Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Oncogene ; 35(38): 5043-55, 2016 09 22.
Artigo em Inglês | MEDLINE | ID: mdl-27041583

RESUMO

Heparan sulfate (HS) proteoglycan chains are key components of the breast tumor microenvironment that critically influence the behavior of cancer cells. It is established that abnormal synthesis and processing of HS play a prominent role in tumorigenesis, albeit mechanisms remain mostly obscure. HS function is mainly controlled by sulfotransferases, and here we report a novel cellular and pathophysiological significance for the 3-O-sulfotransferase 3-OST3A (HS3ST3A), catalyzing the final maturation step of HS, in breast cancer. We show that 3-OST3A is epigenetically repressed in all breast cancer cell lines of a panel representative of distinct molecular subgroups, except in human epidermal growth factor receptor 2-positive (HER2+) sloan-kettering breast cancer (SKBR3) cells. Epigenetic mechanisms involved both DNA methylation and histone modifications, producing different repressive chromatin environments depending on the cell molecular signature. Gain and loss of function experiments by cDNA and siRNA transfection revealed profound effects of 3-OST3A expression on cell behavior including apoptosis, proliferation, response to trastuzumab in vitro and tumor growth in xenografted mice. 3-OST3A exerted dual activities acting as tumor-suppressor in lumA-michigan cancer foundation (MCF)-7 and triple negative-MD Anderson (MDA) metastatic breast (MB)-231 cells, or as an oncogenic factor in HER2+-SKBR3 cells. Mechanistically, fluorescence-resonance energy transfer-fluorescence-lifetime imaging microscopy experiments indicated that the effects of 3-OST3A in MCF-7 cells were mediated by altered interactions between HS and fibroblast growth factor-7 (FGF-7). Further, this interplay between HS and FGF-7 modulated downstream ERK, AKT and p38 cascades, suggesting that altering 3-O-sulfation affects FGFR2IIIb-mediated signaling. Corroborating our cellular data, a clinical study conducted in a cohort of breast cancer patients uncovered that, in HER2+ patients, high level expression of 3-OST3A in tumors was associated with reduced relapse-free survival. Our findings define 3-OST3A as a novel regulator of breast cancer pathogenicity, displaying tumor-suppressive or oncogenic activities in a cell- and tumor-dependent context, and demonstrate the clinical value of the HS-O-sulfotransferase 3-OST3A as a prognostic marker in HER2+ patients.


Assuntos
Biomarcadores Tumorais/genética , Neoplasias da Mama/genética , Receptor ErbB-2/genética , Sulfotransferases/genética , Animais , Neoplasias da Mama/patologia , Metilação de DNA/genética , Feminino , Heparitina Sulfato/genética , Humanos , Células MCF-7 , Camundongos , Prognóstico , Transdução de Sinais , Ensaios Antitumorais Modelo de Xenoenxerto
2.
FEBS Lett ; 580(8): 1953-8, 2006 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-16529747

RESUMO

We investigated the role of the stop transfer sequence of human UGT1A6 in ER assembly and enzyme activity. We found that this sequence was able to address and translocate the upstream lumenal domain into microsomal membranes in vitro co- and posttranslationally. The signal activity of this sequence was further demonstrated in HeLa cells by its ability to target and maintain the CD4 protein deleted from both the N-terminal signal peptide and C-terminal transmembrane domain into the ER. We showed that total or partial deletion of the stop transfer sequence of UGT1A6 severely impaired enzyme activity highlighting its importance in both membrane assembly and function.


Assuntos
Retículo Endoplasmático/metabolismo , Glucuronosiltransferase/química , Glucuronosiltransferase/metabolismo , Proteínas de Membrana/química , Proteínas de Membrana/metabolismo , Processamento de Proteína Pós-Traducional , Sinais Direcionadores de Proteínas/fisiologia , Região 5'-Flanqueadora/genética , Sequência de Aminoácidos , Retículo Endoplasmático/enzimologia , Células HeLa , Humanos , Dados de Sequência Molecular , Pichia/metabolismo , Estrutura Terciária de Proteína , Transporte Proteico , Deleção de Sequência/genética
3.
Proc Natl Acad Sci U S A ; 101(52): 18087-92, 2004 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-15601778

RESUMO

Osteoarthritis is a degenerative joint disease characterized by a progressive loss of articular cartilage components, mainly proteoglycans (PGs), leading to destruction of the tissue. We investigate a therapeutic strategy based on stimulation of PG synthesis by gene transfer of the glycosaminoglycan (GAG)-synthesizing enzyme, beta1,3-glucuronosyltransferase-I (GlcAT-I) to promote cartilage repair. We previously reported that IL-1beta down-regulated the expression and activity of GlcAT-I in primary rat chondrocytes. Here, by using antisense oligonucleotides, we demonstrate that GlcAT-I inhibition impaired PG synthesis and deposition in articular cartilage explants, emphasizing the crucial role of this enzyme in PG anabolism. Thus, primary chondrocytes and cartilage explants were engineered by lipid-mediated gene delivery to efficiently overexpress a human GlcAT-I cDNA. Interestingly, GlcAT-I overexpression significantly enhanced GAG synthesis and deposition as evidenced by (35)S-sulfate incorporation, histology, estimation of GAG content, and fluorophore-assisted carbohydrate electrophoresis analysis. Metabolic labeling and Western blot analyses further suggested that GlcAT-I expression led to an increase in the abundance rather than in the length of GAG chains. Importantly, GlcAT-I delivery was able to overcome IL-1beta-induced PG depletion and maintain the anabolic activity of chondrocytes. Moreover, GlcAT-I also restored PG synthesis to a normal level in cartilage explants previously depleted from endogenous PGs by IL-1beta-treatment. In concert, our investigations strongly indicated that GlcAT-I was able to control and reverse articular cartilage defects in terms of PG anabolism and GAG content associated with IL-1beta. This study provides a basis for a gene therapy approach to promote cartilage repair in degenerative joint diseases.


Assuntos
Cartilagem/patologia , Técnicas de Transferência de Genes , Glucuronosiltransferase/genética , Osteoartrite/terapia , Proteoglicanas/metabolismo , Animais , Western Blotting , Metabolismo dos Carboidratos , Cartilagem/metabolismo , Cartilagem Articular/patologia , Condrócitos/metabolismo , DNA Complementar/metabolismo , Regulação para Baixo , Vetores Genéticos , Humanos , Imuno-Histoquímica , Interleucina-1/metabolismo , Metabolismo dos Lipídeos , Oligonucleotídeos Antissenso/farmacologia , Ratos , Fatores de Tempo , Transfecção , Cicatrização
4.
Arthritis Rheum ; 44(2): 351-60, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11229466

RESUMO

OBJECTIVE: To assess the variations of galactose-beta-1,3-glucuronosyltransferase I (GlcAT-I) expression related to the decrease in proteoglycan synthesis mediated by interleukin-1beta (IL-1beta) in rat chondrocytes, and to evaluate the influence of glucosamine on the effects elicited by this proinflammatory cytokine. METHODS: Rat articular chondrocytes in primary monolayer cultures or encapsulated into alginate beads were treated with recombinant IL-1beta in the absence or presence (1.0-4.5 gm/liter) of glucosamine. Variations of GlcAT-I and expression of stromelysin 1 (matrix metalloproteinase 3 [MMP-3]) messenger RNA (mRNA) were evaluated by quantitative multistandard reverse transcriptase-polymerase chain reaction. In vitro enzymatic activity of GlcAT-I was measured by thin-layer chromatography, with radiolabeled UDP-glucuronic acid and a digalactoside derivative as substrates. Proteoglycan synthesis was determined by ex vivo incorporation of Na2-35SO4. Nitric oxide synthase and cyclooxygenase activities were monitored by the evaluation of nitrite (NO2-) and prostaglandin E2 (PGE2) produced in the culture medium, respectively. RESULTS: IL-1beta treatment resulted in a marked inhibition of GlcAT-I mRNA expression and in vitro catalytic activity, together with a decrease in proteoglycan synthesis. In addition, glucosamine was able to prevent, in a dose-dependent manner, the inhibitory effects of IL-1beta. In the same way, the amino sugar reduced NO2- and PGE2 production induced by IL-1beta. Finally, the up-regulation of stromelysin 1 (MMP-3) mRNA expression by IL-1beta was fully prevented by glucosamine. CONCLUSION: The results of this study suggest that the deleterious effect of IL-1beta on the anabolism of proteoglycan could involve the repression of GlcAT-I, a key enzyme in the biosynthesis of glycosaminoglycan. Glucosamine was highly effective in preventing these IL-1beta-mediated suppressive effects. The amino sugar also prevented the production of inflammatory mediators induced by the cytokine. This action could account for a possible beneficial effect of glucosamine on osteoarthritic articular cartilage.


Assuntos
Glucuronosiltransferase/biossíntese , Glucuronosiltransferase/fisiologia , Glicosaminoglicanos/biossíntese , Interleucina-1/farmacologia , Animais , Células Cultivadas , Condrócitos/efeitos dos fármacos , Condrócitos/metabolismo , Dinoprostona/metabolismo , Regulação para Baixo/efeitos dos fármacos , Glucosamina/farmacologia , Glucuronosiltransferase/genética , Masculino , Metaloproteinase 3 da Matriz/genética , Óxido Nítrico/biossíntese , Osteoartrite/tratamento farmacológico , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar
5.
Mol Pharmacol ; 58(6): 1609-15, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11093802

RESUMO

The human UDP-glucuronosyltransferase isoform UGT1A6 catalyzes the nucleophilic attack of phenolic xenobiotics on glucuronic acid, leading to the formation of water-soluble glucuronides. Based on the irreversible inhibition of the enzyme activity by the histidyl-selective reagent diethyl pyrocarbonate (DEPC), histidine was suggested to play a key role in the glucuronidation reaction. Therefore, the role of four strictly conserved histidine residues (His38, His361, His370, and His485) in the glucuronidation of 4-methylumbelliferone, as reporter substrate, was examined using site-directed mutagenesis. For this purpose, stable heterologous expression of wild-type and mutant UGT1A6 was achieved in the yeast Pichia pastoris. Replacement of histidine residues by alanine or glutamine led to fully inactive H38A, H38Q, and H485A mutants. Substitution of His361 by alanine affected the interaction of the enzyme with the cosubstrate, as indicated by a 4-fold increase in the K(m) value toward UDP-glucuronic acid. Interestingly, H370A mutant presented a severely impaired catalytic efficiency (with a V(max) value approximately 5% that of the wild-type), whereas conservative substitution of His370 by glutamine (H370Q) led to a significant restoration of activity. Whereas H361A was inactivated by DEPC as the wild-type enzyme, this chemical reagent only produced a minor effect on either H370Q or H370A mutant, providing evidence that His370 is probably the reactive histidine residue targeted by DEPC. The dramatic changes in catalytic efficiency on substitution of His370 by alanine and the ability of glutamine to function in place of histidine along with a weak sensitivity of these mutants to DEPC strongly suggest that His370 plays a catalytic role in the glucuronidation reaction.


Assuntos
Glucuronosiltransferase/metabolismo , Histidina/metabolismo , Alanina/genética , Alanina/metabolismo , Sequência de Aminoácidos , Sítios de Ligação , Catálise , Expressão Gênica , Glucuronosiltransferase/genética , Glutamina/genética , Glutamina/metabolismo , Histidina/genética , Humanos , Cinética , Fígado/enzimologia , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Pichia/genética , Homologia de Sequência de Aminoácidos
6.
J Biol Chem ; 275(36): 28254-60, 2000 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-10842173

RESUMO

Galbeta1,3-glucuronosyltransferase (GlcAT-I) that catalyzes the transfer of a glucuronic acid residue onto the trisaccharide primer of the glycosaminoglycan-protein linkage region plays an essential role in the early steps of the biosynthesis of glycosaminoglycans. In order to gain insight into the structure/function of the enzyme, the human recombinant GlcAT-I was successfully expressed in the yeast Pichia pastoris, with an apparent molecular mass of 43 kDa. Analysis of the electrophoretic mobility of the membrane-bound protein in nonreducing and reducing conditions, together with cross-linking studies, indicated that the membrane-bound GlcAT-I formed active disulfide-linked dimers. GlcAT-I expressed without the predicted N-terminal cytoplasmic tail or secreted as a polypeptide lacking the cytoplasmic tail and transmembrane domain was similarly organized as dimers, suggesting that the structural determinants for the dimerization state are localized in the luminal domain of the protein. In addition, the role of Cys(33) and Cys(301) in that process was investigated by site-directed mutagenesis combined with chemical modification of GlcAT-I by N-phenylmaleimide. Replacement of Cys(33) with alanine abolished the formation of dimers with a concomitant decrease in the catalytic efficiency mainly due to a decrease in apparent maximal velocity and in affinity for UDP-glucuronic acid. On the other hand, N-phenylmaleimide treatment or alanine substitution of the Cys(301) residue inactivated the enzyme. Our study demonstrates that GlcAT-I is organized as a homodimer as a result of disulfide bond formation mediated by Cys(33) localized in the stem region, whereas the residue Cys(301) localized in a conserved C-terminal domain is strictly required for the functional integrity of the enzyme.


Assuntos
Cisteína , Glucuronosiltransferase/química , Glucuronosiltransferase/metabolismo , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Caenorhabditis elegans/enzimologia , Clonagem Molecular , Dimerização , Humanos , Cinética , Maleimidas/farmacologia , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Pichia/enzimologia , Ratos , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Schistosoma mansoni/enzimologia , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos
7.
Biochem Pharmacol ; 59(5): 467-70, 2000 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-10660112

RESUMO

Treatment of Caco-2 cells with the antioxidants quercetin or t-butylhydroquinone led to induced protein levels of UDP-glucuronosyltransferase UGT1A6 (ca. 3-fold over controls) and of the apical conjugate export pump multidrug resistance protein 2 (MRP2; 1.9-fold over controls). In contrast to UGT1A6, MRP2 (symbol ABCC2) was not inducible by 2,3,7,8-tetrachlorodibenzo-p-dioxin. Immunocytochemistry demonstrated that MRP2 was only expressed at the brush border domain of Caco-2 cell monolayers. The results indicate that UGT1A6 and MRP2 are coordinately induced by antioxidants, facilitating chemoprotection against phenolic toxins and excretion of conjugates into the intestinal lumen.


Assuntos
Subfamília B de Transportador de Cassetes de Ligação de ATP/biossíntese , Transportadores de Cassetes de Ligação de ATP/biossíntese , Glucuronosiltransferase/biossíntese , Proteínas de Membrana Transportadoras , Antioxidantes/farmacologia , Células CACO-2 , Humanos , Immunoblotting , Proteína 2 Associada à Farmacorresistência Múltipla , Proteínas Associadas à Resistência a Múltiplos Medicamentos , Dibenzodioxinas Policloradas/farmacologia , Quercetina/farmacologia , Receptores de Hidrocarboneto Arílico/agonistas , Teratogênicos/farmacologia
8.
Chirality ; 12(2): 53-62, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10637410

RESUMO

Acyl glucuronides formed from carboxylic acids can undergo hydrolysis, acyl migration, and covalent binding to proteins. In buffers at physiological pH, the degradation of acylglucuronide of a chiral NSAID, carprofen, consisted mainly of acyl migration. Acidic pH reduced hydrolysis and acyl migration, thus stabilizing the carprofen acyl glucuronides. Addition of human serum albumin (HSA) led to an increased hydrolysis of the conjugates of both enantiomers. This protein protected R-carprofen glucuronide from migration and therefore improved its overall stability. Hydrolysis was stereoselective in favor of the S conjugate. The protein domains and the amino acid residues likely to be responsible for the hydrolytic activity of HSA were deduced from the results of various investigations: competition with probes specific of binding sites, effects of pH and of chemical modifications of albumin. Dansylsarcosine (DS), a specific ligand of site II of HSA, impaired the hydrolysis, whereas dansylamide (DNSA) and digoxin, which are specific ligands of sites I and III, respectively, had no effect. The extent of hydrolysis by HSA strongly increased with pH, indicating the participation of basic amino acids in this process. The results obtained with chemically modified HSA suggest the major involvement of Tyr and Lys residues in the hydrolysis of glucuronide of S-carprofen, and of other Lys residues for that of its diastereoisomer.


Assuntos
Aminoácidos/metabolismo , Anti-Inflamatórios não Esteroides/metabolismo , Carbazóis/metabolismo , Albumina Sérica/metabolismo , Animais , Ligação Competitiva , Soluções Tampão , Cães , Estabilidade de Medicamentos , Glucuronídeos/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Hidrólise , Albumina Sérica/química , Estereoisomerismo
9.
J Biol Chem ; 274(44): 31401-9, 1999 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-10531341

RESUMO

The human UDP-glucuronosyltransferase isoform UGT1A6 is predicted to be a type I transmembrane protein anchored in the endoplasmic reticulum by a single C-terminal transmembrane domain, followed by a short cytoplasmic tail. This topology is thought to be established through the sequential action of a cleavable N-terminal signal peptide and of a C-terminal stop transfer/anchor sequence. We found that the deletion of the signal peptide did not prevent membrane targeting and insertion of this protein expressed in an in vitro transcription/translation system or in yeast Pichia pastoris. Interestingly, the same results were obtained when the protein was depleted of both the signal peptide and the C-terminal transmembrane domain/cytoplasmic tail sequences, suggesting the presence of an internal topogenic element able to translocate and retain UGT1A6 in the endoplasmic reticulum membrane in vitro and in yeast cells. To identify such a sequence, the insertion of several N-terminal deletion mutants of UGT1A6 into microsomal membranes was investigated in vitro. The data clearly showed that the deletion of the N-terminal end did not affect endoplasmic reticulum targeting and retention until residues 140-240 were deleted. The signal-like activity of the 140-240 region was demonstrated by the ability of this segment to confer endoplasmic reticulum residency to the cytosolic green fluorescent protein expressed in mammalian cells. Finally, we show that this novel topogenic sequence can posttranslationally mediate the translocation of UGT1A6. This study provides the first evidence that the membrane assembly of the human UGT1A6 involves an internal signal retention sequence.


Assuntos
Retículo Endoplasmático/enzimologia , Glucuronosiltransferase/metabolismo , Sinais Direcionadores de Proteínas , Transporte Biológico , Compartimento Celular , Sistema Livre de Células , Genes Reporter , Glucuronosiltransferase/genética , Proteínas de Fluorescência Verde , Humanos , Proteínas Luminescentes/genética , Microssomos/metabolismo , Pichia/genética , Processamento de Proteína Pós-Traducional , Proteínas Recombinantes/metabolismo , Deleção de Sequência
10.
FEBS Lett ; 454(3): 187-91, 1999 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-10431804

RESUMO

UDP-glucuronosyltransferase 1A6 (UGT1A6) is a membrane glycoprotein of the endoplasmic reticulum playing a key role in drug metabolism. It is synthesized as a precursor with an N-terminal cleavable signal peptide. We demonstrate that deletion of the signal peptide sequence does not prevent membrane targeting and integration of this human isoform when expressed in an in vitro transcription-translation system, as shown by N-glycosylation, resistance to alkaline treatment and protease protection. Furthermore, UGT1A6 lacking the signal peptide (UGT1A6delta sp) was targeted to the endoplasmic reticulum in mammalian cells as shown by immunofluorescence microscopy and was catalytically active with kinetic constants for 4-methylumbelliferone glucuronidation similar to that of the wild-type. These results provide evidence that the signal peptide is not essential for the membrane assembly and activity of UGT1A6 suggesting that additional topogenic element(s) mediate(s) this process.


Assuntos
Glucuronosiltransferase/metabolismo , Fígado/enzimologia , Animais , Transporte Biológico , Linhagem Celular , Cricetinae , Retículo Endoplasmático/metabolismo , Glucuronosiltransferase/genética , Humanos , Fígado/ultraestrutura , Deleção de Sequência
11.
Arch Biochem Biophys ; 368(1): 75-84, 1999 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-10415114

RESUMO

7-Azido-4-methylcoumarin (AzMC) is a fluorescent photoactive compound structurally related to 4-methylumbelliferone (4-MU), a marker substrate of the human liver recombinant UDP-glucuronosyltransferase (UGT) 1A6. AzMC was synthesized and utilized to label the substrate binding site of UGT1A6. AzMC exhibits a fluorescence spectrum with maximum excitation and emission wavelengths of 380 and 442 nm, respectively. Upon irradiation, the probe irreversibly inhibited glucuronidation activity measured with para-nitrophenol (pNP) as substrate and interacted with UGT1A6 according to a saturable process indicative of reversible binding before covalent incorporation of the photoaffinity label. This inhibition was both time and concentration dependent and led to the calculation of an inhibition constant, k(2) = 0.113 mM min(-1), and dissociation constant, K(d) = 2.89 mM, for the reaction. Partial photoinactivation of UGT1A6 with AzMC revealed that the probe decreased the apparent V(max) of the pNP glucuronidation reaction, but not the K(m). Moreover, inhibition was partially prevented by 1-naphthol, a surrogate substrate for the enzyme, or by preincubation with an active-site directed inhibitor, 5'-O-[[(2-decanoylamino-3-phenyl-propyloxycarbonyl)amino]-su lfonyl]-2 ',3'-O-isopropylideneuridine. In contrast, UDP-glucuronic acid (UDP-GlcUA) did not have any protective effect against photoinactivation and AzMC did not affect the photoaffinity labeling of UGT1A6 by 5-[beta-(32)P]N(3)UDP-GlcUA, a photoaffinity analog of UDP-GlcUA. Additionally, in the absence of irradiation, AzMC was found to be a competitive inhibitor of 4MU glucuronidation. Collectively, these results strongly indicate that AzMC specifically binds to the UGT1A6 aglycon binding site. Amino acid alignment of phenol-binding proteins revealed a conserved motif, YXXXKXXPXP. It is possible that this motif is involved in phenol binding to UGT1A6 and other phenol-accepting proteins.


Assuntos
Glucuronosiltransferase/química , Glucuronosiltransferase/metabolismo , Fígado/enzimologia , Marcadores de Afinidade/síntese química , Sequência de Aminoácidos , Animais , Ligação Competitiva , Domínio Catalítico/genética , Linhagem Celular , Sequência Conservada , Cumarínicos/síntese química , Cricetinae , Glucuronosiltransferase/genética , Humanos , Técnicas In Vitro , Cinética , Ligantes , Dados de Sequência Molecular , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Homologia de Sequência de Aminoácidos , Especificidade por Substrato
12.
Mol Pharmacol ; 56(1): 226-34, 1999 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10385704

RESUMO

Acylglucuronides formed from carboxylic acids by UDP-glucuronosyltransferases (UGTs) are electrophilic metabolites able to covalently bind proteins. In this study, we demonstrate the reactivity of the acylglucuronide from the nonsteroidal anti-inflammatory drug, ketoprofen, toward human and rat liver UGTs. Ketoprofen acylglucuronide irreversibly inhibited the glucuronidation of 1-naphthol and 2-naphthol catalyzed by human liver microsomes or by the recombinant rat liver isoform, UGT2B1, which is the main isoform involved in the glucuronidation of the drug. A decrease of about 35% in the glucuronidation of 2-naphthol was observed when ketoprofen acylglucuronide was produced in situ in cultured V79 cells expressing UGT2B1. Inhibition was always associated with the formation of microsomal protein-ketoprofen adducts. The presence of these covalent adducts within the endoplasmic reticulum of cells expressing UGT2B1 was demonstrated following addition of ketoprofen to culture medium by immunofluorescence microscopy with antiketoprofen antibodies. Immunoblots of liver microsomes incubated with ketoprofen acylglucuronide and probed with antiketoprofen antibodies revealed the presence of several protein adducts; among those was a major immunoreactive protein at 56 kDa, in the range of the apparent molecular mass of UGTs. The adduct formation partially prevented the photoincorporation of the UDP-glucuronic acid (UDP-GlcUA) analog, [beta-32P]5N3UDP-GlcUA, on the UGTs, suggesting that ketoprofen glucuronide covalently reacted with the UDP-GlcUA binding domain. Finally, UGT purification from rat liver microsomes incubated with ketoprofen glucuronide led to the isolation of UGT adducts recognized by both anti-UGT and antiketoprofen antibodies, providing strong evidence that UGTs are targets of this metabolite.


Assuntos
Glucuronosiltransferase/antagonistas & inibidores , Cetoprofeno/análogos & derivados , Microssomos Hepáticos/efeitos dos fármacos , Animais , Anti-Inflamatórios não Esteroides/metabolismo , Anti-Inflamatórios não Esteroides/farmacologia , Células Cultivadas , Cricetinae , Inibidores Enzimáticos/farmacologia , Glucuronosiltransferase/metabolismo , Humanos , Cetoprofeno/metabolismo , Cetoprofeno/farmacologia , Microssomos Hepáticos/enzimologia , Ratos
13.
Drug Metab Dispos ; 27(5): 588-95, 1999 May.
Artigo em Inglês | MEDLINE | ID: mdl-10220487

RESUMO

To investigate the glucuronidation on the hydroxyl group of carbohydrate-containing drugs, the in vitro formation of glucuronides on the thioxyloside ring of the antithrombotic drug, LF 4.0212, was followed in rat and human liver microsomes and with recombinant UDP-glucuronosyltransferases (UGT). The reaction revealed a marked regioselectivity in rat and humans. Human liver microsomes glucuronidated the compound mainly on the 2-hydroxyl position of the thioxyloside ring, whereas rat was able to form glucuronide on either the 2-, 3-, or 4- hydroxyl group of the molecule, although to a lower extent. LF 4.0212 was a much better substrate of human UGT than the rat enzyme (Vmax/Km 30.0 and 0.06 microl/min/mg, respectively). Phenobarbital, 3-methylcholanthrene, and clofibrate enhanced the glucuronidation of LF 4.0212 on positions 2, 3, and 4 of the thioxyloside ring, thus indicating that several UGT isoforms were involved in this process. The biosynthesis of the 2-O-glucuronide isomer was catalyzed by the human UGT1A9 and 2B4, but not by UGT1A6 and 2B11. By contrast, the rat liver recombinant UGT1A6 and 2B1 failed to form the 2-O-glucuronide isomers. From all the recombinant UGTs tested, none catalyzed the formation of the 3-O-glucuronide isomer. Interestingly, glucuronidation on the 4-position was found in all the metabolic competent V79 cell lines considered, including the nontransfected V79 cells, suggesting the presence of an endogenous UGT in fibroblasts able to actively glucuronidate the drug. This activity, which was nonsensitive to the inhibitory effect of 7,7,7-triphenylheptanoic acid, a potent UGT inhibitor, could reflect the existence of a different enzyme.


Assuntos
Fibrinolíticos/farmacocinética , Glucuronosiltransferase/química , Glucuronosiltransferase/metabolismo , Glicosídeos/farmacocinética , Tioglicosídeos/farmacologia , Tioglicosídeos/farmacocinética , Animais , Catálise , Cromatografia Líquida de Alta Pressão , Cricetinae , Cricetulus , Fibrinolíticos/metabolismo , Fibroblastos/enzimologia , Fibroblastos/metabolismo , Glucuronatos/metabolismo , Glucuronosiltransferase/genética , Glicosídeos/metabolismo , Ácidos Heptanoicos/farmacologia , Humanos , Isoenzimas/química , Isoenzimas/genética , Isoenzimas/metabolismo , Cinética , Masculino , Microssomos Hepáticos/metabolismo , Ratos , Ratos Wistar , Especificidade por Substrato , Tioglicosídeos/metabolismo
14.
Drug Metab Dispos ; 26(8): 812-7, 1998 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9698297

RESUMO

Sodium periodate reacts with UDP-glucuronic acid (UDP-GlcUA) to generate a reactive derivative [periodate-oxidized UDP-GlcUA (o-UDP-GlcUA)]. The ability of this analog of UDP-GlcUA to inactivate and label the human recombinant UDP-glucuronosyltransferase (UGT) UGT1A6 via the UDP-GlcUA binding site was investigated. At an o-UDP-GlcUA concentration of 20 mM, the enzymatic activity of UGT1A6 was totally inactivated after 30 min of incubation at pH 7.4. Inhibition was irreversible, time-dependent, and concentration-dependent and exhibited pseudo-first order kinetics (kinact = 4.0 M-1.min-1). Cosubstrate protection with UDP-GlcUA was biphasic, with no protection in the first phase and almost total protection in the second phase, suggesting that at least 65% of the cross-linking occurs at the cosubstrate binding site. Partial inactivation by o-UDP-GlcUA led to a decrease in Vmax, suggesting that o-UDP-GlcUA can act as an active site-directed inhibitor. Furthermore, proteins, including the UGTs, from membrane fractions of a recombinant V79 cell line expressing the UGT1A6 enzyme and from rat liver microsomes were cross-linked by in situ periodate oxidation of [beta-32P]UDP-GlcUA. The present results suggest that periodate-oxidized UDP-GlcUA, which inactivates UGT1A6 by the possible formation of a Schiff base adduct with active site lysyl residues, can be used as a new affinity label for the UDP-GlcUA binding site.


Assuntos
Glucuronosiltransferase/efeitos dos fármacos , Glucuronosiltransferase/metabolismo , Fígado/enzimologia , Ácido Periódico/farmacologia , Uridina Difosfato Ácido Glucurônico/farmacologia , Animais , Sítios de Ligação , Ativação Enzimática/efeitos dos fármacos , Humanos , Marcação por Isótopo , Cinética , Oxirredução , Ácido Periódico/química , Ácido Periódico/metabolismo , Radioisótopos de Fósforo , Ratos , Proteínas Recombinantes/metabolismo , Uridina Difosfato Ácido Glucurônico/química , Uridina Difosfato Ácido Glucurônico/metabolismo
15.
J Biochem Mol Toxicol ; 12(1): 19-27, 1998.
Artigo em Inglês | MEDLINE | ID: mdl-9414484

RESUMO

A series of potent and competitive inhibitors of UDP-glucuronosyltransferase derived from 7,7,7-triphenylheptanoic acid has been synthesized in order to probe the active site of the isozyme involved in the glucuronidation of the endogenous toxic compound, bilirubin IX alpha. Like triphenylalkylcarboxylic acids, triphenyl alcohols were found to be very effective competitive inhibitors of the reaction (Ki 12 to 180 microM). Superimposition of the best inhibitors with bilirubin by computer modeling showed a marked spatial similarity, which accounts for the observed competitive-type inhibition. The bulky triphenylmethyl moiety of the inhibitor superimposed well on the part of the bilirubin molecule containing three of the four pyrrole rings. In agreement, substitution of the triphenylmethyl moiety by planar structures such as fluorenyl or indenyl rings completely suppressed the inhibition. In addition, the weak inhibition exerted by the shortest carboxylic acids could be related to the higher acidity of these molecules. The inhibition potency depended on the acidity of the molecules; the more acidic, the less inhibitory, suggesting that the presence of a negative charge on the inhibitor molecule prevents bilirubin glucuronidation. Based on these results, a reaction mechanism for bilirubin glucuronidation is postulated.


Assuntos
Inibidores Enzimáticos/farmacologia , Glucuronosiltransferase/antagonistas & inibidores , Ácidos Heptanoicos/farmacologia , Fígado/efeitos dos fármacos , Animais , Bilirrubina/química , Inibidores Enzimáticos/química , Ácidos Heptanoicos/química , Concentração de Íons de Hidrogênio , Fígado/enzimologia , Masculino , Modelos Moleculares , Ratos , Ratos Wistar
16.
Drug Metab Dispos ; 25(11): 1234-41, 1997 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9351898

RESUMO

This article is a report on a symposium held at the March 1997 meeting of the American Society for Pharmacology and Experimental Therapeutics in San Diego. Current developments in the heterologous expression of cytochrome P450, NADPH-cytochrome P450 reductase, glutathione transferase, and UDP-glucuronosyltransferase enzymes are described. Systems include bacteria, insect cells, and transient and stable mammalian cells. Uses of the products are described for discernment of which enzymes are involved in metabolism of drugs, genotoxicity assays, mutagenesis (for structure-activity relationships), large scale production of enzyme products, antibody production, and production of proteins for biophysical studies.


Assuntos
Enzimas/metabolismo , Preparações Farmacêuticas/metabolismo , Enzimas/biossíntese , Enzimas/química , Humanos , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo
17.
Pharmacogenetics ; 7(4): 255-69, 1997 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9295054

RESUMO

This review represents an update of the nomenclature system for the UDP glucuronosyltransferase gene superfamily, which is based on divergent evolution. Since the previous review in 1991, sequences of many related UDP glycosyltransferases from lower organisms have appeared in the database, which expand our database considerably. At latest count, in animals, yeast, plants and bacteria there are 110 distinct cDNAs/genes whose protein products all contain a characteristic 'signature sequence' and, thus, are regarded as members of the same superfamily. Comparison of a relatedness tree of proteins leads to the definition of 33 families. It should be emphasized that at least six cloned UDP-GlcNAc N-acetylglucosaminyltransferases are not sufficiently homologous to be included as members of this superfamily and may represent an example of convergent evolution. For naming each gene, it is recommended that the root symbol UGT for human (Ugt for mouse and Drosophila), denoting 'UDP glycosyltransferase,' be followed by an Arabic number representing the family, a letter designating the subfamily, and an Arabic numeral denoting the individual gene within the family or subfamily, e.g. 'human UGT2B4' and 'mouse Ugt2b5'. We recommend the name 'UDP glycosyltransferase' because many of the proteins do not preferentially use UDP glucuronic acid, or their nucleotide sugar preference is unknown. Whereas the gene is italicized, the corresponding cDNA, transcript, protein and enzyme activity should be written with upper-case letters and without italics, e.g. 'human or mouse UGT1A1.' The UGT1 gene (spanning > 500 kb) contains at least 12 promoters/first exons, which can be spliced and joined with common exons 2 through 5, leading to different N-terminal halves but identical C-terminal halves of the gene products; in this scheme each first exon is regarded as a distinct gene (e.g. UGT1A1, UGT1A2, ... UGT1A12). When an orthologous gene between species cannot be identified with certainty, as occurs in the UGT2B subfamily, sequential naming of the genes is being carried out chronologically as they become characterized. We suggest that the Human Gene Nomenclature Guidelines (http://www.gene.acl.ac.uk/nomenclature/guidelines.html++ +) be used for all species other than the mouse and Drosophila. Thirty published human UGT1A1 mutant alleles responsible for clinical hyperbilirubinemias are listed herein, and given numbers following an asterisk (e.g. UGT1A1*30) consistent with the Human Gene Nomenclature Guidelines. It is anticipated that this UGT gene nomenclature system will require updating on a regular basis.


Assuntos
Evolução Molecular , Genes , Glucuronosiltransferase/genética , Família Multigênica , Terminologia como Assunto , Sequência de Aminoácidos , Animais , Glucuronosiltransferase/química , Humanos , Dados de Sequência Molecular , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos
18.
Drug Metab Dispos ; 25(4): 406-11, 1997 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9107538

RESUMO

Recombinant human liver UDP-glucuronosyltransferase (UGT), UGT1*6, which catalyzes the glucuronidation of small phenols, previously expressed in a V79 cell line (1) was photolabeled with [beta-32P]5N3UDP-glucuronic acid ([beta-32P]5N3UDP-GlcUA). Two polypeptides with an approximate molecular weight of 54 kDa were extensively photolabeled in the recombinant cell line while the nontransfected cell line showed no photoincorporation in this area. The identity of the two polypeptides as UGTs, which correspond to two different glycosylation forms of the same enzyme, was confirmed by Western blot using a polyclonal monospecific antibody directed against the 120 amino acids of the N-terminal end of UGT1*6. Preincubation with UDP-glucuronic acid (UDP-GlcUA) inhibited the photoincorporation of the probe into the polypeptides indicating competition of both the photoprobe and the nucleotide-sugar for the same binding site. It was further shown that photoincorporation of [beta-32P]5N3UDP-GlcUA into the UDP-GlcUA-binding site was saturable. The lack of photoincorporation of a related photoprobe, [beta-32P]5N3UDP-glucose ([beta-32P]5N3UDP-Glc), into UGT1*6 demonstrated specificity of this enzyme for UDP-GlcUA. In enzymatic assays, unlabeled 5N3UDP-GlcUA was shown to be an effective cosubstrate of the glucuronidation of 4-nitrophenol catalyzed by UGT1*6. The studies were further extended by demonstrating that photolabeling of UGT1*6 was inhibited by several active site-directed inhibitors. Finally, photoaffinity labelling was used in the purification of the labeled UGT1*6 using preparative gel electrophoresis. In conclusion, we have demonstrated that photoaffinity labeling with [beta-32P]5N3UDP-GlcUA is an effective tool for the characterization of enzymes such as recombinant UGTs that use UDP-GlcUA.


Assuntos
Glucuronosiltransferase/química , Uridina Difosfato Ácido Glucurônico/análogos & derivados , Marcadores de Afinidade , Animais , Western Blotting , Linhagem Celular , Cricetinae , Eletroforese em Gel de Poliacrilamida , Glucuronosiltransferase/metabolismo , Humanos , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Especificidade por Substrato , Uridina Difosfato Ácido Glucurônico/química , Açúcares de Uridina Difosfato/metabolismo
19.
Mol Pharmacol ; 51(3): 406-13, 1997 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-9058595

RESUMO

The hepatic UDP-glucuronosyltransferase UGT1*6 is actively involved in the glucuronidation of short and planar phenols in humans. Based on the irreversible inhibition of the enzyme on chemical modification by 2,3-butanedione and diethyl pyrocarbonate, the roles of His54 and Arg52 were investigated by oligonucleotide site-directed mutagenesis. These amino acids belong to a consensus sequence LX2-R52-G-H54-X3-V-L located in a conserved hydrophobic region of the variable amino-terminal domain of UGT. Arg52 was replaced by alanine (mutant R52A), and His54 was replaced by alanine or glutamine (mutants H54A and H54Q). The immunological and catalytic properties of UGT1*6 and mutants were examined after stable expression in V79 cell lines. Immunoblots and immunoprecipitation studies revealed that the mutant and UGT1*6 proteins were expressed in the microsomal membranes in similar amounts. However, replacement of His54 by glutamine led to a complete loss of activity toward 4-methylumbelliferone, and the Vmax value was decreased 4-5-fold in the mutants R52A and H54A compared with the wild-type enzyme. The dissociation constants that characterize the binding of 4-methylumbelliferone and UDP-glucuronic acid to UGT1*6 were not greatly affected by the mutations. Interestingly, H54Q was not recognized by specific antibodies to the amino-terminal portion of UGT1*6, thereby indicating that this amino acid was critical to antibody recognition. In contrast, the mutants R52A and H54A could not be differentiated from the wild-type protein by pH optimum or thermal denaturation. Furthermore, these mutants were still sensitive to irreversible inhibition by diethyl pyrocarbonate and 2,3-butanedione, with second-order inactivation constant values similar to those obtained for UGT1*6. Altogether, the strict conservation of His54 and Arg52 and the mutational analysis of these residues suggest that these amino acids in the hydrophobic amino-terminal consensus sequence LX2-R52-G-H54-X3-V-L are important for the function and the structure required for optimal catalytic efficiency of UGT1*6.


Assuntos
Arginina/fisiologia , Sequência Consenso , Glucuronosiltransferase/química , Glucuronosiltransferase/metabolismo , Histidina/fisiologia , Fígado/enzimologia , Animais , Linhagem Celular , Cricetinae , Cricetulus , Glucuronatos/metabolismo , Humanos , Himecromona/metabolismo , Mutagênese Sítio-Dirigida
20.
Drug Metab Dispos ; 24(10): 1050-7, 1996 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-8894504

RESUMO

We have previously shown that the acyl glucuronide of racemic ketoprofen can irreversibly bind in vitro to plasma proteins (Dubois, N., et ai., Drug Metab. Dispos. 21, 617-623, 1993), but the mechanism of the reaction has not been characterized. In the present study, the reactivity toward albumin of the glucuronide of both ketoprofen enantiomers was investigated. The extent of binding increased with the concentration of both protein and glucuronide. However, the two diastereoisomers showed different reactivities toward human serum albumin (HSA): the maximum yield of adducts with the glucuronide of the S-enantiomer was twice that obtained with the glucuronide of its antipode. The maximum extent of irreversible binding was at 4 hr for the R-ketoprofen conjugate, but was later for the S-form. Chemical modifications of albumin indicated that the glucuronide of the S-isomer reacted only with lysine residues, whereas the R-form linked covalently mainly with tyrosine residues and secondarily with lysine residues. A competition study using specific binding probes and fatty acids showed that the conjugates of S- and R-ketoprofen reacted with amino acids located in sites I and II of HSA, respectively. Taken together, these findings suggest that the irreversible binding of ketoprofen to albumin depends on the stereochemistry of the aglycon: the R-enantiomer binds to site II of the protein probably by a nucleophilic attack by tyrosine and/or lysine residues, whereas adduct formation via the conjugate of the S-enantiomer could occur at site I of HSA by the Schiff base mechanism. This irreversible binding at sites I and II may affect the major function of albumin (i.e. the transport of drugs and endogenous compounds).


Assuntos
Cetoprofeno/análogos & derivados , Albumina Sérica/metabolismo , Animais , Humanos , Cetoprofeno/metabolismo , Ligantes , Microssomos Hepáticos/metabolismo , Ligação Proteica , Ratos , Albumina Sérica/química , Estereoisomerismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...