Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 176
Filtrar
1.
Int J Mol Sci ; 24(9)2023 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-37176103

RESUMO

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.


Assuntos
Citrus , Antioxidantes/farmacologia , Clorofila/metabolismo , Citrus/genética , Citrus/metabolismo , Fotossíntese/genética , Folhas de Planta/metabolismo , Uroporfirinogênios/metabolismo , Uroporfirinogênios/farmacologia , Necrose e Clorose das Plantas
2.
Microb Cell Fact ; 22(1): 102, 2023 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-37198628

RESUMO

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.


Assuntos
Bacillus subtilis , Heme , Bacillus subtilis/genética , Bacillus subtilis/metabolismo , Heme/metabolismo , Engenharia Metabólica/métodos , Fermentação , Uroporfirinogênios/metabolismo
3.
Microbiology (Reading) ; 167(10)2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34661520

RESUMO

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.


Assuntos
Heme/biossíntese , Hidroximetilbilano Sintase/metabolismo , Plasmodium falciparum/enzimologia , Vias Biossintéticas , Escherichia coli/genética , Teste de Complementação Genética , Hidroximetilbilano Sintase/genética , Mutação , Plasmodium falciparum/genética , Proteínas de Protozoários/química , Proteínas de Protozoários/genética , Proteínas de Protozoários/metabolismo , Uroporfirinogênio III Sintetase/genética , Uroporfirinogênio III Sintetase/metabolismo , Uroporfirinogênios/metabolismo
4.
Biochem J ; 478(5): 1023-1042, 2021 03 12.
Artigo em Inglês | MEDLINE | ID: mdl-33600566

RESUMO

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.


Assuntos
Hidroximetilbilano Sintase/química , Hidroximetilbilano Sintase/metabolismo , Porfobilinogênio/metabolismo , Uroporfirinogênios/metabolismo , Catálise , Domínio Catalítico , Cristalografia por Raios X , Humanos , Simulação de Dinâmica Molecular , Conformação Proteica , Domínios Proteicos , Especificidade por Substrato
5.
Photodiagnosis Photodyn Ther ; 25: 309-316, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30639584

RESUMO

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.


Assuntos
Ácido Aminolevulínico/farmacologia , Transportador 1 de Peptídeos/biossíntese , Fotoquimioterapia/métodos , Protoporfirinas/metabolismo , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/biossíntese , Western Blotting , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/secundário , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Ferroquelatase/biossíntese , Heme Oxigenase-1/biossíntese , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/patologia , Metástase Neoplásica , Reação em Cadeia da Polimerase em Tempo Real , Espectrometria de Fluorescência , Uroporfirinogênios/biossíntese
6.
Biochim Biophys Acta Gen Subj ; 1862(9): 1948-1955, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29908816

RESUMO

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.


Assuntos
Hidroximetilbilano Sintase/química , Hidroximetilbilano Sintase/metabolismo , Pirróis/química , Pirróis/metabolismo , Catálise , Domínio Catalítico , Cristalografia por Raios X , Humanos , Polimerização , Porfobilinogênio/química , Porfobilinogênio/metabolismo , Conformação Proteica , Uroporfirinogênios/química , Uroporfirinogênios/metabolismo
7.
Structure ; 26(4): 565-571.e3, 2018 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-29551288

RESUMO

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.


Assuntos
Acetilcolina/química , Acetilcolinesterase/química , Medicamentos Biossimilares/química , Uroporfirinogênio III Sintetase/química , Uroporfirinogênios/química , Acetilcolina/metabolismo , Acetilcolinesterase/metabolismo , Sítios de Ligação , Medicamentos Biossimilares/metabolismo , Humanos , Ligantes , Simulação de Acoplamento Molecular , Ligação Proteica , Especificidade por Substrato , Uroporfirinogênio III Sintetase/metabolismo , Uroporfirinogênios/metabolismo
8.
ACS Chem Biol ; 13(3): 750-760, 2018 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-29360345

RESUMO

Porphobilinogen deaminase (PBGD) is an enzyme that catalyzes the formation of hydroxymethylbilane, a tetrapyrrole intermediate, during heme biosynthesis through the stepwise polymerization of four molecules of porphobilinogen. PBGD from Vibrio cholerae was expressed in Escherichia coli and characterized in this study. Unexpectedly, spectroscopic measurements revealed that PBGD bound one equivalent of heme with a dissociation constant of 0.33 ± 0.01 µM. The absorption and resonance Raman spectra suggested that heme is a mixture of the 5-coordinate and 6-coordinate hemes. Mutational studies indicated that the 5-coordinate heme possessed Cys105 as a heme axial ligand, and His227 was coordinated to form the 6-coordinate heme. Upon heme binding, the deamination activity decreased by approximately 15%. The crystal structure of PBGD revealed that His227 was located near Cys105, but the side chain of His227 did not point toward Cys105. The addition of the cyanide ion to heme-PBGD abolished the effect of heme binding on the enzymatic activity. Therefore, coordination of His227 to heme appeared to induce reorientation of the domains containing Cys105, leading to a decrease in the enzymatic activity. This is the first report indicating that the PBGD activity is controlled by heme, the final product of heme biosynthesis. This finding improves our understanding of the mechanism by which heme biosynthesis is regulated.


Assuntos
Heme/biossíntese , Hidroximetilbilano Sintase/metabolismo , Uroporfirinogênios/biossíntese , Vibrio cholerae/metabolismo , Cristalografia por Raios X , Escherichia coli/genética , Heme/metabolismo , Hidroximetilbilano Sintase/química , Ligação Proteica , Análise Espectral Raman , Vibrio cholerae/enzimologia
9.
Sheng Wu Gong Cheng Xue Bao ; 33(1): 55-67, 2017 Jan 25.
Artigo em Chinês | MEDLINE | ID: mdl-28959863

RESUMO

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.


Assuntos
Metiltransferases/isolamento & purificação , Rhodobacter capsulatus/enzimologia , Escherichia coli , Metionina , Pseudomonas , S-Adenosilmetionina , Uroporfirinogênios , Uroporfirinas
10.
Proteins ; 85(1): 46-53, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27756106

RESUMO

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


Assuntos
Proteínas de Bactérias/química , Thermus thermophilus/química , Uroporfirinogênio III Sintetase/química , Uroporfirinogênios/química , Biocatálise , Entropia , Evolução Molecular , Ligantes , Simulação de Dinâmica Molecular , Movimento (Física) , Ligação Proteica , Domínios Proteicos , Estrutura Secundária de Proteína , Thermus thermophilus/enzimologia
11.
PLoS Comput Biol ; 10(3): e1003484, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24603363

RESUMO

Porphobilinogen deaminase (PBGD) catalyzes the formation of 1-hydroxymethylbilane (HMB), a crucial intermediate in tetrapyrrole biosynthesis, through a step-wise polymerization of four molecules of porphobilinogen (PBG), using a unique dipyrromethane (DPM) cofactor. Structural and biochemical studies have suggested residues with catalytic importance, but their specific role in the mechanism and the dynamic behavior of the protein with respect to the growing pyrrole chain remains unknown. Molecular dynamics simulations of the protein through the different stages of pyrrole chain elongation suggested that the compactness of the overall protein decreases progressively with addition of each pyrrole ring. Essential dynamics showed that domains move apart while the cofactor turn region moves towards the second domain, thus creating space for the pyrrole rings added at each stage. Residues of the flexible active site loop play a significant role in its modulation. Steered molecular dynamics was performed to predict the exit mechanism of HMB from PBGD at the end of the catalytic cycle. Based on the force profile and minimal structural changes the proposed path for the exit of HMB is through the space between the domains flanking the active site loop. Residues reported as catalytically important, also play an important role in the exit of HMB. Further, upon removal of HMB, the structure of PBGD gradually relaxes to resemble its initial stage structure, indicating its readiness to resume a new catalytic cycle.


Assuntos
Escherichia coli/enzimologia , Hidroximetilbilano Sintase/metabolismo , Uroporfirinogênios/biossíntese , Catálise , Domínio Catalítico , Biologia Computacional , Difusão , Heme/química , Conformação Molecular , Simulação de Dinâmica Molecular , Mutação , Pirróis/química
12.
Archaea ; 2014: 327637, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24669201

RESUMO

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.


Assuntos
Proteínas Arqueais/genética , Proteínas Arqueais/metabolismo , Vias Biossintéticas/genética , Heme/biossíntese , Methanosarcina barkeri/genética , Methanosarcina barkeri/metabolismo , Methanosarcina barkeri/enzimologia , Multimerização Proteica , Uroporfirinogênios/metabolismo
13.
Cell Mol Life Sci ; 71(15): 2837-63, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24515122

RESUMO

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.


Assuntos
Vias Biossintéticas , Heme/análogos & derivados , Tetrapirróis/metabolismo , Animais , Heme/química , Heme/metabolismo , Humanos , Modelos Moleculares , Tetrapirróis/química , Uroporfirinogênios/química , Uroporfirinogênios/metabolismo
14.
Appl Microbiol Biotechnol ; 97(16): 7337-44, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23604563

RESUMO

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.


Assuntos
Ácido Aminolevulínico/metabolismo , Escherichia coli/metabolismo , Thermus thermophilus/enzimologia , Uroporfirinogênios/metabolismo , Biotecnologia/métodos , Escherichia coli/genética , Temperatura Alta , Hidroximetilbilano Sintase/genética , Engenharia Metabólica/métodos , Redes e Vias Metabólicas/genética , Sintase do Porfobilinogênio/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Thermus thermophilus/genética , Uroporfirinogênio III Sintetase/genética
15.
Eukaryot Cell ; 10(11): 1536-44, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21908598

RESUMO

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.


Assuntos
Alcaloides/farmacologia , Heme/biossíntese , Saccharomyces cerevisiae/efeitos dos fármacos , Proteases Dependentes de ATP/genética , Proteases Dependentes de ATP/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Humanos , Células Jurkat , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , ATPases Mitocondriais Próton-Translocadoras/genética , ATPases Mitocondriais Próton-Translocadoras/metabolismo , Naftiridinas , Extratos Vegetais/farmacologia , Protoporfirinogênio Oxidase/genética , Protoporfirinogênio Oxidase/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Uroporfirinogênio III Sintetase/biossíntese , Uroporfirinogênio III Sintetase/metabolismo , Uroporfirinogênios/metabolismo
16.
J Biol Chem ; 286(30): 26754-67, 2011 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-21632530

RESUMO

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.


Assuntos
Proteínas de Bactérias/química , Metiltransferases/química , Pseudomonas aeruginosa/enzimologia , Uroporfirinogênios/química , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Catálise , Cristalografia por Raios X , Heme/análogos & derivados , Heme/biossíntese , Heme/química , Heme/metabolismo , Metiltransferases/genética , Metiltransferases/metabolismo , Mutagênese Sítio-Dirigida , Mutação de Sentido Incorreto , Estrutura Terciária de Proteína , Pseudomonas aeruginosa/genética , Uroporfirinogênios/genética , Uroporfirinogênios/metabolismo
17.
Blood ; 118(6): 1443-51, 2011 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-21653323

RESUMO

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.


Assuntos
5-Aminolevulinato Sintetase/genética , Mutação de Sentido Incorreto , Porfiria Eritropoética/genética , Uroporfirinogênio III Sintetase/genética , 5-Aminolevulinato Sintetase/metabolismo , Sequência de Aminoácidos , Anemia Sideroblástica/genética , Anemia Sideroblástica/metabolismo , Anemia Sideroblástica/patologia , Sequência de Bases , Pré-Escolar , Eletroforese em Gel de Poliacrilamida , Saúde da Família , Feminino , Doenças Genéticas Ligadas ao Cromossomo X/genética , Doenças Genéticas Ligadas ao Cromossomo X/metabolismo , Genótipo , Humanos , Lactente , Cinética , Masculino , Dados de Sequência Molecular , Linhagem , Porfiria Eritropoética/metabolismo , Porfiria Eritropoética/patologia , Protoporfiria Eritropoética/genética , Protoporfiria Eritropoética/metabolismo , Homologia de Sequência de Aminoácidos , Índice de Gravidade de Doença , Espectrofotometria , Uroporfirinogênio III Sintetase/metabolismo , Uroporfirinogênios/metabolismo
18.
J Phys Chem B ; 114(27): 8994-9001, 2010 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-20553007

RESUMO

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


Assuntos
Arginina , Oniocompostos , Uroporfirinogênio Descarboxilase/química , Uroporfirinogênio Descarboxilase/metabolismo , Domínio Catalítico , Humanos , Simulação de Dinâmica Molecular , Mutagênese Sítio-Dirigida , Prótons , Uroporfirinogênio Descarboxilase/genética , Uroporfirinogênios/metabolismo
19.
FEBS J ; 276(21): 6399-411, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19796169

RESUMO

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.


Assuntos
Bactérias/metabolismo , Heme/biossíntese , Nitrito Redutases/fisiologia , Proteínas de Transporte de Ânions/fisiologia , Citocromos/fisiologia , Proteínas de Escherichia coli/fisiologia , Heme/análogos & derivados , Paracoccus pantotrophus/genética , Paracoccus pantotrophus/crescimento & desenvolvimento , Uroporfirinogênios/metabolismo , Uroporfirinas/biossíntese
20.
FEBS J ; 276(20): 5973-82, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19754882

RESUMO

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.


Assuntos
Proteínas de Bactérias/metabolismo , Heme/análogos & derivados , Metiltransferases/química , Metiltransferases/metabolismo , Pseudomonas aeruginosa/enzimologia , Pseudomonas aeruginosa/genética , Sequência de Aminoácidos , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Teste de Complementação Genética , Heme/biossíntese , Metiltransferases/genética , Dados de Sequência Molecular , Ligação Proteica , Pseudomonas aeruginosa/crescimento & desenvolvimento , Pseudomonas aeruginosa/metabolismo , S-Adenosilmetionina/metabolismo , Homologia de Sequência de Aminoácidos , Especificidade por Substrato , Uroporfirinogênios/metabolismo , Uroporfirinas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...