

RESUMEN

The Geobacillus stearothermophilus V cobA gene encoding uroporphyrinogen-III C-methyltransferase (also referred to as SUMT) was cloned into Escherichia coli and the recombinant enzyme was overexpressed and purified to homogeneity. The enzyme binds S-adenosyl-L-methionine and catalyzes the production of III methyl uroporphyrinogen in vitro. E. coli cells expressing the G. stearothermophilus V cobA gene exhibited increased resistance to potassium tellurite and potassium tellurate. Site-directed mutagenesis of cobA abolished tellurite resistance of the mesophilic, heterologous host and SUMT activity in vitro. No methylated, volatile derivatives of tellurium were found in the headspace of tellurite-exposed cobA-expressing E. coli, suggesting that the role of SUMT methyltransferase in tellurite(ate) detoxification is not related to tellurium volatilization.


Asunto(s)
Escherichia coli/metabolismo , Geobacillus stearothermophilus/enzimología , Metiltransferasas , Telurio/metabolismo , Secuencia de Aminoácidos , Clonación Molecular , Metiltransferasas/análisis , Metiltransferasas/biosíntesis , Metiltransferasas/genética , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Proteínas Recombinantes/biosíntesis , S-Adenosilmetionina/metabolismo , Uroporfirinógenos/biosíntesis
7.
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
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.
J Bacteriol ; 188(24): 8666-8, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17028275

RESUMEN

The methanogenic archaeon Methanosarcina barkeri synthesizes protoheme via precorrin-2, which is formed from uroporphyrinogen III in two consecutive methylation reactions utilizing S-adenosyl-L-methionine. The existence of this pathway, previously exclusively found in the sulfate-reducing delta-proteobacterium Desulfovibrio vulgaris, was demonstrated for M. barkeri via the incorporation of two methyl groups from methionine into protoheme.


Asunto(s)
Hemo/biosíntesis , Methanosarcina barkeri/metabolismo , Metionina/análogos & derivados , Proteínas Arqueales/genética , Proteínas Arqueales/metabolismo , Medios de Cultivo , Methanosarcina barkeri/genética , Methanosarcina barkeri/crecimiento & desarrollo , Metionina/metabolismo , Metilación , Uroporfirinógenos/metabolismo , Uroporfirinas/metabolismo
10.
Biochem Pharmacol ; 66(3): 405-14, 2003 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-12907239

RESUMEN

Polyhalogenated compounds, such as 2,3,7,8-tetrachlorodibenzo-p-dioxin, are associated with toxic Uroporphyria and cause alleviation of jaundice in the Gunn rat. These effects have been attributed to a microsomal oxidation of uroporphyrinogen and bilirubin for which supportive evidence has been obtained in vitro. CYP1A1 required planar polyhalogenated biphenyls for these oxidative reactions, while CYP1A2 was capable of oxidation in their absence. We have now used rat CYP1A1 and confirmed with the pure enzyme that increased bilirubin oxidation was caused by the addition of 3,4,3',4'-tetrachlorobiphenyl. CYP1A2 was more active than CYP1A1 at oxidizing bilirubin in presence of NADPH alone and reacted to addition of 3,4,3',4'-tetrachlorobiphenyl with a depression rather than a stimulation of bilirubin oxidation. We have also tested a bacterial enzyme, CYP102. Dodecanoic acid and its polyhalogenated analogue (perfluorododecanoic acid) both stimulated NADPH oxidation by CYP102, but only the perfluoro analogue stimulated markedly bilirubin oxidation. The analogue exhibited much greater potency than the normal substrate in stimulating NADPH and bilirubin oxidation and also showed greater affinity for CYP102, as measured by the binding constant, Ks. The molar stoichiometry ratio between NADPH and O(2) consumption was 1 in the case of the substrate, but approximated 2 with the perfluoro analogue. We conclude that halogenated substrate analogues can interact with different CYPs to increase production of oxidative species, probably by an uncoupling mechanism. A role of the ferryl-oxygen intermediate is suggested in the oxidation of biologically important molecules, with possible implications for the therapy of jaundice and for toxic oxidative reactions, such as uroporphyria and cancer.


Asunto(s)
Bilirrubina/metabolismo , Sistema Enzimático del Citocromo P-450/metabolismo , Bifenilos Policlorados/farmacología , Uroporfirinógenos/metabolismo , Animales , Proteínas Bacterianas/metabolismo , Catalasa/metabolismo , Citocromo P-450 CYP1A1/metabolismo , Citocromo P-450 CYP1A2/metabolismo , Peróxido de Hidrógeno/metabolismo , Ácidos Láuricos/química , Ácidos Láuricos/metabolismo , Oxigenasas de Función Mixta/metabolismo , Conformación Molecular , NADP/metabolismo , NADPH-Ferrihemoproteína Reductasa , Oxidación-Reducción , Bifenilos Policlorados/química , Ratas , Superóxido Dismutasa/metabolismo
11.
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
12.
Biochem Soc Trans ; 30(4): 613-20, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12196148

RESUMEN

The biosynthesis of vitamin B(12) is summarized, emphasizing the differences observed between the aerobic and anaerobic pathways. The biosynthetic route to adenosylcobalamin from its five-carbon precursor, 5-aminolaevulinic acid, can be divided into three sections: (1) the biosynthesis of uroporphyrinogen III from 5-aminolaevulinic acid, which is common to both pathways; (2) the conversion of uroporphyrinogen III into the ring-contracted, deacylated intermediate precorrin 6 or cobalt-precorrin 6, which includes the primary differences between the two pathways; and (3) the transformation of this intermediate to form adenosylcobalamin.


Asunto(s)
Bacterias/metabolismo , Vitamina B 12/biosíntesis , Ácido Aminolevulínico/metabolismo , Cobamidas/biosíntesis , Cobamidas/química , Uroporfirinógenos/biosíntesis , Uroporfirinógenos/metabolismo , Vitamina B 12/química
13.
Microbiology (Reading) ; 148(Pt 6): 1845-1853, 2002 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12055304

RESUMEN

A search for genes encoding enzymes involved in cobalamin (vitamin B12) production in the commercially important organism Propionibacterium freudenreichii (P. shermanii) has resulted in the isolation of an additional 14 genes encoding enzymes responsible for 17 steps of the anaerobic B12 pathway in this organism. All of the genes believed to be necessary for the biosynthesis of adenosylcobinamide from uroporphyrinogen III have now been isolated except two (cbiA and an as yet unidentified gene encoding cobalt reductase). Most of the genes are contained in two divergent operons, one of which, in turn, is closely linked to the operon encoding the B12-dependent enzyme methylmalonyl-CoA mutase. The close linkage of the three genes encoding the subunits of transcarboxylase to the hemYHBXRL gene cluster is reported. The functions of the P. freudenreichii B12 pathway genes are discussed, and a mechanism for the regulation of cobalamin and propionic acid production by oxygen in this organism is proposed.


Asunto(s)
Genes Bacterianos/genética , Propionibacterium/genética , Propionibacterium/metabolismo , Vitamina B 12/análogos & derivados , Vitamina B 12/biosíntesis , Anaerobiosis , Secuencia de Bases , Transporte Biológico , Clonación Molecular , Cobalto/metabolismo , Secuencia de Consenso , Metiltransferasas/genética , Datos de Secuencia Molecular , Operón/genética , Propionibacterium/enzimología , Uroporfirinógenos/metabolismo , Vitamina B 12/metabolismo
14.
Toxicol Appl Pharmacol ; 168(3): 177-82, 2000 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-11042089

RESUMEN

We had reported previously that 2.5-5 microM sodium arsenite decreased the phenobarbital-mediated induction of CYP2H activity and protein but not CYP2H1 mRNA in chick-embryo hepatocyte cultures. Induction of a CYP1A activity and protein by 3-methylcholanthrene was also decreased by low arsenite concentrations; however, CYP1A mRNAs were not measured in those studies. We report here that low concentrations of arsenite decreased induction of activities and mRNAs of two chicken cytochromes P450, CYP1A (1A4 and 1A5), by 3-methylcholanthrene in chick-embryo hepatocyte cultures. Arsenite treatment did not affect the turnover of either mRNA, nor did it decrease the superinduction of each mRNA caused by treatment with cycloheximide in addition to 3-methylcholanthrene. Glutathione depletion enhanced the effect of arsenite to decrease induction of CYP1A4. These results indicate the induction of CYP1A4 and 1A5 is inhibited by sodium arsenite at the level of transcription, suggesting that the Ah receptor complex may be involved.


Asunto(s)
Arsenitos/toxicidad , Hidrocarburo de Aril Hidroxilasas , Proteínas Aviares , Sistema Enzimático del Citocromo P-450/biosíntesis , Hepatocitos/enzimología , Oxidorreductasas/biosíntesis , Animales , Células Cultivadas , Embrión de Pollo , Cicloheximida/farmacología , Sistema Enzimático del Citocromo P-450/metabolismo , Inducción Enzimática/efectos de los fármacos , Glutatión/fisiología , Hepatocitos/efectos de los fármacos , Indicadores y Reactivos , Microsomas Hepáticos/efectos de los fármacos , Microsomas Hepáticos/enzimología , Oxidación-Reducción , Oxidorreductasas/metabolismo , Inhibidores de la Síntesis de la Proteína/farmacología , ARN Mensajero/biosíntesis , Uroporfirinógenos/biosíntesis
15.
Toxicol Appl Pharmacol ; 162(1): 60-7, 2000 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-10631128

RESUMEN

Using Cyp1a2(-/-) mice we previously showed that CYP1A2 is absolutely required for hepatic uroporphyrin accumulation caused by iron and 5-aminolevulinate (ALA) treatment, both in the presence and absence of an inducer of CYP1A2. In this study we have used these mice to investigate whether CYP1A2 has an obligatory role in hepatic uroporphyria caused by hexachlorobenzene (HCBZ), an inducer of CYP2B and CYP3A, as well as CYP1A2. Here we treated mice with HCBZ and iron, with and without the porphyrin precursor, ALA, in the drinking water. In iron-loaded wild-type mice given a single dose of HCBZ and ALA, hepatic uroporphyrin (URO) accumulated to 300 nmol/g liver after 37 days, whereas in Cyp1a2(-/-) mice, there was no hepatic URO, even after an additional dose of HCBZ, and a further 29 days of ALA treatment. A similar requirement for CYP1A2 was found in uroporphyria produced in HCBZ and iron-treated mice in the absence of ALA. As detected by Western immunoblotting, HCBZ induced small increases in CYP2B and CYP3A in the livers of all animals. In the wild-type animals, HCBZ also induced CYP1A2 and associated enzyme activities, including uroporphyrinogen oxidation, by about 2-3-fold. In the Cyp1a2(-/-) mice, HCBZ did not increase hepatic microsomal uroporphyrinogen oxidation. These results indicate that, in mice, CYP1A2 is essential in the process leading to HCBZ-induced uroporphyria. Contributions by other CYP forms induced by HCBZ appear to be minimal.


Asunto(s)
Citocromo P-450 CYP1A2/fisiología , Inducción Enzimática/efectos de los fármacos , Imidazoles/toxicidad , Hierro/toxicidad , Microsomas Hepáticos/metabolismo , Uroporfirinas/metabolismo , Ácido Aminolevulínico/farmacología , Animales , Anticuerpos/inmunología , Western Blotting , Enfermedad Hepática Inducida por Sustancias y Drogas , Citocromo P-450 CYP1A2/metabolismo , Sistema Enzimático del Citocromo P-450/metabolismo , Interacciones Farmacológicas , Hígado/química , Masculino , Metilcolantreno/toxicidad , Ratones , Ratones Endogámicos C57BL , Oxazinas/metabolismo , Oxidación-Reducción , Fenobarbital/toxicidad , Factores de Tiempo , Uroporfirinógenos/metabolismo
16.
Biochem Mol Biol Int ; 47(6): 945-56, 1999 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10410240

RESUMEN

Porphyrinogen carboxy-lyase is an enzyme that sequentially decarboxylates uroporphyrinogen III (8-COOH) to yield coproporphyrinogen III (4-COOH). In mammals this enzyme activity is impaired by hexachlorobenzene treatment, through generation of an enzyme inhibitor. The interaction of porphyrinogen carboxy-lyase inhibitor, extracted from the liver of hexachlorobenzene-treated rats, with substrate decarboxylation sites on the enzyme, was studied using four different carboxylated substrates belonging to the isomeric III series of naturally-formed porphyrinogens containing 8-,7-,6- and 5-COOH. Similar inhibitor effects were elicited against all the substrates assayed, with the exception of pentacarboxyporphyrinogen III in which decarboxylation was not inhibited to same extent. Enzyme protection assays in the presence of the different substrates, indicated that each porphyrinogen protects its own decarboxylation from inhibitor action. Preincubation of the inhibitor with normal enzyme increased its inhibitory effect. On the other hand, preincubation of both enzyme and inhibitor with superoxide dismutase or mannitol, did not alter inhibitory activity. Preincubation of the inhibitor with a number of amino acids showed that only arginine and its derivative N alpha-Benzoyl-L-Arginine ethyl ester interact with the inhibitor, noticeably reducing its ability to inhibit porphyrinogen carboxy-lyase. Albumin, histidine, serine, cysteine and imidazol, were unable to quench inhibitor activity. The present results indicate that the inhibitor acts at the binding site of each porphyrinogen. Taking into account that arginine is related to enzyme activity, and that histidine is found at the binding site of the substrates, the results suggest that the inhibitor could bind to arginine residues, blocking the access of substrates to histidine and altering the adequate orientation for decarboxylation by masking the positively charged active site necessary for porphyrinogen binding to the enzyme. In addition an indirect effect of the inhibitor mediated through free radicals could be discarded.


Asunto(s)
Carboxiliasas/antagonistas & inhibidores , Porfirinógenos/metabolismo , Animales , Arginina/análogos & derivados , Arginina/metabolismo , Arginina/farmacología , Sitios de Unión , Inhibidores Enzimáticos/farmacología , Femenino , Hexaclorobenceno/farmacología , Humanos , Hígado/enzimología , Porfirias/inducido químicamente , Ratas , Ratas Wistar , Uroporfirinógenos/metabolismo
17.
Proc Natl Acad Sci U S A ; 95(9): 4853-8, 1998 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-9560192

RESUMEN

Culture of Desulfovibrio vulgaris in a medium supplemented with 5-aminolevulinic acid and L-methionine-methyl-d3 resulted in the formation of porphyrins (sirohydrochlorin, coproporphyrin III, and protoporphyrin IX) in which the methyl groups at the C-2 and C-7 positions were deuterated. A previously unknown hexacarboxylic acid was also isolated, and its structure was determined to be 12, 18-didecarboxysirohydrochlorin by mass spectrometry and 1H NMR. These results indicate a primitive pathway of heme biosynthesis in D. vulgaris consisting of the following enzymatic steps: (i) methylation of the C-2 and C-7 positions of uroporphyrinogen III to form precorrin-2 (dihydrosirohydrochlorin); (ii) decarboxylation of acetate groups at the C-12 and C-18 positions of precorrin-2 to form 12,18-didecarboxyprecorrin-2; (iii) elimination of acetate groups of the C-2 and C-7 positions of 12,18-didecarboxyprecorrin-2 to form coproporphyrinogen III; and (iv) conversion of coproporphyrinogen III to protoporphyrin IX via protoporphyrinogen IX. We isolated the following three enzymatic activities involved in steps i-iii from the soluble fraction of the cells by anion-exchange chromatography: S-adenosyl-L-methionine:uroporphyrinogen III methyltransferase, precorrin-2 12,18-acetate decarboxylase, and 12, 18-didecarboxyprecorrin-2 2,7-decarboxymethylase; all enzymic products were converted into autooxidized methyl esters and analyzed by thin-layer chromatography, UV-visible (UV-VIS) absorption, and mass spectrometry. The enzymatic reactions in D. vulgaris shed new light on porphyrin biosynthesis at an early stage in the evolution of prokaryotes.


Asunto(s)
Desulfovibrio vulgaris/metabolismo , Porfirinas/biosíntesis , Cromatografía en Capa Delgada , Coproporfirinógenos/metabolismo , Medios de Cultivo , Lactonas/química , Espectrometría de Masas , Peso Molecular , Porfirinas/química , Espectrofotometría , Uroporfirinógenos/metabolismo
18.
Biochem J ; 330 ( Pt 1): 149-53, 1998 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-9461503

RESUMEN

In the present study we have investigated the putative requirement for the cytochrome P-450 isoform CYP1A2 in murine uroporphyria, by comparing Cyp1a2(-/-) knockout mice with Cyp1a2(+/+) wild-type mice. Uroporphyria was produced by injecting animals with iron-dextran and giving the porphyrin precursor 5-aminolaevulinic acid in the drinking water. Some animals also received 3-methylcholanthrene (MC) to induce hepatic CYP1A2. In both protocols, uroporphyria was elicited by these treatments in the Cyp1a2(+/+) wild-type mice, but not in the null mutant mice. Uroporphyrinogen oxidation activity in hepatic microsomes from untreated Cyp1a2(+/+) mice was 2.5-fold higher than in Cyp1a2(-/-) mice. Treatment with MC increased hepatic CYP1A1 in both mouse lines and hepatic CYP1A2 only in the Cyp1a2(+/+) line, as determined by Western immunoblotting. MC increased hepatic ethoxy- and methoxy-resorufin O-dealkylase activities in both mouse lines, but increased uroporphyrinogen oxidation activity in the Cyp1a2(+/+) wild-type mice only. These results indicate the absolute requirement for hepatic CYP1A2 in causing experimental uroporphyria under the conditions used.


Asunto(s)
Ácido Aminolevulínico/farmacología , Citocromo P-450 CYP1A2/fisiología , Hierro/farmacología , Porfirias/inducido químicamente , Uroporfirinas/orina , Animales , Citocromo P-450 CYP1A1/metabolismo , Citocromo P-450 CYP1A2/metabolismo , Sistema Enzimático del Citocromo P-450/metabolismo , Humanos , Hígado/metabolismo , Metilcolantreno/farmacología , Ratones , Ratones Noqueados , Microsomas Hepáticos/metabolismo , Oxidorreductasas/metabolismo , Uroporfirinógenos/metabolismo
19.
Arch Biochem Biophys ; 347(1): 69-77, 1997 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-9344466

RESUMEN

Properties of purified porphobilinogen deaminase (PBG-D; EC 4.3.1.8) from rat harderian gland are here presented. The enzyme behaves as a monomer of Mr 38 +/- 2 kDa and is optimally active at pH 8.0-8.2. Its activation energy, determined by an Arrhenius plot, is 76.1 kJ/mol. Initial velocity studies showed a linear progress curve for uroporphyringen I formation and a hyperbolic dependence of the initial rate on substrate concentration, indicating the existence of a sequential displacement mechanism. Apparent kinetic constants, Km and Vm, calculated at 37 degrees C and pH 8.0 were 1.1 microM and 170 pmol/min mg, respectively. The pH dependence of the apparent kinetic parameters revealed the ionization of residues with pKAES and pKBES of 7.4 +/- 0.1 and 8.6 +/- 0.1, respectively, and a pKE value of 8.0 +/- 0.1. Incubation of PBG-D with 5.0 mM N-ethylmaleimide and 5.0 mM 5,5'-dithiobis(2-nitrobenzoic acid) at pH 8.0 led to inhibitions of 70 and 50%, respectively. The effect of pH, as well as the effect of thiol reagents, on enzyme activity strongly suggests the involvement of cysteine residue(s) in the mechanism of uroporphyrinogen I biosynthesis, in both the catalytic reaction and the substrate binding. Rat harderian gland PBG-D activity decreased with increasing concentrations of protoporphyrin IX, reaching a 40% inhibition at the in vivo concentration of the porphyrin and 7 microM PBG. Even at saturating concentrations of substrate, inhibition by protoporphyrin was not completely reversed. So, accumulated porphyrin may act as an regulator of PBG-D activity in rat harderian gland.


Asunto(s)
Glándula de Harder/enzimología , Hidroximetilbilano Sintasa/metabolismo , Protoporfirinas/farmacología , Animales , Sitios de Unión , Cromatografía Líquida de Alta Presión , Inhibidores Enzimáticos/farmacología , Estabilidad de Enzimas , Etilmaleimida/farmacología , Concentración de Iones de Hidrógeno , Hidroximetilbilano Sintasa/química , Cinética , Masculino , Porfirinas/análisis , Porfirinas/aislamiento & purificación , Ratas , Ratas Endogámicas , Compuestos de Sulfhidrilo/metabolismo , Uroporfirinógenos/biosíntesis
20.
Biochem J ; 316 ( Pt 2): 373-6, 1996 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-8687374

RESUMEN

The assembly process of the dipyrromethane cofactor of Escherichia coli porphobilinogen deaminase holoenzyme is initiated by the reaction of the porphobilinogen deaminase apoenzyme with preuroporphyrinogen. The resulting enzyme-bound tetrapyrrole (bilane) is equivalent to the holoenzyme intermediate complex ES2 and yields the dipyrromethane cofactor by reactions of the normal catalytic cycle. These observations indicate that preuroporphyrinogen, rather than porphobilinogen, is the preferred precursor for the dipyrromethane cofactor and explain the existence of the D84A and D84N deaminase mutants as catalytically inactive ES2 complexes.


Asunto(s)
Coenzimas/metabolismo , Escherichia coli/enzimología , Hidroximetilbilano Sintasa/metabolismo , Porfobilinógeno/metabolismo , Uroporfirinógenos/metabolismo , Apoenzimas/metabolismo , Cisteína/metabolismo , Electroforesis en Gel de Poliacrilamida , Activación Enzimática , Escherichia coli/genética , Expresión Génica , Hidroximetilbilano Sintasa/química , Hidroximetilbilano Sintasa/genética , Estructura Molecular , Mutación/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA