Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
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.
Life Sci ; 67(20): 2473-84, 2000 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-11065170

RESUMO

Human UDP-glucuronosyltransferases (UGTs) 1A6 and 1A9 were expressed using Semliki Forest virus (SFV) vectors. Infection of chinese hamster lung fibroblast V79 cells with recombinant SFV-UGT viruses resulted in efficient protein expression as detected by metabolic labeling, Western blot analyses and immunofluorescence microscopy. The expression of UGT 1A6 and UGT1A9 in the SFV-infected cells was approximately two fold higher than in a stable V79 cell line. No UGT signal was detected in noninfected cells. In addition, SFV-UGT viruses also efficiently infected other mammalian cells, such as baby hamster kidney (BHK), chinese hamster ovary (CHO) and human lung (WI-26 VA4) cells leading to high production of recombinant enzyme. The measurement of enzyme activities and kinetic parameters using p-nitrophenol and nitrocatechol (entacapone) as substrates for UGT1A6 and UGT1A9, respectively, showed that the overall kinetic properties of the enzymes produced by the two systems were similar. We conclude that the SFV expression system represents an efficient, fast and versatile method for production of metabolic enzymes for in vitro assays.


Assuntos
Expressão Gênica , Glucuronosiltransferase/biossíntese , Glucuronosiltransferase/genética , Vírus da Floresta de Semliki/genética , Animais , Células CHO/enzimologia , Células CHO/virologia , Catecóis/metabolismo , Células Cultivadas , Cricetinae , Primers do DNA/química , Fibroblastos/enzimologia , Fibroblastos/virologia , Vetores Genéticos , Humanos , Rim/enzimologia , Rim/virologia , Pulmão/enzimologia , Pulmão/virologia , Nitrilas , Nitrofenóis/metabolismo , RNA Mensageiro/biossíntese , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Vírus da Floresta de Semliki/enzimologia , Especificidade por Substrato , Transfecção , UDP-Glucuronosiltransferase 1A
6.
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
7.
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
8.
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
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.
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
12.
FEBS Lett ; 399(3): 215-9, 1996 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-8985148

RESUMO

Lysyl oxidase (LO) is a copper amine oxidase of the extracellular matrix which initiates covalent cross-linking in collagens and elastin. Human LO was expressed in Escherichia coli. At 37 degrees C, large amounts of protein were obtained, but in the form of insoluble aggregates. Lowering the growth temperature, and reducing the amount of inducer, resulted in the production of soluble LO, which was active on a degrees [3H]lysine-labeled elastin substrate. LO was also targeted to the periplasm as a fusion protein with the pelb signal peptide. The periplasmic enzyme was soluble, active and inhibited by beta-aminopropionitrile. Production of the carbonyl co-factor is therefore not a limitation in the expression of active LO in bacteria.


Assuntos
Escherichia coli/genética , Proteína-Lisina 6-Oxidase/genética , Sequência de Aminoácidos , Clonagem Molecular , DNA Complementar , Eletroforese em Gel de Poliacrilamida , Humanos , Dados de Sequência Molecular , Proteína-Lisina 6-Oxidase/isolamento & purificação , Proteína-Lisina 6-Oxidase/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo
14.
Clin Chim Acta ; 226(2): 171-90, 1994 May.
Artigo em Inglês | MEDLINE | ID: mdl-7923812

RESUMO

Many studies on drug metabolism have been carried out during the last decades using protein purification, molecular cloning techniques and analysis of polymorphisms at phenotype and genotype levels. These researchers led to a better understanding of the role of drug metabolizing enzymes in the biotransformation of drugs, pollutants or foreign compounds and of their use in laboratory medicine. The metabolic processes commonly involved in the biotransformation of xenobiotics have been classified into functionalization reaction (phase I reactions), which implicate lipophilic compounds. These molecules are modified via monooxygenation, dealkylation, reduction, aromatization, hydrolysis and can be substrates for the phase II reactions, often called conjugation reactions as they conjugate a functional group with a polar, endogenous compound. This review, devoted to cytochromes P-450 (CYP) and UDP-glucuronosyltransferases (UGT), describes essentially the genetic polymorphisms found in humans, their clinical consequences and the methods to assess the phenotypes or genotypes, with a view to studying the interindividual differences in drug monooxygenation and drug glucuronidation. Variations in drug glucuronidation reported here focused essentially on variations due to physiological factors, induction, drug interactions and genetic factors in disorders such as Gilbert's Syndrome and Crigler-Najjar type I and II diseases.


Assuntos
Sistema Enzimático do Citocromo P-450/genética , Sistema Enzimático do Citocromo P-450/metabolismo , Glucuronosiltransferase/genética , Glucuronosiltransferase/metabolismo , Preparações Farmacêuticas/metabolismo , Genótipo , Humanos , Isoenzimas , Polimorfismo Genético
15.
Arch Biochem Biophys ; 310(1): 196-204, 1994 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-8161205

RESUMO

Characterization of human UDP-glucuronyltransferases (UGTs) has been limited by the unavailability of probes selective for each of several highly related isoforms. To better understand the role of this superfamily in the metabolism of drugs and xenobiotics, we describe a molecular/immunological strategy for discriminating the implication of each human isoenzyme in this process. Specific polyclonal antibodies were generated against the divergent amino-terminal domain of the UGT isoform UGT1*6 which is involved in the detoxification of nucleophilic compounds related to phenols and naphthols in human liver. The novel approach consists of the expression of a N-terminal UGT polypeptide fused to Staphylococcus aureus protein A in Escherichia coli and a single step purification of the fusion protein by immunoaffinity chromatography. Immunoblot and immunoinhibition analysis showed that the antibodies raised against the fusion protein selectively recognized both the denaturated and the native forms of UGT1*6, when expressed in V79 cell lines, but not three other recombinant UGT isoenzymes. In human liver microsomes, specific immunoinhibition analysis demonstrated that glucuronidation by UGT1*6 represented 20 to 50% of the total 1-naphthol UGT activity with a good correlation with the amount of protein selectively quantified on immunoblot. The specific expression of UGT1*6 was found to be significantly reduced in tumoral tissues but enhanced in cholestatic livers, when compared with healthy hepatic tissues. Interestingly, in human kidney microsomes, antibodies revealed a high level of UGT1*6 expression on immunoblot and inhibited 1-naphthol glucuronidation up to 55%, indicating that this isoform is also expressed in kidney and extensively contributes to phenol glucuronidation in this tissue.


Assuntos
Glucuronosiltransferase/fisiologia , Isoenzimas/fisiologia , Fígado/enzimologia , Adulto , Animais , Especificidade de Anticorpos , Pré-Escolar , Cricetinae , Escherichia coli/genética , Feminino , Glucuronosiltransferase/imunologia , Humanos , Lactente , Isoenzimas/imunologia , Fígado/patologia , Masculino , Microssomos Hepáticos/enzimologia , Pessoa de Meia-Idade , Naftóis/metabolismo , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/imunologia , Proteína Estafilocócica A/biossíntese , Proteína Estafilocócica A/genética , Transfecção
16.
Arch Biochem Biophys ; 309(2): 266-72, 1994 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-8135537

RESUMO

Chemical modification with diethyl pyrocarbonate (DEPC) of the recombinant human liver UDP-glucuronosyltransferase UGT1*6 in enriched membrane fractions from a V79 cell line resulted in a rapid inactivation of the glucuronidation reaction, measured with 4-methyl-umbelliferone as aglycone substrate, with a second-order rate constant of 3110 M-1.min-1 at pH 6.0 and 25 degrees C. The enzymatic activity was restored by hydroxylamine. Chemical modification with 0.2 mM DEPC for 60 s decreased the apparent Vmax 2.4-fold without significantly affecting the apparent Km toward 4-methylumbelliferone and UDP-glucuronic acid. Similarly, the binding of the photoactivatable cosubstrate analog [beta-32P]5-azido-UDP-glucuronic acid to the active site was not affected by the chemical modification. The enzyme was protected against this inactivation by 4-methylumbelliferone, suggesting that the modified residue was located in or near the aglycone binding site. In contrast, the cosubstrate UDP-glucuronic acid potentiated the irreversible inhibition, indicating a conformational change in the protein upon binding. The pH-dependence of the inactivation was in agreement with the modification of an amino acid residue with a pKa of 6.1. On the other hand, analysis of the variation of Vmax and Vmax/Km values of the glucuronidation reaction as a function of the pH revealed the presence of two essential residues with a pKa within the range 5.7-6.0. The data of the chemical modification of the recombinant enzyme together with that of the pH dependence of the activity strongly suggest the involvement of a histidine residue, highly reactive toward DEPC, which could be the base catalyst of the glucuronidation reaction supported by human UGT1*6.


Assuntos
Dietil Pirocarbonato/farmacologia , Glucuronosiltransferase/antagonistas & inibidores , Histidina/química , Marcadores de Afinidade , Animais , Catálise , Linhagem Celular , Cricetinae , Cricetulus , Ativação Enzimática/efeitos dos fármacos , Glucuronatos/metabolismo , Glucuronosiltransferase/química , Glucuronosiltransferase/metabolismo , Concentração de Íons de Hidrogênio , Hidroxilamina , Hidroxilaminas/farmacologia , Himecromona/metabolismo , Himecromona/farmacologia , Cinética , Pulmão , Proteínas Recombinantes , Uridina Difosfato Ácido Glucurônico/análogos & derivados , Uridina Difosfato Ácido Glucurônico/metabolismo
17.
FEBS Lett ; 339(1-2): 195-9, 1994 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-8313973

RESUMO

The membrane-bound human liver UDP-glucuronosyltransferase UGT1*6 was expressed in Escherichia coli. Exchange of the natural signal peptide by the bacterial signal peptides of pclB or OmpT proteins considerably increased the level of expression and, as the natural signal peptide, targeted the protein to the membranes. The extent of maturation of SpelB-UGT1*6 precursor was about 30%. No processing of sOmpT-UGT1*6 occurred but the processing rate of this precursor could be significantly increased by mutagenesis of the first two amino acid residues of the mature sequence. These expression vectors allowed us to produce high levels of recombinant mature UGT1*6 required for further structural studies.


Assuntos
Proteínas de Bactérias/genética , Escherichia coli/genética , Expressão Gênica/efeitos dos fármacos , Glucuronosiltransferase/genética , Precursores de Proteínas/genética , Sinais Direcionadores de Proteínas/genética , Sequência de Aminoácidos , Proteínas de Bactérias/fisiologia , Sequência de Bases , Membrana Celular/enzimologia , Clonagem Molecular , Vetores Genéticos , Glucuronosiltransferase/biossíntese , Humanos , Dados de Sequência Molecular , Peso Molecular , Plasmídeos , Sinais Direcionadores de Proteínas/fisiologia , Proteínas Recombinantes de Fusão , Proteínas Recombinantes/biossíntese
18.
Biochem Biophys Res Commun ; 197(2): 785-91, 1993 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-8267616

RESUMO

The interactions between UDP-glucuronic acid and two human liver UDP-glucuronosyltransferase 2B4 peptides (14-150 and 299-446) purified from E. coli as Staphylococcus aureus protein A fusion proteins have been investigated. Photoaffinity labeling with azidonucleotides ([beta-32P]5N3UDP-Glucuronic acid and [beta-32P]5N3UDP-Glucose) and competition experiments with UDP-glucuronic acid and structurally related compounds emphasized the presence of a specific UDP binding site between amino acids 299 and 446. Moreover, competition experiments strongly suggested an interaction between the amino terminal part of the protein and glucuronic acid. It would involve an electrostatic bond in the binding of the cosubstrate via the carboxyl group of UDP-glucuronic acid and a positively charged amino acid of the N-terminal domain of the enzyme.


Assuntos
Glucuronosiltransferase/química , Fígado/enzimologia , Uridina Difosfato Ácido Glucurônico/metabolismo , Marcadores de Afinidade , Clonagem Molecular , Escherichia coli , Glucuronosiltransferase/isolamento & purificação , Glucuronosiltransferase/metabolismo , Humanos , Radioisótopos de Fósforo , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/isolamento & purificação , Proteínas Recombinantes de Fusão/metabolismo , Mapeamento por Restrição , Proteína Estafilocócica A/isolamento & purificação , Proteína Estafilocócica A/metabolismo , Staphylococcus aureus/metabolismo
19.
J Biol Chem ; 268(34): 25636-42, 1993 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-8244999

RESUMO

Monospecific polyclonal antibodies were raised against a variable amino-terminal domain (amino acids 14-150) of a human liver form of UDP-glucuronosyl-transferase conjugating bile acids, UGT2B4 (Jackson, M. R., McCarthy, L. R., Harding, D., Wilson, S., Coughtrie, M. W., and Burchell, B. (1987) Biochem. J. 242, 581-588), expressed as a fusion protein in Escherichia coli. The antibodies were able to recognize the protein, stably expressed in a genetically engineered eukaryotic V79 cell line, against which they were directed. The specificity of these antibodies allowed their use for analyzing the substrate specificity of this isoform in human liver, as well as for determining its contribution to the total hepatic and extra-hepatic glucuronidation of hyodeoxycholic acid. Western blot analysis of microsomal proteins demonstrated the presence of UGT2B4 exclusively in human liver and not in human kidney. In human liver microsomes, the antibodies were able to inhibit and precipitate up to 90% of the total hyodeoxycholic acid 6-O-glucuronidation activity, but had no effect on activities toward several other substrates, such as phenols, bilirubin, or other bile acids, especially hyocholic acid and the steroids 4-hydroxyesterone and estriol. Moreover, Western blot analysis and immunoinhibition studies of human liver microsomes from healthy patients and from patients presenting liver diseases revealed a good correlation between the glucuronidation rate of hyodeoxycholic acid and the UGT2B4 expression level. The absence of immunoinhibition of hyodeoxycholic acid conjugation with UDP sugars other than UDP-glucuronic acid suggests the involvement of different enzymatic systems in the glucosidation and xylosylation of hyodeoxycholic acid. Altogether, the results provided strong evidence for the specific and predominant involvement of UGT2B4 in the 6-O-glucuronidation of this bile acid via a UDP-glucuronic acid-dependent mechanism.


Assuntos
Ácido Desoxicólico/metabolismo , Glucuronosiltransferase/metabolismo , Isoenzimas/metabolismo , Fígado/enzimologia , Microssomos Hepáticos/enzimologia , Animais , Anticorpos , Linhagem Celular , Criança , Pré-Escolar , Clonagem Molecular , Escherichia coli , Vetores Genéticos , Glucuronatos/metabolismo , Glucuronosiltransferase/biossíntese , Glucuronosiltransferase/isolamento & purificação , Humanos , Immunoblotting , Isoenzimas/biossíntese , Isoenzimas/isolamento & purificação , Rim/enzimologia , Cinética , Masculino , Microssomos/enzimologia , Pessoa de Meia-Idade , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/isolamento & purificação , Proteínas Recombinantes de Fusão/metabolismo , Mapeamento por Restrição
20.
Biochem Biophys Res Commun ; 196(1): 473-9, 1993 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-8216331

RESUMO

The purification and the characterization of functional human liver UDP-glucuronosyltransferase 2B4 produced as a Staphylococcus aureus protein A fusion protein in E. coli are described. The purified fusion protein was able to catalyze the glucuronidation of hyodeoxycholic acid, the major substrate described for this isoform to date. The effects of the amount and the nature of the phospholipids upon reconstitution into phospholipid micelles were investigated. Apparent determined Km values for hyodeoxycholic acid and UDP-glucuronic acid were 0.55 and 0.43 mM, respectively. Moreover, photoaffinity labelling of the fusion protein with a photoactivatable analog of UDP-glucuronic acid strongly suggested that this recombinant protein exhibited similar binding properties as the microsomal protein, which emphasizes its use for further structural analyses.


Assuntos
Glucuronosiltransferase/biossíntese , Glucuronosiltransferase/genética , Fígado/enzimologia , Marcadores de Afinidade , Ácido Desoxicólico/metabolismo , Escherichia coli/genética , Vetores Genéticos/genética , Glucuronosiltransferase/metabolismo , Humanos , Cinética , Micelas , Fosfolipídeos , Proteínas Recombinantes de Fusão/biossíntese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA