Network


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

Hotspot


Dive into the research topics where Robert P. Davis is active.

Publication


Featured researches published by Robert P. Davis.


Journal of Neurochemistry | 2003

Pharmacological and immunohistochemical characterization of the APJ receptor and its endogenous ligand apelin

Andrew D. Medhurst; Carol A. Jennings; Melanie J. Robbins; Robert P. Davis; Catherine E. Ellis; Kim Winborn; Kenneth W. M. Lawrie; Guillaume Hervieu; Graham J. Riley; Jane E. Bolaky; Nicole C. Herrity; Paul R. Murdock; John G. Darker

Apelin peptides have recently been identified to be the endogenous ligands for the G protein‐coupled receptor APJ. However, little is known about the physiological roles of this ligand‐receptor pairing. In the present study we investigated the pharmacology of several apelin analogues at the human recombinant APJ receptor using radioligand binding and functional assays. This has led to the identification of key residues in the apelin peptide required for functional potency and binding affinity through structure–activity studies. In particular, we have identified that replacement of leucine in position 5, or arginine in position 2 and 4 of the C‐terminal apelin peptide, apelin‐13, resulted in significant changes in pharmacology. We also investigated the detailed localization of pre‐proapelin and APJ receptor mRNA in a wide range of human, rat and mouse tissues using quantitative RT–PCR, and carried out a detailed immunohistochemical study of the distribution of the APJ receptor in rat brain and spinal cord. Interestingly, the APJ receptor was not only co‐localized in white matter with GFAP in the spinal cord, but was also clearly localized on neurones in the brain, suggesting that this receptor and its peptide may be involved in a wide range of biological process yet to be determined.


Journal of Pharmacology and Experimental Therapeutics | 2007

GSK189254, a Novel H3 Receptor Antagonist That Binds to Histamine H3 Receptors in Alzheimer's Disease Brain and Improves Cognitive Performance in Preclinical Models

Andrew D. Medhurst; Alan R. Atkins; Isabel J. M. Beresford; Kim Brackenborough; Michael A. Briggs; Andrew R. Calver; Jackie Cilia; Jane E. Cluderay; Barry Crook; John B. Davis; Rebecca K. Davis; Robert P. Davis; Lee A. Dawson; Andrew G. Foley; Jane Gartlon; M. Isabel Gonzalez; Teresa Heslop; Warren D. Hirst; Carol A. Jennings; Declan N.C. Jones; Laurent P. Lacroix; Abbe Martyn; Sandrine Ociepka; Alison M. Ray; Ciaran M. Regan; Jennifer C. Roberts; Joanne Schogger; Eric Southam; Tania O. Stean; Brenda K. Trail

6-[(3-Cyclobutyl-2,3,4,5-tetrahydro-1H-3-benzazepin-7-yl)oxy]-N-methyl-3-pyridinecarboxamide hydrochloride (GSK189254) is a novel histamine H3 receptor antagonist with high affinity for human (pKi = 9.59 –9.90) and rat (pKi = 8.51–9.17) H3 receptors. GSK189254 is >10,000-fold selective for human H3 receptors versus other targets tested, and it exhibited potent functional antagonism (pA2 = 9.06 versus agonist-induced changes in cAMP) and inverse agonism [pIC50 = 8.20 versus basal guanosine 5′-O-(3-[35S]thio)triphosphate binding] at the human recombinant H3 receptor. In vitro autoradiography demonstrated specific [3H]GSK189254 binding in rat and human brain areas, including cortex and hippocampus. In addition, dense H3 binding was detected in medial temporal cortex samples from severe cases of Alzheimers disease, suggesting for the first time that H3 receptors are preserved in late-stage disease. After oral administration, GSK189254 inhibited cortical ex vivo R-(–)-α-methyl[imidazole-2,5(n)-3H]histamine dihydrochloride ([3H]R-α-methylhistamine) binding (ED50 = 0.17 mg/kg) and increased c-Fos immunoreactivity in prefrontal and somatosensory cortex (3 mg/kg). Microdialysis studies demonstrated that GSK189254 (0.3–3 mg/kg p.o.) increased the release of acetylcholine, noradrenaline, and dopamine in the anterior cingulate cortex and acetylcholine in the dorsal hippocampus. Functional antagonism of central H3 receptors was demonstrated by blockade of R-α-methylhistamine-induced dipsogenia in rats (ID50 = 0.03 mg/kg p.o.). GSK189254 significantly improved performance of rats in diverse cognition paradigms, including passive avoidance (1 and 3 mg/kg p.o.), water maze (1 and 3 mg/kg p.o.), object recognition (0.3 and 1 mg/kg p.o.), and attentional set shift (1 mg/kg p.o.). These data suggest that GSK189254 may have therapeutic potential for the symptomatic treatment of dementia in Alzheimers disease and other cognitive disorders.


Molecular Brain Research | 2000

The use of quantitative RT-PCR to measure mRNA expression in a rat model of focal ischemia--caspase-3 as a case study.

David C. Harrison; Andrew D. Medhurst; Brian C Bond; Colin A. Campbell; Robert P. Davis; Karen L. Philpott

Quantitative reverse transcription and polymerisation chain reaction (RT-PCR) using Taqman¿trade mark omitted¿ fluorogenic probes has been used to measure changes in gene expression in the cerebral cortex of rats in the permanent middle cerebral artery occlusion (pMCAO) model of focal ischemia. The mRNA levels of three housekeeping genes have been analysed in this model to determine which gene showed least change following experimental insult. In the lesioned cortex, beta-actin mRNA increased at 24 h, while the levels of cyclophilin and glyceraldehyde-3-phosphate dehydrogenase (GAPDH) did not change. We have also used this methodology to examine modulations in the level of caspase-3 mRNA during focal ischemia in the rat. Caspase-3 mRNA showed a 41% increase at 6 h post-MCAO, which was specific to the lesioned cortex. This change became more pronounced with time, showing an increase of 220% at 24 h. This methodology enables changes in mRNA expression to be analysed more sensitively and quantitatively than other available techniques and highlights the need for careful choice of control or housekeeping genes used for RNA comparisons.


Molecular Brain Research | 2001

Caspase mRNA expression in a rat model of focal cerebral ischemia

David C. Harrison; Robert P. Davis; Brian C Bond; Colin A. Campbell; Michael F. James; Andrew A. Parsons; Karen L. Philpott

Proteins of the caspase family are involved in the signalling pathway that ultimately leads to programmed cell death (apoptosis), which has been reported to occur in some experimental models of stroke. In a previous paper we used quantitative reverse transcription and polymerase chain reaction (RT-PCR) to characterise changes in the mRNA expression of one member of this family, caspase-3, in a rat model of permanent focal ischemia. Here we have used this technique to study the expression of a further three caspases which are involved in different aspects of caspase signalling. Caspase-8, involved in Fas-mediated apoptosis, was upregulated in the cortex of ischemic rats. Caspase-11, which leads to the synthesis of the functional form of the cytokine interleukin-1 beta, also showed increased expression, but with a different temporal profile from caspase-8. In contrast, caspase-9, which forms part of the pathway signalling through the mitochondria, showed a decrease in expression. The expression of a further four caspases (1, 2, 6 and 7) has also been characterised in a simpler experiment. These caspases all showed distinctive patterns of expression following the induction of ischemia. These data lead us to conclude that caspase expression as a whole is under very strict transcriptional control in this model. Certain elements of caspase signalling, such as the Fas-induced pathway and the events upstream of IL-1 beta processing, are upregulated, while others are not. This may be due to some form of genetic program activated in response to ischemia in the brain and may highlight which biological pathways are modulated.


Bioorganic & Medicinal Chemistry Letters | 2011

Discovery of GSK143, a highly potent, selective and orally efficacious spleen tyrosine kinase inhibitor.

John Liddle; Francis Louis Atkinson; Michael David Barker; Paul S. Carter; Neil R. Curtis; Robert P. Davis; Clement Douault; Marion C. Dickson; Dorothy Elwes; Neil Stuart Garton; Matthew Gray; Thomas G. Hayhow; Clare I. Hobbs; Emma Jones; Stuart G. Leach; Karen Leavens; Huw D. Lewis; Scott McCleary; Margarete Neu; Vipulkumar Kantibhai Patel; Alex G.S. Preston; Cesar Ramirez-Molina; Tracy Jane Shipley; Philip Alan Skone; Nick Smithers; Donald O. Somers; Ann Louise Walker; Robert J. Watson; Gordon G. Weingarten

The lead optimisation of the diaminopyrimidine carboxamide series of spleen tyrosine kinase inhibitors is described. The medicinal chemistry strategy was focused on optimising the human whole blood activity whilst achieving a sufficient margin over liability kinases and hERG activity. GSK143 is a potent and highly selective SYK inhibitor showing good efficacy in the rat Arthus model.


Neurodegenerative Diseases | 2011

Dynamics of Aβ42 reduction in plasma, CSF and brain of rats treated with the γ-secretase modulator, GSM-10h.

Julie Hawkins; David C. Harrison; Sharlin Ahmed; Robert P. Davis; Trevor D. Chapman; Ian Marshall; Beverley Smith; Tania L. Mead; Andrew D. Medhurst; Ged M.P. Giblin; Adrian Hall; Maria I. Gonzalez; Jill C. Richardson; Ishrut Hussain

Background:Allosteric modulation of γ-secretase is an attractive therapeutic approach for the treatment of Alzheimer’s disease. We recently identified a novel γ-secretase modulator, GSM-10h, which effectively lowers Aβ42 production in cells and in amyloid precursor protein transgenic mice. Objective: Here, we describe the in vivo characterization of GSM-10h in a model of endogenous Aβ production. Methods: Rats were administered orally with GSM-10h, and the effect on Aβ levels in peripheral and central compartments was determined. In addition, the effect of GSM-10h on Notch processing was assessed. Results: Acute administration of GSM-10h to rats causes a dose-dependent decrease in the level of Aβ42 in plasma, CSF and brain, with little effect on the level of Aβ40 in these compartments. The magnitude of Aβ42 lowering in the CSF and brain was further enhanced upon sub-chronic administration of GSM-10h. No deleterious effect on Notch processing was evident in either of these studies. To further explore the dynamics of Aβ42 reduction in peripheral and CNS compartments, a time course study was conducted. In all compartments, the decrease in Aβ42 was greatest at 6 h after administration of GSM-10h. This decrease in Aβ42 was maintained for 9–15 h, after which time Aβ42 levels returned to baseline levels. Encouragingly, no rebound in Aβ42 levels beyond baseline levels was observed. Conclusions: These findings support the γ-secretase modulator profile of GSM-10h, and highlight the utility of the rat for assessing the pre-clinical efficacy of γ-secretase modulators.


Bioorganic & Medicinal Chemistry Letters | 2008

The identification of potent, selective and CNS penetrant furan-based inhibitors of B-Raf kinase

Andrew K. Takle; Mark J. Bamford; Susannah Davies; Robert P. Davis; David Kenneth Dean; Alessandra Gaiba; Elaine Irving; Frank D. King; Antoinette Naylor; Christopher A. Parr; Alison M. Ray; Alastair D. Reith; Beverley Smith; Penelope C. Staton; Jon Graham Anthony Steadman; Tania O. Stean; David M. Wilson

Modification of the potent imidazole-based B-Raf inhibitor SB-590885 resulted in the identification of a series of furan-based derivatives with enhanced CNS penetration. One such compound, SB-699393 (17), was examined in vivo to challenge the hypothesis that selective B-Raf inhibitors may be of value in the treatment of stroke.


Bioorganic & Medicinal Chemistry Letters | 2013

Identification of clinical candidates from the benzazepine class of histamine H3 receptor antagonists.

David M. Wilson; James Apps; Nicholas Bailey; Mark J. Bamford; Isabel J. Beresford; Kim Brackenborough; Michael A. Briggs; Stephen J Brough; Andrew R. Calver; Barry Crook; Rebecca K. Davis; Robert P. Davis; Susannah Davis; David Kenneth Dean; Leanne Harris; Teresa Heslop; Vicky Holland; Phillip Jeffrey; Terrance A. Panchal; Christopher A. Parr; Nigel Quashie; Joanne Schogger; Sanjeet Singh Sehmi; Tania O. Stean; Jon Graham Anthony Steadman; Brenda K. Trail; Jeffrey Wald; Angela Worby; Andrew K. Takle; Jason Witherington

This Letter describes the discovery of GSK189254 and GSK239512 that were progressed as clinical candidates to explore the potential of H3 receptor antagonists as novel therapies for the treatment of Alzheimers disease and other dementias. By carefully controlling the physicochemical properties of the benzazepine series and through the implementation of an aggressive and innovative screening strategy that employed high throughput in vivo assays to efficiently triage compounds, the medicinal chemistry effort was able to rapidly progress the benzazepine class of H3 antagonists through to the identification of clinical candidates with robust in vivo efficacy and excellent developability properties.


Neuroscience Letters | 1999

Site-specific splice variation of the human P2X4 receptor

David O. Carpenter; Helen Jane Meadows; Stephen J Brough; Gayle Chapman; Catherine E. Clarke; Martyn C. Coldwell; Robert P. Davis; David C. Harrison; Jackie Meakin; Mark Thomas Mchale; Simon Q.J Rice; W.Jeff Tomlinson; Martyn D. Wood; Gareth J. Sanger

P2X4 receptors are expressed in specific brain areas. We now describe site-specific splice variations of the human P2X4 receptor subunit, occurring at residue [YVIG / WVFV(W)] near the end of the first predicted transmembrane domain. p2X4(b) is formed by the insertion of an additional 16 amino acids. p2X4(C) is formed by deleting a cassette of 130 amino acids, including six of the 10 conserved extracellular cysteine residues. Transfection of P2X4(a), but not p2x4(c), formed functional channels in Xenopus oocytes and human 1321N1 cells. After transfection of p2X4(b) small, inconsistent ATP-evoked responses were detected only in the human cells, but when co-expressed, p2x4(b) may alter the function of P2X4(a) in oocytes. The distribution of splice variant RNA within human brain suggests regionally-dependent expression. These data indicate that the functions of the human P2X4 receptor may be altered by alternative splicing.


Bioorganic & Medicinal Chemistry Letters | 2013

The discovery of the benzazepine class of histamine H3 receptor antagonists

David M. Wilson; James Apps; Nicholas Bailey; Mark J. Bamford; Isabel J. Beresford; Michael A. Briggs; Andrew R. Calver; Barry Crook; Robert P. Davis; Susannah Davis; David Kenneth Dean; Leanne Harris; Tom D. Heightman; Terry Panchal; Christopher A. Parr; Nigel Quashie; Jon Graham Anthony Steadman; Joanne Schogger; Sanjeet Singh Sehmi; Tania O. Stean; Andrew K. Takle; Brenda K. Trail; Trevor White; Jason Witherington; Angela Worby; Andrew D. Medhurst

This Letter describes the discovery of a novel series of H3 receptor antagonists. The initial medicinal chemistry strategy focused on deconstructing and simplifying an early screening hit which rapidly led to the discovery of a novel series of H3 receptor antagonists based on the benzazepine core. Employing an H3 driven pharmacodynamic model, the series was then further optimised through to a lead compound that showed robust in vivo functional activity and possessed overall excellent developability properties.

Collaboration


Dive into the Robert P. Davis's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

David M. Wilson

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge