Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
Add more filters










Publication year range
1.
Biol Pharm Bull ; 39(9): 1432-6, 2016.
Article in English | MEDLINE | ID: mdl-27582324

ABSTRACT

Dextrorphan, an active metabolite of the antitussive dextromethorphan, has been shown to be subjected to sulfation by several zebrafish cytosolic sulfotransferases (SULTs). We were interested in finding out which of the human SULT(s) is(are) capable of catalyzing the sulfation of dextrorphan, and to verify whether sulfation of dextrorphan may occur in cultured human cells and human organ cytosols. Data from the enzymatic assays showed that, of all thirteen known human SULTs, SULT1A3 displayed the strongest dextrorphan-sulfating activity. Cell culture experiments using HepG2 human hepatoma cells and Caco-2 human colon carcinoma cells incubated with [(35)S]sulfate together with varying concentrations of dextrorphan revealed indeed the production and release of [(35)S]sulfated dextrorphan in a concentration-dependent manner. Additionally, significant dextrorphan-sulfating activity was detected in human liver, small intestine and lung cytosols. Taken together, these results provided a biochemical basis for the sulfation of dextrorphan in humans.


Subject(s)
Arylsulfotransferase/metabolism , Dextrorphan/pharmacology , Caco-2 Cells , Excitatory Amino Acid Antagonists/pharmacology , Hep G2 Cells , Humans
2.
J Appl Toxicol ; 36(9): 1090-4, 2016 09.
Article in English | MEDLINE | ID: mdl-26663444

ABSTRACT

The aim of the present study was to identify human cytosolic sulfotransferases (SULTs) that are capable of sulfating benzyl alcohol and to examine whether benzyl alcohol sulfation may occur in cultured human cells as well as in human organ homogenates. A systematic analysis revealed that of the 13 known human SULTs, SULT1A1 SULT1A2, SULTA3, and SULT1B1 are capable of mediating the sulfation of benzyl alcohol. The kinetic parameters of SULT1A1 that showed the strongest benzyl alcohol-sulfating activity were determined. HepG2 human hepatoma cells were used to demonstrate the generation and release of sulfated benzyl alcohol under the metabolic settings. Moreover, the cytosol or S9 fractions of human liver, lung, kidney and small intestine were examined to verify the presence of benzyl alcohol sulfating activity in vivo. Copyright © 2015 John Wiley & Sons, Ltd.


Subject(s)
Arylsulfotransferase/metabolism , Benzyl Alcohol/toxicity , Cytosol/drug effects , Caco-2 Cells , Cytosol/metabolism , Hep G2 Cells , Humans , Intestine, Small/drug effects , Intestine, Small/metabolism , Kidney/drug effects , Kidney/metabolism , Liver/drug effects , Liver/metabolism , Lung/drug effects , Lung/metabolism , Sulfates/metabolism
3.
J Biochem ; 158(6): 497-504, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26067475

ABSTRACT

Sulphation is known to be critically involved in the metabolism of acetaminophen in vivo. This study aimed to systematically identify the major human cytosolic sulfotransferase (SULT) enzyme(s) responsible for the sulphation of acetaminophen. A systematic analysis showed that three of the twelve human SULTs, SULT1A1, SULT1A3 and SULT1C4, displayed the strongest sulphating activity towards acetaminophen. The pH dependence of the sulphation of acetaminophen by each of these three SULTs was examined. Kinetic parameters of these three SULTs in catalysing acetaminophen sulphation were determined. Moreover, sulphation of acetaminophen was shown to occur in HepG2 human hepatoma cells and Caco-2 human intestinal epithelial cells under the metabolic setting. Of the four human organ samples tested, liver and intestine cytosols displayed considerably higher acetaminophen-sulphating activity than those of lung and kidney. Collectively, these results provided useful information concerning the biochemical basis underlying the metabolism of acetaminophen in vivo previously reported.


Subject(s)
Acetaminophen/pharmacokinetics , Analgesics, Non-Narcotic/pharmacokinetics , Arylsulfotransferase/metabolism , Cytosol/enzymology , Sulfotransferases/metabolism , Biocatalysis , Caco-2 Cells , Hep G2 Cells , Humans , Hydrogen-Ion Concentration , Intestines/enzymology , Kidney/enzymology , Kinetics , Liver/enzymology , Lung/enzymology
4.
Drug Metab Lett ; 8(2): 96-100, 2014.
Article in English | MEDLINE | ID: mdl-25429512

ABSTRACT

Previous studies had demonstrated that sulfation constituted a major pathway for the metabolism of phenylephrine in vivo. The current study was designed to identify the major human SULT(s) responsible for the sulfation of phenylephrine. Of the twelve human SULTs analyzed, SULT1A3 displayed the strongest sulfating activity toward phenylephrine. The enzyme exhibited a pH optimum spanning 7 - 10.5. Kinetic analysis revealed that SULT1A3- mediated sulfation of phenylephrine occurred in the same order of magnitude compared with that previously reported for SULT1A3-mediated sulfation of dopamine. Moreover, sulfation of phenylephrine was shown to occur in HepG2 cells under metabolic setting. Collectively, these results provided useful information concerning the biochemical basis underlying the metabolism of phenylephrine in vivo as previously reported.


Subject(s)
Arylsulfotransferase/metabolism , Cytosol/enzymology , Phenylephrine/metabolism , Sulfotransferases/metabolism , Adrenergic alpha-1 Receptor Agonists/metabolism , Hep G2 Cells , Humans , Hydrogen-Ion Concentration , Kinetics , Sulfates/metabolism
5.
Eur J Pharm Sci ; 62: 40-8, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-24832963

ABSTRACT

The current study was designed to examine the sulfation of eight opioid drugs, morphine, hydromorphone, oxymorphone, butorphanol, nalbuphine, levorphanol, nalorphine, and naltrexone, in HepG2 human hepatoma cells and human organ samples (lung, liver, kidney, and small intestine) and to identify the human SULT(s) responsible for their sulfation. Analysis of the spent media of HepG2 cells, metabolically labeled with [35S]sulfate in the presence of each of the eight opioid drugs, showed the generation and release of corresponding [35S]sulfated derivatives. Five of the eight opioid drugs, hydromorphone, oxymorphone, butorphanol, nalorphine, and naltrexone, appeared to be more strongly sulfated in HepG2 cells than were the other three, morphine, nalbuphine, and levorphanol. Differential sulfating activities toward the opioid drugs were detected in cytosol or S9 fractions of human lung, liver, small intestine, and kidney, with the highest activities being found for the liver sample. A systematic analysis using eleven known human SULTs and kinetic experiment revealed SULT1A1 as the major responsible SULTs for the sulfation of oxymorphone, nalbuphine, nalorphine, and naltrexone, SULT1A3 for the sulfation of morphine and hydromorphone, and SULT2A1 for the sulfation of butorphanol and levorphanol. Collectively, the results obtained imply that sulfation may play a significant role in the metabolism of the tested opioid drugs in vivo.


Subject(s)
Analgesics, Opioid/metabolism , Narcotic Antagonists/metabolism , Sulfotransferases/metabolism , Cytosol/metabolism , Hep G2 Cells , Humans , Intestine, Small/metabolism , Kidney/metabolism , Liver/metabolism , Lung/metabolism , Sulfates/metabolism , Sulfur Radioisotopes
6.
Biol Pharm Bull ; 35(12): 2180-5, 2012.
Article in English | MEDLINE | ID: mdl-23207770

ABSTRACT

Ethyl sulfate, a minor and direct ethanol metabolite in adult human body, has been implicated as a biomarker for alcohol consumption and in utero exposure to ethanol. To understand better the physiological relevance of the sulfation of ethanol, it is important to clarify the cytosolic sulfotransferase (SULT) enzymes that are responsible for ethanol sulfation. The present study aimed to identify the major ethanol-sulfating human SULTs and to investigate the sulfation of ethanol under the metabolic setting. A systematic analysis revealed four ethanol-sulfating SULTs, SULT1A1, SULT1A2, SULT1A3, and SULT1C4, among the eleven human SULT enzymes previously prepared and purified. A metabolic labeling study demonstrated the generation and release of ethyl [(35)S]sulfate in a concentration-dependent manner by HepG2 human hepatoma cells labeled with [(35)S]sulfate in the presence of different concentrations of ethanol. Cytosol or S9 fractions of human lung, liver, and small intestine were examined to verify the presence of ethanol-sulfating activity in vivo. Of the three human organs, the small intestine displayed the highest activity.


Subject(s)
Alcohol Drinking/metabolism , Ethanol/metabolism , Intestine, Small/enzymology , Liver/enzymology , Lung/enzymology , Sulfotransferases/metabolism , Sulfuric Acid Esters/metabolism , Adult , Biomarkers/metabolism , Cytosol/metabolism , Dose-Response Relationship, Drug , Hep G2 Cells , Humans , Staining and Labeling
7.
Drug Metab Lett ; 6(2): 109-15, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22946908

ABSTRACT

Buprenorphine, pentazocine, and naloxone are opioid drugs used for the treatment of pain and opioid dependence or overdose. Sulfation as catalyzed by the cytosolic sulfotransferases (SULTs) is involved in the metabolism of a variety of xenobiotics including drug compounds. Sulfation of opioid drugs has not been well investigated. The current study was designed to examine the sulfation of three opioid drugs, buprenorphine, pentazocine, and naloxone, in HepG2 human hepatoma cells and to identify the human SULT(s) responsible for their sulfation. Analysis of the spent media of HepG2 cells, metabolically labeled with [(35)S]sulfate in the presence of each of the three opioid drugs, showed the generation and release of their [(35)S]sulfated derivatives. A systematic analysis using eleven known human SULTs revealed SULT1A3 and SULT2A1 as the major responsible SULTs for the sulfation of, respectively, pentazocine and buprenorphine; whereas three other SULTs, SULT1A1, SULT1A2, and SULT1C4, were capable of sulfating naloxone. Enzymatic assays using combinations of these opioid drugs as substrates showed significant inhibitory effects in the sulfation of buprenorphine and pentazocine by naloxone. Differential sulfating activities toward the three opioid drugs were detected in cytosol or S9 fractions of human lung, liver, kidney, and small intestine. Collectively, these results imply that sulfation may play a role in the metabolism of buprenorphine, pentazocine, and naloxone in vivo.


Subject(s)
Buprenorphine/metabolism , Naloxone/metabolism , Pentazocine/metabolism , Sulfotransferases/metabolism , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/metabolism , Buprenorphine/administration & dosage , Carcinoma, Hepatocellular/metabolism , Cytosol/metabolism , Hep G2 Cells , Humans , Liver Neoplasms/metabolism , Naloxone/administration & dosage , Narcotic Antagonists/administration & dosage , Narcotic Antagonists/metabolism , Pentazocine/administration & dosage , Sulfates/metabolism
8.
J Bacteriol ; 194(22): 6206-16, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22984263

ABSTRACT

Branched-chain aminotransferases (BCAT), which utilize pyridoxal 5'-phosphate (PLP) as a cofactor, reversibly catalyze the transfer of the α-amino groups of three of the most hydrophobic branched-chain amino acids (BCAA), leucine, isoleucine, and valine, to α-ketoglutarate to form the respective branched-chain α-keto acids and glutamate. The BCAT from Deinococcus radiodurans (DrBCAT), an extremophile, was cloned and expressed in Escherichia coli for structure and functional studies. The crystal structures of the native DrBCAT with PLP and its complexes with L-glutamate and α-ketoisocaproate (KIC), respectively, have been determined. The DrBCAT monomer, comprising 358 amino acids, contains large and small domains connected with an interdomain loop. The cofactor PLP is located at the bottom of the active site pocket between two domains and near the dimer interface. The substrate (L-glutamate or KIC) is bound with key residues through interactions of the hydrogen bond and the salt bridge near PLP inside the active site pocket. Mutations of some interaction residues, such as Tyr71, Arg145, and Lys202, result in loss of the specific activity of the enzymes. In the interdomain loop, a dynamic loop (Gly173 to Gly179) clearly exhibits open and close conformations in structures of DrBCAT without and with substrates, respectively. DrBCAT shows the highest specific activity both in nature and under ionizing radiation, but with lower thermal stability above 60 °C, than either BCAT from Escherichia coli (eBCAT) or from Thermus thermophilus (HB8BCAT). The dimeric molecular packing and the distribution of cysteine residues at the active site and the molecular surface might explain the resistance to radiation but small thermal stability of DrBCAT.


Subject(s)
Deinococcus/enzymology , Glutamic Acid/chemistry , Keto Acids/chemistry , Transaminases/chemistry , Transaminases/metabolism , Amino Acid Sequence , Catalysis , Catalytic Domain , Crystallization , Crystallography, X-Ray , Enzyme Stability , Gene Expression Regulation, Bacterial , Gene Expression Regulation, Enzymologic , Models, Molecular , Molecular Sequence Data , Protein Conformation , Spectrophotometry , Substrate Specificity
9.
Biochem Pharmacol ; 84(9): 1186-95, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-22917559

ABSTRACT

Catecholic drugs had been reported to be metabolized through conjugation reactions, particularly methylation and sulfation. Whether and how these two Phase II conjugation reactions may occur in a concerted manner, however, remained unclear. The current study was designed to investigate the methylation and/or sulfation of five catecholic drugs. Analysis of the spent media of HepG2 cells metabolically labeled with [(35)S]sulfate in the presence of individual catecholic drugs revealed the presence of two [(35)S]sulfated metabolites for dopamine, epinephrine, isoproterenol, and isoetharine, but only one [(35)S]sulfated metabolite for apomorphine. Further analyses using tropolone, a catechol O-methyltransferase (COMT) inhibitor, indicated that one of the two [(35)S]sulfated metabolites of dopamine, epinephrine, isoproterenol, and isoetharine was a doubly conjugated (methylated and sulfated) product, since its level decreased proportionately with increasing concentrations of tropolone added to the labeling media. Moreover, while the inhibition of methylation resulted in a decrease of the total amount of [(35)S]sulfated metabolites, sulfation appeared to be capable of compensating the suppressed methylation in the metabolism of these four catecholic drugs. A two-stage enzymatic assay showed the sequential methylation and sulfation of dopamine, epinephrine, isoproterenol, and isoetharine mediated by, respectively, the COMT and the cytosolic sulfotransferase SULT1A3. Collectively, the results from the present study implied the concerted actions of the COMT and SULT1A3 in the metabolism of catecholic drugs.


Subject(s)
Catechol O-Methyltransferase/metabolism , Catechols/metabolism , Sulfotransferases/metabolism , Arylsulfotransferase , Catechol O-Methyltransferase Inhibitors , Hep G2 Cells , Humans , Methylation , Solubility , Sulfuric Acid Esters/metabolism , Tropolone/pharmacology
10.
J Biochem ; 152(3): 275-83, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22763752

ABSTRACT

Feed additives such as ractopamine and salbutamol are pharmacologically active compounds, acting primarily as ß-adrenergic agonists. This study was designed to investigate whether the sulfation of ractopamine and salbutamol may occur under the metabolic conditions and to identify the human cytosolic sulfotransferases (SULTs) that are capable of sulfating two major feed additive compounds, ractopamine and salbutamol. A metabolic labelling study showed the generation and release of [(35)S]sulfated ractopamine and salbutamol by HepG2 human hepatoma cells labelled with [(35)S]sulfate in the presence of these two compounds. A systematic analysis using 11 purified human SULTs revealed SULT1A3 as the major SULT responsible for the sulfation of ractopamine and salbutamol. The pH dependence and kinetic parameters were analyzed. Moreover, the inhibitory effects of ractopamine and salbutamol on SULT1A3-mediated dopamine sulfation were investigated. Cytosol or S9 fractions of human lung, liver, kidney and small intestine were examined to verify the presence of ractopamine-/salbutamol-sulfating activity in vivo. Of the four human organs, the small intestine displayed the highest activity towards both compounds. Collectively, these results imply that the sulfation mediated by SULT1A3 may play an important role in the metabolism and detoxification of ractopamine and salbutamol.


Subject(s)
Albuterol/metabolism , Cytosol/enzymology , Phenethylamines/metabolism , Sulfotransferases/metabolism , Sulfur/metabolism , Albuterol/chemistry , Albuterol/pharmacology , Cytosol/drug effects , Dopamine/metabolism , Hep G2 Cells , Humans , Hydrogen-Ion Concentration/drug effects , Isotope Labeling , Kinetics , Organ Specificity/drug effects , Phenethylamines/chemistry , Phenethylamines/pharmacology , Substrate Specificity/drug effects , Sulfates/metabolism , Sulfur Isotopes
11.
Aquat Toxicol ; 112-113: 11-8, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22360938

ABSTRACT

By searching the GenBank database, we identified sequences encoding three new zebrafish cytosolic sulfotransferases (SULTs). These three new zebrafish SULTs, designated SULT1 ST9, SULT3 ST4, and SULT3 ST5, were cloned, expressed, purified, and characterized. SULT1 ST9 appeared to be mostly involved in the metabolism and detoxification of xenobiotics such as ß-naphthol, ß-naphthylamine, caffeic acid and gallic acid. SULT3 ST4 showed strong activity toward endogenous compounds such as dehydroepiandrosterone (DHEA), pregnenolone, and 17ß-estradiol. SULT3 ST5 showed weaker, but significant, activities toward endogenous compounds such as DHEA and corticosterone, as well as xenobiotics including mestranol, ß-naphthylamine, ß-naphthol, and butylated hydroxyl anisole (BHA). pH-dependency and kinetic constants of these three enzymes were determined with DHEA, ß-naphthol, and 17ß-estradiol as substrates. Reverse transcription-polymerase chain reaction (RT-PCR) was performed to examine the expression of these three new zebrafish SULTs at different developmental stages during embryogenesis, through larval development, and on to maturity.


Subject(s)
Sulfotransferases/genetics , Sulfotransferases/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Zebrafish/genetics , Zebrafish/metabolism , Amino Acid Sequence , Animals , Cluster Analysis , Cytosol/enzymology , Gene Expression Regulation, Developmental , Hydrogen-Ion Concentration , Kinetics , Molecular Sequence Data , Protein Binding , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Substrate Specificity , Sulfotransferases/chemistry , Sulfotransferases/isolation & purification , Zebrafish/classification , Zebrafish Proteins/chemistry , Zebrafish Proteins/isolation & purification
12.
J Steroid Biochem Mol Biol ; 127(3-5): 307-14, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21839837

ABSTRACT

The current study was designed to examine the sulfation of bile acids and bile alcohols by the Zebra danio (Danio rerio) SULTs in comparison with human SULTs. A systematic analysis using the fifteen Zebra danio SULTs revealed that SULT3 ST2 and SULT3 ST3 were the major bile acid/alcohol-sulfating SULTs. Among the eleven human SULTs, only SULT2A1 was found to be capable of sulfating bile acids and bile alcohols. To further investigate the sulfation of bile acids and bile alcohols by the two Zebra danio SULT3 STs and the human SULT2A1, pH-dependence and kinetics of the sulfation of bile acids/alcohols were analyzed. pH-dependence experiments showed that the mechanisms underlying substrate recognition for the sulfation of lithocholic acid (a bile acid) and 5α-petromyzonol (a bile alcohol) differed between the human SULT2A1 and the Zebra danio SULT3 ST2 and ST3. Kinetic analysis indicated that both the two Zebra danio SULT3 STs preferred petromyzonol as substrate compared to bile acids. In contrast, the human SULT2A1 was more catalytically efficient toward lithocholic acid than petromyzonol. Collectively, the results imply that the Zebra danio and human SULTs have evolved to serve for the sulfation of, respectively, bile alcohols and bile acids, matching the cholanoid profile in these two vertebrate species.


Subject(s)
Bile Acids and Salts/metabolism , Cholestanols/metabolism , Cytosol/enzymology , Sulfates/metabolism , Sulfotransferases/metabolism , Animals , Humans , Hydrogen-Ion Concentration , Kinetics , Substrate Specificity , Zebrafish
13.
Aquat Toxicol ; 102(1-2): 18-23, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21371608

ABSTRACT

To establish the zebrafish as a model for investigating the methylation pathway of drug metabolism, we embarked on the molecular cloning of the zebrafish catechol O-methyltransferase (COMT). By searching the GenBank database, a zebrafish nucleotide sequence encoding a putative COMT was identified. Based on the sequence information, we designed and synthesized oligonucleotides corresponding to its 5'- and 3'-coding regions of this zebrafish COMT. Using the first-strand cDNA reverse-transcribed from the total RNA isolated from a 3-month-old adult female zebrafish as the template, the cDNA encoding the zebrafish COMT was PCR-amplified. The recombinant zebrafish COMT protein was subsequently expressed in and purified from BL21 (DE3) Escherichia coli cells transformed with the pGEX-2TK expression vector harboring the zebrafish COMT cDNA. Upon enzymatic characterization, purified COMT displayed methylating activity toward dopamine, dopa, and catecholestrogens, as well as three representative catechol drugs, methyldopa, dobutamine, and isoproterenol. A reverse transcription-polymerase chain reaction (RT-PCR) analysis revealed developmental stage-dependent expression of the zebrafish COMT during embryonic development and throughout the larval stage onto maturity. These results provide a foundation for investigating the involvement of COMT-mediated methylation in protection against the adverse effects of catechol drugs and other xenobiotic catechols during the developmental process.


Subject(s)
Catechol O-Methyltransferase/genetics , Catechol O-Methyltransferase/metabolism , Cloning, Molecular , Metabolic Detoxication, Phase II/physiology , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Animals , Base Sequence , Catechols/metabolism , Enzyme Assays , Gene Expression , Gene Expression Regulation, Developmental , Metabolic Detoxication, Phase II/genetics , Methylation , Models, Animal , Molecular Sequence Data , Polymerase Chain Reaction , Xenobiotics/metabolism , Zebrafish/genetics
14.
Toxicol Appl Pharmacol ; 251(2): 104-9, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21168432

ABSTRACT

During inflammation, potent reactive oxidants formed may cause chlorination and nitration of both free and protein-bound tyrosine. In addition to serving as biomarkers of inflammation-mediated oxidative stress, elevated levels of chlorotyrosine and nitrotyrosine have been linked to the pathogenesis of lung and vascular disorders. The current study was designed to investigate whether the lung cells are equipped with mechanisms for counteracting these tyrosine derivatives. By metabolic labeling, chlorotyrosine O-[³5S]sulfate and nitrotyrosine O-[³5S]sulfate were found to be generated and released into the labeling media of human lung endothelial and epithelial cells labeled with [³5S]sulfate in the presence of added chlorotyrosine and nitrotyrosine. Enzymatic assays using the eleven known human cytosolic sulfotransferases (SULTs) revealed SULT1A3 as the enzyme responsible for catalyzing the sulfation of chlorotyrosine and nitrotyrosine. Reverse transcription-polymerase chain reaction (RT-PCR) analysis demonstrated the expression of SULT1A3 in the lung endothelial and epithelial cells used in this study. Kinetic constants of the sulfation of chlorotyrosine and nitrotyrosine by SULT1A3 were determined. Collectively, these results suggest that sulfation by SULT1A3 in lung endothelial and epithelial cells may play a role in the inactivation and/or disposal of excess chlorotyrosine and nitrotyrosine generated during inflammation.


Subject(s)
Endothelial Cells/metabolism , Inflammation Mediators/physiology , Lung/metabolism , Respiratory Mucosa/metabolism , Sulfates/metabolism , Sulfotransferases/physiology , Tyrosine/analogs & derivatives , Arylsulfotransferase , Cell Line , Endothelial Cells/pathology , Humans , Lung/pathology , Respiratory Mucosa/pathology , Sulfates/chemistry , Tyrosine/antagonists & inhibitors , Tyrosine/chemistry , Tyrosine/metabolism
15.
Mol Microbiol ; 78(5): 1101-16, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21059110

ABSTRACT

The crystal structures of two active forms of dissimilatory sulphite reductase (Dsr) from Desulfovibrio gigas, Dsr-I and Dsr-II, are compared at 1.76 and 2.05 Å resolution respectively. The dimeric α2ß2γ2 structure of Dsr-I contains eight [4Fe-4S] clusters, two saddle-shaped sirohaems and two flat sirohydrochlorins. In Dsr-II, the [4Fe-4S] cluster associated with the sirohaem in Dsr-I is replaced by a [3Fe-4S] cluster. Electron paramagnetic resonance (EPR) of the active Dsr-I and Dsr-II confirm the co-factor structures, whereas EPR of a third but inactive form, Dsr-III, suggests that the sirohaem has been demetallated in addition to its associated [4Fe-4S] cluster replaced by a [3Fe-4S] centre. In Dsr-I and Dsr-II, the sirohydrochlorin is located in a putative substrate channel connected to the sirohaem. The γ-subunit C-terminus is inserted into a positively charged channel formed between the α- and ß-subunits, with its conserved terminal Cys104 side-chain covalently linked to the CHA atom of the sirohaem in Dsr-I. In Dsr-II, the thioether bond is broken, and the Cys104 side-chain moves closer to the bound sulphite at the sirohaem pocket. These different forms of Dsr offer structural insights into a mechanism of sulphite reduction that can lead to S3O6(2-), S2O3(2-) and S2-.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Desulfovibrio gigas/enzymology , Hydrogensulfite Reductase/chemistry , Hydrogensulfite Reductase/metabolism , Amino Acid Sequence , Bacterial Proteins/genetics , Catalysis , Catalytic Domain , Desulfovibrio gigas/chemistry , Desulfovibrio gigas/genetics , Hydrogensulfite Reductase/genetics , Molecular Conformation , Molecular Sequence Data
16.
Biol Pharm Bull ; 33(9): 1633-7, 2010.
Article in English | MEDLINE | ID: mdl-20823587

ABSTRACT

Prolonged exposure to high level of estrogen is a known risk factor for breast carcinogenesis. It has been suggested recently that nitrative stress may be an etiologic factor for breast carcinogenesis. Since sulfation plays a major role in the homeostasis of estrogens and their metabolites, we attempted in the present study to find out whether nitrative stress may affect the homeostasis of estrogens through sulfation. Metabolic labeling experiments revealed that the amount of sulfated 17beta-estradiol or 4-methoxyestradiol decreased dramatically in MCF-10A mammary epithelial cells incubated in the presence of 3-morpholinosydnonimine (SIN-1) or diethylenetriamine NONOate (DETA NONOate), two nitric oxide donors commonly used to simulate nitrative stress conditions. In searching for the mechanism underlying the decrease of the sulfation of 17beta-estradiol and 4-methoxyestradiol, we demonstrated in an in vitro nitration experiment, that the human cytosolic sulfotransferase isoform 1E1 (SULT1E1), a major estrogen-sulfating enzyme, lost its estrogen-sulfating activity proportionately to the degree of nitration on tyrosine residues. Moreover, cell lysates prepared from MCF-10A cells treated with SIN-1 or DETA NONOate also showed much lower 4-methoxyestradiol-sulfating activities, compared with those determined with cell lysate prepared from control MCF-10A cells.


Subject(s)
Epithelial Cells/enzymology , Estradiol/analogs & derivatives , Estradiol/metabolism , Mammary Glands, Human/enzymology , Nitric Oxide Donors/pharmacology , Sulfotransferases/metabolism , Cell Line , Dose-Response Relationship, Drug , Epithelial Cells/drug effects , Humans , Mammary Glands, Human/drug effects , Nitro Compounds/pharmacology , Oxidative Stress/drug effects , Oxidative Stress/physiology , Sulfotransferases/antagonists & inhibitors
17.
Curr Drug Metab ; 11(6): 538-46, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20545621

ABSTRACT

Cytosolic sulfotransferases (SULTs) are traditionally known as the Phase II drug-metabolizing or detoxifying enzymes that serve for the detoxification of drugs and other xenobiotics. These enzymes in general catalyze the transfer of a sulfonate group from the active sulfate, 3'-phosphoadenosine 5'-phosphosulfate (PAPS), to low-molecular weight substrate compounds containing hydroxyl or amino group(s). Despite considerable efforts made in recent years, some fundamental aspects of the SULTs, particularly their ontogeny, cell type/tissue/organ-specific distribution, and physiological relevance, particularly their involvement in drug metabolism and detoxification, still remain poorly understood. To better understand these fundamental issues, we have embarked on developing the zebrafish as a model for studies concerning the SULTs. To date, fifteen zebrafish SULTs have been cloned, expressed, purified, and characterized. These zebrafish SULTs, which fall into four major SULT gene families, exhibited differential substrate specificities and distinct patterns of expression at different stages during embryogenesis, through larval development, and on to maturity. The information obtained, as summarized in this review, provides a foundation for further investigation into the physiological and pharmacological involvement of the SULTs using the zebrafish as a model.


Subject(s)
Cytosol/enzymology , Models, Animal , Sulfotransferases/metabolism , Zebrafish/metabolism , Amino Acid Sequence , Animals , Humans , Molecular Sequence Data , Multigene Family , Sulfotransferases/biosynthesis , Sulfotransferases/genetics , Zebrafish/genetics
18.
Drug Metab Lett ; 4(2): 62-8, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20446911

ABSTRACT

To establish the zebrafish as a model to investigate drug metabolism through sulfation, we had previous cloned, expressed, and purified fourteen distinct zebrafish cytosolic sulfotransferases (SULTs). In the present study, we carried a systematic analysis of the sulfating activities of these fourteen zebrafish SULTs toward a panel of drug compounds. Results showed that four of the fourteen zebrafish SULTs showed no detectable activities toward any of the tested drugs. Among the other ten zebrafish SULTs, three SULT1 enzymes (SULT1 ST1, SULT1 ST2, and SULT1 ST3) displayed considerably stronger activities than the others toward the majority of the drug compounds tested. Specifically, SULT1 ST1, SULT1 ST2, and SULT1 ST3 showed the highest specific activities, at 26.9, 29.3, and 31.5 nmol/min/mg, toward aesculetin, 4-methylembelliferone, and dobutamine, respectively. To further investigate the sulfation of tested drugs by the responsible zebrafish SULT enzymes, the kinetics of the sulfation reactions were analyzed. Kinetic constants determined indicated that the sulfation of these drugs by the SULT enzymes tested is likely to be physiologically relevant. A metabolic labeling experiment using cultured zebrafish liver cells and HepG2 human hepatoma cells was performed. Results showed that zebrafish liver cells displayed a similar pattern of sulfation of the drugs tested as that of HepG2 cells, implying that human and zebrafish liver cells may share considerable similarities with regard to their constituent drug-sulfating SULT enzymes.


Subject(s)
Cytosol/enzymology , Pharmaceutical Preparations/metabolism , Sulfotransferases/metabolism , Zebrafish/metabolism , Animals , Cell Line, Tumor , Hepatocytes/enzymology , Hepatocytes/metabolism , Humans , Kinetics , Recombinant Proteins , Sulfates/metabolism , Sulfotransferases/genetics
19.
Biochem Biophys Res Commun ; 396(2): 429-34, 2010 May 28.
Article in English | MEDLINE | ID: mdl-20417180

ABSTRACT

The cytosolic sulfotransferases (SULTs) in vertebrates catalyze the sulfonation of endogenous thyroid/steroid hormones and catecholamine neurotransmitters, as well as a variety of xenobiotics, using 3'-phosphoadenosine 5'-phosphosulfate (PAPS) as the sulfonate donor. In this study, we determined the structures of SULT1A2 and an allozyme of SULT1A1, SULT1A1 *3, bound with 3'-phosphoadenosine 5'-phosphate (PAP), at 2.4 and 2.3A resolution, respectively. The conformational differences between the two structures revealed a plastic substrate-binding pocket with two channels and a switch-like substrate selectivity residue Phe247, providing clearly a structural basis for the substrate inhibition. In SULT1A2, Tyr149 extends approximately 2.1A further to the inside of the substrate-binding pocket, compared with the corresponding His149 residue in SULT1A1 *3. Site-directed mutagenesis study showed that, compared with the wild-type SULT1A2, mutant Tyr149Phe SULT1A2 exhibited a 40 times higher K(m) and two times lower V(max) with p-nitrophenol as substrate. These latter data imply a significant role of Tyr149 in the catalytic mechanism of SULT1A2.


Subject(s)
Arylsulfotransferase/antagonists & inhibitors , Arylsulfotransferase/chemistry , Arylsulfotransferase/genetics , Catalysis , Crystallography, X-Ray , Humans , Mutagenesis, Site-Directed , Mutation , Nitrophenols/chemistry , Phosphoadenosine Phosphosulfate/chemistry , Protein Conformation , Substrate Specificity , Tyrosine/chemistry , Tyrosine/genetics
20.
Article in English | MEDLINE | ID: mdl-20124706

ABSTRACT

The crystal structure of Bacillus amyloliquefaciens alpha-amylase (BAA) at 1.4 A resolution revealed ambiguities in the thermal adaptation of homologous proteins in this family. The final model of BAA is composed of two molecules in a back-to-back orientation, which is likely to be a consequence of crystal packing. Despite a high degree of identity, comparison of the structure of BAA with those of other liquefying-type alpha-amylases indicated moderate discrepancies at the secondary-structural level. Moreover, a domain-displacement survey using anisotropic B-factor and domain-motion analyses implied a significant contribution of domain B to the total flexibility of BAA, while visual inspection of the structure superimposed with that of B. licheniformis alpha-amylase (BLA) indicated higher flexibility of the latter in the central domain A. Therefore, it is suggested that domain B may play an important role in liquefying alpha-amylases, as its rigidity offers a substantial improvement in thermostability in BLA compared with BAA.


Subject(s)
Bacillus/enzymology , alpha-Amylases/chemistry , Amino Acid Sequence , Conserved Sequence , Crystallography, X-Ray , Enzyme Stability , Models, Molecular , Molecular Sequence Data , Protein Structure, Quaternary , Protein Structure, Tertiary , Sequence Alignment , Static Electricity , Structural Homology, Protein , Temperature , alpha-Amylases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...