Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters










Publication year range
1.
Nat Commun ; 11(1): 864, 2020 02 13.
Article in English | MEDLINE | ID: mdl-32054833

ABSTRACT

Siroheme is the central cofactor in a conserved class of sulfite and nitrite reductases that catalyze the six-electron reduction of sulfite to sulfide and nitrite to ammonia. In Salmonella enterica serovar Typhimurium, siroheme is produced by a trifunctional enzyme, siroheme synthase (CysG). A bifunctional active site that is distinct from its methyltransferase activity catalyzes the final two steps, NAD+-dependent dehydrogenation and iron chelation. How this active site performs such different chemistries is unknown. Here, we report the structures of CysG bound to precorrin-2, the initial substrate; sirohydrochlorin, the dehydrogenation product/chelation substrate; and a cobalt-sirohydrochlorin product. We identified binding poses for all three tetrapyrroles and tested the roles of specific amino acids in both activities to give insights into how a bifunctional active site catalyzes two different chemistries and acts as an iron-specific chelatase in the final step of siroheme synthesis.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Heme/analogs & derivatives , Methyltransferases/chemistry , Methyltransferases/metabolism , Amino Acid Substitution , Bacterial Proteins/genetics , Catalytic Domain/genetics , Electrochemistry , Ferrochelatase/chemistry , Ferrochelatase/genetics , Ferrochelatase/metabolism , Heme/biosynthesis , Heme/chemistry , Methyltransferases/genetics , Models, Molecular , Mutagenesis, Site-Directed , Oxidoreductases/chemistry , Oxidoreductases/genetics , Oxidoreductases/metabolism , Salmonella typhimurium/genetics , Salmonella typhimurium/metabolism , Substrate Specificity , Tetrapyrroles/chemistry , Tetrapyrroles/metabolism , Uroporphyrins/chemistry , Uroporphyrins/metabolism
2.
Angew Chem Int Ed Engl ; 58(31): 10756-10760, 2019 07 29.
Article in English | MEDLINE | ID: mdl-31115943

ABSTRACT

The B12 cofactors instill a natural curiosity regarding the primordial selection and evolution of their corrin ligand. Surprisingly, this important natural macrocycle has evaded molecular scrutiny, and its specific role in predisposing the incarcerated cobalt ion for organometallic catalysis has remained obscure. Herein, we report the biosynthesis of the cobalt-free B12 corrin moiety, hydrogenobyric acid (Hby), a compound crafted through pathway redesign. Detailed insights from single-crystal X-ray and solution structures of Hby have revealed a distorted helical cavity, redefining the pattern for binding cobalt ions. Consequently, the corrin ligand coordinates cobalt ions in desymmetrized "entatic" states, thereby promoting the activation of B12 -cofactors for their challenging chemical transitions. The availability of Hby also provides a route to the synthesis of transition metal analogues of B12 .


Subject(s)
Corrinoids/biosynthesis , Uroporphyrins/metabolism , Vitamin B 12/metabolism , Biocatalysis , Cobalt/chemistry , Cobalt/metabolism , Corrinoids/chemistry , Ligands , Molecular Structure , Uroporphyrins/chemistry , Vitamin B 12/chemistry
3.
Dalton Trans ; 48(18): 6083-6090, 2019 May 07.
Article in English | MEDLINE | ID: mdl-30778451

ABSTRACT

The crystal structure of Bacillus subtilis SirB, which catalyses the insertion of Fe2+ into the substrate sirohydrochlorin (SHC) in siroheme biosynthesis, is reported herein as the last of the structures of class II chelatases. The structure of SirB with Co2+ showed that the active site of SirB is located at the N-terminal domain with metal-binding amino acid residues His10, Glu43, and His76, which was also predicted for CbiX, but is distinct from the C-terminal active sites of CbiK and HemH. The biosynthetic model reactions using SirB, Co2+ and uroporphyrin I or protoporphyrin IX as a SHC analogue revealed that SirB showed chelatase activity for uroporphyrin I, but not for protoporphyrin IX. Simulations of tetrapyrroles docking to SirB provided an insight into its tetrapyrrole substrate recognition: SHC and uroporphyrin I were suitably bound beside the Co2+ ion-binding site at the active site cavity; protoporphyrin IX was also docked to the active site but its orientation was different from those of the other two tetrapyrroles. Summarizing the present data, it was proposed that the key structural features for substrate recognition of SirB could be the hydrophobic area at the active site as well as the substituents of the tetrapyrroles.


Subject(s)
Bacterial Proteins/chemistry , Cobalt/chemistry , Ferrochelatase/chemistry , Uroporphyrins/chemistry , Amino Acids/chemistry , Bacillus subtilis/metabolism , Biosynthetic Pathways , Catalytic Domain , Crystallization , Hydrophobic and Hydrophilic Interactions , Molecular Docking Simulation , Protein Binding , Protein Conformation , Tetrapyrroles/chemistry
4.
Methods Mol Biol ; 1876: 125-140, 2019.
Article in English | MEDLINE | ID: mdl-30317478

ABSTRACT

Nitrogenase-like enzymes play a vital role in the reduction of the conjugated ring systems of diverse tetrapyrrole molecules. The biosynthesis of all bacteriochlorophylls involves the two-electron reduction of the C7-C8 double bond of the green pigment chlorophyllide, which is catalyzed by the nitrogenase-like two-component metalloenzyme chlorophyllide oxidoreductase (COR); whereas in all methanogenic microbes, another nitrogenase-like system, CfbC/D, is responsible for the sophisticated six-electron reduction of Ni2+-sirohydrochlorin a,c-diamide in the course of coenzyme F430 biosynthesis. The first part of this chapter describes the production and purification of the COR components (BchY/BchZ)2 and BchX2, the measurement of COR activity, and the trapping of the ternary COR complex; and the second part describes the strategy for obtaining homogenous and catalytically active preparations of CfbC2 and CfbD2 and a suitable method for extracting the reaction product Ni2+-hexahydrosirohydrochlorin a,c-diamide.


Subject(s)
Metalloproteins/isolation & purification , Metalloproteins/metabolism , Uroporphyrins/chemistry , Alcohol Oxidoreductases/chemistry , Alcohol Oxidoreductases/isolation & purification , Alcohol Oxidoreductases/metabolism , Catalytic Domain , Chlorophyll/biosynthesis , Metalloporphyrins/metabolism , Metalloproteins/chemistry , Multienzyme Complexes , Nickel/chemistry , Oxidation-Reduction
5.
Pediatr Res ; 84(1): 80-84, 2018 07.
Article in English | MEDLINE | ID: mdl-29795201

ABSTRACT

BACKGROUND: Heme is the prosthetic group of numerous proteins involved in vital processes such as oxygen transport, oxidative stress, and energetic mitochondrial metabolism. Free heme also plays a significant role at early stages of development and in cell differentiation processes. The metabolism of heme by the fetal placenta unit is not well-established in humans. METHODS: In a retrospective study, we measured heme precursors in the amniotic fluid (AF) of 51 healthy women, and 10 AF samples from pregnancies with either upper or lower intestinal atresia or ileus were also analyzed. RESULTS: We showed that the porphyrin precursors aminolevulinic acid, porphobilinogen, and protoporphyrin IX are present at the limit of detection in the AF. Total porphyrin levels decreased progressively from week 13 to week 33 (p < 0.01). Interestingly, uroporphyrin, initially detected as traces, increased with maturation, in contrast to coproporphyrin. Uro- and coproporphyrins were type I immature isomers (>90%), suggesting a lack of maturity in the fetal compartment of the heme pathway. Finally, the differential analysis of AF from normal and pathological pregnancies demonstrated the predominant hepatic origin of fetal porphyrins excreted in the AF. CONCLUSION: This study gives the first insight into heme metabolism in the AF during normal and pathological pregnancies.


Subject(s)
Amniotic Fluid/chemistry , Heme/chemistry , Intestinal Atresia/metabolism , Prenatal Diagnosis/methods , Adult , Amniocentesis/methods , Cell Differentiation , Coproporphyrins/chemistry , Female , Humans , Ileus/pathology , Intestinal Atresia/pathology , Karyotyping , Maternal Age , Mitochondria/metabolism , Oxidative Stress , Oxygen/metabolism , Placenta/metabolism , Porphyrias/diagnosis , Pregnancy , Protoporphyrins/chemistry , Retrospective Studies , Uroporphyrins/chemistry
6.
Nature ; 543(7643): 78-82, 2017 03 02.
Article in English | MEDLINE | ID: mdl-28225763

ABSTRACT

Methane biogenesis in methanogens is mediated by methyl-coenzyme M reductase, an enzyme that is also responsible for the utilization of methane through anaerobic methane oxidation. The enzyme uses an ancillary factor called coenzyme F430, a nickel-containing modified tetrapyrrole that promotes catalysis through a methyl radical/Ni(ii)-thiolate intermediate. However, it is unclear how coenzyme F430 is synthesized from the common primogenitor uroporphyrinogen iii, incorporating 11 steric centres into the macrocycle, although the pathway must involve chelation, amidation, macrocyclic ring reduction, lactamization and carbocyclic ring formation. Here we identify the proteins that catalyse the biosynthesis of coenzyme F430 from sirohydrochlorin, termed CfbA-CfbE, and demonstrate their activity. The research completes our understanding of how the repertoire of tetrapyrrole-based pigments are constructed, permitting the development of recombinant systems to use these metalloprosthetic groups more widely.


Subject(s)
Biocatalysis , Biosynthetic Pathways , Coenzymes/biosynthesis , Metalloporphyrins/metabolism , Methane/biosynthesis , Methanosarcina barkeri/enzymology , Tetrapyrroles/biosynthesis , Amidohydrolases/genetics , Amidohydrolases/metabolism , Biosynthetic Pathways/genetics , Coenzymes/chemistry , Lyases/genetics , Lyases/metabolism , Metalloporphyrins/chemistry , Methane/analogs & derivatives , Methane/metabolism , Methanosarcina barkeri/genetics , Methanosarcina barkeri/metabolism , Multigene Family , Nickel/metabolism , Oxidoreductases/genetics , Oxidoreductases/metabolism , Tetrapyrroles/chemistry , Uroporphyrins/chemistry , Uroporphyrins/metabolism
7.
FEMS Microbiol Lett ; 363(19)2016 10.
Article in English | MEDLINE | ID: mdl-27609233

ABSTRACT

Upon transition of Mycobacterium smegmatis into the dormant state, accumulation of a dark brown fluorescent pigment was observed. This pigment gave bright red fluorescence in both cells and the culture medium. Based on 1H-NMR, MALDI and UV spectra, the fluorescent compounds, extracted from the culture medium as well as from the dormant cells, were concluded to be a mixture of free coproporphyrin III and uroporphyrin III and their corresponding methyl esters. A possible significance of porphyrin pigment accumulation in the dormant cells is discussed.


Subject(s)
Mycobacterium smegmatis/chemistry , Pigments, Biological/chemistry , Pigments, Biological/isolation & purification , Porphyrins/chemistry , Coproporphyrins/chemistry , Coproporphyrins/isolation & purification , Culture Media/chemistry , Fluorescence , Mycobacterium smegmatis/physiology , Porphyrins/isolation & purification , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Uroporphyrins/chemistry , Uroporphyrins/isolation & purification
8.
PLoS One ; 11(3): e0151149, 2016.
Article in English | MEDLINE | ID: mdl-26974652

ABSTRACT

In order to maximize the production of biologically-derived chemicals, kinetic analyses are first necessary for predicting the role of enzyme components and coordinating enzymes in the same reaction system. Precorrin-2 is a key precursor of cobalamin and siroheme synthesis. In this study, we sought to optimize the concentrations of several molecules involved in precorrin-2 synthesis in vitro: porphobilinogen synthase (PBGS), porphobilinogen deaminase (PBGD), uroporphyrinogen III synthase (UROS), and S-adenosyl-l-methionine-dependent urogen III methyltransferase (SUMT). Response surface methodology was applied to develop a kinetic model designed to maximize precorrin-2 productivity. The optimal molar ratios of PBGS, PBGD, UROS, and SUMT were found to be approximately 1:7:7:34, respectively. Maximum precorrin-2 production was achieved at 0.1966 ± 0.0028 µM/min, agreeing with the kinetic model's predicted value of 0.1950 µM/min. The optimal concentrations of the cofactor S-adenosyl-L-methionine (SAM) and substrate 5-aminolevulinic acid (ALA) were also determined to be 200 µM and 5 mM, respectively, in a tandem-enzyme assay. By optimizing the relative concentrations of these enzymes, we were able to minimize the effects of substrate inhibition and feedback inhibition by S-adenosylhomocysteine on SUMT and thereby increase the production of precorrin-2 by approximately five-fold. These results demonstrate the effectiveness of kinetic modeling via response surface methodology for maximizing the production of biologically-derived chemicals.


Subject(s)
Bacillus megaterium/enzymology , Bacterial Proteins/chemistry , Enzymes/chemistry , Pseudomonas/enzymology , Uroporphyrins/chemical synthesis , Bacillus megaterium/genetics , Bacterial Proteins/genetics , Enzymes/genetics , Pseudomonas/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Uroporphyrins/chemistry
9.
Sci Rep ; 5: 16943, 2015 Nov 30.
Article in English | MEDLINE | ID: mdl-26616290

ABSTRACT

CobK catalyzes the essential reduction of the precorrin ring in the cobalamin biosynthetic pathway. The crystal structure of CobK reveals that the enzyme, despite not having the signature sequence, comprises two Rossmann fold domains which bind coenzyme and substrate respectively. The two parallel ß-sheets have swapped their last ß-strands giving a novel sheet topology which is an interesting variation on the Rossmann-fold. The trapped ternary complex with coenzyme and product reveals five conserved basic residues that bind the carboxylates of the tetrapyrrole tightly anchoring the product. A loop, disordered in both the apoenzyme and holoenzyme structures, closes around the product further tightening binding. The structure is consistent with a mechanism involving protonation of C18 and pro-R hydride transfer from NADPH to C19 of precorrin-6A and reveals the interactions responsible for the specificity of CobK. The almost complete burial of the reduced precorrin product suggests a remarkable form of metabolite channeling where the next enzyme in the biosynthetic pathway triggers product release.


Subject(s)
Models, Molecular , Oxidoreductases/chemistry , Protein Conformation , Protein Interaction Domains and Motifs , Uroporphyrins/chemistry , Binding Sites , Catalysis , Coenzymes/chemistry , Coenzymes/metabolism , Crystallography, X-Ray , Holoenzymes/chemistry , Holoenzymes/metabolism , Hydrogen Bonding , Molecular Docking Simulation , NADP/chemistry , NADP/metabolism , Oxidoreductases/metabolism , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Structure-Activity Relationship , Substrate Specificity , Uroporphyrins/metabolism
10.
J Mol Biol ; 426(19): 3272-3286, 2014 Sep 23.
Article in English | MEDLINE | ID: mdl-25083922

ABSTRACT

The isobacteriochlorin heme d1 serves as an essential cofactor in the cytochrome cd1 nitrite reductase NirS that plays an important role for denitrification. During the biosynthesis of heme d1, the enzyme siroheme decarboxylase catalyzes the conversion of siroheme to 12,18-didecarboxysiroheme. This enzyme was discovered recently (Bali S, Lawrence AD, Lobo SA, Saraiva LM, Golding BT, Palmer DJ et al. Molecular hijacking of siroheme for the synthesis of heme and d1 heme. Proc Natl Acad Sci USA 2011;108:18260-5) and is only scarcely characterized. Here, we present the crystal structure of the siroheme decarboxylase from Hydrogenobacter thermophilus representing the first three-dimensional structure for this type of enzyme. The overall structure strikingly resembles those of transcriptional regulators of the Lrp/AsnC family. Moreover, the structure of the enzyme in complex with a substrate analog reveals first insights into its active-site architecture. Through site-directed mutagenesis and subsequent biochemical characterization of the enzyme variants, two conserved histidine residues within the active site are identified to be involved in substrate binding and catalysis. Based on our results, we propose a potential catalytic mechanism for the enzymatic reaction catalyzed by the siroheme decarboxylase.


Subject(s)
Bacteria/enzymology , Bacterial Proteins/chemistry , Carboxy-Lyases/chemistry , Histidine/chemistry , Iron/chemistry , Uroporphyrins/chemistry , Amino Acid Sequence , Bacterial Proteins/genetics , Bacterial Proteins/ultrastructure , Binding Sites , Carboxy-Lyases/ultrastructure , Catalytic Domain , Decarboxylation , Heme/analogs & derivatives , Heme/biosynthesis , Heme/chemistry , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Binding , Sequence Alignment
11.
BMC Struct Biol ; 13: 10, 2013 May 20.
Article in English | MEDLINE | ID: mdl-23688113

ABSTRACT

BACKGROUND: In the anaerobic pathway of cobalamin (vitamin B12) synthesis, the CbiT enzyme plays two roles, as a cobalt-precorrin-7 C15-methyltransferase and a C12-decarboxylase, to produce the intermediate, cobalt-precorrin 8. RESULTS: The primary structure of the hypothetical protein MJ0391, from Methanocaldococcus jannaschii, suggested that MJ0391 is a putative CbiT. Here, we report the crystal structure of MJ0391, solved by the MAD procedure and refined to final R-factor and R-free values of 19.8 & 27.3%, respectively, at 2.3 Å resolution. The asymmetric unit contains two NCS molecules, and the intact tetramer generated by crystallographic symmetry may be functionally important. The overall tertiary structure and the tetrameric arrangements are highly homologous to those found in MT0146/CbiT from Methanobacterium thermoautotrophicum. CONCLUSIONS: The conservation of functional residues in the binding site for the co-factor, AdoMet, and in the putative precorrin-7 binding pocket suggested that MJ0391 may also possess CbiT activity. The putative function of MJ0391 is discussed, based on structural homology.


Subject(s)
Archaeal Proteins/chemistry , Methanocaldococcus/enzymology , Methyltransferases/chemistry , Vitamin B 12/biosynthesis , Amino Acid Sequence , Archaeal Proteins/metabolism , Binding Sites , Crystallography, X-Ray , Methyltransferases/metabolism , Molecular Sequence Data , Protein Structure, Quaternary , Protein Structure, Tertiary , S-Adenosylmethionine/chemistry , S-Adenosylmethionine/metabolism , Sequence Alignment , Uroporphyrins/chemistry , Uroporphyrins/metabolism
12.
Nat Chem Biol ; 8(11): 933-40, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23042036

ABSTRACT

The biosynthesis of many vitamins and coenzymes has often proven difficult to elucidate owing to a combination of low abundance and kinetic lability of the pathway intermediates. Through a serial reconstruction of the cobalamin (vitamin B(12)) pathway in Escherichia coli and by His tagging the terminal enzyme in the reaction sequence, we have observed that many unstable intermediates can be isolated as tightly bound enzyme-product complexes. Together, these approaches have been used to extract intermediates between precorrin-4 and hydrogenobyrinic acid in their free acid form and permitted the delineation of the overall reaction catalyzed by CobL, including the formal elucidation of precorrin-7 as a metabolite. Furthermore, a substrate-carrier protein, CobE, that can also be used to stabilize some of the transient metabolic intermediates and enhance their onward transformation, has been identified. The tight association of pathway intermediates with enzymes provides evidence for a form of metabolite channeling.


Subject(s)
Methyltransferases/metabolism , Vitamin B 12/biosynthesis , Biocatalysis , Escherichia coli/enzymology , Escherichia coli/metabolism , Methyltransferases/chemistry , Models, Molecular , Molecular Structure , Uroporphyrins/chemistry , Uroporphyrins/isolation & purification , Uroporphyrins/metabolism , Vitamin B 12/chemistry , Vitamin B 12/metabolism
13.
Biochem J ; 444(2): 227-37, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22414210

ABSTRACT

Sirohaem is a cofactor of nitrite and sulfite reductases, essential for assimilation of nitrogen and sulfur. Sirohaem is synthesized from the central tetrapyrrole intermediate uroporphyrinogen III by methylation, oxidation and ferrochelation reactions. In Arabidopsis thaliana, the ferrochelation step is catalysed by sirohydrochlorin ferrochelatase (SirB), which, unlike its counterparts in bacteria, contains an [Fe-S] cluster. We determined the cluster to be a [4Fe-4S] type, which quickly oxidizes to a [2Fe-2S] form in the presence of oxygen. We also identified the cluster ligands as four conserved cysteine residues located at the C-terminus. A fifth conserved cysteine residue, Cys(135), is not involved in ligating the cluster directly, but influences the oxygen-sensitivity of the [4Fe-4S] form, and possibly the affinity for the substrate metal. Substitution mutants of the enzyme lacking the Fe-S cluster or Cys(135) retain the same specific activity in vitro and dimeric quaternary structure as the wild-type enzyme. The mutant variants also rescue a defined Escherichia coli sirohaem-deficient mutant. However, the mutant enzymes cannot complement Arabidopsis plants with a null AtSirB mutation, which exhibits post-germination arrest. These observations suggest an important physiological role for the Fe-S cluster in Planta, highlighting the close association of iron, sulfur and tetrapyrrole metabolism.


Subject(s)
Arabidopsis/enzymology , Evolution, Molecular , Ferrochelatase/chemistry , Iron-Sulfur Proteins/chemistry , Uroporphyrins/chemistry , Amino Acid Sequence , Arabidopsis/genetics , Catalysis , Conserved Sequence , Iron-Sulfur Proteins/genetics , Molecular Sequence Data , Mutation , Plant Extracts/chemistry , Plant Extracts/genetics , Plant Leaves/enzymology , Plant Leaves/genetics , Uroporphyrins/genetics
14.
Biometals ; 22(2): 345-51, 2009 Apr.
Article in English | MEDLINE | ID: mdl-18956144

ABSTRACT

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.


Subject(s)
Aminolevulinic Acid/pharmacology , Iron/metabolism , Methylcholanthrene/pharmacology , Porphyrias/chemically induced , Uroporphyrins/metabolism , Animals , Cytochrome P-450 CYP1A2/genetics , Iron/chemistry , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxygen/chemistry , Porphyrias/metabolism , Time Factors , Uroporphyrinogens/chemistry , Uroporphyrinogens/metabolism , Uroporphyrins/chemistry
15.
Biochemistry ; 47(21): 5851-7, 2008 May 27.
Article in English | MEDLINE | ID: mdl-18457416

ABSTRACT

The sulfate-reducing bacterium Desulfovibrio vulgaris Hildenborough possesses a large number of porphyrin-containing proteins whose biosynthesis is poorly characterized. In this work, we have studied two putative CbiK cobaltochelatases present in the genome of D. vulgaris. The assays revealed that both enzymes insert cobalt and iron into sirohydrochlorin, with specific activities with iron lower than that measured with cobalt. Nevertheless, the two D. vulgaris chelatases complement an E. coli cysG mutant strain showing that, in vivo, they are able to load iron into sirohydrochlorin. The results showed that the functional cobaltochelatases have distinct roles with one, CbiK(C), likely to be the enzyme associated with cytoplasmic cobalamin biosynthesis, while the other, CbiK(P), is periplasmic located and possibly associated with an iron transport system. Finally, the ability of D. vulgaris to produce vitamin B 12 was also demonstrated in this work.


Subject(s)
Bacterial Proteins/chemistry , Desulfovibrio vulgaris/enzymology , Lyases/chemistry , Amino Acid Sequence , Biological Transport , Cytoplasm/metabolism , Escherichia coli/metabolism , Genome, Bacterial , Iron/metabolism , Magnetic Resonance Spectroscopy , Molecular Sequence Data , Mutation , Sequence Homology, Amino Acid , Spectrophotometry, Ultraviolet/methods , Uroporphyrins/chemistry , Vitamin B 12/metabolism
16.
Chem Pharm Bull (Tokyo) ; 54(7): 1004-9, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16819220

ABSTRACT

The pH-dependence of the ability of coproporphyrin (CP) and uroporphyrin (UP) to photogenerate hydrogen peroxide (H2O2) in aqueous solution was investigated, with special attention to the structure-activity relationship related to the aggregation of the porphyrins. It was found that the efficiency was strongly dependent on the aggregation of CP and UP mediated by changes in the pH of the solution, and a dimeric form had a weak ability to produce H2O2, while a highly aggregated form had a good ability. The increased efficiency of the highly aggregated porphyrin to produce H2O2 was further demonstrated using a different type of aggregate formed by the electrostatic interaction of cationic tetrakis-5,10,15,20-(N-methyl-4-pyridyl)porphin (TMPyP) with anionic tetrakis-5,10,15,20-(4-sulfonatophenyl)porphin (TSPP). The present results demonstrated the importance of the state of aggregation of porphyrin to photogenerate H2O2, and the results may help to develop a new type of medicine for photodynamic therapy.


Subject(s)
Coproporphyrins/chemistry , Hydrogen Peroxide/chemical synthesis , Uroporphyrins/chemistry , Water/chemistry , Coproporphyrins/physiology , Dimerization , Hydrogen Peroxide/chemistry , Hydrogen-Ion Concentration , Photochemistry , Porphyrins/chemistry , Solutions/chemistry , Structure-Activity Relationship , Uroporphyrins/physiology
17.
J Am Chem Soc ; 128(30): 9971-8, 2006 Aug 02.
Article in English | MEDLINE | ID: mdl-16866557

ABSTRACT

Two new cobalt corrinoid intermediates, cobalt-precorrin 5A and cobalt-precorrin 5B, have been synthesized with the aid of overexpressed enzymes of the vitamin B(12) pathway of Salmonella entericaserovar typhimurium. These compounds were made in several regioselectively (13)C-labeled forms, and their structures have been established by multidimensional NMR spectroscopy. The addition of CbiF to the enzymes known to synthesize cobalt-precorrin 4 resulted in the formation of cobalt-precorrin 5A, and the inclusion of CbiG with CbiF produced cobalt-precorrin 5B, which has allowed us to define the role of these enzymes in the anaerobic biosynthetic pathway. CbiF is the C-11 methylase, and CbiG, an enzyme which shows homology with CobE of the aerobic pathway, is the gene product responsible for the opening of the ring A delta-lactone and extrusion of the "C(2)" unit. The discovery of these long-sought intermediates paves the way for defining the final stages of the anaerobic pathway. It is of considerable evolutionary interest that nature uses two distinct pathways to vitamin B(12), both conserved over several billion years and featuring completely different mechanisms for ring-contraction of the porphyrinoid to the corrinoid ring system. Thus the aerobic pathway utilizes molecular oxygen to trigger the events at C-20 leading to contraction and expulsion of the "C(2)" unit as acetic acid from a metal-free intermediate, whereas the anaerobic route features internal delivery of oxygen from a carboxylic acid terminus to C-20 followed by extrusion of the "C(2)" unit as acetaldehyde, using cobalt complexes as substrates.


Subject(s)
Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Genetic Engineering , Organometallic Compounds/chemistry , Salmonella typhimurium/metabolism , Uroporphyrins/chemistry , Vitamin B 12/biosynthesis , Anaerobiosis , Models, Molecular , Molecular Structure , Vitamin B 12/chemistry
18.
Anal Biochem ; 353(2): 257-65, 2006 Jun 15.
Article in English | MEDLINE | ID: mdl-16564489

ABSTRACT

Few methods for assessing total antioxidant capacity (TAC) include both the percentage of inhibition and the length of inhibition in the measurement. Available methods require above ambient constant temperature incubation, reaction preheating, and/or separate assays for testing hydrophilic and hydrophobic samples. We describe a high-throughput method, antioxidant inhibition of oxygen radicals (AIOR), that overcomes these difficulties. AIOR uses peroxyl radicals to trigger a decrease in fluorescence of the indicator molecule, uroporphyrin I, which is delayed by the presence of antioxidants. The area under the curve is measured by a fluorescence spectrophotometer in a 96-well microplate format, and TAC results are expressed as millimole/liter Trolox equivalents. AIOR is performed at ambient temperature and is applicable to samples in either aqueous or common organic solvents. The reaction between uroporphyrin I and the peroxyl radicals generated from 2,2'-azobis(2-amidinopropane) dihydrochloride (AAPH) was found to be of first-order kinetics with a mean rate constant (k) of 0.0254. Applications to measure antioxidant capacity are demonstrated on individual chemicals and biological samples. The method has good linearity, within- and between-assay precision, and recovery.


Subject(s)
Antioxidants/analysis , Beverages/analysis , Reactive Oxygen Species/antagonists & inhibitors , Antioxidants/metabolism , Humans , Kinetics , Reactive Oxygen Species/metabolism , Sulfhydryl Compounds/analysis , Sulfhydryl Compounds/blood , Uric Acid/analysis , Uric Acid/blood , Uroporphyrins/chemistry , Uroporphyrins/metabolism , Vitamins/analysis , Vitamins/blood
19.
Biotechnol Bioeng ; 93(5): 989-97, 2006 Apr 05.
Article in English | MEDLINE | ID: mdl-16489629

ABSTRACT

A common problem encountered when using fluorescence detection in real samples analysis is that the matrix may contain compounds that autofluorescence or that can be excited at the wavelengths of commonly employed fluorescent reporter molecules. This causes an increase in background fluorescence, which in turn tends to compromise the detection limits of the system. To address this issue, we investigated the use of a reporter enzyme that produces fluorescent compounds, which can be excited at wavelengths that are not commonly encountered in compounds present in real samples. For that, a whole cell-based sensing system for arsenite that employs cobA as the reporter gene was developed. The system utilizes genetically engineered bacteria that incorporate the specificity of the ars operon with the sensitivity of the cobA gene. The cobA gene codes for uroporphyrinogen III methyltransferase that converts the substrate uroporphyrinogen (urogen) III into two fluorescent compounds sirohydrochlorin and trimethylpyrrocorphin. Urogen III is ubiquitous within the cell, however, because the cells use it for vitamin B12 and siroheme biosynthesis, this sensing system is limited by substrate availability. By supplementing the media with ALA, a precursor of urogen III, a more stable and reproducible response was obtained. We observed three excitation maxima at 357, 378, and 498 nm, with a single emission maximum at 605 nm. Excitation at 498 nm was selected because it results in less background interference as most endogenous substances are not active at this wavelength. Advantages and limitations of using the cobA gene in whole-cell sensing applications are presented.


Subject(s)
Genes, Reporter/genetics , Methyltransferases/genetics , Uroporphyrinogens/metabolism , Uroporphyrins/metabolism , Aminolevulinic Acid/pharmacology , Arsenites/pharmacology , Escherichia coli/drug effects , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Fluorescence , Gene Expression/drug effects , Ion Pumps/genetics , Methyltransferases/metabolism , Multienzyme Complexes/genetics , Operon/genetics , Plasmids/genetics , Sodium Compounds/pharmacology , Spectrometry, Fluorescence , Transfection , Uroporphyrins/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...