Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Food Funct ; 15(10): 5439-5449, 2024 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-38650575

RESUMO

Barley, rich in bioactive components including dietary fiber, polyphenolic compounds and functional proteins, exhibits health benefits such as regulating glucose and lipid metabolism. Previous studies have found that the content and composition of free phenolic acids in barley may be significantly changed by fermentation with the laboratory patented strain Lactobacillus plantarum dy-1 (L. p dy-1), but the mechanism of enzymatic release of phenolic acid remains to be elucidated. Based on this, this study aimed to identify the key enzyme in L. p dy-1 responsible for releasing the bound phenolic acid and to further analyze its enzymatic properties. The Carbohydrate-Active enZYmes database revealed that L. p dy-1 encodes 7 types of auxiliary enzymes, among which we have identified a membrane sulfatase. The enzyme gene LPMS05445 was heterologous to that expressed in E. coli, and a recombinant strain was induced to produce the target protein and purified. The molecular weight of the purified enzyme was about 59.9 kDa, with 578.21 U mg-1 enzyme activity. The optimal temperature and pH for LPMS05445 expression were 40 °C and 7.0, respectively. Furthermore, enzymatic hydrolysis by LPMS05445 can obviously change the surface microstructure of dietary fiber from barley bran and enhance the release of bound phenolic acid, thereby increasing the free phenolic acid content and improving its physiological function. In conclusion, sulfatase produced by Lactobacillus plantarum dy-1 plays a key role in releasing bound phenolic acids during the fermentation of barley.


Assuntos
Lactobacillus plantarum , Sulfatases , Lactobacillus plantarum/enzimologia , Lactobacillus plantarum/metabolismo , Lactobacillus plantarum/genética , Sulfatases/metabolismo , Sulfatases/genética , Sulfatases/química , Hordeum , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/química , Fermentação , Hidroxibenzoatos/metabolismo , Concentração de Íons de Hidrogênio , Escherichia coli/genética , Temperatura , Fibras na Dieta/metabolismo
2.
Org Lett ; 25(12): 2001-2005, 2023 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-36662590

RESUMO

α-Formylglycine (fGly) is a rare residue located in the active site of sulfatases and serves as a precursor to pharmaceutically relevant motifs. The installation of fGly motifs into peptides is currently challenging due to degradation under the acidic and nucleophile-rich conditions accompanying resin cleavage during solid-phase peptide synthesis. We report the synthesis of acid- and nucleophile-tolerant α-formylglycine building blocks from vitamin C and use them to prepare callyaerin A, a macrocyclic peptide containing an fGly-derived motif.


Assuntos
Alanina , Técnicas de Síntese em Fase Sólida , Alanina/química , Glicina/química , Sulfatases/química , Sulfatases/metabolismo , Peptídeos/química
3.
Carbohydr Polym ; 271: 118449, 2021 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-34364583

RESUMO

Fucoidans are a class of sulfated fucose-containing bioactive polysaccharides produced by brown algae. The biological effects exhibited by fucoidans are thought to be related to their sulfation. However, the lack of methods for sulfation control does not allow for a reliable conclusion about the influence of the position of certain sulfate groups on the observed biological effects. We identified the gene encoding the endo-acting fucoidan sulfatase swf5 in the marine bacterium Wenyingzhuangia fucanilytica CZ1127T. This is the first report on the sequence of fucoidan endo-sulfatase. Sulfatase SWF5 belongs to the subfamily S1_22 of the family S1. SWF5 was shown to remove 4O-sulfation in fucoidans composed from the alternating α-(1→3)- and α-(1→4)-linked residues of sulfated L-fucose but not from fucoidans with the α-(1→3)-linked backbone. The endo-sulfatase was used to selectively prepare 4O-desulfated fucoidan derivatives. It was shown that the 4O-desulfated fucoidans inhibit colony formation of DLD-1 and MCF-7 cells less effectively than unmodified fucoidans. Presumably, 4O-sulfation makes a significant contribution to the anticancer activity of fucoidans.


Assuntos
Antineoplásicos/farmacologia , Polissacarídeos/farmacologia , Sulfatases/química , Sequência de Aminoácidos , Antineoplásicos/síntese química , Proteínas de Bactérias/química , Proteínas de Bactérias/isolamento & purificação , Linhagem Celular Tumoral , Ensaios de Seleção de Medicamentos Antitumorais , Flavobacteriaceae/enzimologia , Humanos , Estrutura Molecular , Polissacarídeos/síntese química , Especificidade por Substrato , Sulfatases/isolamento & purificação
4.
Angew Chem Int Ed Engl ; 60(36): 19957-19964, 2021 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-34164914

RESUMO

Aminomalonate (Ama) is a widespread structural motif in Nature, whereas its biosynthetic route is only partially understood. In this study, we show that a radical S-adenosylmethionine (rSAM) enzyme involved in cyclophane biosynthesis exhibits remarkable catalytic promiscuity. This enzyme, named three-residue cyclophane forming enzyme (3-CyFE), mainly produces cyclophane in vivo, whereas it produces formylglycine (FGly) as a major product and barely produce cyclophane in vitro. Importantly, the enzyme can further oxidize FGly to produce Ama. Bioinformatic study revealed that 3-CyFEs have evolved from a common ancestor with anaerobic sulfatase maturases (anSMEs), and possess a similar set of catalytic residues with anSMEs. Remarkably, the enzyme does not need leader peptide for activity and is fully active on a truncated peptide containing only 5 amino acids of the core sequence. Our work discloses the first ribosomal path towards Ama formation, providing a possible hint for the rich occurrence of Ama in Nature.


Assuntos
Malonatos/metabolismo , Peptídeos/metabolismo , S-Adenosilmetionina/metabolismo , Sulfatases/metabolismo , Radicais Livres/química , Radicais Livres/metabolismo , Malonatos/química , Estrutura Molecular , Peptídeos/química , Processamento de Proteína Pós-Traducional , S-Adenosilmetionina/química , Sulfatases/química
5.
J Med Chem ; 64(12): 8599-8606, 2021 06 24.
Artigo em Inglês | MEDLINE | ID: mdl-34096701

RESUMO

Glioblastoma multiforme (GBM) is a highly invasive and aggressive malignant glioma. Current treatment modalities are unable to significantly prolong survival in patients diagnosed with glioblastoma, so more effective strategies of antitumor treatments are in urgent demand. Here, we found that lysosomal sulfatase expression was significantly correlated with poor prognosis of GBM. Hence, a new probe, MNG, was developed with a new protocol utilizing glucose groups to detect lysosomal sulfatase. It also exhibits potential for monitoring GBM cells, depending on the hyperactive lysosomal sulfatase expression of tumor cells. Meantime, we identified that sulbactam as the first reported lysosomal sulfatase inhibitor inhibits the tumor growth of GBM. Collectively, our work highlights that lysosomal sulfatase was detected using a new protocol and its potential as a therapeutic target in GBM and reveals a distinct mechanism that sulbactam inhibits cell proliferation related to lysosomal sulfatase, indicating that sulbactam could be a promising therapeutic agent against GBM.


Assuntos
Antineoplásicos/farmacologia , Proliferação de Células/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Glioblastoma/tratamento farmacológico , Sulbactam/farmacologia , Sulfatases/antagonistas & inibidores , Linhagem Celular Tumoral , Corantes Fluorescentes/química , Glioblastoma/diagnóstico , Glioblastoma/enzimologia , Glucosídeos/química , Humanos , Lisossomos/enzimologia , Naftalimidas/química , Prognóstico , Sulfatases/análise , Sulfatases/química
6.
Sci Rep ; 11(1): 6727, 2021 03 24.
Artigo em Inglês | MEDLINE | ID: mdl-33762621

RESUMO

Direct delivery of therapeutic enzymes to the Central Nervous System requires stringent formulation design. Not only should the formulation design consider the delicate balance of existing ions, proteins, and osmolality in the cerebrospinal fluid, it must also provide long term efficacy and stability for the enzyme. One fundamental approach to this predicament is designing formulations with no buffering species. In this study, we report a high concentration, saline-based formulation for a human sulfatase for its delivery into the intrathecal space. A high concentration formulation (≤ 40 mg/mL) was developed through a series of systematic studies that demonstrated the feasibility of a self-buffered formulation for this molecule. The self-buffering capacity phenomenon was found to be a product of both the protein itself and potentially the residual phosphates associated with the protein. To date, the self-buffered formulation for this molecule has been stable for up to 4 years when stored at 5 ± 3 °C, with no changes either in the pH values or other quality attributes of the molecule. The high concentration self-buffered protein formulation was also observed to be stable when exposed to multiple freeze-thaw cycles and was robust during in-use and agitation studies.


Assuntos
Sistema Nervoso Central/metabolismo , Sistemas de Liberação de Medicamentos , Sulfatases/administração & dosagem , Soluções Tampão , Sistema Nervoso Central/efeitos dos fármacos , Humanos , Peptídeos/química , Peptídeos/metabolismo , Fosfatos , Estabilidade Proteica , Proteínas/química , Proteínas/metabolismo , Soluções , Sulfatases/química
7.
Biochem J ; 477(20): 3963-3983, 2020 10 30.
Artigo em Inglês | MEDLINE | ID: mdl-33120425

RESUMO

Sulfatases constitute a family of enzymes that specifically act in the hydrolytic degradation of sulfated metabolites by removing sulfate monoesters from various substrates, particularly glycolipids and glycosaminoglycans. A common essential feature of all known eukaryotic sulfatases is the posttranslational modification of a critical cysteine residue in their active site by oxidation to formylglycine (FGly), which is mediated by the FGly-generating enzyme in the endoplasmic reticulum and is indispensable for catalytic activity. The majority of the so far described sulfatases localize intracellularly to lysosomes, where they act in different catabolic pathways. Mutations in genes coding for lysosomal sulfatases lead to an accumulation of the sulfated substrates in lysosomes, resulting in impaired cellular function and multisystemic disorders presenting as lysosomal storage diseases, which also cover the mucopolysaccharidoses and metachromatic leukodystrophy. Bioinformatics analysis of the eukaryotic genomes revealed, besides the well described and long known disease-associated sulfatases, additional genes coding for putative enzymes with sulfatases activity, including arylsulfatase G as well as the arylsulfatases H, I, J and K, respectively. In this article, we review current knowledge about lysosomal sulfatases with a special focus on the just recently characterized family members arylsulfatase G and arylsulfatase K.


Assuntos
Doenças por Armazenamento dos Lisossomos/enzimologia , Lisossomos/metabolismo , Sulfatases/genética , Sulfatases/metabolismo , Animais , Domínio Catalítico , Modelos Animais de Doenças , Retículo Endoplasmático/metabolismo , Glicina/análogos & derivados , Glicina/química , Humanos , Lisossomos/enzimologia , Filogenia , Processamento de Proteína Pós-Traducional , Sulfatases/química , Sulfatases/deficiência
8.
Artigo em Inglês | MEDLINE | ID: mdl-31195190

RESUMO

Metachromatic Leukodystrophy (MLD) and Multiple Sulfatase Deficiency (MSD) are rare and ultra-rare lysosomal storage diseases. Due to enzyme defects, patients are unable to split the sulfategroup from the respective substrates. In MSD all sulfatases are affected due to a defect of the Sulfatase Modifying Factor 1 (SUMF1) gene coding for the formylglycine generating enzyme (FGE) necessary for the modification of the active site of sulfatases. In MLD mutations in the arylsulfatase A (ARSA) gene cause ARSA deficiency with subsequent accumulation of 3-sulfogalactocerebroside especially in oligodendrocytes. The clinical consequence is demyelination and a devastating neurological disease. Enzyme replacement therapy (ERT) with recombinant human arylsulfatase A (rhARSA), gene therapy, and stem cell transplantation are suggested as new therapeutic options. The aim of our study was to characterize rhARSA concerning its substrate specificity using analytical isotachophoresis (ITP). Substrate specificity could be demonstrated by sulfate splitting from the natural substrates 3-sulfogalactocerebroside and ascorbyl-2-sulfate and the artificial substrate p-nitrocatecholsulfate, whereas galactose-6-sulfate, a substrate of galactose-6­sulfurylase, was totally resistant. In contrast, leukocyte extracts of healthy donors were able to split sulfate also from galactose-6-sulfate. The ITP method allows therefore a rapid and simple differentiation between samples of MLD and MSD patients and healthy donors. Therefore, the isotachophoretic diagnostic assay from leukocyte extracts described here provides a fast and efficient way for the diagnosis of MLD and MSD patients and an elegant system to differentiate between these diseases in one assay.


Assuntos
Cerebrosídeo Sulfatase/química , Ensaios Enzimáticos/métodos , Isotacoforese/métodos , Leucócitos/enzimologia , Leucodistrofia Metacromática/enzimologia , Doença da Deficiência de Múltiplas Sulfatases/enzimologia , Sulfatases/química , Cerebrosídeo Sulfatase/genética , Cerebrosídeo Sulfatase/metabolismo , Humanos , Cinética , Leucodistrofia Metacromática/diagnóstico , Leucodistrofia Metacromática/genética , Doença da Deficiência de Múltiplas Sulfatases/diagnóstico , Doença da Deficiência de Múltiplas Sulfatases/genética , Proteínas Recombinantes/análise , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Sulfatases/genética , Sulfatases/metabolismo , Sulfatos/química , Sulfatos/metabolismo
9.
Biol Chem ; 400(3): 289-297, 2019 02 25.
Artigo em Inglês | MEDLINE | ID: mdl-30291781

RESUMO

Site-specific bioconjugation strategies offer many possibilities for directed protein modifications. Among the various enzyme-based conjugation protocols, formylglycine-generating enzymes allow to posttranslationally introduce the amino acid Cα-formylglycine (FGly) into recombinant proteins, starting from cysteine or serine residues within distinct consensus motifs. The aldehyde-bearing FGly-residue displays orthogonal reactivity to all other natural amino acids and can, therefore, be used for site-specific labeling reactions on protein scaffolds. In this review, the state of research on catalytic mechanisms and consensus motifs of different formylglycine-generating enzymes, as well as labeling strategies and applications of FGly-based bioconjugations are presented.


Assuntos
Glicina/análogos & derivados , Sulfatases/metabolismo , Glicina/biossíntese , Glicina/química , Glicina/metabolismo , Humanos , Modelos Moleculares , Estrutura Molecular , Sulfatases/química
10.
Insect Sci ; 25(6): 946-958, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28569426

RESUMO

The diamondback moth, Plutella xylostella (L.), uses sulfatases (SULF) to counteract the glucosinolate-myrosinase defensive system that cruciferous plants have evolved to deter insect feeding. Sulfatase activity is regulated by post-translational modification of a cysteine residue by sulfatase modifying factor 1 (SUMF1). We identified 12 SULF genes (PxylSulfs) and two SUMF1 genes (PxylSumf1s) in the P. xylostella genome. Phylogenetic analysis of SULFs and SUMFs from P. xylostella, Bombyx mori, Manduca sexta, Heliconius melpomene, Danaus plexippus, Drosophila melanogaster, Tetranychus urticae and Homo sapiens showed that the SULFs were clustered into five groups, and the SUMFs could be divided into two groups. Profiling of the expression of PxylSulfs and PxylSumfs by RNA-seq and by quantitative real-time polymerase chain reaction showed that two glucosinolate sulfatase genes (GSS), PxylSulf2 and PxylSulf3, were primarily expressed in the midgut of 3rd- and 4th-instar larvae. Moreover, expression of sulfatases PxylSulf2, PxylSulf3 and PxylSulf4 were correlated with expression of the sulfatases modifying factor PxylSumf1a. The findings from this study provide new insights into the structure and expression of SUMF1 and PxylSulf genes that are considered to be key factors for the evolutionary success of P. xylostella as a specialist herbivore of cruciferous plants.


Assuntos
Regulação Enzimológica da Expressão Gênica , Proteínas de Insetos/química , Proteínas de Insetos/metabolismo , Mariposas/enzimologia , Sulfatases/química , Sulfatases/metabolismo , Sequência de Aminoácidos , Animais , Sequência Conservada , Proteínas de Insetos/genética , Mariposas/metabolismo , Especificidade de Órgãos , Filogenia , Domínios Proteicos , Sulfatases/genética
11.
Gene ; 634: 53-61, 2017 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-28882567

RESUMO

Maturation of type I sulfatases requires the conversion of the cysteine (Cys) or serine (Ser) present in the active site to formylglycine (FGly). This activation represents a limiting step during the production of recombinant sulfatases in bacteria and eukaryotic hosts. AslB, YdeM and YidF have been proposed to participate in the activation of sulfatases in Escherichia coli. In this study, we combined in-silico and experimental approaches to study the interaction between Escherichia coli BL21(DE3) AslB and human sulfatases, more specifically iduronate-2-sulfate sulfatase (IDS) and N-acetylgalactosamine-6-sulfate sulfatase (GALNS). In-silico results show that AslB has a higher affinity for the residual motif of GALNS (-9.4kcalmol-1), Cys- and Ser-type, than for the one of IDS (-8.0kcalmol-1). However, the distance between the AslB active residue and the target motif favors the interaction with IDS (4.4Å) more than with GALNS (5.5Å). Experimental observations supported in-silico results where the co-expression of AslB with GALNS Cys- and Ser-type presented an activity increment of 2.0- and 1.5-fold compared to the control cultures, lacking overexpressed AslB. Similarly, IDS activity was increased in 4.6-fold when co-expressed with AslB. The higher sulfatase activity of AslB-IDS suggests that the distance between the AslB active residue and the motif target is a key parameter for the in-silico search of potential sulfatase activators. In conclusion, our results suggest that AslB is involve in the maturation of heterologous human sulfatases in E. coli BL21(DE3), and that it can have important implications in the production of recombinant sulfatases for therapeutic purposes and research.


Assuntos
Condroitina Sulfatases/metabolismo , Escherichia coli/enzimologia , Glicoproteínas/metabolismo , Sulfatases/química , Sulfatases/metabolismo , Domínio Catalítico , Condroitina Sulfatases/química , Cisteína/metabolismo , Ativação Enzimática , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Glicoproteínas/química , Humanos , Modelos Moleculares , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Ligação Proteica , Proteínas Recombinantes/metabolismo , Serina/metabolismo
12.
Protein Eng Des Sel ; 30(7): 477-488, 2017 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-28651356

RESUMO

Sulfatases are a family of enzymes (sulfuric ester hydrolases, EC 3.1.6.-) that catalyze the hydrolysis of a wide array of sulfate esters. To date, despite the discovery of many sulfatase genes and the accumulation of data on numerous sulfated molecules, the number of characterized enzymes that are key players in sulfur metabolism remains extremely limited. While mammalian sulfatases are well studied due to their involvement in a wide range of normal and pathological biological processes, lower eukaryotic sulfatases, especially fungal sulfatases, have not been thoroughly investigated at the biochemical and structural level. In this paper, we describe the molecular cloning of Fusarium proliferatum sulfatase (F.p.Sulf-6His), its recombinant expression in Pichia pastoris as a soluble and active cytosolic enzyme and its detailed characterization. Gel filtration and native electrophoretic experiments showed that this recombinant enzyme exists as a tetramer in solution. The enzyme is thermo-sensitive, with an optimal temperature of 25°C. The optimal pH value for the hydrolysis of sulfate esters and stability of the enzyme was 6.0. Despite the absence of the post-translational modification of cysteine into Cα-formylglycine, the recombinant F.p.Sulf-6His has remarkably stable catalytic activity against p-nitrophenol sulfate, with kcat = 0.28 s-1 and Km = 2.45 mM, which indicates potential use in the desulfating processes. The currently proposed enzymatic mechanisms of sulfate ester hydrolysis do not explain the appearance of catalytic activity for the unmodified enzyme. According to the available models, the unmodified enzyme is not able to perform multiple catalytic acts; therefore, the enzymatic mechanism of sulfate esters hydrolysis remains to be fully elucidated.


Assuntos
Sequência de Aminoácidos/genética , Fusarium/enzimologia , Processamento de Proteína Pós-Traducional/genética , Sulfatases/genética , Sítios de Ligação , Clonagem Molecular , Regulação Enzimológica da Expressão Gênica , Pichia/genética , Estrutura Quaternária de Proteína , Especificidade por Substrato , Sulfatases/biossíntese , Sulfatases/química
13.
Food Funct ; 8(7): 2419-2424, 2017 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-28573284

RESUMO

Phenolic compounds are present in human fluids (plasma and urine) mainly as glucuronidated and sulfated metabolites. Up to now, due to the unavailability of standards, enzymatic hydrolysis has been the method of choice in analytical chemistry to quantify these phase II phenolic metabolites. Enzymatic hydrolysis procedures vary in enzyme concentration, pH and temperature; however, there is a lack of knowledge about the stability of polyphenols in their free form during the process. In this study, we evaluated the stability of 7 phenolic acids, 2 flavonoids and 3 prenylflavanoids in urine during enzymatic hydrolysis to assess the suitability of this analytical procedure, using three different concentrations of ß-glucuronidase/sulfatase enzymes from Helix pomatia. The results indicate that enzymatic hydrolysis negatively affected the recovery of the precursor and free-form polyphenols present in the sample. Thus, enzymatic hydrolysis does not seem an ideal analytical strategy to quantify glucuronidated and sulfated polyphenol metabolites.


Assuntos
Técnicas de Química Analítica/métodos , Glucuronidase/química , Caracois Helix/enzimologia , Polifenóis/química , Sulfatases/química , Sulfatos/química , Animais , Biocatálise , Humanos , Hidrólise , Polifenóis/urina
14.
Chembiochem ; 18(2): 161-165, 2017 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-27862795

RESUMO

Formylglycine-generating enzyme (FGE) is an O2 -utilizing oxidase that converts specific cysteine residues of client proteins to formylglycine. We show that CuI is an integral cofactor of this enzyme and binds with high affinity (KD =of 10-17 m) to a pair of active-site cysteines. These findings establish FGE as a novel type of copper enzyme.


Assuntos
Cobre/metabolismo , Glicina/análogos & derivados , Sulfatases/metabolismo , Actinobacteria/enzimologia , Biocatálise , Domínio Catalítico , Cobre/química , Cisteína/química , Cisteína/metabolismo , Espectroscopia de Ressonância de Spin Eletrônica , Glicina/química , Glicina/metabolismo , Humanos , Cinética , Simulação de Dinâmica Molecular , Sulfatases/química
15.
Chembiochem ; 16(15): 2147-50, 2015 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-26403223

RESUMO

Formylglycine-generating enzymes (FGEs) catalyze O2 -dependent conversion of specific cysteine residues of arylsulfatases and alkaline phosphatases into formylglycine. The ability also to introduce unique aldehyde functions into recombinant proteins makes FGEs a powerful tool for protein engineering. One limitation of this technology is poor in vitro activity of reconstituted FGEs. Although FGEs have been characterized as cofactor-free enzymes we report that the addition of one equivalent of Cu(I) increases catalytic efficiency more than 20-fold and enables the identification of stereoselective C-H bond cleavage at the substrate as the rate-limiting step. These findings remove previous limitations of FGE-based protein engineering and also pose new questions about the catalytic mechanism of this O2 -utilizing enzyme.


Assuntos
Cobre/metabolismo , Glicina/análogos & derivados , Sulfatases/metabolismo , Actinobacteria/enzimologia , Biocatálise , Cobre/química , Glicina/biossíntese , Estrutura Molecular , Mycobacterium smegmatis/enzimologia , Oxigênio/química , Oxigênio/metabolismo , Sulfatases/química
16.
FEBS J ; 282(17): 3262-74, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26077311

RESUMO

C α-formylglycine (FGly) is the catalytic residue of sulfatases in eukaryotes. It is generated by a unique post-translational modification catalysed by the FGly-generating enzyme (FGE) in the endoplasmic reticulum. FGE oxidizes a cysteine residue within the conserved CxPxR sequence motif of nascent sulfatase polypeptides to FGly. Here we show that this oxidation is strictly dependent on molecular oxygen (O2) and consumes 1 mol O2 per mol FGly formed. For maximal activity FGE requires an O2 concentration of 9% (105 µM). Sustained FGE activity further requires the presence of a thiol-based reductant such as DTT. FGly is also formed in the absence of DTT, but its formation ceases rapidly. Thus inactivated FGE accumulates in which the cysteine pair Cys336/Cys341 in the catalytic site is oxidized to form disulfide bridges between either Cys336 and Cys341 or Cys341 and the CxPxR cysteine of the sulfatase. These results strongly suggest that the Cys336/Cys341 pair is directly involved in the O2 -dependent conversion of the CxPxR cysteine to FGly. The available data characterize eukaryotic FGE as a monooxygenase, in which Cys336/Cys341 disulfide bridge formation donates the electrons required to reduce one oxygen atom of O2 to water while the other oxygen atom oxidizes the CxPxR cysteine to FGly. Regeneration of a reduced Cys336/Cys341 pair is accomplished in vivo by a yet unknown reductant of the endoplasmic reticulum or in vitro by DTT. Remarkably, this monooxygenase reaction utilizes O2 without involvement of any activating cofactor.


Assuntos
Alanina/análogos & derivados , Glicina/análogos & derivados , Oxigenases de Função Mista/metabolismo , Oxigênio/metabolismo , Sulfatases/metabolismo , Alanina/química , Alanina/metabolismo , Animais , Baculoviridae/genética , Biocatálise , Domínio Catalítico , Cisteína/química , Cisteína/metabolismo , Dissulfetos/química , Ditiotreitol/química , Ensaios Enzimáticos , Expressão Gênica , Glicina/química , Glicina/metabolismo , Humanos , Cinética , Oxigenases de Função Mista/química , Oxigenases de Função Mista/genética , Oxirredução , Oxirredutases atuantes sobre Doadores de Grupo Enxofre , Oxigênio/química , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Células Sf9 , Spodoptera , Sulfatases/química , Sulfatases/genética
17.
J Biol Chem ; 290(25): 15730-15745, 2015 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-25931126

RESUMO

To further our aim of synthesizing aldehyde-tagged proteins for research and biotechnology applications, we developed methods for recombinant production of aerobic formylglycine-generating enzyme (FGE) in good yield. We then optimized the FGE biocatalytic reaction conditions for conversion of cysteine to formylglycine in aldehyde tags on intact monoclonal antibodies. During the development of these conditions, we discovered that pretreating FGE with copper(II) is required for high turnover rates and yields. After further investigation, we confirmed that both aerobic prokaryotic (Streptomyces coelicolor) and eukaryotic (Homo sapiens) FGEs contain a copper cofactor. The complete kinetic parameters for both forms of FGE are described, along with a proposed mechanism for FGE catalysis that accounts for the copper-dependent activity.


Assuntos
Proteínas de Bactérias/química , Coenzimas/química , Cobre/química , Streptomyces coelicolor/enzimologia , Sulfatases/química , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Coenzimas/metabolismo , Cobre/metabolismo , Cisteína/química , Cisteína/metabolismo , Humanos , Oxirredutases atuantes sobre Doadores de Grupo Enxofre , Streptomyces coelicolor/genética , Sulfatases/genética , Sulfatases/metabolismo
18.
J Org Chem ; 79(5): 1995-2005, 2014 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-24555731

RESUMO

Type I sulfatases catalyze the hydrolysis of sulfate esters through S-O bond cleavage and possess a catalytically essential formylglycine (FGly) active-site residue that is post-translationally derived from either cysteine or serine. Type I sulfatases are inactivated by aryl sulfamates in a time-dependent, irreversible, and active-site directed manner consistent with covalent modification of the active site. We report a theoretical (SCS-MP2//B3LYP) and experimental study of the uncatalyzed and enzyme-catalyzed hydrolysis of aryl sulfates and sulfamates. In solution, aryl sulfate monoanions undergo hydrolysis by an S(N)2 mechanism whereas aryl sulfamate monoanions follow an S(N)1 pathway with SO2NH as an intermediate; theory traces this difference to the markedly greater stability of SO2NH versus SO3. For Pseudomonas aeruginosa arylsulfatase-catalyzed aryl sulfate hydrolysis, Brønsted analysis (log(V(max)/K(M)) versus leaving group pK(a) value) reveals ß(LG) = -0.86 ± 0.23, consistent with an S(N)2 at sulfur reaction but substantially smaller than that reported for uncatalyzed hydrolysis (ß(LG) = -1.81). Common to all proposed mechanisms of sulfatase catalysis is a sulfated FGly intermediate. Theory indicates a ≥26 kcal/mol preference for the intermediate to release HSO4(-) by an E2 mechanism, rather than alkaline phosphatase-like S(N)2 substitution by water. An evaluation of the stabilities of various proposed end-products of sulfamate-induced sulfatase inactivation highlights that an imine N-sulfate derived from FGly is the most likely irreversible adduct.


Assuntos
Glicina/análogos & derivados , Sulfatases/química , Sulfatos/química , Ácidos Sulfônicos/química , Ésteres do Ácido Sulfúrico/química , Sítios de Ligação , Catálise , Ésteres , Glicina/química , Concentração de Íons de Hidrogênio , Hidrólise , Modelos Teóricos
19.
Anal Bioanal Chem ; 405(23): 7259-67, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23857141

RESUMO

Alkyl esters of p-hydroxybenzoic acid (parabens) are a family of compounds that have been in use since the 1920s as preservatives in cosmetic formulations, with one of the lowest rates of skin problems reported in dermatological patients. However, in the last few years, many scientific publications have demonstrated that parabens are weak endocrine disruptors, meaning that they can interfere with the function of endogenous hormones, increasing the risk of breast cancer. In the present work, a new sample treatment method is introduced based on dispersive liquid-liquid microextraction for the extraction of the most commonly used parabens (methyl-, ethyl-, propyl-, and butylparaben) from human serum samples followed by separation and quantification using ultrahigh performance liquid chromatography-tandem mass spectrometry. The method involves an enzymatic treatment to quantify the total content of parabens. The extraction parameters (solvent and disperser solvent, extractant and dispersant volume, pH of the sample, salt addition, and extraction time) were accurately optimized using multivariate optimization strategies. Ethylparaben ring (13)C6-labeled was used as surrogate. Limits of quantification ranging from 0.2 to 0.7 ng mL(-1) and an interday variability (evaluated as relative standard deviations) from 3.8 to 11.9 % were obtained. The method was validated using matrix-matched calibration standard and a spike recovery assay. Recovery rates for spiked samples ranged from 96 to 106 %, and a good linearity up to concentrations of 100 ng mL(-1) was obtained. The method was satisfactorily applied for the determination of target compounds in human serum samples.


Assuntos
Microextração em Fase Líquida/métodos , Parabenos/análise , Cromatografia Líquida de Alta Pressão/métodos , Glucuronidase/química , Humanos , Concentração de Íons de Hidrogênio , Limite de Detecção , Conservantes Farmacêuticos/química , Soro/química , Solventes , Sulfatases/química
20.
Biomed Chromatogr ; 27(10): 1280-95, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23649485

RESUMO

Glucuronidation and sulfation represent two major pathways in phase II drug metabolism in humans and other mammalian species. The great majority of drugs, for example, polyphenols, flavonoids and anthraquinones, could be transformed into sulfated and glucuronidated conjugates simultaneously and extensively in vivo. The pharmacological activities of drug conjugations are normally decreased compared with those of their free forms. However, some drug conjugates may either bear biological activities themselves or serve as excellent sources of biologically active compounds. As the bioactivities of drugs are thought to be relevant to the kinetics of their conjugates, it is essential to study the pharmacokinetic behaviors of the conjugates in more detail. Unfortunately, the free forms of drugs cannot be detected directly in most cases if their glucuronides and sulfates are the predominant forms in biological samples. Nevertheless, an initial enzymatic hydrolysis step using ß-glucuronidase and/or sulfatase is usually performed to convert the glucuronidated and/or sulfated conjugates to their free forms prior to the extraction, purification and other subsequent analysis steps in the literature. This review provides fundamental information on drug metabolism pathways, the bio-analytical strategies for the quantification of various drug conjugates, and the applications of the analytical methods to pharmacokinetic studies.


Assuntos
Glucuronidase/análise , Preparações Farmacêuticas/análise , Preparações Farmacêuticas/metabolismo , Sulfatases/análise , Animais , Fracionamento Químico , Cromatografia Líquida de Alta Pressão , Cromatografia Gasosa-Espectrometria de Massas , Glucuronidase/química , Glucuronidase/isolamento & purificação , Glucuronidase/metabolismo , Humanos , Preparações Farmacêuticas/química , Preparações Farmacêuticas/isolamento & purificação , Sulfatases/química , Sulfatases/isolamento & purificação , Sulfatases/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA