Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 18 de 18
1.
Nat Commun ; 15(1): 4214, 2024 May 17.
Article En | MEDLINE | ID: mdl-38760332

The liver gene expression of the peroxisomal ß-oxidation enzyme acyl-coenzyme A oxidase 1 (ACOX1), which catabolizes very long chain fatty acids (VLCFA), increases in the context of obesity, but how this pathway impacts systemic energy metabolism remains unknown. Here, we show that hepatic ACOX1-mediated ß-oxidation regulates inter-organ communication involved in metabolic homeostasis. Liver-specific knockout of Acox1 (Acox1-LKO) protects mice from diet-induced obesity, adipose tissue inflammation, and systemic insulin resistance. Serum from Acox1-LKO mice promotes browning in cultured white adipocytes. Global serum lipidomics show increased circulating levels of several species of ω-3 VLCFAs (C24-C28) with previously uncharacterized physiological role that promote browning, mitochondrial biogenesis and Glut4 translocation through activation of the lipid sensor GPR120 in adipocytes. This work identifies hepatic peroxisomal ß-oxidation as an important regulator of metabolic homeostasis and suggests that manipulation of ACOX1 or its substrates may treat obesity-associated metabolic disorders.


Acyl-CoA Oxidase , Liver , Mice, Knockout , Obesity , Animals , Liver/metabolism , Mice , Acyl-CoA Oxidase/metabolism , Acyl-CoA Oxidase/genetics , Obesity/metabolism , Obesity/genetics , Male , Insulin Resistance , Mice, Inbred C57BL , Oxidation-Reduction , Lipid Metabolism , Adipose Tissue/metabolism , Diet, High-Fat , Energy Metabolism , Fatty Acids/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/genetics
2.
JACS Au ; 3(4): 1076-1088, 2023 Apr 24.
Article En | MEDLINE | ID: mdl-37124302

G protein-coupled receptors (GPCRs) are the largest superfamily of human membrane target proteins for approved drugs. GPCR ligands can have a complex array of pharmacological activities. Among these activities, biased agonists have potential to serve as both chemical probes to understand specific aspects of receptor signaling and therapeutic leads with more specific, desired activity. Challenges exist, however, in the development of new biased activators due, in part, to the low throughput of traditional screening approaches. DNA-encoded chemical libraries (DELs) dramatically improve the throughput of drug discovery by allowing a collective selection, rather than discrete screening, of large compound libraries. The use of DELs has been largely limited to affinity-based selections against purified protein targets, which identify binders only. Herein, we report a split protein complementation approach that allows direct identification of DNA-linked molecules that induce the dimerization of two proteins. We used this selection with a DEL against opioid receptor GPCRs on living cells for the identification of small molecules that possess the specific function of activation of either ß-arrestin or G protein signaling pathways. This approach was applied to δ-, µ-, and κ-opioid receptors and enabled the discovery of compound [66,66], a selective, G-protein-biased agonist of the κ-opioid receptor (EC50 = 100 nM, E max = 82%, Gi bias factor = 6.6). This approach should be generally applicable for the direct selection of chemical inducers of dimerization from DELs and expand the utility of DELs to enrich molecules with a specific and desired biochemical function.

3.
Nat Commun ; 14(1): 1338, 2023 03 11.
Article En | MEDLINE | ID: mdl-36906681

The κ-opioid receptor (KOR) has emerged as an attractive drug target for pain management without addiction, and biased signaling through particular pathways of KOR may be key to maintaining this benefit while minimizing side-effect liabilities. As for most G protein-coupled receptors (GPCRs), however, the molecular mechanisms of ligand-specific signaling at KOR have remained unclear. To better understand the molecular determinants of KOR signaling bias, we apply structure determination, atomic-level molecular dynamics (MD) simulations, and functional assays. We determine a crystal structure of KOR bound to the G protein-biased agonist nalfurafine, the first approved KOR-targeting drug. We also identify an arrestin-biased KOR agonist, WMS-X600. Using MD simulations of KOR bound to nalfurafine, WMS-X600, and a balanced agonist U50,488, we identify three active-state receptor conformations, including one that appears to favor arrestin signaling over G protein signaling and another that appears to favor G protein signaling over arrestin signaling. These results, combined with mutagenesis validation, provide a molecular explanation of how agonists achieve biased signaling at KOR.


Morphinans , Receptors, Opioid, kappa , Receptors, Opioid, kappa/metabolism , GTP-Binding Proteins/metabolism , Arrestin/metabolism , Analgesics, Opioid
4.
Nature ; 613(7945): 767-774, 2023 01.
Article En | MEDLINE | ID: mdl-36450356

Mu-opioid receptor (µOR) agonists such as fentanyl have long been used for pain management, but are considered a major public health concern owing to their adverse side effects, including lethal overdose1. Here, in an effort to design safer therapeutic agents, we report an approach targeting a conserved sodium ion-binding site2 found in µOR3 and many other class A G-protein-coupled receptors with bitopic fentanyl derivatives that are functionalized via a linker with a positively charged guanidino group. Cryo-electron microscopy structures of the most potent bitopic ligands in complex with µOR highlight the key interactions between the guanidine of the ligands and the key Asp2.50 residue in the Na+ site. Two bitopics (C5 and C6 guano) maintain nanomolar potency and high efficacy at Gi subtypes and show strongly reduced arrestin recruitment-one (C6 guano) also shows the lowest Gz efficacy among the panel of µOR agonists, including partial and biased morphinan and fentanyl analogues. In mice, C6 guano displayed µOR-dependent antinociception with attenuated adverse effects, supporting the µOR sodium ion-binding site as a potential target for the design of safer analgesics. In general, our study suggests that bitopic ligands that engage the sodium ion-binding pocket in class A G-protein-coupled receptors can be designed to control their efficacy and functional selectivity profiles for Gi, Go and Gz subtypes and arrestins, thus modulating their in vivo pharmacology.


Drug Design , Fentanyl , Morphinans , Receptors, Opioid, mu , Animals , Mice , Analgesics, Opioid/chemistry , Analgesics, Opioid/metabolism , Arrestins/metabolism , Cryoelectron Microscopy , Fentanyl/analogs & derivatives , Fentanyl/chemistry , Fentanyl/metabolism , Ligands , Morphinans/chemistry , Morphinans/metabolism , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/chemistry , Receptors, Opioid, mu/metabolism , Receptors, Opioid, mu/ultrastructure , Binding Sites , Nociception
5.
Molecules ; 27(3)2022 Jan 18.
Article En | MEDLINE | ID: mdl-35163856

In our society today, pain has become a main source of strain on most individuals. It is crucial to develop novel treatments against pain while focusing on decreasing their adverse effects. Throughout the extent of development for new pain therapies, the nociceptin/orphanin FQ receptor (NOP receptor) has appeared to be an encouraging focal point. Concentrating on NOP receptor to treat chronic pain with limited range of unwanted effects serves as a suitable alternative to prototypical opioid morphine that could potentially lead to life-threatening effects caused by respiratory depression in overdose, as well as generate abuse and addiction. In addition to these harmful effects, the uprising opioid epidemic is responsible for becoming one of the most disastrous public health issues in the US. In this article, the contributing molecular and cellular structure in controlling the cellular trafficking of NOP receptor and studies that support the role of NOP receptor and its ligands in pain management are reviewed.


Narcotic Antagonists/pharmacology , Pain/drug therapy , Receptors, Opioid/chemistry , Animals , Humans , Ligands , Pain/metabolism , Pain/pathology , Receptors, Opioid/metabolism , Nociceptin Receptor
6.
Methods Cell Biol ; 166: 161-177, 2021.
Article En | MEDLINE | ID: mdl-34752331

Nanobodies have emerged as useful tools to study G protein-coupled receptor (GPCR) structure, dynamic, and subcellular localization. Initially, several nanobodies have been developed as chaperones to facilitate GPCR crystallization. To explore their potential as biosensors to monitor receptor activation and dynamics, we here described protocols to characterize nanobody's interaction with GPCRs and their application as probes for protein identification and visualization on the cellular level. We also introduced a chimeric approach to enable a kappa-opioid receptor derived nanobody to bind to other GPCRs, including orphan GPCRs whose endogenous ligand or intracellular transducers are unknown. This approach provides a reporter assay to identify tool molecules to study the function of orphan GPCRs.


Biosensing Techniques , Single-Domain Antibodies , Ligands , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Single-Domain Antibodies/chemistry , Single-Domain Antibodies/genetics
7.
Expert Opin Drug Metab Toxicol ; 17(7): 767-784, 2021 Jul.
Article En | MEDLINE | ID: mdl-34107842

INTRODUCTION: Cytosolic sulfotransferases (SULTs)-mediated sulfation is critically involved in the metabolism of key endogenous compounds, such as catecholamines and thyroid/steroid hormones, as well as a variety of drugs and other xenobiotics. Studies performed in the past three decades have yielded a good understanding about the enzymology of the SULTs and their structural biology, phylogenetic relationships, tissue/organ-specific/developmental expression, as well as the regulation of the SULT gene expression. An emerging area is related to the functional impact of the SULT genetic polymorphisms. AREAS COVERED: The current review aims to summarize our current knowledge about the above-mentioned aspects of the SULT research. An emphasis is on the information concerning the effects of the polymorphisms of the SULT genes on the functional activity of the SULT allozymes and the associated physiological, pharmacological, and clinical implications. EXPERT OPINION: Elucidation of how SULT SNPs may influence the drug-sulfating activity of SULT allozymes will help understand the differential drug metabolism and eventually aid in formulating personalized drug regimens. Moreover, the information concerning the differential sulfating activities of SULT allozymes toward endogenous compounds may allow for the development of strategies for mitigating anomalies in the metabolism of these endogenous compounds in individuals with certain SULT genotypes.


Pharmaceutical Preparations/metabolism , Sulfotransferases/genetics , Animals , Cytosol/metabolism , Gene Expression Regulation, Enzymologic , Genotype , Humans , Polymorphism, Single Nucleotide , Sulfates/metabolism , Sulfotransferases/metabolism
8.
J Biochem ; 170(3): 419-426, 2021 Oct 12.
Article En | MEDLINE | ID: mdl-33950190

Doxorubicin is a chemotherapeutic drug widely utilized in cancer treatment. An enzyme critical to doxorubicin metabolism is the cytosolic sulfotransferase (SULT) SULT1C4. This study investigated the functional impact of SULT1C4 single nucleotide polymorphisms (SNPs) on the sulfation of doxorubicin by SULT1C4 allozymes. A comprehensive database search was performed to identify various SULT1C4 SNPs. Ten nonsynonymous SULT1C4 SNPs were selected, and the corresponding cDNAs, packaged in pGEX-2TK expression vector, were generated via site-directed mutagenesis. Respective SULT1C4 allozymes were bacterially expressed and purified by affinity chromatography. Purified SULT1C4 allozymes, in comparison with the wild-type enzyme, were analysed for sulphating activities towards doxorubicin and 4-nitrophenol, a prototype substrate. Results obtained showed clearly differential doxorubicin-sulphating activity of SULT1C4 allozymes, implying differential metabolism of doxorubicin through sulfation in individuals with distinct SULT1C4 genotypes.


Doxorubicin/metabolism , Polymorphism, Single Nucleotide , Sulfotransferases/genetics , Sulfotransferases/metabolism , Cytosol/metabolism , Genotype , Humans , Isoenzymes/metabolism , Kinetics , Mutagenesis, Site-Directed , Nitrophenols/metabolism , Sulfates/metabolism
9.
Eur J Drug Metab Pharmacokinet ; 46(1): 105-118, 2021 Jan.
Article En | MEDLINE | ID: mdl-33064293

BACKGROUND AND OBJECTIVES: Previous studies have revealed that sulfation, as mediated by the estrogen-sulfating cytosolic sulfotransferase (SULT) SULT1E1, is involved in the metabolism of 17ß-estradiol (E2), 4-hydroxytamoxifen (4OH-tamoxifen), and diethylstilbestrol in humans. It is an interesting question whether the genetic polymorphisms of SULT1E1, the gene that encodes the SULT1E1 enzyme, may impact on the metabolism of E2 and these two drug compounds through sulfation. METHODS: In this study, five missense coding single nucleotide polymorphisms of the SULT1E1 gene were selected to investigate the sulfating activity of the coded SULT1E1 allozymes toward E2, 4OH-tamoxifen, and diethylstilbestrol. Corresponding cDNAs were generated by site-directed mutagenesis, and recombinant SULT1E1 allozymes were bacterially expressed, affinity-purified, and characterized using enzymatic assays. RESULTS: Purified SULT1E1 allozymes were shown to display differential sulfating activities toward E2, 4OH-tamoxifen, and diethylstilbestrol. Kinetic analysis revealed further distinct Km (reflecting substrate affinity) and Vmax (reflecting catalytic activity) values of the five SULT1E1 allozymes with E2, 4OH-tamoxifen, and diethylstilbestrol as substrates. CONCLUSIONS: Taken together, these findings highlighted the significant differences in E2-, as well as the drug-sulfating activities of SULT1E1 allozymes, which may have implications in the differential metabolism of E2, 4OH-tamoxifen, and diethylstilbestrol in individuals with different SULT1E1 genotypes.


Diethylstilbestrol/metabolism , Estradiol/metabolism , Polymorphism, Single Nucleotide/genetics , Sulfotransferases/genetics , Sulfotransferases/metabolism , Tamoxifen/analogs & derivatives , Diethylstilbestrol/pharmacology , Dose-Response Relationship, Drug , Estradiol/pharmacology , Estrogen Antagonists/metabolism , Estrogen Antagonists/pharmacology , Estrogens/metabolism , Estrogens/pharmacology , Estrogens, Non-Steroidal/metabolism , Estrogens, Non-Steroidal/pharmacology , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Protein Structure, Secondary , Sulfotransferases/chemistry , Tamoxifen/metabolism , Tamoxifen/pharmacology
10.
Biochemistry ; 60(18): 1420-1429, 2021 05 11.
Article En | MEDLINE | ID: mdl-33274929

Pain management devoid of serious opioid adverse effects is still far from reach despite vigorous research and development efforts. Alternatives to classical opioids have been sought for years, and mounting reports of individuals finding pain relief with kratom have recently intensified research on this natural product. Although the composition of kratom is complex, the pharmacological characterization of its most abundant alkaloids has drawn attention to three molecules in particular, owing to their demonstrated antinociceptive activity and limited side effects in vivo. These three molecules are mitragynine (MG), its oxidized active metabolite, 7-hydroxymitragynine (7OH), and the indole-to-spiropseudoindoxy rearrangement product of MG known as mitragynine pseudoindoxyl (MP). Although these three alkaloids have been shown to preferentially activate the G protein signaling pathway by binding and allosterically modulating the µ-opioid receptor (MOP), a molecular level understanding of this process is lacking and yet important for the design of improved therapeutics. The molecular dynamics study and experimental validation reported here provide an atomic level description of how MG, 7OH, and MP bind and allosterically modulate the MOP, which can eventually guide structure-based drug design of improved therapeutics.


Analgesics, Opioid/pharmacology , Mitragyna/chemistry , Receptors, Opioid, mu/agonists , Secologanin Tryptamine Alkaloids/pharmacology , Allosteric Regulation , Analgesics, Opioid/chemistry , Humans , Models, Molecular , Molecular Docking Simulation , Molecular Structure , Phytotherapy , Protein Binding , Protein Conformation , Secologanin Tryptamine Alkaloids/chemistry , Structure-Activity Relationship
11.
Nat Prod Res ; 34(6): 797-803, 2020 Mar.
Article En | MEDLINE | ID: mdl-30398375

Previous studies have revealed sulfation as a major pathway for the metabolism of hesperetin, naringenin and apigenin. The current study was designed to identify the human cytosolic sulfotransferase (SULT) enzyme(s) capable of sulfating these flavonoid compounds. Of the thirteen human SULTs, six (1A1, 1A2, 1A3, 1B2, 1C4, 1E1) displayed significant sulfating activity toward hesperetin, five (1A1, 1A2, 1A3, 1B2, 1C4) displayed sulfating activity towards naringenin, and four (1A1, 1A2, 1A3, 1C4) showed sulfating activity towards apigenin. Of the four human organ specimens tested, liver and intestine cytosols displayed much higher hesperetin-, naringenin- and apigenin-sulfating activity than lung and kidney cytosols. Moreover, sulfation of hesperetin, naringenin and apigenin was shown to take place in HepG2 human hepatoma cells and Caco-2 human colon adenocarcinoma cells under cultured conditions. Taken together, these results provided a biochemical basis underlying the metabolism of hesperetin, naringenin and apigenin through sulfation in humans.[Formula: see text].


Apigenin/metabolism , Flavanones/metabolism , Hesperidin/metabolism , Metabolic Networks and Pathways , Sulfates/metabolism , Sulfotransferases/metabolism , Cell Line, Tumor , Cytosol/enzymology , Humans , Intestinal Mucosa/metabolism , Kidney/metabolism , Liver/metabolism , Lung/metabolism
12.
Mol Cell Endocrinol ; 496: 110535, 2019 10 01.
Article En | MEDLINE | ID: mdl-31400397

Pregnenolone and dehydroepiandrosterone (DHEA) are hydroxysteroids that serve as biosynthetic precursors for steroid hormones in human body. SULT2B1b has been reported to be critically involved in the sulfation of pregnenolone and DHEA, particularly in the sex steroid-responsive tissues. The current study was designed to investigate the impact of the genetic polymorphisms of SULT2B1 on the sulfation of DHEA and pregnenolone by SULT2B1b allozymes. Ten SULT2B1b allozymes previously prepared were shown to exhibit differential sulfating activities toward DHEA and pregnenolone in comparison to the wild-type enzyme. Kinetic studies revealed further significant changes in their substrate-binding affinity and catalytic activity toward DHEA and pregnenolone. Taken together, these results indicated clearly a profound effect of SULT2B1 genetic polymorphisms on the sulfating activity of SULT2B1b allozymes toward DHEA and pregnenolone, which may have implications in inter-individual variations in the homeostasis of these two important steroid precursors.


Dehydroepiandrosterone/chemistry , Polymorphism, Single Nucleotide , Pregnenolone/chemistry , Sulfotransferases/chemistry , Humans , Isoenzymes , Sulfotransferases/genetics
13.
Pharmacogenet Genomics ; 29(5): 99-105, 2019 07.
Article En | MEDLINE | ID: mdl-31145702

OBJECTIVES: Phenylephrine and salbutamol are drugs that are used widely to treat diseases/disorders, such as nasal congestion, hypotension, and asthma, in individuals of different age groups. Human cytosolic sulfotransferase (SULT) SULT1A3 has been shown to be critically involved in the metabolism of these therapeutic agents. This study was carried out to investigate the effects of single nucleotide polymorphisms of human SULT1A3 and SULT1A4 genes on the sulfation of phenylephrine and salbutamol by SULT1A3 allozymes. MATERIALS AND METHODS: Wild-type and SULT1A3 allozymes, prepared previously by site-directed mutagenesis in conjunction with bacterial expression and affinity purification, were analyzed for sulfating activity using an established assay procedure. RESULTS: Purified SULT1A3 allozymes, in comparison with the wild-type enzyme, showed differential sulfating activities toward phenylephrine and salbutamol. Kinetic studies showed further significant variations in their substrate-binding affinity and catalytic activity toward phenylephrine and salbutamol. CONCLUSION: The results obtained showed clearly the differential enzymatic characteristics of SULT1A3 allozymes in mediating the sulfation of phenylephrine and salbutamol. This information may contribute toward a better understanding of the pharmacokinetics of these two drugs in individuals with distinct SULT1A3 and/or SULT1A4 genotypes.


Albuterol/metabolism , Arylsulfotransferase/genetics , Phenylephrine/metabolism , Sulfotransferases/genetics , Albuterol/therapeutic use , Arylsulfotransferase/chemistry , Arylsulfotransferase/metabolism , Asthma/drug therapy , Asthma/genetics , Genotype , Humans , Hypotension/drug therapy , Hypotension/genetics , Isoenzymes/genetics , Isoenzymes/metabolism , Kinetics , Mutagenesis, Site-Directed , Phenylephrine/therapeutic use , Polymorphism, Single Nucleotide/genetics , Sulfates/metabolism , Sulfotransferases/chemistry , Sulfotransferases/metabolism
14.
Pharmacol Rep ; 71(2): 257-265, 2019 Apr.
Article En | MEDLINE | ID: mdl-30822619

BACKGROUND: Non-opioid and opioid analgesics, as over-the-counter or prescribed medications, are widely used for the management of a diverse array of pathophysiological conditions. Previous studies have demonstrated the involvement of human cytosolic sulfotransferase (SULT) SULT1A1 in the sulfation of acetaminophen, O-desmethylnaproxen (O-DMN), and tapentadol. The current study was designed to investigate the impact of single nucleotide polymorphisms (SNPs) of the human SULT1A1 gene on the sulfation of these analgesic compounds by SULT1A1 allozymes. METHODS: Human SULT1A1 genotypes were identified by database search. cDNAs corresponding to nine SULT1A1 nonsynonymous missense coding SNPs (cSNPs) were generated by site-directed mutagenesis. Recombinant wild-type and SULT1A1 allozymes were bacterially expressed and affinity-purified. Purified SULT1A1 allozymes were analyzed for sulfation activity using an established assay procedure. RESULTS: Compared with the wild-type enzyme, SULT1A1 allozymes were shown to display differential sulfating activities toward three analgesic compounds, acetaminophen, O-desmethylnaproxen (O-DMN), and tapentadol, as well as the prototype substrate 4NP. CONCLUSION: Results obtained indicated clearly the impact of genetic polymorphisms on the drug-sulfation activity of SULT1A1 allozymes. Such information may contribute to a better understanding about the differential metabolism of acetaminophen, O-DMN, and tapentadol in individuals with different SULT1A1 genotypes.


Acetaminophen/metabolism , Arylsulfotransferase/genetics , Naproxen/analogs & derivatives , Tapentadol/metabolism , Analgesics, Non-Narcotic/metabolism , Analgesics, Opioid/metabolism , Cytosol/metabolism , Escherichia coli/cytology , Genotype , Humans , Isoenzymes , Mutagenesis, Site-Directed , Naproxen/metabolism , Polymorphism, Single Nucleotide , Sulfates/metabolism
15.
Arch Biochem Biophys ; 648: 44-52, 2018 06 15.
Article En | MEDLINE | ID: mdl-29705271

Sulfoconjugation has been shown to be critically involved in the metabolism of acetaminophen (APAP), morphine, tapentadol and O-desmethyl tramadol (O-DMT). The objective of this study was to investigate the effects of single nucleotide polymorphisms (SNPs) of human SULT1A3 and SULT1A4 genes on the sulfating activity of SULT1A3 allozymes toward these analgesic compounds. Twelve non-synonymous coding SNPs (cSNPs) of SULT1A3/SULT1A4 were investigated, and the corresponding cDNAs were generated by site-directed mutagenesis. SULT1A3 allozymes, bacterially expressed and purified, exhibited differential sulfating activity toward each of the four analgesic compounds tested as substrates. Kinetic analyses of SULT1A3 allozymes further revealed significant differences in binding affinity and catalytic activity toward the four analgesic compounds. Collectively, the results derived from the current study showed clearly the impact of cSNPs of the coding genes, SULT1A3 and SULT1A4, on the sulfating activity of the coded SULT1A3 allozymes toward the tested analgesic compounds. These findings may have implications in the pharmacokinetics as well as the toxicity profiles of these analgesics administered in individuals with distinct SULT1A3 and/or SULT1A4 genotypes.


Acetaminophen/metabolism , Analgesics, Opioid/metabolism , Arylsulfotransferase/genetics , Arylsulfotransferase/metabolism , Cytosol/enzymology , Polymorphism, Single Nucleotide , Sulfates/metabolism , Sulfotransferases/genetics , Arylsulfotransferase/chemistry , Humans , Kinetics , Models, Molecular , Protein Conformation
16.
Biochem Cell Biol ; 96(5): 655-662, 2018 10.
Article En | MEDLINE | ID: mdl-29671343

The cytosolic sulfotransferase (SULT) SULT2A1 is known to mediate the sulfation of DHEA as well as some other hydroxysteroids such as pregnenolone. The present study was designed to investigate how genetic polymorphisms of the human SULT2A1 gene may affect the sulfation of DHEA and pregnenolone. Online databases were systematically searched to identify human SULT2A1 single nucleotide polymorphisms (SNPs). Of the 98 SULT2A1 non-synonymous coding SNPs identified, seven were selected for further investigation. Site-directed mutagenesis was used to generate cDNAs encoding these seven SULT2A1 allozymes, which were expressed in BL21 Escherichia coli cells and purified by glutathione-Sepharose affinity chromatography. Enzymatic assays revealed that purified SULT2A1 allozymes displayed differential sulfating activity toward both DHEA and pregnenolone. Kinetic analyses showed further differential catalytic efficiency and substrate affinity of the SULT2A1 allozymes, in comparison with wild-type SULT2A1. These findings provided useful information concerning the effects of genetic polymorphisms on the sulfating activity of SULT2A1 allozymes.


Dehydroepiandrosterone/chemistry , Polymorphism, Single Nucleotide , Pregnenolone/chemistry , Sulfotransferases/chemistry , Sulfotransferases/genetics , Humans , Kinetics , Mutagenesis, Site-Directed , Recombinant Proteins , Sulfotransferases/metabolism
17.
J Biochem ; 164(3): 215-221, 2018 Sep 01.
Article En | MEDLINE | ID: mdl-29701841

Sulphated cholesterol, like its unsulphated counterpart, is known to be biologically active and serves a myriad of biochemical/physiological functions. Of the 13 human cytosolic sulphotransferases (SULTs), SULT2B1b has been reported as the main enzyme responsible for the sulphation of cholesterol. As such, SULT2B1b may play the role as a key regulator of cholesterol metabolism. Variations in the sulphating activity of SULT2B1b may affect the sulphation of cholesterol and, consequently, the related physiological events. This study was designed to evaluate the impact of the genetic polymorphisms on the sulphation of cholesterol by SULT2B1b. Ten recombinant SULT2B1b allozymes were generated, expressed, and purified. Purified SULT2B1b allozymes were shown to display differential cholesterol-sulphating activities, compared with the wild-type enzyme. Kinetic studies revealed further their distinct substrate affinity and catalytic efficiency toward cholesterol. These findings showed clearly the impact of genetic polymorphisms on the cholesterol-sulphating activity of SULT2B1b allozymes, which may underscore the differential metabolism of cholesterol in individuals with different SULT2B1b genotypes.


Cholesterol/metabolism , Cytosol/enzymology , Polymorphism, Single Nucleotide , Sulfates/metabolism , Sulfotransferases/metabolism , Catalysis , Genotype , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Kinetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Substrate Specificity , Sulfotransferases/genetics
18.
Biochem Pharmacol ; 151: 104-113, 2018 05.
Article En | MEDLINE | ID: mdl-29524394

Previous studies have demonstrated the involvement of sulfoconjugation in the metabolism of catecholamines and serotonin. The current study aimed to clarify the effects of single nucleotide polymorphisms (SNPs) of human SULT1A3 and SULT1A4 genes on the enzymatic characteristics of the sulfation of dopamine, epinephrine, norepinephrine and serotonin by SULT1A3 allozymes. Following a comprehensive search of different SULT1A3 and SULT1A4 genotypes, twelve non-synonymous (missense) coding SNPs (cSNPs) of SULT1A3/SULT1A4 were identified. cDNAs encoding the corresponding SULT1A3 allozymes, packaged in pGEX-2T vector were generated by site-directed mutagenesis. SULT1A3 allozymes were expressed, and purified. Purified SULT1A3 allozymes exhibited differential sulfating activity toward catecholamines and serotonin. Kinetic analyses demonstrated differences in both substrate affinity and catalytic efficiency of the SULT1A3 allozymes. Collectively, these findings provide useful information relevant to the differential metabolism of dopamine, epinephrine, norepinephrine and serotonin through sulfoconjugation in individuals having different SULT1A3/SULT1A4 genotypes.


Arylsulfotransferase/genetics , Dopamine/metabolism , Epinephrine/metabolism , Norepinephrine/metabolism , Polymorphism, Single Nucleotide , Serotonin/metabolism , Amino Acid Sequence , Humans , Isoenzymes/metabolism , Kinetics , Models, Molecular , Mutagenesis, Site-Directed , Substrate Specificity
...