Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 109
Filtrar
1.
Int J Mol Sci ; 24(9)2023 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-37176103

RESUMEN

In autumn and spring, citrus leaves with a Ponkan (Citrus reticulata Blanco cv. Ponkan) genetic background (Harumi, Daya, etc.) are prone to abnormal physiological chlorosis. The effects of different degrees of chlorosis (normal, mild, moderate and severe) on photosynthesis and the chlorophyll metabolism of leaves of Citrus cultivar (Harumi) were studied via field experiment. Compared with severe chlorotic leaves, the results showed that chlorosis could break leaf metabolism balance, including reduced chlorophyll content, photosynthetic parameters, antioxidant enzyme activity and enzyme activity related to chlorophyll synthesis, increased catalase and decreased enzyme activity. In addition, the content of chlorophyll synthesis precursors showed an overall downward trend expected for uroporphyrinogen III. Furthermore, the relative expression of genes for chlorophyll synthesis (HEMA1, HEME2, HEMG1 and CHLH) was down-regulated to some extent and chlorophyll degradation (CAO, CLH, PPH, PAO and SGR) showed the opposite trend with increased chlorosis. Changes in degradation were more significant. In general, the chlorosis of Harumi leaves might be related to the blocked transformation of uroporphyrinogen III (Urogen III) to coproporphyrinogen III (Coprogen III), the weakening of antioxidant enzyme system activity, the weakening of chlorophyll synthesis and the enhancement in degradation.


Asunto(s)
Citrus , Antioxidantes/farmacología , Clorofila/metabolismo , Citrus/genética , Citrus/metabolismo , Fotosíntesis/genética , Hojas de la Planta/metabolismo , Uroporfirinógenos/metabolismo , Uroporfirinógenos/farmacología , Necrosis y Clorosis de las Plantas
2.
Microb Cell Fact ; 22(1): 102, 2023 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-37198628

RESUMEN

BACKGROUND: Heme is an iron/porphyrin complex compound, widely used in the health care, food, and pharmaceutical industries. It is more advantageous and attractive to develop microbial cell factories to produce heme by fermentation, with lower production costs and environmentally more friendly procedures than those of the traditional extraction based on animal blood. In this study, Bacillus subtilis, a typical industrial model microorganism of food safety grade, was used for the first time as the host to synthesize heme. RESULTS: The heme biosynthetic pathway was engineered as four modules, the endogenous C5 pathway, the heterologous C4 pathway, the uroporphyrinogen (urogen) III synthesis pathway, and the downstream synthesis pathway. Knockout of hemX encoding the negative effector of the concentration of HemA, overexpression of hemA encoding glutamyl-tRNA reductase, and knockout of rocG encoding the major glutamate dehydrogenase in the C5 pathway, resulted in an increase of 427% in heme production. Introduction of the heterologous C4 pathway showed a negligible effect on heme biosynthesis. Overexpression of hemCDB, which encoded hydroxymethylbilane synthase, urogen III synthase, and porphobilinogen synthase participating in the urogen III synthesis pathway, increased heme production by 39%. Knockouts of uroporphyrinogen methyltransferase gene nasF and both heme monooxygenase genes hmoA and hmoB in the downstream synthesis pathway increased heme production by 52%. The engineered B. subtilis produced 248.26 ± 6.97 mg/L of total heme with 221.83 ± 4.71 mg/L of extracellular heme during the fed-batch fermentation in 10 L fermenter. CONCLUSIONS: Strengthening endogenous C5 pathway, urogen III synthesis pathway and downstream synthesis pathway promoted the biosynthesis of heme in B. subtilis. The engineered B. subtilis strain has great potential as a microbial cell factory for efficient industrial heme production.


Asunto(s)
Bacillus subtilis , Hemo , Bacillus subtilis/genética , Bacillus subtilis/metabolismo , Hemo/metabolismo , Ingeniería Metabólica/métodos , Fermentación , Uroporfirinógenos/metabolismo
3.
Microbiology (Reading) ; 167(10)2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34661520

RESUMEN

Uroporphyrinogen III, the universal progenitor of macrocyclic, modified tetrapyrroles, is produced from aminolaevulinic acid (ALA) by a conserved pathway involving three enzymes: porphobilinogen synthase (PBGS), hydroxymethylbilane synthase (HmbS) and uroporphyrinogen III synthase (UroS). The gene encoding uroporphyrinogen III synthase has not yet been identified in Plasmodium falciparum, but it has been suggested that this activity is housed inside a bifunctional hybroxymethylbilane synthase (HmbS). Additionally, an unknown protein encoded by PF3D7_1247600 has also been predicted to possess UroS activity. In this study it is demonstrated that neither of these proteins possess UroS activity and the real UroS remains to be identified. This was demonstrated by the failure of codon-optimized genes to complement a defined Escherichia coli hemD- mutant (SASZ31) deficient in UroS activity. Furthermore, HPLC analysis of the oxidized reaction product from recombinant, purified P. falciparum HmbS showed that only uroporphyrin I could be detected (corresponding to hydroxymethylbilane production). No uroporphyrin III was detected, showing that P. falciparum HmbS does not have UroS activity and can only catalyze the formation of hydroxymethylbilane from porphobilinogen.


Asunto(s)
Hemo/biosíntesis , Hidroximetilbilano Sintasa/metabolismo , Plasmodium falciparum/enzimología , Vías Biosintéticas , Escherichia coli/genética , Prueba de Complementación Genética , Hidroximetilbilano Sintasa/genética , Mutación , Plasmodium falciparum/genética , Proteínas Protozoarias/química , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo , Uroporfirinógeno III Sintetasa/genética , Uroporfirinógeno III Sintetasa/metabolismo , Uroporfirinógenos/metabolismo
4.
Biochem J ; 478(5): 1023-1042, 2021 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-33600566

RESUMEN

Hydroxymethylbilane synthase (HMBS), which is involved in the heme biosynthesis pathway, has a dipyrromethane cofactor and combines four porphobilinogen (PBG) molecules to form a linear tetrapyrrole, hydroxymethylbilane. Enzyme kinetic study of human HMBS using a PBG-derivative, 2-iodoporphobilinogen (2-I-PBG), exhibited noncompetitive inhibition with the inhibition constant being 5.4 ± 0.3 µM. To elucidate the reaction mechanism of HMBS in detail, crystal structure analysis of 2-I-PBG-bound holo-HMBS and its reaction intermediate possessing two PBG molecules (ES2), and inhibitor-free ES2 was performed at 2.40, 2.31, and 1.79 Šresolution, respectively. Their overall structures are similar to that of inhibitor-free holo-HMBS, and the differences are limited near the active site. In both 2-I-PBG-bound structures, 2-I-PBG is located near the terminus of the cofactor or the tetrapyrrole chain. The propionate group of 2-I-PBG interacts with the side chain of Arg173, and its acetate group is associated with the side chains of Arg26 and Ser28. Furthermore, the aminomethyl group and pyrrole nitrogen of 2-I-PBG form hydrogen bonds with the side chains of Gln34 and Asp99, respectively. These amino acid residues form a single substrate-binding site, where each of the four PBG molecules covalently binds to the cofactor (or oligopyrrole chain) consecutively, ultimately forming a hexapyrrole chain. Molecular dynamics simulation of the ES2 intermediate suggested that the thermal fluctuation of the lid and cofactor-binding loops causes substrate recruitment and oligopyrrole chain shift needed for consecutive condensation. Finally, the hexapyrrole chain is hydrolyzed self-catalytically to produce hydroxymethylbilane.


Asunto(s)
Hidroximetilbilano Sintasa/química , Hidroximetilbilano Sintasa/metabolismo , Porfobilinógeno/metabolismo , Uroporfirinógenos/metabolismo , Catálisis , Dominio Catalítico , Cristalografía por Rayos X , Humanos , Simulación de Dinámica Molecular , Conformación Proteica , Dominios Proteicos , Especificidad por Sustrato
5.
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
6.
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
7.
Archaea ; 2014: 327637, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24669201

RESUMEN

In living organisms heme is formed from the common precursor uroporphyrinogen III by either one of two substantially different pathways. In contrast to eukaryotes and most bacteria which employ the so-called "classical" heme biosynthesis pathway, the archaea use an alternative route. In this pathway, heme is formed from uroporphyrinogen III via the intermediates precorrin-2, sirohydrochlorin, siroheme, 12,18-didecarboxysiroheme, and iron-coproporphyrin III. In this study the heme biosynthesis proteins AhbAB, AhbC, and AhbD from Methanosarcina barkeri were functionally characterized. Using an in vivo enzyme activity assay it was shown that AhbA and AhbB (Mbar_A1459 and Mbar_A1460) together catalyze the conversion of siroheme into 12,18-didecarboxysiroheme. The two proteins form a heterodimeric complex which might be subject to feedback regulation by the pathway end-product heme. Further, AhbC (Mbar_A1793) was shown to catalyze the formation of iron-coproporphyrin III in vivo. Finally, recombinant AhbD (Mbar_A1458) was produced in E. coli and purified indicating that this protein most likely contains two [4Fe-4S] clusters. Using an in vitro enzyme activity assay it was demonstrated that AhbD catalyzes the conversion of iron-coproporphyrin III into heme.


Asunto(s)
Proteínas Arqueales/genética , Proteínas Arqueales/metabolismo , Vías Biosintéticas/genética , Hemo/biosíntesis , Methanosarcina barkeri/genética , Methanosarcina barkeri/metabolismo , Methanosarcina barkeri/enzimología , Multimerización de Proteína , Uroporfirinógenos/metabolismo
8.
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
9.
Appl Microbiol Biotechnol ; 97(16): 7337-44, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23604563

RESUMEN

Uroporphyrinogen III (urogen III) was produced from 5-aminolevulinic acid (ALA), which is a common precursor of all metabolic tetrapyrroles, using thermostable ALA dehydratase (ALAD), porphobilinogen deaminase (PBGD), and urogen III synthase (UROS) of Thermus thermophilus HB8. The UROS-coding gene (hemD2) of T. thermophilus HB8 was identified by examining the gene product for its ability to produce urogen III in a coupled reaction with ALAD and PBGD. The genes encoding ALAD, PBGD, and UROS were separately expressed in Escherichia coli BL21 (DE3). To inactivate indigenous mesophilic enzymes, the E. coli transformants were heated at 70 °C for 10 min. The bioconversion of ALA to urogen III was performed using a mixture of heat-treated E. coli transformants expressing ALAD, PBGD, and UROS at a cell ratio of 1:1:1. When the total cell concentration was 7.5 g/l, the mixture of heat-treated E. coli transformants could convert about 88 % 10 mM ALA to urogen III at 60 °C after 4 h. Since eight ALA molecules are required for the synthesis of one porphyrin molecule, approximately 1.1 mM (990 mg/l) urogen III was produced from 10 mM ALA. The present technology has great potential to supply urogen III for the biocatalytic production of vitamin B12.


Asunto(s)
Ácido Aminolevulínico/metabolismo , Escherichia coli/metabolismo , Thermus thermophilus/enzimología , Uroporfirinógenos/metabolismo , Biotecnología/métodos , Escherichia coli/genética , Calor , Hidroximetilbilano Sintasa/genética , Ingeniería Metabólica/métodos , Redes y Vías Metabólicas/genética , Porfobilinógeno Sintasa/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Thermus thermophilus/genética , Uroporfirinógeno III Sintetasa/genética
10.
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
11.
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
12.
Blood ; 118(6): 1443-51, 2011 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-21653323

RESUMEN

Mutations in the uroporphyrinogen III synthase (UROS) gene cause congenital erythropoietic porphyria (CEP), an autosomal-recessive inborn error of erythroid heme biosynthesis. Clinical features of CEP include dermatologic and hematologic abnormalities of variable severity. The discovery of a new type of erythroid porphyria, X-linked dominant protoporphyria (XLDPP), which results from increased activity of 5-aminolevulinate synthase 2 (ALAS2), the rate-controlling enzyme of erythroid heme synthesis, led us to hypothesize that the CEP phenotype may be modulated by sequence variations in the ALAS2 gene. We genotyped ALAS2 in 4 unrelated CEP patients exhibiting the same C73R/P248Q UROS genotype. The most severe of the CEP patients, a young girl, proved to be heterozygous for a novel ALAS2 mutation: c.1757 A > T in exon 11. This mutation is predicted to affect the highly conserved and penultimate C-terminal amino acid of ALAS2 (Y586). The rate of 5-aminolevulinate release from Y586F was significantly increased over that of wild-type ALAS2. The contribution of the ALAS2 gain-of-function mutation to the CEP phenotype underscores the importance of modifier genes underlying CEP. We propose that ALAS2 gene mutations should be considered not only as causative of X-linked sideroblastic anemia (XLSA) and XLDPP but may also modulate gene function in other erythropoietic disorders.


Asunto(s)
5-Aminolevulinato Sintetasa/genética , Mutación Missense , Porfiria Eritropoyética/genética , Uroporfirinógeno III Sintetasa/genética , 5-Aminolevulinato Sintetasa/metabolismo , Secuencia de Aminoácidos , Anemia Sideroblástica/genética , Anemia Sideroblástica/metabolismo , Anemia Sideroblástica/patología , Secuencia de Bases , Preescolar , Electroforesis en Gel de Poliacrilamida , Salud de la Familia , Femenino , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Enfermedades Genéticas Ligadas al Cromosoma X/metabolismo , Genotipo , Humanos , Lactante , Cinética , Masculino , Datos de Secuencia Molecular , Linaje , Porfiria Eritropoyética/metabolismo , Porfiria Eritropoyética/patología , Protoporfiria Eritropoyética/genética , Protoporfiria Eritropoyética/metabolismo , Homología de Secuencia de Aminoácido , Índice de Severidad de la Enfermedad , Espectrofotometría , Uroporfirinógeno III Sintetasa/metabolismo , Uroporfirinógenos/metabolismo
13.
J Phys Chem B ; 114(27): 8994-9001, 2010 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-20553007

RESUMEN

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.


Asunto(s)
Arginina , Compuestos Onio , Uroporfirinógeno Descarboxilasa/química , Uroporfirinógeno Descarboxilasa/metabolismo , Dominio Catalítico , Humanos , Simulación de Dinámica Molecular , Mutagénesis Sitio-Dirigida , Protones , Uroporfirinógeno Descarboxilasa/genética , Uroporfirinógenos/metabolismo
14.
FEBS J ; 276(21): 6399-411, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19796169

RESUMEN

The synthesis of the modified tetrapyrrole known as d(1) haem requires several dedicated proteins which are coded for by a set of genes that are often found adjacent to the structural gene, nirS, for cytochrome cd(1) nitrite reductase. NirE, the product of the first gene in the nir biogenesis operon, was anticipated to catalyse the conversion of uroporphyrinogen III into precorrin-2; this was confirmed, but it was shown that this enzyme is less sensitive to product inhibition than similar enzymes that function in other biosynthetic pathways. Sequence analysis suggesting that one of these proteins, NirN, is a c-type cytochrome, and has similarity to the part of cytochrome cd(1) that binds d(1), was validated by recombinant production and characterization of NirN. A NirN-d(1) haem complex was demonstrated to release the cofactor to a semi-apo form of cytochrome cd(1) from which d(1) was extracted, suggesting a role for NirN in the assembly of cytochrome cd(1) (NirS). However, inactivation of nirN surprisingly led to only a marginal attenuation of growth of Paracoccus pantotrophus under anaerobic denitrifying conditions. As predicted, NirC is a c-type cytochrome; it was shown in vitro to be an electron donor to the NirN-d(1) complex.


Asunto(s)
Bacterias/metabolismo , Hemo/biosíntesis , Nitrito Reductasas/fisiología , Proteínas de Transporte de Anión/fisiología , Citocromos/fisiología , Proteínas de Escherichia coli/fisiología , Hemo/análogos & derivados , Paracoccus pantotrophus/genética , Paracoccus pantotrophus/crecimiento & desarrollo , Uroporfirinógenos/metabolismo , Uroporfirinas/biosíntesis
15.
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
16.
Cell Mol Biol (Noisy-le-grand) ; 55(2): 46-54, 2009 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-19656451

RESUMEN

Most rodent models of porphyria cutanea tarda (PCT) share in common the administration of iron and agents that induce transcription of cytochrome P450s. Dissection of changes related to porphyrin accumulation required generation of a genetic model free from exogenous precipitants. Mice heterozygous for a null Urod mutation and homozygous for null Hfe alleles spontaneously develop major increases in hepatic and urinary porphyrins several months after weaning but the high % uroporphyrin signature of PCT is established earlier, before total hepatic and urinary porphyrins rise. Total porphyrin levels eventually plateau at higher levels in females than in males. Porphyrinogens were the dominant tetrapyrroles accumulating in hepatocytes. Hepatic Urod activity is markedly reduced but total hepatic heme content does not diminish. Microsomal heme, however, is reduced and in vitro metabolism of prototype substrates showed that some but not all cytochrome P450 activities are reduced. High hepatic levels of uroporphyrinogen are also associated with increased glutathione S-transferase activity and elevated mRNA of 2 transporters, Abcc1 and Abcc4. This murine model of familial PCT affords the opportunity to study changes in porphyrinogen and porphyrin accumulation and transport in the absence of exogenous factors that alter P450 activity and transmembrane transporters.


Asunto(s)
Porfiria Cutánea Tardía/genética , Animales , Sistema Enzimático del Citocromo P-450/metabolismo , Modelos Animales de Enfermedad , Femenino , Genotipo , Glutatión Transferasa/metabolismo , Hemo/análisis , Proteína de la Hemocromatosis , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/metabolismo , Hígado/metabolismo , Estudios Longitudinales , Masculino , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/antagonistas & inhibidores , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Porfiria Cutánea Tardía/patología , Porfirinas/sangre , Porfirinas/orina , Uroporfirinógeno Descarboxilasa/deficiencia , Uroporfirinógeno Descarboxilasa/genética , Uroporfirinógeno Descarboxilasa/metabolismo , Uroporfirinógenos/metabolismo
17.
J Mol Biol ; 389(2): 306-14, 2009 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-19362562

RESUMEN

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.


Asunto(s)
Coproporfirinógenos/biosíntesis , Uroporfirinógeno Descarboxilasa/metabolismo , Dominio Catalítico , Cristalografía por Rayos X , Conformación Proteica , Multimerización de Proteína , Subunidades de Proteína , Uroporfirinógeno Descarboxilasa/química , Uroporfirinógenos/metabolismo
18.
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
19.
Bioorg Chem ; 36(5): 241-51, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18760440

RESUMEN

The structurally related tetrapyrrolic pigments are a group of natural products that participate in many of the fundamental biosynthetic and catabolic processes of living organisms. Hydroxymethylbilane synthase catalyzes a rate-limiting step for the biosyntheses of tetrapyrrolic natural products. We carried out extensive studies of rat hydroxymethylbilane synthase in the present investigation. The enzymatic reaction rate of the holoenzyme was found to be lower than those of the enzyme-intermediate complexes, which corrected the previous theoretical analysis result. Several mutants were constructed, purified and characterized. D44 was found to play an important role in the disassembly of the enzyme-intermediate complexes. E63 and H78 were important for maintaining the activity of the enzyme at high temperature. Four substrate analogs with variation of porphobilinogen side-chain were synthesized and incubated with the enzyme. Three analogs were found to be weak substrates of the enzyme. All four analogs can be used for the preparation of uroporphyrin I analogs.


Asunto(s)
Histidina/química , Hidroximetilbilano Sintasa/metabolismo , Hígado/enzimología , Porfobilinógeno/química , Secuencia de Aminoácidos , Animales , Cromatografía de Afinidad , Clonación Molecular , Estabilidad de Enzimas , Biblioteca de Genes , Concentración de Iones de Hidrógeno , Hidroximetilbilano Sintasa/genética , Hidroximetilbilano Sintasa/aislamiento & purificación , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Ratas , Homología de Secuencia de Aminoácido , Especificidad por Sustrato , Temperatura , Uroporfirinógenos/metabolismo
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA