Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Transl Med ; 10(459)2018 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-30232228

RESUMO

Congenital erythropoietic porphyria is a rare autosomal recessive disease produced by deficient activity of uroporphyrinogen III synthase, the fourth enzyme in the heme biosynthetic pathway. The disease affects many organs, can be life-threatening, and currently lacks curative treatments. Inherited mutations most commonly reduce the enzyme's stability, altering its homeostasis and ultimately blunting intracellular heme production. This results in uroporphyrin by-product accumulation in the body, aggravating associated pathological symptoms such as skin photosensitivity and disfiguring phototoxic cutaneous lesions. We demonstrated that the synthetic marketed antifungal ciclopirox binds to the enzyme, stabilizing it. Ciclopirox targeted the enzyme at an allosteric site distant from the active center and did not affect the enzyme's catalytic role. The drug restored enzymatic activity in vitro and ex vivo and was able to alleviate most clinical symptoms of congenital erythropoietic porphyria in a genetic mouse model of the disease at subtoxic concentrations. Our findings establish a possible line of therapeutic intervention against congenital erythropoietic porphyria, which is potentially applicable to most of deleterious missense mutations causing this devastating disease.


Assuntos
Ciclopirox/uso terapêutico , Reposicionamento de Medicamentos , Porfiria Eritropoética/tratamento farmacológico , Sítio Alostérico , Animais , Fenômenos Biofísicos , Linhagem Celular , Ciclopirox/farmacocinética , Modelos Animais de Doenças , Homeostase , Camundongos , Fenótipo , Porfiria Eritropoética/enzimologia , Porfiria Eritropoética/patologia , Uroporfirinogênio III Sintetase/antagonistas & inibidores , Uroporfirinogênio III Sintetase/química , Uroporfirinogênio III Sintetase/metabolismo
2.
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
3.
Hum Mol Genet ; 26(8): 1565-1576, 2017 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-28334762

RESUMO

Congenital erythropoietic porphyria (CEP) is an inborn error of heme biosynthesis characterized by uroporphyrinogen III synthase (UROS) deficiency resulting in deleterious porphyrin accumulation in blood cells responsible for hemolytic anemia and cutaneous photosensitivity. We analyzed here the molecular basis of UROS impairment associated with twenty nine UROS missense mutations actually described in CEP patients. Using a computational and biophysical joint approach we predicted that most disease-causing mutations would affect UROS folding and stability. Through the analysis of enhanced green fluorescent protein-tagged versions of UROS enzyme we experimentally confirmed these data and showed that thermodynamic instability and premature protein degradation is a major mechanism accounting for the enzymatic deficiency associated with twenty UROS mutants in human cells. Since the intracellular loss in protein homeostasis is in excellent agreement with the in vitro destabilization, we used molecular dynamic simulation to rely structural 3D modification with UROS disability. We found that destabilizing mutations could be clustered within three types of mechanism according to side chain rearrangements or contact alterations within the pathogenic UROS enzyme so that the severity degree correlated with cellular protein instability. Furthermore, proteasome inhibition using bortezomib, a clinically available drug, significantly enhanced proteostasis of each unstable UROS mutant. Finally, we show evidence that abnormal protein homeostasis is a prevalent mechanism responsible for UROS deficiency and that modulators of UROS proteolysis such as proteasome inhibitors or chemical chaperones may represent an attractive therapeutic option to reduce porphyrin accumulation and prevent skin photosensitivity in CEP patients when the genotype includes a missense variant.


Assuntos
Mutação de Sentido Incorreto/genética , Porfiria Eritropoética/genética , Relação Estrutura-Atividade , Uroporfirinogênio III Sintetase/genética , Biologia Computacional , Homeostase , Humanos , Porfiria Eritropoética/metabolismo , Porfiria Eritropoética/patologia , Complexo de Endopeptidases do Proteassoma/efeitos dos fármacos , Complexo de Endopeptidases do Proteassoma/genética , Inibidores de Proteassoma/química , Inibidores de Proteassoma/uso terapêutico , Dobramento de Proteína , Uroporfirinogênio III Sintetase/química
4.
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
5.
Hum Mol Genet ; 23(21): 5805-13, 2014 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-24925316

RESUMO

Congenital erythropoietic porphyria (CEP) results from a deficiency in uroporphyrinogen III synthase enzyme (UROIIIS) activity that ultimately stems from deleterious mutations in the uroS gene. C73 is a hotspot for these mutations and a C73R substitution, which drastically reduces the enzyme activity and stability, is found in almost one-third of all reported CEP cases. Here, we have studied the structural basis, by which mutations in this hotspot lead to UROIIIS destabilization. First, a strong interdependency is observed between the volume of the side chain at position 73 and the folded protein. Moreover, there is a correlation between the in vitro half-life of the mutated proteins and their expression levels in eukaryotic cell lines. Molecular modelling was used to rationalize the results, showing that the mutation site is coupled to the hinge region separating the two domains. Namely, mutations at position 73 modulate the inter-domain closure and ultimately affect protein stability. By incorporating residues capable of interacting with R73 to stabilize the hinge region, catalytic activity was fully restored and a moderate increase in the kinetic stability of the enzyme was observed. These results provide an unprecedented rationale for a destabilizing missense mutation and pave the way for the effective design of molecular chaperones as a therapy against CEP.


Assuntos
Homeostase , Porfiria Eritropoética/metabolismo , Engenharia de Proteínas , Uroporfirinogênio III Sintetase/metabolismo , Substituição de Aminoácidos , Catálise , Ativação Enzimática , Estabilidade Enzimática , Humanos , Espaço Intracelular/metabolismo , Cinética , Modelos Moleculares , Mutação , Porfiria Eritropoética/enzimologia , Porfiria Eritropoética/genética , Conformação Proteica , Uroporfirinogênio III Sintetase/química , Uroporfirinogênio III Sintetase/genética
6.
Proc Natl Acad Sci U S A ; 110(45): 18238-43, 2013 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-24145442

RESUMO

Congenital erythropoietic porphyria (CEP) is a rare autosomal recessive disorder characterized by uroporphyrinogen III synthase (UROS) deficiency resulting in massive porphyrin accumulation in blood cells, which is responsible for hemolytic anemia and skin photosensitivity. Among the missense mutations actually described up to now in CEP patients, the C73R and the P248Q mutations lead to a profound UROS deficiency and are usually associated with a severe clinical phenotype. We previously demonstrated that the UROS(C73R) mutant protein conserves intrinsic enzymatic activity but triggers premature degradation in cellular systems that could be prevented by proteasome inhibitors. We show evidence that the reduced kinetic stability of the UROS(P248Q) mutant is also responsible for increased protein turnover in human erythroid cells. Through the analysis of EGFP-tagged versions of UROS enzyme, we demonstrate that both UROS(C73R) and UROS(P248Q) are equally destabilized in mammalian cells and targeted to the proteasomal pathway for degradation. We show that a treatment with proteasomal inhibitors, but not with lysosomal inhibitors, could rescue the expression of both EGFP-UROS mutants. Finally, in CEP mice (Uros(P248Q/P248Q)) treated with bortezomib (Velcade), a clinically approved proteasome inhibitor, we observed reduced porphyrin accumulation in circulating RBCs and urine, as well as reversion of skin photosensitivity on bortezomib treatment. These results of medical importance pave the way for pharmacologic treatment of CEP disease by preventing certain enzymatically active UROS mutants from early degradation by using proteasome inhibitors or chemical chaperones.


Assuntos
Modelos Moleculares , Porfiria Eritropoética/tratamento farmacológico , Inibidores de Proteassoma/uso terapêutico , Uroporfirinogênio III Sintetase/genética , Uroporfirinogênio III Sintetase/metabolismo , Animais , Western Blotting , Ácidos Borônicos/farmacologia , Ácidos Borônicos/uso terapêutico , Bortezomib , Dicroísmo Circular , Primers do DNA/genética , Células Eritroides/metabolismo , Humanos , Camundongos , Mutação de Sentido Incorreto/genética , Porfiria Eritropoética/genética , Porfirinas/sangue , Porfirinas/urina , Dobramento de Proteína , Pirazinas/farmacologia , Pirazinas/uso terapêutico , Reação em Cadeia da Polimerase em Tempo Real , Espectrometria de Fluorescência , Uroporfirinogênio III Sintetase/química
7.
Artigo em Inglês | MEDLINE | ID: mdl-21570665

RESUMO

Congenital erythropoietic porphyria (CEP) is a rare autosomal disease ultimately related to deleterious mutations in uroporphyrinogen III synthase (UROIIIS), the fourth enzyme of the biosynthetic route of the heme group. UROIIIS catalyzes the cyclization of the linear tetrapyrrol hydroxymethylbilane (HMB), inverting the configuration in one of the aromatic rings. In the absence of the enzyme (or when ill-functioning), HMB spontaneously degrades to the by-product uroporphyrinogen I, which cannot lead to the heme group and accumulates in the body, producing some of the symptoms observed in CEP patients. In the present chapter, clinical, biochemical, and biophysical information has been compiled to provide an integrative view on the molecular basis of CEP. The high-resolution structure of UROIIIS sheds light on the enzyme reaction mechanism while thermodynamic analysis revealed that the protein is thermolabile. Pathogenic missense mutations are found throughout the primary sequence of the enzyme. All but one of these is rarely found in patients, whereas C73R is responsible for more than one-third of the reported cases. Most of the mutant proteins (C73R included) retain partial catalytic activity but the mutations often reduce the enzyme's stability. The stabilization of the protein in vivo is discussed in the context of a new line of intervention to complement existing treatments such as bone marrow transplantation and gene therapy.


Assuntos
Porfiria Eritropoética/enzimologia , Uroporfirinogênio III Sintetase/química , Uroporfirinogênio III Sintetase/metabolismo , Animais , Biocatálise , Humanos , Porfiria Eritropoética/terapia , Conformação Proteica , Termodinâmica , Uroporfirinogênio III Sintetase/genética
8.
Biochem Biophys Res Commun ; 408(4): 576-81, 2011 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-21527255

RESUMO

Uroporphyrinogen III synthase (U3S) is one of the key enzymes in the biosynthesis of tetrapyrrole compounds. It catalyzes the cyclization of the linear hydroxymethylbilane (HMB) to uroporphyrinogen III (uro'gen III). We have determined the crystal structure of U3S from Pseudomonas syringae pv. tomato DC3000 (psU3S) at 2.5Å resolution by the single wavelength anomalous dispersion (SAD) method. Each psU3S molecule consists of two domains interlinked by a two-stranded antiparallel ß-sheet. The conformation of psU3S is different from its homologous proteins because of the flexibility of the linker between the two domains, which might be related to this enzyme's catalytic properties. Based on mutation and activity analysis, a key residue, Arg219, was found to be important for the catalytic activity of psU3S. Mutation of Arg219 to Ala caused a decrease in enzymatic activity to about 25% that of the wild type enzyme. Our results provide the structural basis and biochemical evidence to further elucidate the catalytic mechanism of U3S.


Assuntos
Pseudomonas syringae/enzimologia , Uroporfirinogênio III Sintetase/química , Domínio Catalítico , Cristalografia por Raios X , Estrutura Secundária de Proteína , Uroporfirinogênio III Sintetase/genética
9.
Biotechnol Lett ; 33(8): 1625-32, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21484341

RESUMO

PURPOSE OF WORK: To clone, express and characterize uroporphyrinogen III synthase/methyltransferase gene (cobA/hemD) from Lactobacillus reuteri. Some strains of Lb. reuteri produce cobalamin (vitamin B(12)). Cobalamin biosynthesis relies on the sequential action of more than 25 enzymes in a complex metabolic pathway. We have cloned, expressed and characterized the gene in Lb. reuteri that codes for the S-adenosy L: -methionine uroprophyrinogen III methyltransferase/synthase (CobA/HemD), a key bifunctional enzyme in the biosynthesis of cobalamin and other tetrapyrrols.


Assuntos
Limosilactobacillus reuteri/enzimologia , Metiltransferases/biossíntese , Metiltransferases/genética , Uroporfirinogênio III Sintetase/biossíntese , Uroporfirinogênio III Sintetase/genética , Sequência de Aminoácidos , Proteínas de Bactérias/biossíntese , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Sequência de Bases , Clonagem Molecular , Simulação por Computador , Escherichia coli/genética , Escherichia coli/metabolismo , Limosilactobacillus reuteri/genética , Metiltransferases/química , Dados de Sequência Molecular , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Espectrometria de Fluorescência , Uroporfirinogênio III Sintetase/química
10.
Mol Med ; 16(9-10): 381-8, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20485863

RESUMO

The first feline model of human congenital erythropoietic porphyria (CEP) due to deficient uroporphyrinogen III synthase (URO-synthase) activity was identified by its characteristic clinical phenotype, and confirmed by biochemical and molecular genetic studies. The proband, an adult domestic shorthair cat, had dark-red urine and brownish discolored teeth with red fluorescence under ultraviolet light. Biochemical studies demonstrated markedly increased uroporphyrinogen I in urine and plasma (2,650- and 10,700-fold greater than wild type, respectively), whereas urinary 5-aminolevulinic acid and porphobilinogen were lower than normal. Erythrocytic URO-synthase activity was <1% of mean wild-type activity, confirming the diagnosis and distinguishing it from feline phenocopies having acute intermittent porphyria. Sequencing of the affected cat's UROS gene revealed two missense mutations, c.140C>T (p.S47F) in exon 3 and c.331G>A (p.G111S) in exon 6, both of which were homozygous, presumably owing to parental consanguinity. Neither was present in 100 normal cat alleles. Prokaryotic expression and thermostability studies of the purified monomeric wild-type, p.S47F, p.G111S, and p.S47F/G111S enzymes showed that the p.S47F enzyme had 100% of wild-type specific activity but ~50% decreased thermostability, whereas the p.G111S and p.S47F/G111S enzymes had about 60% and 20% of wild-type specific activity, respectively, and both were markedly thermolabile. Molecular modeling results indicated that the less active/less stable p.G111S enzyme was further functionally impaired by a structural interaction induced by the presence of the S47F substitution. Thus, the synergistic interaction of two rare amino acid substitutions in the URO-synthase polypeptide caused the feline model of human CEP.


Assuntos
Doenças do Gato/enzimologia , Doenças do Gato/genética , Homozigoto , Mutação de Sentido Incorreto/genética , Porfiria Eritropoética/veterinária , Porfirinas/metabolismo , Uroporfirinogênio III Sintetase/genética , Animais , Doenças do Gato/sangue , Doenças do Gato/urina , Gatos , Eritrócitos/metabolismo , Masculino , Modelos Moleculares , Dados de Sequência Molecular , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Porfiria Eritropoética/sangue , Porfiria Eritropoética/enzimologia , Porfiria Eritropoética/urina , Porfirinas/sangue , Porfirinas/urina , Uroporfirinogênio III Sintetase/química , Uroporfirinogênio III Sintetase/metabolismo
11.
Biochemistry ; 47(33): 8648-55, 2008 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-18651750

RESUMO

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.


Assuntos
Uroporfirinogênio III Sintetase/química , Uroporfirinogênio III Sintetase/metabolismo , Uroporfirinogênios/química , Uroporfirinogênios/metabolismo , Cristalização , Modelos Moleculares , Estrutura Molecular , Thermus thermophilus/enzimologia
12.
J Phys Chem B ; 112(10): 3144-8, 2008 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-18281969

RESUMO

The asymmetric cyclic tetrapyrrole uroporphyrinogen III is the common precursor of heme, chlorophyll, siroheme, and other biological tetrapyrroles. In vivo, it is synthesized from a linear symmetric precursor (hydroxymethylbilane) by uroporphyrinogen III synthase, which catalyzes the inversion of one of the four heterocyclic rings present in the substrate. Two mechanisms have been proposed to explain this puzzling ring inversion, either through sigmatropic shifts or through the direct formation of a spirocyclic pyrrolenine intermediate. We performed the first high-level quantum mechanical calculations on model systems of this enzyme to analyze these contrasting reaction mechanisms. The results allow us to discard the sigmatropic shift mechanism and suggest that the D-ring of the hydroxymethylbilane substrate binds to the enzyme in a conformation that shields its terminal portion from reacting with ring A and prevents the formation of the biologically useless uroporphyrinogen I, whose accumulation (in individuals lacking functional uroporphyrinogen III synthase) leads to severe cutaneous dermatosis.


Assuntos
Modelos Biológicos , Uroporfirinogênio III Sintetase/química , Uroporfirinogênio III Sintetase/metabolismo , Catálise , Elétrons , Modelos Moleculares , Estrutura Molecular , Compostos de Espiro/química
13.
Proteins ; 71(2): 855-73, 2008 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-18004775

RESUMO

Uroporphyrinogen III synthase (URO-synthase) catalyzes the cyclization and D-ring isomerization of hydroxymethylbilane (HMB) to uroporphyrinogen (URO'gen) III, the cyclic tetrapyrrole and physiologic precursor of heme, chlorophyl, and corrin. The deficient activity of human URO-synthase results in the autosomal recessive cutaneous disorder, congenital erythropoietic porphyria. Mapping of the structural determinants that specify catalysis and, potentially, protein-protein interactions is lacking. To map the active site and assess the enzyme's possible interaction in a complex with hydroxymethylbilane-synthase (HMB-synthase) and/or uroporphyrinogen-decarboxylase (URO-decarboxylase) by NMR, an efficient expression and purification procedure was developed for these cytosolic enzymes of heme biosynthesis that enabled preparation of special isotopically-labeled protein samples for NMR characterization. Using an 800 MHz instrument, assignment of the URO-synthase backbone (13)C(alpha) (100%), (1)H(alpha) (99.6%), and nonproline (1)H(N) and (15)N resonances (94%) was achieved as well as 85% of the side-chain (13)C and (1)H resonances. NMR analyses of URO-synthase titrated with competitive inhibitors N(D)-methyl-1-formylbilane (NMF-bilane) or URO'gen III, revealed resonance perturbations of specific residues lining the cleft between the two major domains of URO synthase that mapped the enzyme's active site. In silico docking of the URO-synthase crystal structure with NMF-bilane and URO'gen III was consistent with the perturbation results and provided a 3D model of the enzyme-inhibitor complex. The absence of chemical shift changes in the (15)N spectrum of URO-synthase mixed with the homogeneous HMB-synthase holoenzyme or URO-decarboxylase precluded occurrence of a stable cytosolic enzyme complex.


Assuntos
Sítios de Ligação , Uroporfirinogênio III Sintetase/química , Sequência de Aminoácidos , Isótopos de Carbono , Simulação por Computador , Humanos , Hidroximetilbilano Sintase/isolamento & purificação , Cinética , Modelos Moleculares , Dados de Sequência Molecular , Isótopos de Nitrogênio , Ressonância Magnética Nuclear Biomolecular , Proteínas Recombinantes/química , Proteínas Recombinantes/isolamento & purificação , Termodinâmica , Uroporfirinogênio Descarboxilase/isolamento & purificação , Uroporfirinogênio III Sintetase/antagonistas & inibidores , Uroporfirinogênio III Sintetase/isolamento & purificação , Uroporfirinogênios/farmacologia
14.
Protein J ; 26(8): 569-76, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17763925

RESUMO

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. Urogen III synthase catalyzes a key step in the formation of urogen III, a common intermediate for tetrapyrrolic natural products. In the present study, we cloned, purified, and characterized His-tagged rat urogen III synthase. The mechanism of enzymatic reaction was studied through site-directed mutagenesis of eight highly conserved residues with functional side chains around the active site followed with activity tests. Lys10, Asp17, Glu68, Tyr97, Asn121, Lys147, and His173 have not been studied previously, which were found to be unessential for enzymatic reaction. Tyr168 was identified as an important residue for enzymatic reaction catalyzed by rat urogen III synthase. Molecular modeling suggests the hydroxyl group of Tyr168 side chain is 3.5 A away from the D ring, and is within hydrogen bond distance (1.9 A) with acetate side chain of the D ring.


Assuntos
Histidina/química , Fígado/enzimologia , Uroporfirinogênio III Sintetase/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Catálise , Clonagem Molecular , Cristalografia por Raios X , Biblioteca Gênica , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Conformação Proteica , Ratos , Homologia de Sequência de Aminoácidos , Especificidade por Substrato , Uroporfirinogênio III Sintetase/química , Uroporfirinogênio III Sintetase/genética
15.
DNA Seq ; 18(1): 54-60, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17364814

RESUMO

Heme is synthesized from glycine and succinyl CoA by eight heme synthesis enzymes. Although genetic defects in any of these enzymes are known to cause severe human blood diseases, their developmental expression in mammals is unknown. In this paper, we report two zebrafish heme synthesis enzymes, uroporphyrinogen III synthase (UROS) and protoporphyrinogen oxidase (PPO) that are well conserved in comparison to their human counterparts. Both UROS and PPO formed pairs of bilateral stripes in the lateral plate mesoderm at the 15-somite stage. At 24 h post-fertilization (hpf), UROS and PPO were predominantly expressed in the intermediate cell mass (ICM) that is the major site of primitive hematopoiesis. The expression of UROS and PPO was drastically suppressed in the bloodless mutants cloche and vlad tepes/gata 1 from 15-somite to 24hpf stages, indicating that both cloche and vlad tepes/gata 1 are required for the induction and maintenance of UROS and PPO expression in the ICM.


Assuntos
Heme/biossíntese , Protoporfirinogênio Oxidase/genética , Peixe-Zebra/genética , Animais , Sequência de Bases , Clonagem Molecular , Primers do DNA , Regulação da Expressão Gênica no Desenvolvimento , Regulação Enzimológica da Expressão Gênica , Hibridização In Situ , Protoporfirinogênio Oxidase/química , Uroporfirinogênio III Sintetase/química , Uroporfirinogênio III Sintetase/genética
16.
Biochem Soc Trans ; 30(4): 595-600, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12196144

RESUMO

All tetrapyrroles are synthesized through a branched pathway, and although each tetrapyrrole receives unique modifications around the ring periphery, they all share the unifying feature of a central metal ion. Each pathway maintains a unique metal ion chelatase, and several tertiary structures have been determined, including those of the protoporphyrin ferrochelatase from both human and Bacillus subtilus, and the cobalt chelatase CbiK. These enzymes exhibit strong structural similarity and appear to function by a similar mechanism. Met8p, from Saccharomyces cerevisiae, catalyses ferrochelation during the synthesis of sirohaem, and the structure reveals a novel chelatase architecture whereby both ferrochelation and NAD(+)-dependent dehydrogenation take place in a single bifunctional active site. Asp-141 appears to participate in both catalytic reactions. The final common biosynthetic step in tetrapyrrole biosynthesis is the generation of uroporphyrinogen by uroporphyrinogen III synthase, whereby the D ring of hydroxymethylbilane is flipped during ring closure to generate the asymmetrical structure of uroporphyrinogen III. The recently derived structure of uroporphyrinogen III synthase reveals a bi-lobed structure in which the active site lies between the domains.


Assuntos
Quelantes , Metais , Uroporfirinogênio III Sintetase/química , Bacillus subtilis/enzimologia , Humanos , Modelos Moleculares , Estrutura Secundária de Proteína , Pirróis/química , Saccharomyces cerevisiae/enzimologia , Tetrapirróis
18.
FEBS Lett ; 525(1-3): 25-8, 2002 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-12163155

RESUMO

Uroporphyrinogen III synthase from the cyanobacterium Anacystis nidulans was overproduced in Escherichia coli and analyzed by site specific mutagenesis. Of the nine conserved amino acids altered, only a single tyrosine mutant (Y166F) showed any significant decrease in activity suggesting this residue is critical for proper substrate binding and/or catalysis.


Assuntos
Cianobactérias/enzimologia , Tirosina/metabolismo , Uroporfirinogênio III Sintetase/química , Uroporfirinogênio III Sintetase/metabolismo , Sequência de Aminoácidos , Substituição de Aminoácidos , Catálise , Cromatografia Líquida de Alta Pressão , Sequência Conservada , Eletroforese em Gel de Poliacrilamida , Ativação Enzimática/fisiologia , Dados de Sequência Molecular , Estrutura Molecular , Mutagênese Sítio-Dirigida , Relação Estrutura-Atividade , Tirosina/genética , Uroporfirinogênio III Sintetase/genética
19.
EMBO J ; 20(21): 5832-9, 2001 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-11689424

RESUMO

Uroporphyrinogen III synthase, U3S, the fourth enzyme in the porphyrin biosynthetic pathway, catalyzes cyclization of the linear tetrapyrrole, hydroxymethylbilane, to the macrocyclic uroporphyrino gen III, which is used in several different pathways to form heme, siroheme, chlorophyll, F(430) and vitamin B(12). U3S activity is essential in all organisms, and decreased activity in humans leads to the autosomal recessive disorder congenital erythropoetic porphyria. We have determined the crystal structure of recombinant human U3S at 1.85 A resolution. The protein folds into two alpha/beta domains connected by a beta-ladder. The active site appears to be located between the domains, and variations in relative domain positions observed between crystallographically independent molecules indicates the presence of flexibility that may be important in the catalytic cycle. Possible mechanisms of catalysis were probed by mutating each of the four invariant residues in the protein that have titratable side chains. Additionally, six other highly conserved and titratable side chains were also mutated. In no case, however, did one of these mutations abolish enzyme activity, suggesting that the mechanism does not require acid/base catalysis.


Assuntos
Modelos Moleculares , Uroporfirinogênio III Sintetase/química , Sítios de Ligação/fisiologia , Cristalografia por Raios X , Humanos , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Mutação/genética , Porfiria Eritropoética/genética , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína/fisiologia , Proteínas Recombinantes/química , Homologia de Sequência de Aminoácidos , Relação Estrutura-Atividade , Uroporfirinogênio III Sintetase/genética , Uroporfirinogênio III Sintetase/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...