Network


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

Hotspot


Dive into the research topics where Anthony W. Ford-Hutchinson is active.

Publication


Featured researches published by Anthony W. Ford-Hutchinson.


Nature | 1999

Characterization of the human cysteinyl leukotriene CysLT1 receptor

Kevin R. Lynch; Gary P. O'Neill; Qingyun Liu; D.-S. Im; N. Sawyer; K. M. Metters; Nathalie Coulombe; Mark Abramovitz; David J. Figueroa; Zhizhen Zeng; Brett Connolly; Chang Bai; Christopher P. Austin; A. Chateauneuf; R. Stocco; G. M. Greig; S. Kargman; S. B. Hooks; E. Hosfield; David L. Williams; Anthony W. Ford-Hutchinson; C. T. Caskey; Jilly F. Evans

The cysteinyl leukotrienes—leukotriene C4(LTC4), leukotriene D4(LTD4) and leukotriene E4(LTE 4)—are important mediators of human bronchial asthma,. Pharmacological studies have determined that cysteinyl leukotrienes activate at least two receptors, designated CysLT1 and CysLT2 (refs 4,5,6). The CysLT1-selective antagonists, such as montelukast (Singulair), zafirlukast (Accolate) and pranlukast (Onon), are important in the treatment of asthma. Previous biochemical characterization of CysLT1 antagonists and the CysLT1 receptor has been in membrane preparations from tissues enriched for this receptor. Here we report the molecular and pharmacological characterization of the cloned human CysLT1 receptor. We describe the functional activation (calcium mobilization) of this receptor by LTD4 and LTC4, and competition for radiolabelled LTD4 binding to this receptor by the cysteinyl leukotrienes and three structurally distinct classes of CysLT1-receptor antagonists. We detected CysLT1-receptor messenger RNA in spleen, peripheral blood leukocytes and lung. In normal human lung, expression of the CysLT 1-receptor mRNA was confined to smooth muscle cells and tissue macrophages. Finally, we mapped the human CysLT1-receptor gene to the X chromosome.


FEBS Letters | 1993

Expression of mRNA for cyclooxygenase‐1 and cyclooxygenase‐2 in human tissues

Gary P. O'Neill; Anthony W. Ford-Hutchinson

The rate‐limiting step in the formation of prostanoids is the conversion of arachidonic acid to prostaglandin H2 by cyclooxygenase, also known as prostaglandin G/H synthase/cyclooxygenase. Two forms of cyclooxygenase have been characterized: a ubiquitously expressed form (COX‐1) and a recently described second form (COX‐2) inducible by various factors including mitogens, hormones, serum and cytokines. Here we quantitate by the reverse transcriptase‐polymerase chain reaction (RT‐PCR) the expression of COX‐1 and COX‐2 mRNA in human tissues including lung, uterus, testis, brain, pancreas, kidney, liver, thymus, prostate, mammary gland, stomach and small intestine. All tissues examined contained both COX‐1 and COX‐2 mRNA and could be grouped according to the level of COX mRNA expression. The highest levels of COX mRNAs were detected in the prostate where approximately equal levels of COX‐1 and COX‐2 transcripts were present. In the lung high levels of COX‐2 were observed whereas COX‐1 mRNA levels were about 2‐fold lower. An intermediate level of expression of both COX‐1 and COX‐2 mRNA was observed in the mammary gland, stomach, small intestine, and uterus. The lowest levels of COX‐1 and COX‐2 mRNA were observed in the testis, pancreas, kidney, liver, thymus, and brain.


Bioorganic & Medicinal Chemistry Letters | 1999

The discovery of rofecoxib, [MK 966, VIOXX®, 4-(4′-methylsulfonylphenyl)-3-phenyl-2(5H)-furanone], an orally active cyclooxygenase-2 inhibitor

Petpiboon Prasit; Zhaoyin Wang; Christine Brideau; Chi-Chung Chan; S. Charleson; Wanda Cromlish; Diane Ethier; Jilly F. Evans; Anthony W. Ford-Hutchinson; Jacques-Yves Gauthier; Robert Gordon; Jocelyne Guay; M Gresser; Stacia Kargman; Brian P. Kennedy; Yves Leblanc; Serge Leger; Joseph A. Mancini; Gary P. O'Neill; Marc Ouellet; M.D Percival; Helene Perrier; Denis Riendeau; Ian W. Rodger; Philip Tagari; Michel Therien; Philip J. Vickers; E.H.F. Wong; Lijing Xu; Robert N. Young

The development of a COX-2 inhibitor rofecoxib (MK 966, Vioxx) is described. It is essentially equipotent to indomethacin both in vitro and in vivo but without the ulcerogenic side effect due to COX-1 inhibition.


British Journal of Pharmacology | 1997

Biochemical and pharmacological profile of a tetrasubstituted furanone as a highly selective COX-2 inhibitor

Denis Riendeau; M.D Percival; Susan Boyce; Christine Brideau; S. Charleson; Wanda Cromlish; Diane Ethier; Jilly F. Evans; Jean-Pierre Falgueyret; Anthony W. Ford-Hutchinson; Robert Gordon; Gillian Greig; M Gresser; Jocelyne Guay; Stacia Kargman; Serge Leger; Joseph A. Mancini; Gary P. O'Neill; Marc Ouellet; Ian W. Rodger; Michel Therien; Zhaoyin Wang; J.K. Webb; E.H.F. Wong; Lijing Xu; Robert N. Young; Robert Zamboni; Petpiboon Prasit; Chi-Chung Chan

DFU (5,5‐dimethyl‐3‐(3‐fluorophenyl)‐4‐(4‐methylsulphonyl)phenyl‐2(5H)‐furanone) was identified as a novel orally active and highly selective cyclo‐oxygenase‐2 (COX‐2) inhibitor. In CHO cells stably transfected with human COX isozymes, DFU inhibited the arachidonic acid‐dependent production of prostaglandin E2 (PGE2) with at least a 1,000 fold selectivity for COX‐2 (IC50=41±14 nM) over COX‐1 (IC50>50 μM). Indomethacin was a potent inhibitor of both COX‐1 (IC50=18±3 nM) and COX‐2 (IC50=26±6 nM) under the same assay conditions. The large increase in selectivity of DFU over indomethacin was also observed in COX‐1 mediated production of thromboxane B2 (TXB2) by Ca2+ ionophore‐challenged human platelets (IC50>50 μM and 4.1±1.7 nM, respectively). DFU caused a time‐dependent inhibition of purified recombinant human COX‐2 with a Ki value of 140±68 μM for the initial reversible binding to enzyme and a k2 value of 0.11±0.06 s−1 for the first order rate constant for formation of a tightly bound enzyme‐inhibitor complex. Comparable values of 62±26 μM and 0.06±0.01 s−1, respectively, were obtained for indomethacin. The enzyme‐inhibitor complex was found to have a 1 : 1 stoichiometry and to dissociate only very slowly (t1/2=1–3 h) with recovery of intact inhibitor and active enzyme. The time‐dependent inhibition by DFU was decreased by co‐incubation with arachidonic acid under non‐turnover conditions, consistent with reversible competitive inhibition at the COX active site. Inhibition of purified recombinant human COX‐1 by DFU was very weak and observed only at low concentrations of substrate (IC50=63±5 μM at 0.1 μM arachidonic acid). In contrast to COX‐2, inhibition was time‐independent and rapidly reversible. These data are consistent with a reversible competitive inhibition of COX‐1. DFU inhibited lipopolysaccharide (LPS)‐induced PGE2 production (COX‐2) in a human whole blood assay with a potency (IC50=0.28±0.04 μM) similar to indomethacin (IC50=0.68±0.17 μM). In contrast, DFU was at least 500 times less potent (IC50>97 μM) than indomethacin at inhibiting coagulation‐induced TXB2 production (COX‐1) (IC50=0.19±0.02 μM). In a sensitive assay with U937 cell microsomes at a low arachidonic acid concentration (0.1 μM), DFU inhibited COX‐1 with an IC50 value of 13±2 μM as compared to 20±1 nM for indomethacin. CGP 28238, etodolac and SC‐58125 were about 10 times more potent inhibitors of COX‐1 than DFU. The order of potency of various inhibitors was diclofenac>indomethacin∼naproxen>nimesulide∼ meloxicam∼piroxicam>NS‐398∼SC‐57666>SC‐58125>CGP 28238∼etodolac>L‐745,337>DFU. DFU inhibited dose‐dependently both the carrageenan‐induced rat paw oedema (ED50 of 1.1 mg kg−1 vs 2.0 mg kg−1 for indomethacin) and hyperalgesia (ED50 of 0.95 mg kg−1 vs 1.5 mg kg−1 for indomethacin). The compound was also effective at reversing LPS‐induced pyrexia in rats (ED50=0.76 mg kg−1 vs 1.1 mg kg−1 for indomethacin). In a sensitive model in which 51Cr faecal excretion was used to assess the integrity of the gastrointestinal tract in rats, no significant effect was detected after oral administration of DFU (100 mg kg−1, b.i.d.) for 5 days, whereas chromium leakage was observed with lower doses of diclofenac (3 mg kg−1), meloxicam (3 mg kg−1) or etodolac (10–30 mg kg−1). A 5 day administration of DFU in squirrel monkeys (100 mg kg−1) did not affect chromium leakage in contrast to diclofenac (1 mg kg−1) or naproxen (5 mg kg−1). The results indicate that COX‐1 inhibitory effects can be detected for all selective COX‐2 inhibitors tested by use of a sensitive assay at low substrate concentration. The novel inhibitor DFU shows the lowest inhibitory potency against COX‐1, a consistent high selectivity of inhibition of COX‐2 over COX‐1 (>300 fold) with enzyme, whole cell and whole blood assays, with no detectable loss of integrity of the gastrointestinal tract at doses >200 fold higher than efficacious doses in models of inflammation, pyresis and hyperalgesia. These results provide further evidence that prostanoids derived from COX‐1 activity are not important in acute inflammatory responses and that a high therapeutic index of anti‐inflammatory effect to gastropathy can be achieved with a selective COX‐2 inhibitor.


Journal of Biological Chemistry | 1997

Identification and Characterization of a Novel Microsomal Enzyme with Glutathione-dependent Transferase and Peroxidase Activities

Per-Johan Jakobsson; Joseph A. Mancini; Denis Riendeau; Anthony W. Ford-Hutchinson

5-Lipoxygenase activating protein (FLAP), leukotriene-C4 (LTC4) synthase, and microsomal glutathione S-transferase II (microsomal GST-II) are all members of a common gene family that may also include microsomal GST-I. The present work describes the identification and characterization of a novel member of this family termed microsomal glutathione S-transferase III (microsomal GST-III). The open reading frame encodes a 16.5-kDa protein with a calculated pI of 10.2. Microsomal GST-III has 36, 27, 22, and 20% amino acid identity to microsomal GST-II, LTC4 synthase, microsomal GST-I, and FLAP, respectively. Microsomal GST-III also has a similar hydrophobicity pattern to FLAP, LTC4 synthase, and microsomal GST-I. Fluorescent in situ hybridization mapped microsomal GST-III to chromosomal localization 1q23. Like microsomal GST-II, microsomal GST-III has a wide tissue distribution (at the mRNA level) and is predominantly expressed in human heart, skeletal muscle, and adrenal cortex, and it is also found in brain, placenta, liver, and kidney tissues. Expression of microsomal GST-III mRNA was also detected in several glandular tissues such as pancreas, thyroid, testis, and ovary. In contrast, microsomal GST-III mRNA expression was very low (if any) in lung, thymus, and peripheral blood leukocytes. Microsomal GST-III protein was expressed in a baculovirus insect cell system, and microsomes from Sf9 cells containing either microsomal GST-II or microsomal GST-III were both found to possess glutathione-dependent peroxidase activity as shown by their ability to reduce 5-HPETE to 5-HETE in the presence of reduced glutathione. The apparent K m of 5-HPETE was determined to be approximately 7 μm for microsomal GST-II and 21 μm for microsomal GST-III. Microsomal GST-III was also found to catalyze the production of LTC4 from LTA4 and reduced glutathione. Based on these catalytic activities it is proposed that this novel membrane protein is a member of the microsomal glutathione S-transferase super family, which also includes microsomal GST-I, LTC4 synthase, FLAP, and microsomal GST-II.


Bioorganic & Medicinal Chemistry Letters | 1995

Discovery of MK-0476, a potent and orally active leukotriene D4 receptor antagonist devoid of peroxisomal enxyme induction

Marc Labelle; Michel Belley; Yves Gareau; Jacques-Yves Gauthier; Daniel Guay; Robert Gordon; S.G. Grossman; Thomas R. Jones; Yves Leblanc; M. McAuliffe; C. McFarlane; Paul Masson; Kathleen M. Metters; Nathalie Ouimet; D.H. Patrick; H. Piechuta; C. Rochette; Nicole Sawyer; Y.B. Xiang; C.B. Pickett; Anthony W. Ford-Hutchinson; Robert Zamboni; Robert N. Young

Abstract Structure-activity studies leading to the discovery of 1 (MK-0476) are described. The initial compound of this series, 2, was a potent leukotriene D4 (LTD4) antagonist, but was also a peroxisomal enzyme inducer in the mouse. Structure-activity relationships around the thioether chain were explored to remove this undesirable feature. It was found that alkyl substituents in the s position relative to the carboxylic acid reduce the potency as a peroxisomal enzyme inducer while preserving the LTD4 antagonistic properties. Dialkyl substitution essentially eliminates the enzyme induction. The optimal styryl quinoline 1 exhibited high in vitro potency and in vivo activity on oral dosing without significant liver enzyme induction in the mouse.


Prostaglandins | 1982

Differing mechanisms for leukotriene D4-induced bronchoconstriction in guinea pigs following intravenous and aerosol administration

R. Hamel; Paul Masson; Anthony W. Ford-Hutchinson; Thomas R. Jones; G. Brunet; H. Piechuta

Leukotriene D4 (LTD4) when administered intravenously or by aerosol to guinea pigs produced changes in pulmonary mechanics including a decrease in dynamic compliance and an increase in pulmonary resistance. The effects of intravenous LTD4 (0.5 microgram kg-1) were short lived and abolished by pretreatment of the animal with either cyclooxygenase inhibitors, a thromboxane synthetase inhibitor (OKY 1555) or an SRS-A antagonist (FPL 55712). These findings suggest that bronchoconstriction produced by the intravenous infusion of LTD4 at 0.5 microgram kg-1 is due to the release of thromboxane A2. However, in animals treated with indomethacin, LTD4 at higher doses (greater than 0.8 microgram kg-1) still elicited a bronchoconstriction which could be blocked by FPL 55712. Nebulization of 0.1 - 1.0 microgram of LTD4 into the lung produced prolonged changes in pulmonary mechanics which were inhibited by FPL 55712 and were potentiated by indomethacin. LTD4, therefore, when administered by aerosol produced effects on the lung which were not mediated by cyclooxygenase products. Responses to nebulized rather than intravenous LTD4 in the guinea pig may more closely resemble those seen in human tissues.


British Journal of Pharmacology | 1998

Characterization of the recombinant human prostanoid DP receptor and identification of L-644,698, a novel selective DP agonist.

D. Hamish Wright; Kathleen M. Metters; Mark Abramovitz; Anthony W. Ford-Hutchinson

A human embryonic kidney cell line [HEK 293(EBNA)] stably expressing the human recombinant prostaglandin D2 (PGD2) receptor (hDP) has been characterized with respect to radioligand binding and signal transduction properties by use of prostanoids and prostanoid analogues. Radioligand binding studies included saturation analyses, the effects of nucleotide analogues, the initial rate of ligand‐receptor association and equilibrium competition assays. In addition, adenosine 3′:5′‐cyclic monophosphate (cyclic AMP) generation in response to ligand challenge was also measured, as this is the predominant hDP signalling pathway. L‐644,698 ((4‐(3‐(3‐(3‐hydroxyoctyl)‐4‐oxo‐2‐thiazolidinyl) propyl) benzoic acid) (racemate)) was identified as a novel ligand having high affinity for hDP with an inhibitor constant (Ki) of 0.9 nm. This Ki value was comparable to the Ki values obtained in this study for ligands that have previously shown high affinity for DP: PGD2 (0.6 nm), ZK 110841 (0.3 nm), BW245C (0.4 nm), and BW A868C (2.3 nm). L‐644,698 was found to be a full agonist with an EC50 value of 0.5 nm in generating cyclic AMP following activation of hDP. L‐644,698 is, therefore, comparable to those agonists with known efficacy at the DP receptor (EC50): PGD2 (0.5 nm), ZK 110841 (0.2 nm) and BW245C (0.3 nm). L‐644,698 displayed a high degree of selectivity for hDP when compared to the family of cloned human prostanoid receptors: EP1 (>25,400 fold), EP2 (∼300 fold), EP3‐III (∼4100 fold), EP4 (∼10000 fold), FP (>25,400 fold), IP (>25,400 fold) and TP (>25,400 fold). L‐644,698 is, therefore, one of the most selective DP agonists as yet described. PGJ2 and Δ12‐PGJ2, two endogenous metabolites of PGD2, were also tested in this system and shown to be effective agonists with Ki and EC50 values in the nanomolar range for both compounds. In particular, PGJ2 was equipotent to known DP specific agonists with a Ki value of 0.9 nm and an EC50 value of 1.2 nm.


Prostaglandins | 1983

Comparative biological activities of synthetic leukotriene b4 and its ω-oxidation products

Anthony W. Ford-Hutchinson; A. Rackham; Robert Zamboni; Joshua Rokach; S. Roy

Abstract Synthetic leukotriene B4 (LTB4) and its ω-oxidation products, 20 OH-LT4 and 20 COOH-LTB4, were tested for their ability to induce the aggregation of rat neutrophils in vitro , to contract the guinea pig parenchymal strip in vitro and to cause vascular permeability changes in rabbit skin in vivo . 20 OH-LTB4 had 10, 100 and 20% of the activity of LTB4 in the neutrophil aggregation, parenchymal strip and vascular permeability assays respectively. 20 C00H-LTB4 was inactive in vivo and showed in vitro . These results show that while ω-oxidation is a route for biological inactivation of LTB4, 20 OH-LTB4 still retains significant biological activity.


Biochimica et Biophysica Acta | 1985

Purification and characterisation of leukotriene A4 hydrolase from rat neutrophils

Jillian F. Evans; P. Dupuis; Anthony W. Ford-Hutchinson

Leukotriene A4 hydrolase was rapidly and extensively purified from rat neutrophils using anion exchange and gel filtration high-pressure liquid chromatography. The enzyme which converts the allylic epoxide leukotriene A4 to the 5,12-dihydroxyeicosatetraenoic acid leukotriene B4 was localized in the cytosolic fraction and exhibited an optimum activity at pH 7.8 and an apparent Km for leukotriene A4 between 2 X 10(-5) and 3 X 10(-5) M. The purified leukotriene A4 hydrolase was shown to have a molecular weight of 68 000 on sodium dodecylsulfate polyacrylamide gel electrophoresis and of 50 000 by gel filtration. The molecular weight and monomeric native form of this enzyme are unique characteristics which distinguish leukotriene A4 hydrolase from previously purified epoxide hydrolases.

Collaboration


Dive into the Anthony W. Ford-Hutchinson's collaboration.

Researchain Logo
Decentralizing Knowledge