Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Int J Biochem Cell Biol ; 40(12): 2686-91, 2008.
Article in English | MEDLINE | ID: mdl-18248844

ABSTRACT

In this review, we highlight the physical and enzymatic properties of the novel human sulfotransferase, SULT4A1. The gene is most highly expressed in selective regions of the brain, although work to date has failed to identify any specific endogenous substrate for the enzyme. SULT4A1 shares low homology with other human sulfotransferases. Nevertheless, it is highly conserved between species. Despite the low homology, it is structurally very similar to other cytosolic sulfotransferases with a conserved substrate binding domain, dimerization site and partial cofactor binding sites. However, the catalytic cavity is much smaller, and it has been suggested that the cofactor may not be accommodated within it. A recent link between variability in the 5'UTR of the SULT4A1 gene and schizophrenia has heightened interest in the endogenous function of the enzyme and its possible role in human disease.


Subject(s)
Sulfotransferases/chemistry , Sulfotransferases/metabolism , Amino Acid Motifs , Amino Acid Sequence , Binding Sites , Cloning, Molecular , Cytosol/enzymology , Cytosol/metabolism , DNA, Complementary/genetics , DNA, Complementary/metabolism , Dimerization , Humans , Models, Molecular , Molecular Sequence Data , Sequence Homology, Amino Acid , Substrate Specificity , Sulfotransferases/genetics
2.
Int J Biochem Cell Biol ; 39(4): 685-9, 2007.
Article in English | MEDLINE | ID: mdl-17110154

ABSTRACT

Sulfonation is an important conjugation reaction required for a range of biological processes including phase II metabolism, whereby sulfo-conjugation renders a compound more hydrophilic to aid its excretion. The major enzyme responsible for xenobiotic sulfonation is the widely expressed cytosolic sulfotransferase SULT1A1. The SULT1A1 crystal structure has provided insights into this enzyme's substrate specificity and catalytic function, including its role in the sulfonation of endogenous substrates such as oestrogens. Contrary to its metabolic role, SULT1A1 can also bioactivate compounds; it is known to sulfonate pro-carcinogens such as hydroxymethyl polycyclic aromatic hydrocarbons leading to highly reactive intermediates capable of forming DNA adducts, potentially resulting in mutagenesis. Given the role of SULT1A1 in these diverse functions and the discovery of allelic variants with differing catalytic activities, this enzyme has been the focus of numerous polymorphic studies investigating the link between inter-individual SULT1A1 variance and the etiology of a variety of cancers.


Subject(s)
Arylsulfotransferase/chemistry , Arylsulfotransferase/metabolism , Cytosol/enzymology , Catalysis , Humans , Models, Molecular , Protein Structure, Secondary , Protein Structure, Tertiary , Substrate Specificity
3.
Toxicol Sci ; 90(1): 5-22, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16322073

ABSTRACT

Sulfonation is an important reaction in the metabolism of numerous xenobiotics, drugs, and endogenous compounds. A supergene family of enzymes called sulfotransferases (SULTs) catalyze this reaction. In most cases, the addition of a sulfonate moiety to a compound increases its water solubility and decreases its biological activity. However, many of these enzymes are also capable of bioactivating procarcinogens to reactive electrophiles. In humans three SULT families, SULT1, SULT2, and SULT4, have been identified that contain at least thirteen distinct members. SULTs have a wide tissue distribution and act as a major detoxification enzyme system in adult and the developing human fetus. Nine crystal structures of human cytosolic SULTs have now been determined, and together with site-directed mutagenesis experiments and molecular modeling, we are now beginning to understand the factors that govern distinct but overlapping substrate specificities. These studies have also provided insight into the enzyme kinetics and inhibition characteristics of these enzymes. The regulation of human SULTs remains as one of the least explored areas of research in the field, though there have been some recent advances on the molecular transcription mechanism controlling the individual SULT promoters. Interindividual variation in sulfonation capacity may be important in determining an individual's response to xenobiotics, and recent studies have begun to suggest roles for SULT polymorphism in disease susceptibility. This review aims to provide a summary of our present understanding of the function of human cytosolic sulfotransferases.


Subject(s)
Sulfotransferases/metabolism , Xenobiotics/pharmacokinetics , Animals , Biotransformation , Crystallization , Humans , Isoenzymes , Models, Molecular , Protein Conformation , Sulfotransferases/chemistry , Sulfotransferases/genetics
4.
J Biol Chem ; 280(50): 41482-6, 2005 Dec 16.
Article in English | MEDLINE | ID: mdl-16221673

ABSTRACT

Human SULT1A1 belongs to the supergene family of sulfotransferases (SULTs) involved in the sulfonation of xeno- and endobiotics. The enzyme is also one of the SULTs responsible for metabolic activation of mutagenic and carcinogenic compounds and therefore is implicated in various cancer forms. Further, it is not well understood how substrate inhibition takes place with rigid fused multiring substrates such as 17beta-estradiol (E2) at high substrate concentrations when subcellular fractions or recombinant enzymes are used. To investigate how estradiol binds to SULT1A1, we co-crystallized SULT1A1 with sulfated estradiol and the cofactor product, PAP (3'-phosphoadenosine 5'-phosphate). The crystal structure of SULT1A1 that we present here has PAP and one molecule of E2 bound in a nonproductive mode in the active site. The structure reveals how the SULT1A1 binding site undergoes conformational changes to accept fused ring substrates such as steroids. In agreement with previous reports, the enzyme shows partial substrate inhibition at high concentrations of E2. A model to explain these kinetics is developed based on the formation of an enzyme x PAP x E2 dead-end complex during catalysis. This model provides a very good quantitative description of the rate versus the [E2] curve. This dead-end complex is proposed to be that described by the observed structure, where E2 is bound in a nonproductive mode.


Subject(s)
Arylsulfotransferase/chemistry , Estradiol/chemistry , Adenosine Diphosphate/chemistry , Animals , Arylsulfotransferase/metabolism , Binding Sites , Carcinogens , Catalysis , Crystallography, X-Ray , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Humans , Kinetics , Mice , Models, Chemical , Models, Molecular , Mutagenesis , Protein Binding , Protein Conformation , Protein Structure, Secondary , Protein Structure, Tertiary , Substrate Specificity , X-Ray Diffraction
5.
Methods Enzymol ; 400: 147-65, 2005.
Article in English | MEDLINE | ID: mdl-16399348

ABSTRACT

The three human SULT1A sulfotransferase enzymes are closely related in amino acid sequence (>90%), yet differ in their substrate preference and tissue distribution. SULT1A1 has a broad tissue distribution and metabolizes a range of xenobiotics as well as endogenous substrates such as estrogens and iodothyronines. While the localization of SULT1A2 is poorly understood, it has been shown to metabolize a number of aromatic amines. SULT1A3 is the major catecholamine sulfonating form, which is consistent with it being expressed principally in the gastrointestinal tract. SULT1A proteins are encoded by three separate genes, located in close proximity to each other on chromosome 16. The presence of differential 5'-untranslated regions identified upon cloning of the SULT1A cDNAs suggested the utilization of differential transcriptional start sites and/or differential splicing. This chapter describes the methods utilized by our laboratory to clone and assay the activity of the promoters flanking these different untranslated regions found on SULT1A genes. These techniques will assist investigators in further elucidating the differential mechanisms that control regulation of the human SULT1A genes. They will also help reveal how different cellular environments and polymorphisms affect the activity of SULT1A gene promoters.


Subject(s)
Arylsulfotransferase/genetics , Gene Expression Regulation, Enzymologic/genetics , Promoter Regions, Genetic , Animals , Arylsulfotransferase/classification , Cell Line , Cloning, Molecular/methods , Drosophila , Enzyme Activation , Humans , Polymorphism, Genetic , Protein Isoforms/genetics
6.
Mol Pharmacol ; 66(6): 1690-701, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15383623

ABSTRACT

Human sulfotransferase SULT1A1 is an important phase II xenobiotic metabolizing enzyme that is highly expressed in the liver and mediates the sulfonation of drugs, carcinogens, and steroids. Until this study, the transcriptional regulation of the SULT1A subfamily had been largely unexplored. Preliminary experiments in primary human hepatocytes showed that SULT1A mRNA levels were not changed in response to nuclear receptor activators, such as dexamethasone and 3-methylcolanthrene, unlike other metabolizing enzymes. Using HepG2 cells, the high activity of the TATA-less SULT1A1 promoter was shown to be dependent on the presence of Sp1 and Ets transcription factor binding sites (EBS), located within -112 nucleotides from the transcriptional start site. The homologous promoter of the closely related SULT1A3 catecholamine sulfotransferase, which is expressed at negligible levels in the adult liver, displayed 70% less activity than SULT1A1. This was shown to be caused by a two-base pair difference in the EBS. The Ets transcription factor GA binding protein (GABP) was shown to bind the SULT1A1 EBS and could transactivate the SULT1A1 promoter in Drosophila melanogaster S2 cells. Cotransfection of Sp1 could synergistically enhance GABP-mediated activation by 10-fold. Although Sp1 and GABP alone could induce SULT1A3 promoter activity, the lack of the EBS on this promoter prevented a synergistic interaction between the two factors. This study reports the first insight into the transcriptional regulation of the SULT1A1 gene and identifies a crucial difference in regulation of the closely related SULT1A3 gene, which accounts for the two enzymes' differential expression patterns observed in the adult liver.


Subject(s)
Arylsulfotransferase/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Enzymologic/genetics , Hepatocytes/enzymology , Sp1 Transcription Factor/metabolism , Transcription Factors/metabolism , 5' Untranslated Regions/genetics , Base Sequence , DNA Primers , GA-Binding Protein Transcription Factor , Humans , Molecular Sequence Data , Promoter Regions, Genetic , Random Amplified Polymorphic DNA Technique/methods , Reverse Transcriptase Polymerase Chain Reaction , Sulfotransferases/genetics
7.
J Biol Chem ; 279(18): 18799-805, 2004 Apr 30.
Article in English | MEDLINE | ID: mdl-14871892

ABSTRACT

Human SULT1A1 is primarily responsible for sulfonation of xenobiotics, including the activation of promutagens, and it has been implicated in several forms of cancer. Human SULT1A3 has been shown to be the major sulfotransferase that sulfonates dopamine. These two enzymes shares 93% amino acid sequence identity and have distinct but overlapping substrate preferences. The resolution of the crystal structures of these two enzymes has enabled us to elucidate the mechanisms controlling their substrate preferences and inhibition. The presence of two p-nitrophenol (pNP) molecules in the crystal structure of SULT1A1 was postulated to explain cooperativity at low and inhibition at high substrate concentrations, respectively. In SULT1A1, substrate inhibition occurs with pNP as the substrate but not with dopamine. For SULT1A3, substrate inhibition is found for dopamine but not with pNP. We investigated how substrate inhibition occurs in these two enzymes using molecular modeling, site-directed mutagenesis, and kinetic analysis. The results show that residue Phe-247 of SULT1A1, which interacts with both p-nitrophenol molecules in the active site, is important for substrate inhibition. Mutation of phenylalanine to leucine at this position in SULT1A1 results in substrate inhibition by dopamine. We also propose, based on modeling and kinetic studies, that substrate inhibition by dopamine in SULT1A3 is caused by binding of two dopamine molecules in the active site.


Subject(s)
Mutation, Missense , Sulfotransferases/antagonists & inhibitors , Sulfotransferases/genetics , Arylsulfotransferase , Binding Sites/genetics , Dopamine/metabolism , Enzyme Inhibitors , Humans , Kinetics , Mutagenesis, Site-Directed , Nitrophenols/metabolism , Protein Binding
8.
J Histochem Cytochem ; 51(12): 1655-64, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14623933

ABSTRACT

Cytosolic sulfotransferases are believed to play a role in the neuromodulation of certain neurotransmitters and drugs. To date, four cytosolic sulfotransferases have been shown to be expressed in human brain. Recently, a novel human brain sulfotransferase has been identified and characterized, although its role and localization in the brain are unknown. Here we present the first immunohistochemical (IHC) localization of SULT4A1 in human brain using an affinity-purified polyclonal antibody raised against recombinant human SULT4A1. These results are supported and supplemented by the IHC localization of SULT4A1 in rat brain. In both human and rat brains, strong reactivity was found in several brain regions, including cerebral cortex, cerebellum, pituitary, and brainstem. Specific signal was entirely absent on sections for which preimmune serum from the corresponding animal, processed in the same way as the postimmune serum, was used in the primary screen. The findings from this study may assist in determining the physiological role of this SULT isoform.


Subject(s)
Brain/metabolism , Sulfotransferases/metabolism , Animals , Antibodies , Brain/anatomy & histology , Escherichia coli/metabolism , Humans , Immunohistochemistry , Rats , Recombinant Proteins/immunology , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism
9.
J Biol Chem ; 278(9): 7655-62, 2003 Feb 28.
Article in English | MEDLINE | ID: mdl-12471039

ABSTRACT

Sulfonation catalyzed by sulfotransferase enzymes plays an important role in chemical defense mechanisms against various xenobiotics but also bioactivates carcinogens. A major human sulfotransferase, SULT1A1, metabolizes and/or bioactivates many endogenous compounds and is implicated in a range of cancers because of its ability to modify diverse promutagen and procarcinogen xenobiotics. The crystal structure of human SULT1A1 reported here is the first sulfotransferase structure complexed with a xenobiotic substrate. An unexpected finding is that the enzyme accommodates not one but two molecules of the xenobiotic model substrate p-nitrophenol in the active site. This result is supported by kinetic data for SULT1A1 that show substrate inhibition for this small xenobiotic. The extended active site of SULT1A1 is consistent with binding of diiodothyronine but cannot easily accommodate beta-estradiol, although both are known substrates. This observation, together with evidence for a disorder-order transition in SULT1A1, suggests that the active site is flexible and can adapt its architecture to accept diverse hydrophobic substrates with varying sizes, shapes and flexibility. Thus the crystal structure of SULT1A1 provides the molecular basis for substrate inhibition and reveals the first clues as to how the enzyme sulfonates a wide variety of lipophilic compounds.


Subject(s)
Arylsulfotransferase , Sulfotransferases/chemistry , Amino Acid Sequence , Binding Sites , Carcinogens , Crystallography, X-Ray , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Humans , Kinetics , Ligands , Models, Chemical , Models, Molecular , Molecular Sequence Data , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Substrate Specificity , Xenobiotics/pharmacology
10.
Clin Exp Pharmacol Physiol ; 29(9): 754-8, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12165038

ABSTRACT

1. Sulphotransferases are a superfamily of enzymes involved in both detoxification and bioactivation of endogenous and exogenous compounds. The arylsulphotransferase SULT1A1 has been implicated in a decreased activity and thermostability when the wild-type arginine at position 213 of the coding sequence is substituted by a histidine. SULT1A1 is the isoform primarily associated with the conversion of dietary N-OH arylamines to DNA binding adducts and is therefore of interest to determine whether this polymorphism is linked to colorectal cancer. 2. Genotyping, using polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) analysis, was performed using DNA samples of healthy control subjects (n = 402) and patients with histologically proven colorectal cancer (n = 383). Both control and test populations possessed similar frequencies for the mutant allele (32.1 and 31%, respectively; P = 0.935). Results were not altered when age and gender were considered as potential confounders in a logistic regression analysis. 3. Examination of the sulphonating ability of the two allozymes with respect to the substrates p-nitrophenol and paracetamol showed that the affinity and rate of sulphonation was unaffected by substitution of arginine to histidine at position 213 of the amino acid sequence. 4. From this study, we conclude that the SULT1A1 R213H polymorphism is not linked with colorectal cancer in this elderly Australian population.


Subject(s)
Arylsulfotransferase , Colorectal Neoplasms/genetics , Polymorphism, Genetic/genetics , Sulfotransferases/genetics , Aged , Amino Acid Substitution/genetics , Arginine/genetics , Case-Control Studies , Chi-Square Distribution , Female , Gene Frequency/genetics , Genotype , Histidine/genetics , Humans , Logistic Models , Male , Odds Ratio
SELECTION OF CITATIONS
SEARCH DETAIL
...