Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Glyn B. Steventon is active.

Publication


Featured researches published by Glyn B. Steventon.


Neurology | 1989

Xenobiotic metabolism in Parkinson's disease

Glyn B. Steventon; M.T.E. Heafield; Rosemary H. Waring; A. C. Williams

We studied 68 patients with Parkinsons disease (PD) with probe drugs to determine whether a defect in metabolism might be an etiologic factor and found no difference between patients and controls in their ability to form the 4 hydroxy metabolite of debrisoquin. However, using S-carboxymethyl-L-cysteine, 63.2% (43/68) of PD patients had reduced S-oxidation capacity, while 35.3% (24/68) produced no sulfoxides (controls, 35.2% and 2.5%). When we studied acetaminophen (paracetamol) metabolism, only 29.6% of PD patients excreted >,5% of the dose as the sulfate conjugate; the corresponding figure for controls was 83.9%. These results suggest a deficiency in detoxication pathways involving sulfur metabolism. PD patients may be unusually susceptible to exogenous or even endogenous toxins.


Neurology | 1990

Xenobiotic metabolism in Alzheimer's disease

Glyn B. Steventon; M.T.E. Heafield; S. Sturman; Rosemary H. Waring; A. C. Williams

Using 5 methods, we assessed the ability of patients with a clinical diagnosis of Alzheimers disease (AD) to handle xenobiotics. Patients with AD, compared with controls, have reduced sulfoxidation of the probe drug S-carboxymethyl-l-cysteine; they also form less of the sulfate conjugate of acetaminophen. In addition, they have lower activity of the enzyme thiolmethyltransferase. In contrast, the capacity to oxidize debrisoquin and to acetylate sulfamethazine was normal. These findings suggest that a major risk factor for the development of AD is a skewed capacity for xenobiotic metabolism especially of compounds containing sulfur.


Journal of Pharmacy and Pharmacology | 2001

Characterization and purification of the vitamin K1 2,3 epoxide reductase system from rat liver

Louise A. Begent; Anthony P. Hill; Glyn B. Steventon; Andrew J. Hutt; Christopher J. Pallister; David C. Cowell

The enzyme vitamin K1 2,3 epoxide reductase is responsible for converting vitamin K1 2,3 epoxide to vitamin K1 quinone thus completing the vitamin K cycle. The enzyme is also the target of inhibition by the oral anticoagulant, R,S‐warfarin. Purification of this protein would enable the interaction of the inhibitor with its target to be elucidated. To date a single protein possessing vitamin K1 2,3 epoxide reductase activity and binding R,S‐warfarin has yet to be purified to homogeneity, but recent studies have indicated that the enzyme is in fact at least two interacting proteins. We report on the attempted purification of the vitamin K1 2,3 epoxide reductase complex from rat liver microsomes by ion exchange and size exclusion chromatography techniques. The intact system consisted of a warfarin‐binding factor, which possessed no vitamin K1 2,3 epoxide reductase activity and a catalytic protein. This catalytic protein was purified 327‐fold and was insensitive to R,S‐warfarin inhibition at concentrations up to 5 mm. The addition of the S‐200 size exclusion chromatography fraction containing the inhibitor‐binding factor resulted in the return of R,S‐warfarin inhibition. Thus, to function normally, the rat liver endoplasmic reticulum vitamin K1 2,3 epoxide reductase system requires the association of two components, one with catalytic activity for the conversion of the epoxide to the quinone and the second, the inhibitor binding factor. This latter enzyme forms the thiol‐disulphide redox centre that in the oxidized form binds R,S‐warfarin.


Cephalalgia | 1997

Platelet sulphotransferase activity, plasma sulphate levels and sulphation capacity in patients with migraine and tension headache

Z Alam; N Coombes; Rosemary H. Waring; Ac Williams; Glyn B. Steventon

Activity of both the M- and P-forms of sulphotransferase (ST) was measured in platelets from patients with migraine, tension headache and controls. Mean PST values were 0.065±0.023 and 0.057±0.052 mnol/mg protein/min for migraine patients with and without aura. The corresponding values for tension headache and controls were 0.122±0.059 and 0.127:0.093 nmol/mg protein/min respectively (p<0.05). Mean MST values were not different for any of the groups, and MST and PST activities measured in two patients during a migraine attack were not significantly altered from baseline levels. Mean plasma inorganic sulphate concentrations and paracetamol metabolites were not significantly different in any of the groups studied. The results suggest that PST activity may be a factor in the aetiology of migraine.


Drug metabolism and drug interactions | 1996

Human Metabolism of Paracetamol (Acetaminophen) at Different Dose Levels

Glyn B. Steventon; Stephen C. Mitchell; Rosemary H. Waring

Urine (0-24 h) was collected from five subjects on separate occasions following the ingestion of paracetamol at five different dose levels (500, 750, 1000, 1250, 1500 mg) which spanned the normal therapeutic range. The major urinary metabolites were sulphuric and glucuronic acid conjugates which together accounted for around 50% of the administered dose. Unchanged paracetamol excretion was low (5-20%). This situation was similar over the entire dose range. These findings are discussed in relation to previous single dose studies reported in the literature.


Drug metabolism and drug interactions | 2004

Phenylalanine 4-Monooxygenase and the S-Oxidation of S-Carboxymethyl-L-cysteine

Abir H. Goreish; Sandra Bednar; Hefin Jones; Stephen C. Mitchell; Glyn B. Steventon

The identity of the enzyme responsible for the S-oxidation of the mucolytic S-substituted L-cysteine drug, S-carboxymethyl-L-cysteine (SCMC), has been actively investigated for the last 10 years. A genetic polymorphism exists in the oxidation of the thioether moiety that has been identified as a disease susceptibility factor in a number of degenerative diseases. This polymorphism has also been implicated in the wide variation in clinical response to SCMC therapy in man. To date little is known about the molecular enzymology of this reaction but a previous investigation revealed that rat activated phenylalanine 4-monooxygenase (PAH) could S-oxidise both Met- and S-methyl-L-cysteine (SMC) to their S-oxide metabolites. We have investigated the hypothesis that SCMC was also a substrate for activated PAH in the cytosolic faction of the Wistar rat. 1. Substrate and inhibitor investigation revealed that SCMC was a substrate for activated PAH activity in vitro. 2. The large aromatic amino acid hydroxylase monoclonal antibody and the Fe3+ chelator, deferoxamine, completely inhibited both Phe and SCMC oxidation to their respective metabolites. 3. Analysis of the Dixon plots revealed that both Phe and SCMC competitively inhibited each others oxidation. 4. Correlation studies showed that the rate of production of Tyr was positively correlated to the production of both SCMC and SMC S-oxides in 20 female Wistar rat hepatic cytosolic fractions. These results strongly support the hypothesis that PAH is the enzyme responsible for SCMC S-oxidation in the rat.


Journal of Pharmacy and Pharmacology | 2006

In‐vitro effect of flavonoids from Solidago canadensis extract on glutathione S‐transferase

P. Apáti; Peter J. Houghton; Geoffrey C. Kite; Glyn B. Steventon; Ágnes Kéry

Solidago canadensis is typical of a flavonoid‐rich herb and the effect of an aqueous ethanol extract on glutathione‐S‐transferase (GST) activity using HepG2 cells was compared with those of the flavonol quercetin and its glycosides quercitrin and rutin, found as major constituents. The composition of the extract was determined by HPLC and rutin was found to be the major flavonoidal component of the extract. Total GST activity was assessed using 1‐chloro‐2,4‐dinitrobenzene as a substrate. The glycosides rutin and quercitrin gave dose‐dependent increases in GST activity, with a 50% and 24.5% increase at 250 mm, respectively, while the aglycone quercetin inhibited the enzyme by 30% at 250 mm. The total extract of the herb gave an overall dose‐dependent increase, the fractions corresponding to the flavonoids showed activating effects while those containing caffeic acid derivatives were inhibitory. The activity observed corresponds to that reported for similar compounds in‐vivo using rats, thus the HepG2 cell line could serve as a more satisfactory method of assessing the effects of extracts and compounds on GST.


Drug metabolism and drug interactions | 2005

Phenylalanine 4-monooxygenase and the S-oxidation of S-carboxymethyl-L-cysteine in HepG2 cells.

Panayotis Panagopoulos; Hevfin Jones; Stephen C. Mitchell; Ben Forbes; Glyn B. Steventon

The role of phenylalanine 4-monooxygenase (PAH) in the S-oxidation of S-carboxymethyl-L-cysteine (SCMC) in the rat has now been well established in rat cytosolic fractions in vitro. However, the role of PAH in the S-oxidation of SCMC in human cytosolic fractions or hepatocytes has yet to be investigated. The aim of this investigation was to analyse the kinetic parameters of PAH oxidation of both L-phenylalanine (Phe) and SCMC in the human HepG2 cell line in order to investigate the use of these cells as a model for the cellular regulation of SCMC S-oxidation. The experimentally determined Km and V(max) were 7.14 +/- 0.32 mM and 0.85 +/- 0.32 nmole Tyr formed min(-1) x mg protein(-1) using Phe as substrate. For SCMC the values were 25.24 +/- 5.91 mM and 0.79 +/- 0.09 nmole SCMC (RIS) S-oxides formed min(-1) x mg protein(-1). The experimentally determined Km and V(max) for the cofactor BH4 were 6.81 +/- 0.21 microM and 0.41 +/- 0.004 nmole Tyr formed min(-1) x mg protein(-1) for Phe and 7.24 +/- 0.19 microM and 0.42 +/- 0.002 nmole SCMC (R/S) S-oxides formed min(-1) x mg protein(-1) for SCMC. The use of various PAH inhibitors confirmed that HepG2 cells contained PAH and that the enzyme was capable of converting SCMC to its (R) and (S) S-oxide metabolites in an in vitro PAH assay. Thus HepG2 cells have become a useful additional tool for the investigation of the cellular regulation of PAH in the S-oxidation of SCMC.


Chromatographia | 1998

A methodological and metabolite identification study of the metabolism of S-carboxymethyl-L-cysteine in man

Glyn B. Steventon

SummaryThe analysis of 08:00–16:00 (0–8) hour urine collections following oral S-carboxymethyl-L-cysteine administration of 750 mg to 30 individuals identified S-carboxymethyl-L-cysteine, S-carboxymethyl-L-cysteine S-oxide, S-methyl-L-cysteine and S-methyl-L-cysteine S-oxide as the major urinary drug related compounds. No S-(carboxymethylthio)-L-cysteine mixed disulphide metabolite was found in the 08:00–16:00 hour urine collection but the metabolite was detected in the 16:00–00:00 (8–16) hour urune collection by paper chromatography, TLC and HPLC. The production of the S-oxide metabolites 08:00–16:00 hour urine collection) and the mixed disulphide metabolite (16:00–00:00 hour urine collection) were both shown to be biomodally distributed. A significant linear correlation of the S-oxides recovered following 08:00–16:00 hour urine collection as analysed by paper chromatography, TLC and HPLC is reported.


Molecular Genetics and Metabolism | 2009

The activity of wild type and mutant phenylalanine hydroxylase with respect to the C-oxidation of phenylalanine and the S-oxidation of S-carboxymethyl-L-cysteine.

Glyn B. Steventon; Stephen C. Mitchell; Belén Pérez; Lourdes R. Desviat; Magdalena Ugarte

The involvement of the enzyme, phenylalanine hydroxylase (PAH), in the S-oxidation of S-carboxymethyl-L-cysteine (SCMC) is now firmly established in man and rat. However, the underlying role of the molecular genetics of PAH in dictating and influencing the S-oxidation polymorphism of SCMC metabolism is as yet unknown. In this work we report that the S-oxidation of SCMC was dramatically reduced in the tetrahydrobiopterin (BH(4)) responsive mutant PAH proteins (I65T, R68S, R261Q, V388M and Y414C) with these enzymes possessing between 1.2% and 2.0% of the wild type PAH activity when SCMC was used as substrate. These same mutant proteins express between 23% and 76% of the wild type PAH activity when phenylalanine was used as the substrate. The PAH mutant proteins (R158Q, I174T and R408W) that result in the classical phenylketonuria (PKU) phenotype expressing 0.2-1.8% of the wild type PAH activity when using phenylalanine as substrate were found to have <0.1% of the wild type PAH activity when SCMC was used as the substrate. Mutations that result in PAH proteins retaining some residual PAH activity with phenylalanine as substrate have <2.0% residual activity when SCMC was used as a substrate. This investigation has led to the hypothesis that the S-oxidation polymorphism in man is a consequence of an individual carrying one mutant PAH allele which has resulted in the loss of the ability of the residual PAH protein to undertake the S-oxidation of SCMC in vivo.

Collaboration


Dive into the Glyn B. Steventon's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

S. Sturman

University of Birmingham

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge