Network


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

Hotspot


Dive into the research topics where Arthur Tucker is active.

Publication


Featured researches published by Arthur Tucker.


Human Molecular Genetics | 2012

Functional analyses of coronary artery disease associated variation on chromosome 9p21 in vascular smooth muscle cells

Anna Motterle; Xiangyuan Pu; Harriet Wood; Qingzhong Xiao; Shivani Gor; Fu Liang Ng; Kenneth Chan; Frank Cross; Beski Shohreh; Robin N. Poston; Arthur Tucker; Mark J. Caulfield; Shu Ye

Variation on chromosome 9p21 is associated with risk of coronary artery disease (CAD). This genomic region contains the CDKN2A and CDKN2B genes which encode the cell cycle regulators p16(INK4a), p14(ARF) and p15(INK4b) and the ANRIL gene which encodes a non-coding RNA. Vascular smooth muscle cell (VSMC) proliferation plays an important role in the pathogenesis of atherosclerosis which causes CAD. We ascertained whether 9p21 genotype had an influence on CDKN2A/CDKN2B/ANRIL expression levels in VSMCs, VSMC proliferation and VSMC content in atherosclerotic plaques. Immunohistochemical examination showed that VSMCs in atherosclerotic lesions expressed p16(INK4a), p14(ARF) and p15(INK4b). Analyses of primary cultures of VSMCs showed that the 9p21 risk genotype was associated with reduced expression of p16(INK4a), p15(INK4b) and ANRIL (P = 1.2 × 10(-5), 1.4 × 10(-2) and 3.1 × 10(-9)) and with increased VSMC proliferation (P = 1.6 × 10(-2)). Immunohistochemical analyses of atherosclerotic plaques revealed an association of the risk genotype with reduced p15(INK4b) levels in VSMCs (P = 3.7 × 10(-2)) and higher VSMC content (P = 5.6 × 10(-4)) in plaques. The results of this study indicate that the 9p21 variation has an impact on CDKN2A and CDKN2B expression in VSMCs and influences VMSC proliferation, which likely represents an important mechanism for the association between this genetic locus and susceptibility to CAD.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2009

Antiplatelet Actions of Statins and Fibrates Are Mediated by PPARs

Ferhana Y. Ali; Paul C. J. Armstrong; Al-Rehan Dhanji; Arthur Tucker; Mark J. Paul-Clark; Jane A. Mitchell; Timothy D. Warner

Objectives—Statins and fibrates are hypolipidemic drugs which decrease cardiac events in individuals without raised levels of cholesterol. These drugs inhibit platelet function, but the mechanisms by which this pleiotropic effect is exerted are not known. Methods and Results—We used a range of approaches to show statins inhibit human platelet activation in vitro while engaging PPAR&agr; and PPAR&ggr;. The effects of simvastatin were prevented by the PPAR&ggr; antagonist GW9662 or the PPAR&agr; antagonist GW6471. In a small-scale human study fluvastatin activated PPAR&agr; and PPAR&ggr; in platelets and reduced aggregation in response to arachidonic acid ex vivo. The effects of fenofibrate were prevented by PPAR&agr; antagonism with GW6471. Fenofibrate increased bleeding time in wild-type, but not in PPAR&agr;−/− mice. The inhibitory effect of fenofibrate, but not simvastatin, on aggregation was prevented by deletion of PPAR&agr; in murine platelets. PKC&agr;, which influences platelet activation, associated and immune-precipitated with PPAR&ggr; in platelets stimulated with statins and with PPAR&agr; in platelets stimulated with fenofibrate. Conclusions—This study is the first to provide a unifying explanation of how fibrates and statins reduce thrombotic and cardiovascular risk. Our findings that PPARs associate with PKC&agr; in platelets also provide a mechanism by which these effects are mediated.


American Journal of Human Genetics | 2013

ADAMTS7 Cleavage and Vascular Smooth Muscle Cell Migration Is Affected by a Coronary-Artery-Disease-Associated Variant

Xiangyuan Pu; Qingzhong Xiao; Stefan Kiechl; Kenneth Chan; Fu Liang Ng; Shivani Gor; Robin N. Poston; Changcun Fang; Ashish Patel; Ece C. Senver; Sue Shaw-Hawkins; Johann Willeit; Chuan-ju Liu; Jianhua Zhu; Arthur Tucker; Qingbo Xu; Mark J. Caulfield; Shu Ye

Recent genome-wide association studies have revealed an association between variation at the ADAMTS7 locus and susceptibility to coronary artery disease (CAD). Furthermore, in a population-based study cohort, we observed an inverse association between atherosclerosis prevalence and rs3825807, a nonsynonymous SNP (A to G) leading to a Ser-to-Pro substitution in the prodomain of the protease ADAMTS7. In light of these data, we sought a mechanistic explanation for this association. We found that ADAMTS7 accumulated in smooth muscle cells in coronary and carotid atherosclerotic plaques. Vascular smooth muscle cells (VSMCs) of the G/G genotype for rs3825807 had reduced migratory ability, and conditioned media of VSMCs of the G/G genotype contained less of the cleaved form of thrombospondin-5, an ADAMTS7 substrate that had been shown to be produced by VSMCs and inhibit VSMC migration. Furthermore, we found that there was a reduction in the amount of cleaved ADAMTS7 prodomain in media conditioned by VSMCs of the G/G genotype and that the Ser-to-Pro substitution affected ADAMTS7 prodomain cleavage. The results of our study indicate that rs3825807 has an effect on ADAMTS7 maturation, thrombospondin-5 cleavage, and VSMC migration, with the variant associated with protection from atherosclerosis and CAD rendering a reduction in ADAMTS7 function.


Circulation | 2015

Evidence That Links Loss of Cyclooxygenase-2 With Increased Asymmetric Dimethylarginine Novel Explanation of Cardiovascular Side Effects Associated With Anti-Inflammatory Drugs

Blerina Ahmetaj-Shala; Nicholas S. Kirkby; Rebecca Knowles; Malak Al-Yamani; Sarah Mazi; Zhen Wang; Arthur Tucker; Louise Susan MacKenzie; Paul C. J. Armstrong; Rolf M. Nüsing; James Tomlinson; Timothy D. Warner; James Leiper; Jane A. Mitchell

Background— Cardiovascular side effects associated with cyclooxygenase-2 inhibitor drugs dominate clinical concern. Cyclooxygenase-2 is expressed in the renal medulla where inhibition causes fluid retention and increased blood pressure. However, the mechanisms linking cyclooxygenase-2 inhibition and cardiovascular events are unknown and no biomarkers have been identified. Methods and Results— Transcriptome analysis of wild-type and cyclooxygenase-2−/− mouse tissues revealed 1 gene altered in the heart and aorta, but >1000 genes altered in the renal medulla, including those regulating the endogenous nitric oxide synthase inhibitors asymmetrical dimethylarginine (ADMA) and monomethyl-L-arginine. Cyclo-oxygenase-2−/− mice had increased plasma levels of ADMA and monomethyl-L-arginine and reduced endothelial nitric oxide responses. These genes and methylarginines were not similarly altered in mice lacking prostacyclin receptors. Wild-type mice or human volunteers taking cyclooxygenase-2 inhibitors also showed increased plasma ADMA. Endothelial nitric oxide is cardio-protective, reducing thrombosis and atherosclerosis. Consequently, increased ADMA is associated with cardiovascular disease. Thus, our study identifies ADMA as a biomarker and mechanistic bridge between renal cyclooxygenase-2 inhibition and systemic vascular dysfunction with nonsteroidal anti-inflammatory drug usage. Conclusions— We identify the endogenous endothelial nitric oxide synthase inhibitor ADMA as a biomarker and mechanistic bridge between renal cyclooxygenase-2 inhibition and systemic vascular dysfunction.


Transplant International | 2011

A survey to determine the views of renal transplant patients on generic substitution in the UK

Mubarak Nasser Al Ameri; Clare Whittaker; Arthur Tucker; Magdi Yaqoob; Atholl Johnston

Rising healthcare costs promote the generic substitution among patients because it is identifiable costs. A key concern is that patients should be involved in the decision of switching. The aim of this study was to examine renal transplant patients’ views on generic substitution in the UK. A total of 163 renal patients were surveyed using 36 multiple‐choice questions at Barts and The London Renal Transplant Clinic, in the UK. Transplant recipients over 18 years, able to read and write English and willing to fill in the questionnaire were targeted; 84% of patients were conscious of the availability of generic medicines, 70% understood the terms “generic” and “branded” in relation to medicines and 54% were aware of generic substitution practice. However, 75% did not know if they were taking generics and 84% felt that generics are not equivalent or only equivalent sometimes and they were uncertain that generics had the same quality as branded medicines. Moreover, many patients admitted that they would not accept the generic substitution of ciclosporin when become available in the UK. A number of factors such as patients’ education, knowledge, severity of the disease, efficacy of generic medicines and patients’ involvement in decisions regarding their health appear to drive patients’ attitudes towards generic substitution.


Cardiovascular Research | 2012

Pregnane X receptor regulates drug metabolism and transport in the vasculature and protects from oxidative stress

Karen E. Swales; Rick Moore; N. J. Truss; Arthur Tucker; Timothy D. Warner; Masahiko Negishi; David Bishop-Bailey

Aims Circulating endogenous, dietary, and foreign chemicals can contribute to vascular dysfunction. The mechanism by which the vasculature protects itself from these chemicals is unknown. This study investigates whether the pregnane X receptor (PXR), the major transcriptional regulator of hepatic drug metabolism and transport that responds to such xenobiotics, mediates vascular protection by co-ordinating a defence gene programme in the vasculature. Methods and results PXR was detected in primary human and rat aortic endothelial and smooth muscle cells (SMC) and blood vessels including the human and rat aorta. Metabolic PXR target genes cytochrome P450 3A, 2B, 2C, and glutathione S-transferase mRNA and activity were induced by PXR ligands in rodent and human vascular cells and absent in the aortas from PXR-null mice stimulated in vivo or in rat aortic SMC expressing dominant-negative PXR. Activation of aortic PXR by classical agonists had several protective effects: increased xenobiotic metabolism demonstrated by bioactivation of the pro-drug clopidogrel, which reduced adenosine diphosphate-induced platelet aggregation; increased expression of multidrug resistance protein 1, mediating chemical efflux from the vasculature; and protection from reactive oxygen species-mediated cell death. Conclusion PXR co-ordinately up-regulates drug metabolism, transport, and antioxidant genes to protect the vasculature from endogenous and exogenous insults, thus representing a novel gatekeeper for vascular defence.


Thrombosis and Haemostasis | 2009

Utility of 96-well plate aggregometry and measurement of thrombi adhesion to determine aspirin and clopidogrel effectiveness

Paul C. J. Armstrong; Al-Rehan Dhanji; N. J. Truss; Zetty N. Zain; Arthur Tucker; Jane A. Mitchell; Timothy D. Warner

Aspirin and clopidogrel are key anti-thrombotic therapies. Results from platelet reactivity testing during therapy, have been shown to correlate with future events and would allow for the optimisation of therapy. However, there is little agreement among current tests and there remains a clear clinical need for a universal standardised test. It was the objective of this study to explore the potential of 96-well plate aggregometry as a definitive clinical test of platelet reactivity with respect to aspirin and clopidogrel. A small non-blinded trial of 16 healthy male volunteers assigned to seven days of aspirin (75mg/day) or clopidogrel (75mg/day) therapy. Blood was collected before and on day 7 of treatment. Platelet aggregation was measured using a 96-well plate based aggregation method, and thrombi adhesion measured by colourimetric assay. Platelet agonists used were ADP (0.1-30microM), arachidonic acid (0.03-1.3mM), collagen (0.1-30microg/ml), adrenaline (0.001-100microM), ristocetin (0.2-3mg/ml), TRAP6 amide (0.130microM) and U46619 (0.130microM). Concentration response curves were constructed to each agonist under the various conditions and used to extract data such as log EC(50), Hill slope, and area under the curve. These demonstrated low intra- and inter-assay variability and strong discrimination of drug effects. This study demonstrates the ability of the 96-well plate based aggregation and adhesion method to detect and differentiate between stable aspirin and clopidogrel treatment in healthy volunteers. Moreover, this assay marries the ability to test subjects or patients using a range of platelet agonists with more rapidity and ease than the current gold standard platelet assay, traditional light transmission aggregometry, making it a serious alternative assay for use in clinical settings.


Medicinal Chemistry | 2012

Capecitabine: AnIn-vitro Comparison between the Branded Xelodaî 500 Mg nd its Intended Copy Capeda 500 Mg

Mubarak Nasser AlAmeri; Simon Abi Aad; Arthur Tucker; Atholl Johnston

Introduction: Dissolution is an example of in-vitro test which can be used to identify formulations that may present potential bioequivalence problems. It is defined as the amount of substance that goes into solution per unit time under standardised conditions of liquid/solid interface, solvent composition and temperature. It is considered one of the most important tools to predict the in-vivo bioavailability and in some cases replacing clinical studies to determine bioequivalence. Aim: To compare the differences in the dissolution behaviour between two anticancer formulations, Xeloda® 500 mg (reference product) and Capeda 500 mg (test product). Methods: Four replicates for each batch of the tested medicines were carried out using a PT-DT70 dissolution tester (Pharma Test) to detect any differences in their dissolution behaviour. Samples at nine time intervals were tested according to the US Pharmacopeia with the rate of dissolution determined by ultra-violet spectrophotometery. Results: All the tested medicines complied with the pharmacopoeial specifications, the EMA and the FDA guidance for industry when achieved 85% dissolution in 60 minutes. However, Capeda 500 mg (test product) showed slower, different and incomplete dissolution rate compared to Xeloda® 500 mg (reference product) at both 60 and 120 minutes. Other visual differences in the weight, size, clarity of solution, presence of un-dissolved residue and particles during the dissolution test were also detected. Conclusion: Results in this study clearly raise a question about the interchangeability between Xeloda® 500 mg and its Intended copy Capeda 500 mg. Awareness of these scientific concerns should be considered when a clinical choice between these two drugs is required. Differences between the innovator and copy medicines with regard to pharmacokinetics, clinical efficacy and safety may exist. Thereby, patients’ monitoring after performing drug substitution of these two medicines is strongly recommended.


British Journal of Clinical Pharmacology | 2016

P2Y12 receptor blockade synergizes strongly with nitric oxide and prostacyclin to inhibit platelet activation

Melissa V. Chan; Rebecca Knowles; Martina H. Lundberg; Arthur Tucker; Nura A. Mohamed; Nicholas S. Kirkby; Paul C. J. Armstrong; Jane A. Mitchell; Timothy D. Warner

Aims In vivo platelet function is a product of intrinsic platelet reactivity, modifiable by dual antiplatelet therapy (DAPT), and the extrinsic inhibitory endothelial mediators, nitric oxide (NO) and prostacyclin (PGI2), that are powerfully potentiated by P2Y12 receptor blockade. This implies that for individual patients endothelial mediator production is an important determinant of DAPT effectiveness. Here, we have investigated this idea using platelets taken from healthy volunteers treated with anti‐platelet drugs. Methods Three groups of male volunteers (n = 8) received either prasugrel (10 mg), aspirin (75 mg) or DAPT (prasugrel + aspirin) once daily for 7 days. Platelet reactivity in the presence of diethylammonium (Z)‐1‐(N,N‐diethylamino)diazen‐1‐ium‐1,2‐diolate (DEA/NONOate) and PGI2 was studied before and following treatment. Results Ex vivo, PGI2 and/or DEA/NONOate had little inhibitory effect on TRAP‐6‐induced platelet reactivity in control conditions. However, in the presence of DAPT, combination of DEA/NONOate + PGI2 reduced platelet aggregation (74 ± 3% to 19 ± 6%, P < 0.05). In vitro studies showed even partial (25%) P2Y12 receptor blockade produced a significant (67 ± 2% to 39 ± 10%, P < 0.05) inhibition when DEA/NONOate + PGI2 was present. Conclusions We have demonstrated that PGI2 and NO synergize with P2Y12 receptor antagonists to produce powerful platelet inhibition. Furthermore, even with submaximal P2Y12 blockade the presence of PGI2 and NO greatly enhances platelet inhibition. Our findings highlight the importance of endothelial mediator in vivo modulation of P2Y12 inhibition and introduces the concept of refining ex vivo platelet function testing by incorporating an assessment of endothelial function to predict thrombotic outcomes better and adjust therapy to prevent adverse outcomes in individual patients.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2017

Newly Formed Reticulated Platelets Undermine Pharmacokinetically Short-Lived Antiplatelet Therapies

Paul C. J. Armstrong; Thomas Hoefer; Rebecca Knowles; Arthur Tucker; Melissa A. Hayman; Plinio M. Ferreira; Melissa V. Chan; Timothy D. Warner

Objective— Aspirin together with thienopyridine P2Y12 inhibitors, commonly clopidogrel, is a cornerstone of antiplatelet therapy. However, many patients receiving this therapy display high on-treatment platelet reactivity, which is a major therapeutic hurdle to the prevention of recurrent thrombotic events. The emergence of uninhibited platelets after thrombopoiesis has been proposed as a contributing factor to high on-treatment platelet reactivity. Here, we investigate the influences of platelet turnover on platelet aggregation in the face of different dual-antiplatelet therapy strategies. Approach and Results— Traditional light transmission aggregometry, cytometry, advanced flow cytometric imaging, and confocal microscopy were used to follow the interactions of populations of platelets from healthy volunteers and patients with stable cardiovascular disease. Newly formed, reticulated platelets overproportionately contributed to, and clustered at, the core of forming aggregates. This phenomenon was particularly observed in samples from patients treated with aspirin plus a thienopyridine, but was absent in samples taken from patients treated with aspirin plus ticagrelor. Conclusions— Reticulated platelets are more reactive than older platelets and act as seeds for the formation of platelet aggregates even in the presence of antiplatelet therapy. This is coherent with the emergence of an uninhibited subpopulation of reticulated platelets during treatment with aspirin plus thienopyridine, explained by the short pharmacokinetic half-lives of these drugs. This phenomenon is absent during treatment with ticagrelor, because of its longer half-life and ability to act as a circulating inhibitor. These data highlight the important influences of pharmacokinetics on antiplatelet drug efficacies, especially in diseases associated with increased platelet turnover.

Collaboration


Dive into the Arthur Tucker's collaboration.

Top Co-Authors

Avatar

Atholl Johnston

Queen Mary University of London

View shared research outputs
Top Co-Authors

Avatar

Timothy D. Warner

Queen Mary University of London

View shared research outputs
Top Co-Authors

Avatar

Rebecca Knowles

Queen Mary University of London

View shared research outputs
Top Co-Authors

Avatar

Duncan Bain

Queen Mary University of London

View shared research outputs
Top Co-Authors

Avatar

Paul C. J. Armstrong

Queen Mary University of London

View shared research outputs
Top Co-Authors

Avatar

Nigel Benjamin

Queen Mary University of London

View shared research outputs
Top Co-Authors

Avatar

Jane A. Mitchell

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Nicholas S. Kirkby

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Melissa V. Chan

Queen Mary University of London

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge