Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
Sci Rep ; 14(1): 8050, 2024 04 05.
Article in English | MEDLINE | ID: mdl-38580665

ABSTRACT

Pregnenolone is a key intermediate in the biosynthesis of many steroid hormones and neuroprotective steroids. Sulfotransferase family cytosolic 2B member 1 (SULT2B1a) has been reported to be highly selective to sulfate pregnenolone. This study aimed to clarify the effect of missense single nucleotide polymorphisms (SNPs) of the human SULT2B1 gene on the sulfating activity of coded SULT2B1a allozymes toward Pregnenolone. To investigate the effects of single nucleotide polymorphisms of the SULT2B1 gene on the sulfation of pregnenolone by SULT2B1a allozymes, 13 recombinant SULT2B1a allozymes were generated, expressed, and purified using established procedures. Human SULT2B1a SNPs were identified by a comprehensive database search. 13 SULT2B1a nonsynonymous missense coding SNPs (cSNPs) were selected, and site-directed mutagenesis was used to generate the corresponding cDNAs, packaged in pGEX-2TK expression vector, encoding these 13 SULT2B1a allozymes, which were bacterially expressed in BL21 E. coli cells and purified by glutathione-Sepharose affinity chromatography. Purified SULT2B1a allozymes were analyzed for sulfating activities towards pregnenolone. In comparison with the wild-type SULT2B1a, of the 13 allozymes, 11 showed reduced activity toward pregnenolone at 0.1 µM. Specifically, P134L and R259Q allozymes, reported to be involved in autosomal-recessive congenital ichthyosis, displayed low activity (1-10%) toward pregnenolone. The findings of this study may demonstrate the impact of genetic polymorphism on the sulfation of pregnenolone in individuals with different SULT2B1 genotypes.


Subject(s)
Isoenzymes , Pregnenolone , Humans , Isoenzymes/metabolism , Escherichia coli/metabolism , Sulfotransferases/metabolism , Polymorphism, Single Nucleotide
2.
Expert Opin Drug Metab Toxicol ; 17(7): 767-784, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34107842

ABSTRACT

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.


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

ABSTRACT

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.


Subject(s)
Doxorubicin/metabolism , Polymorphism, Single Nucleotide , Sulfotransferases/genetics , Sulfotransferases/metabolism , Cytosol/metabolism , Genotype , Humans , Isoenzymes/metabolism , Kinetics , Mutagenesis, Site-Directed , Nitrophenols/metabolism , Sulfates/metabolism
4.
Eur J Drug Metab Pharmacokinet ; 46(1): 105-118, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33064293

ABSTRACT

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.


Subject(s)
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
5.
Arch Insect Biochem Physiol ; 104(3): e21671, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32227386

ABSTRACT

Sulfoconjugation plays a vital role in the detoxification of xenobiotics and in the metabolism of endogenous compounds. In this study, we aimed to identify new members of the sulfotransferase (SULT) superfamily in the silkworm Bombyx mori. Based on amino acid sequence and phylogenetic analyses, two new enzymes, swSULT ST1 and swSULT ST2, were identified that appear to belong to a distinct group of SULTs including several other insect SULTs. We expressed, purified, and characterized recombinant SULTs. While swSULT ST1 sulfated xanthurenic acid and pentachlorophenol, swSULT ST2 exclusively utilized xanthurenic acid as a substrate. Based on these results, and those concerning the tissue distribution and substrate specificity toward pentachlorophenol analyses, we hypothesize that swSULT ST1 plays a role in the detoxification of xenobiotics, including insecticides, in the silkworm midgut and in the induction of gametogenesis in silkworm ovary and testis. Collectively, the data obtained herein contribute to a better understanding of SULT enzymatic functions in insects.


Subject(s)
Bombyx/enzymology , Inactivation, Metabolic , Sulfotransferases/chemistry , Amino Acid Sequence , Animals , Bombyx/growth & development , Bombyx/metabolism , Female , Gametogenesis , Gastrointestinal Tract/enzymology , Insect Proteins , Larva/enzymology , Male , Ovary , Pentachlorophenol/metabolism , Phylogeny , Sulfotransferases/metabolism , Testis , Xanthurenates/metabolism
6.
J Laryngol Otol ; 133(12): 1038-1040, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31708001

ABSTRACT

BACKGROUND: Human immunodeficiency virus infected patients have a three-fold increased risk of head and neck squamous cell carcinoma. The British HIV Association recommends human immunodeficiency virus testing in all new diagnoses of head and neck squamous cell carcinoma. OBJECTIVES: This observational study aimed to examine the current routine practice of human immunodeficiency virus testing in patients with newly diagnosed head and neck squamous cell carcinoma, and to address the importance of this test in promoting the early diagnosis and treatment of human immunodeficiency virus. METHODS: All head and neck cancer multidisciplinary teams in England were questioned on their protocol for human immunodeficiency virus testing in new diagnoses of head and neck squamous cell carcinoma. RESULTS: Only 1 out of 30 hospitals leading head and neck multidisciplinary teams (3.3 per cent) routinely offered human immunodeficiency virus testing in this high-risk patient group. CONCLUSION: This observational study highlights that head and neck specialists are not aware of, and are consequently not complying with, routine human immunodeficiency virus testing as recommended by the British HIV Association guidelines.


Subject(s)
HIV Infections/diagnosis , Head and Neck Neoplasms/diagnosis , Mass Screening/statistics & numerical data , Otolaryngology/statistics & numerical data , Practice Patterns, Physicians'/statistics & numerical data , Squamous Cell Carcinoma of Head and Neck/diagnosis , Adult , Clinical Protocols , England , Female , HIV , HIV Infections/complications , Head and Neck Neoplasms/virology , Humans , Male , Middle Aged , Squamous Cell Carcinoma of Head and Neck/virology
7.
Mol Cell Endocrinol ; 496: 110535, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31400397

ABSTRACT

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.


Subject(s)
Dehydroepiandrosterone/chemistry , Polymorphism, Single Nucleotide , Pregnenolone/chemistry , Sulfotransferases/chemistry , Humans , Isoenzymes , Sulfotransferases/genetics
8.
Pharmacogenet Genomics ; 29(5): 99-105, 2019 07.
Article in English | MEDLINE | ID: mdl-31145702

ABSTRACT

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.


Subject(s)
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
9.
Pharmacol Rep ; 71(2): 257-265, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30822619

ABSTRACT

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.


Subject(s)
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
10.
Biochem Cell Biol ; 96(5): 655-662, 2018 10.
Article in English | MEDLINE | ID: mdl-29671343

ABSTRACT

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.


Subject(s)
Dehydroepiandrosterone/chemistry , Polymorphism, Single Nucleotide , Pregnenolone/chemistry , Sulfotransferases/chemistry , Sulfotransferases/genetics , Humans , Kinetics , Mutagenesis, Site-Directed , Recombinant Proteins , Sulfotransferases/metabolism
11.
J Biochem ; 164(3): 215-221, 2018 Sep 01.
Article in English | MEDLINE | ID: mdl-29701841

ABSTRACT

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.


Subject(s)
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
12.
Arch Biochem Biophys ; 648: 44-52, 2018 06 15.
Article in English | MEDLINE | ID: mdl-29705271

ABSTRACT

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.


Subject(s)
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
13.
Biochem Pharmacol ; 151: 104-113, 2018 05.
Article in English | MEDLINE | ID: mdl-29524394

ABSTRACT

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.


Subject(s)
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
14.
Pharmacol Rep ; 69(5): 953-958, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28802998

ABSTRACT

BACKGROUND: Previous studies have demonstrated that sulfate conjugation is involved in the metabolism of the active metabolite of tramadol, O-desmethyltramadol (O-DMT). The current study aimed to systematically identify the human cytosolic sulfotransferases (SULTs) that are capable of mediating the sulfation of O-DMT. METHODS: The sulfation of O-DMT under metabolic conditions was demonstrated using HepG2 hepatoma cells and Caco-2 human colon carcinoma cells. O-DMT-sulfating activity of thirteen known human SULTs and four human organ specimens was examined using an established sulfotransferase assay. pH-Dependency and kinetic parameters were also analyzed using, respectively, buffers at different pHs and varying O-DMT concentrations in the assays. RESULTS: Of the thirteen human SULTs tested, only SULT1A3 and SULT1C4 were found to display O-DMT-sulfating activity, with different pH-dependency profiles. Kinetic analysis revealed that SULT1C4 was 60 times more catalytically efficient in mediating the sulfation of O-DMT than SULT1A3 at respective optimal pH. Of the four human organ specimens tested, the cytosol prepared from the small intestine showed much higher O-DMT-sulfating activity than cytosols prepared from liver, lung, and kidney. Both cultured HepG2 and Caco-2 cells were shown to be capable of sulfating O-DMT and releasing sulfated O-DMT into cultured media. CONCLUSION: SULT1A3 and SULT1C4 were the major SULTs responsible for the sulfation of O-DMT. Collectively, the results obtained provided a molecular basis underlying the sulfation of O-DMT and contributed to a better understanding about the pharmacokinetics and pharmacodynamics of tramadol in humans.


Subject(s)
Sulfotransferases/metabolism , Tramadol/analogs & derivatives , Caco-2 Cells , Cytosol/enzymology , Hep G2 Cells , Humans , Hydrogen-Ion Concentration , Intestine, Small/cytology , Kidney/cytology , Liver/cytology , Lung/cytology , Molecular Structure , Tramadol/chemistry , Tramadol/metabolism
15.
Eur J Drug Metab Pharmacokinet ; 42(5): 793-800, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28070880

ABSTRACT

BACKGROUND AND OBJECTIVES: Previous studies reported that tapentadol-sulfate represented one of the major metabolites of tapentadol excreted in urine. The current study aimed to identify the human cytosolic sulfotransferases (SULTs) that is(are) capable of sulfating tapentadol and to examine whether human cells and human organ specimens are capable of sulfating tapentadol. METHODS: Thirteen human SULTs, previously expressed and purified, as well as human organ cytosols, were analyzed for tapentadol-sulfating activity using an established sulfotransferase assay. Cultured HepG2 human hepatoma cells and Caco-2 human colon carcinoma cells were labeled with [35S]sulfate in the presence of different concentrations of tapentadol. RESULTS: Three of the thirteen human SULTs, SULT1A1, SULT1A3, and SULT1C4, were found to display sulfating activity toward tapentadol. Kinetic analysis revealed that SULT1A3 displayed the highest catalytic efficiency in mediating the sulfation of tapentadol, followed by SULT1A1 and SULT1C4. Using cultured HepG2 and Caco-2 cells, the generation and release of sulfated tapentadol under metabolic conditions was demonstrated. Moreover, of the four human organ specimens (kidney, liver, lung, and small intestine) tested, the cytosols prepared from small intestine and liver showed significant tapentadol-sulfating capacity (at 0.0203 and 0.0054 nmol/min/mg, respectively). CONCLUSION: Taken together, the results derived from the current study provided a molecular basis underlying the sulfation of tapentadol in humans.


Subject(s)
Phenols/metabolism , Sulfates/metabolism , Caco-2 Cells , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Colonic Neoplasms/metabolism , Cytosol/metabolism , Hep G2 Cells , Humans , Intestine, Small/metabolism , Kinetics , Liver/metabolism , Liver Neoplasms/metabolism , Sulfotransferases/metabolism , Tapentadol
16.
Ann R Coll Surg Engl ; 98(8): e152-e153, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27388547

ABSTRACT

The left recurrent laryngeal nerve is at increased risk of compression by oesophageal pathology due to its long course through the neck and thorax. Here we report a case of left vocal cord palsy secondary to displacement of a gastric band, resulting in oesophageal dilatation and neuropraxia of the left recurrent laryngeal nerve. Vocal cord function partially improved following removal of the gastric band.


Subject(s)
Gastroplasty/adverse effects , Prosthesis Failure/adverse effects , Vocal Cord Paralysis/etiology , Female , Humans , Middle Aged , Obesity, Morbid/surgery , Tomography, X-Ray Computed
17.
J Thromb Haemost ; 13(9): 1545-56, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26074048

ABSTRACT

Thrombotic occlusion of the coronary artery, which triggers acute myocardial infarction, is one of the major causes of death in the USA. Currently, arterial occlusions are treated with intravenous plasminogen activators (PAs), which dissolve the clot by activating plasminogen. However, PAs indiscriminately generate plasmin, which depletes critical clotting factors (fibrinogen, factor V, and factor VIII), precipitates a lytic state in the blood, and produces bleeding complications in a large patient population. PAs have been extensively investigated to achieve thrombus specificity, to attenuate the bleeding risk, and to widen their clinical applications. In this review, we discuss various strategies that have been pursued since the beginning of thrombolytic therapy. We review the biotechnological approaches that have been used to develop mutant and chimeric PAs for thrombus selectivity, including the use of specific antibodies for targeting thrombi. We discuss particulate carrier-based systems and triggered-release concepts. We propose new hypotheses and strategies to spur future studies in this research arena. Overall, we describe the approaches and accomplishments in the development of patient-friendly and workable delivery systems for thrombolytic drugs.


Subject(s)
Fibrinolytic Agents/therapeutic use , Plasminogen Activators/genetics , Protein Engineering/methods , Thrombolytic Therapy , Thrombosis/drug therapy , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Blood Platelets/drug effects , Blood Platelets/immunology , Fibrin/metabolism , Fibrinolytic Agents/metabolism , Heparin/chemistry , Heparin/therapeutic use , Humans , Mice , Plasminogen Activators/metabolism , Plasminogen Activators/therapeutic use , Protamines/pharmacology , Protamines/therapeutic use , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/therapeutic use
18.
Int J Oral Maxillofac Surg ; 44(1): 34-6, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25457834

ABSTRACT

Mönckeberg's arteriosclerosis is often an incidental finding, identified either clinically or on plain radiography. It can occasionally be associated with diabetes mellitus or chronic kidney disease. It differs from the more common atherosclerosis in that the tunica intima remains largely unaffected and the diameter of the vessel lumen is preserved. Despite such vessels appearing hard and pulseless throughout their affected length, they deliver relatively normal distal perfusion, indeed there is often a bounding pulse at the end of the calcified zone. They appear unremarkable on magnetic resonance angiography but visibly calcified on plain radiography. Mönckeberg's arteriosclerosis has a prevalence of < 1% of the population, but when it does occur it can cause consternation at the prospect of using these vessels for microvascular anastamosis. We report our experience of deliberately using these vessels in an osseocutaneous radial forearm free flap reconstruction. Although there are some technical considerations to bear in mind, we would suggest that unlike vessels affected by atherosclerosis, anastomosis of arteries affected by Mönckeberg's arteriosclerosis has little or no impact on free flap survival.


Subject(s)
Carcinoma, Squamous Cell/surgery , Mandibular Neoplasms/surgery , Monckeberg Medial Calcific Sclerosis/complications , Monckeberg Medial Calcific Sclerosis/diagnosis , Radial Artery/transplantation , Surgical Flaps/blood supply , Arm/blood supply , Carcinoma, Squamous Cell/pathology , Humans , Leg/blood supply , Magnetic Resonance Angiography , Male , Mandibular Neoplasms/pathology , Middle Aged
19.
Pancreas ; 41(3): 353-7, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22415665

ABSTRACT

The initial injury in acute pancreatitis is characteristically sterile and results in acinar cells necrosis. Intracellular contents released from damaged cells into the extracellular space serve as damage-associated molecular patterns (DAMPs) that trigger inflammation. There is increasing evidence that this sterile inflammatory response mediated through DAMPs released from necrotic acinar cells is a key determinant of further pancreatic injury, remote organ injury, and disease resolution in experimental models. A number of DAMPS, including high-mobility group box protein 1, DNA, adenosine triphosphate and heat shock protein 70, have been shown to have a role in experimental pancreatitis. Many of these DAMPs are also detectable in the human pancreatitis. Genetic deletion and pharmacologic antagonism demonstrate that specific DAMP receptors, including Toll-like receptor (TLR) 4, TLR9, and P2X7, are also required for inflammation in experimental acute pancreatitis. Downstream DAMP-sensing components include nod-like receptor protein 3, caspase 1, interleukin-1ß (IL-1), IL-18, and IL-1 receptor, and also are required for full experimental pancreatitis. These DAMP-mediated pathways provide novel therapeutic targets using antagonists of TLRs and other receptors.


Subject(s)
Inflammation Mediators/metabolism , Inflammation/etiology , Pancreas/immunology , Pancreatitis/complications , Signal Transduction , Acute Disease , Animals , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Inflammation/therapy , Pancreas/pathology , Pancreatitis/genetics , Pancreatitis/immunology , Pancreatitis/pathology , Pancreatitis/therapy , Signal Transduction/genetics
20.
Am J Physiol Gastrointest Liver Physiol ; 302(10): G1171-9, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22383490

ABSTRACT

Inflammation contributes to liver injury in acetaminophen (APAP) hepatotoxicity in mice and is triggered by stimulation of immune cells. The purinergic receptor P2X7 is upstream of the nod-like receptor family, pryin domain containing-3 (NLRP3) inflammasome in immune cells and is activated by ATP and NAD that serve as damage-associated molecular patterns. APAP hepatotoxicity was assessed in mice genetically deficient in P2X7, the key inflammatory receptor for nucleotides (P2X7-/-), and in wild-type mice. P2X7-/- mice had significantly decreased APAP-induced liver necrosis. In addition, APAP-poisoned mice were treated with the specific P2X7 antagonist A438079 or etheno-NAD, a competitive antagonist of NAD. Pre- or posttreatment with A438079 significantly decreased APAP-induced necrosis and hemorrhage in APAP liver injury in wild-type but not P2X7-/- mice. Pretreatment with etheno-NAD also significantly decreased APAP-induced necrosis and hemorrhage in APAP liver injury. In addition, APAP toxicity in mice lacking the plasma membrane ecto-NTPDase CD39 (CD39-/-) that metabolizes ATP was examined in parallel with the use of soluble apyrase to deplete extracellular ATP in wild-type mice. CD39-/- mice had increased APAP-induced hemorrhage and mortality, whereas apyrase also decreased APAP-induced mortality. Kupffer cells were treated with extracellular ATP to assess P2X7-dependent inflammasome activation. P2X7 was required for ATP-stimulated IL-1ß release. In conclusion, P2X7 and exposure to the ligands ATP and NAD are required for manifestations of APAP-induced hepatotoxicity.


Subject(s)
Acetaminophen/adverse effects , Antipyretics/adverse effects , Chemical and Drug Induced Liver Injury/metabolism , Receptors, Purinergic P2X7/physiology , Acetaminophen/antagonists & inhibitors , Animals , Antigens, CD/metabolism , Apyrase/metabolism , Cells, Cultured , Chemical and Drug Induced Liver Injury/pathology , Hemorrhage/chemically induced , Hemorrhage/drug therapy , Hemorrhage/pathology , Inflammasomes/metabolism , Interleukin-1beta/metabolism , Kupffer Cells/drug effects , Kupffer Cells/pathology , Male , Mice , Mice, Inbred C57BL , NAD/analogs & derivatives , NAD/metabolism , Necrosis/metabolism , Pyridines/pharmacology , Receptors, Purinergic P2X7/genetics , Signal Transduction/drug effects , Tetrazoles/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...