Network


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

Hotspot


Dive into the research topics where Kiyoyuki Omoto is active.

Publication


Featured researches published by Kiyoyuki Omoto.


Journal of Medicinal Chemistry | 2013

Ion Channels as Therapeutic Targets: A Drug Discovery Perspective

Sharan K. Bagal; Alan Daniel Brown; Peter J. Cox; Kiyoyuki Omoto; Robert M. Owen; David C. Pryde; Benjamin Sidders; Sarah Elizabeth Skerratt; Edward B. Stevens; R. Ian Storer; Nigel Alan Swain

Ion channels are membrane proteins expressed in almost all living cells. The sequencing of the human genome has identified more than 400 putative ion channels, but only a fraction of these have been cloned and functionally tested. The widespread tissue distribution of ion channels, coupled with the plethora of physiological consequences of their opening and closing, makes ion-channel-targeted drug discovery highly compelling. However, despite some important drugs in clinical use today, as a class, ion channels remain underexploited in drug discovery and many existing drugs are poorly selective with significant toxicities or suboptimal efficacy. This Perspective seeks to review the ion channel family, its structural and functional features, and the diseases that are known to be modulated by members of the family. In particular, we will explore the structure and properties of known ligands and consider the future prospects for drug discovery in this challenging but high potential area.


Molecular Pharmacology | 2014

Influence of the N-terminus on the Biophysical Properties and Pharmacology of TREK1 Potassium Channels

Emma L. Veale; Ehab Al-Moubarak; Naina Bajaria; Kiyoyuki Omoto; Lishuang Cao; Stephen J. Tucker; Edward B. Stevens; Alistair Mathie

TWIK-related K+ 1 (TREK1) potassium channels are members of the two-pore domain potassium channel family and contribute to background potassium conductances in many cell types, where their activity can be regulated by a variety of physiologic and pharmacologic mediators. Fenamates such as FFA (flufenamic acid; 2-{[3-(trifluoromethyl)phenyl]amino}benzoic acid), MFA [mefenamic acid; 2-(2,3-dimethylphenyl)aminobenzoic acid], NFA [niflumic acid; 2-{[3-(trifluoromethyl)phenyl]amino}nicotinic acid], and diclofenac [2-(2-(2,6-dichlorophenylamino)phenyl)acetic acid] and the related experimental drug BL-1249 [(5,6,7,8-tetrahydro-naphthalen-1-yl)-[2-(1H-tetrazol-5-yl)-phenyl]-amine] enhance the activity of TREK1 currents, and we show that BL-1249 is the most potent of these compounds. Alternative translation initiation produces a shorter, N terminus truncated form of TREK1 with a much reduced open probability and a proposed increased permeability to sodium compared with the longer form. We show that both forms of TREK1 can be activated by fenamates and that a number of mutations that affect TREK1 channel gating occlude the action of fenamates but only in the longer form of TREK1. Furthermore, fenamates produce a marked enhancement of current through the shorter, truncated form of TREK1 and reveal a K+-selective channel, like the long form. These results provide insight into the mechanism of TREK1 channel activation by fenamates, and, given the role of TREK1 channels in pain, they suggest a novel analgesic mechanism for these compounds.


Bioorganic & Medicinal Chemistry Letters | 2012

Medicinal chemistry approaches to avoid aldehyde oxidase metabolism.

David C. Pryde; Thien-Duc Tran; Peter Jones; Jonathan Duckworth; Martin Howard; Iain Gardner; Ruth Hyland; Rob Webster; Tracey Wenham; Sharan K. Bagal; Kiyoyuki Omoto; Richard P. Schneider; Jian Lin

Aldehyde oxidase (AO) is a molybdenum-containing enzyme distributed throughout the animal kingdom and capable of metabolising a wide range of aldehydes and N-heterocyclic compounds. Although metabolism by this enzyme in man is recognised to have significant clinical impact where human AO activity was not predicted by screening in preclinical species, there is very little reported literature offering real examples where drug discoverers have successfully designed away from AO oxidation. This article reports on some strategies adopted in the Pfizer TLR7 agonist programme to successfully switch off AO metabolism that was seen principally in the rat.


Bioorganic & Medicinal Chemistry Letters | 2011

Design and pharmacological evaluation of PF-4840154, a non-electrophilic reference agonist of the TrpA1 channel.

Thomas Ryckmans; Aisah A. Aubdool; Jennifer V. Bodkin; Peter Cox; Susan D. Brain; Thomas Dupont; Emma Fairman; Yoshinobu Hashizume; Naoko Ishii; Teruhisa Kato; Linda Kitching; Julie Newman; Kiyoyuki Omoto; David James Rawson; Jade Strover

TrpA1 is an ion channel involved in nociceptive and inflammatory pain. It is implicated in the detection of chemical irritants through covalent binding to a cysteine-rich intracellular region of the protein. While performing an HTS of the Pfizer chemical collection, a class of pyrimidines emerged as a non-reactive, non-covalently binding family of agonists of the rat and human TrpA1 channel. Given the issues identified with the reference agonist Mustard Oil (MO) in screening, a new, non-covalently binding agonist was optimized and proved to be a superior agent to MO for screening purposes. Compound 16a (PF-4840154) is a potent, selective agonist of the rat and human TrpA1 channel and elicited TrpA1-mediated nocifensive behaviour in mouse.


Bioorganic & Medicinal Chemistry Letters | 2008

The SAR studies of novel CB2 selective agonists, benzimidazolone derivatives

Hirofumi Omura; Makoto Kawai; Akiko Shima; Yasuhiro Iwata; Fumitaka Ito; Tsutomu Masuda; Atsuko Ohta; Naoya Makita; Kiyoyuki Omoto; Hiromi Sugimoto; Akira Kikuchi; Hiroshi Iwata; Kazuo Ando

Benzimidazolone derivatives were discovered as novel CB2 selective agonists. Structure Activity Relationship (SAR) studies around them were examined to improve metabolic stability. Compound 39 exhibited excellent metabolic stability in human liver microsomes (HLM) and significant attenuation of the chronic colonic allodynia in the TNBS-treated rats by po administration.


Human Mutation | 2017

A Comprehensive Functional Analysis of NTRK1 Missense Mutations Causing Hereditary Sensory and Autonomic Neuropathy Type IV (HSAN IV).

Samiha S. Shaikh; Yaoyao Chen; Sally-Anne Halsall; Michael S. Nahorski; Kiyoyuki Omoto; Gareth T. Young; Anne Phelan; Christopher Geoffrey Woods

Hereditary sensory and autonomic neuropathy type IV (HSAN IV) is an autosomal recessive disorder characterized by a complete lack of pain perception and anhidrosis. Here, we studied a cohort of seven patients with HSAN IV and describe a comprehensive functional analysis of seven novel NTRK1 missense mutations, c.1550G >A, c.1565G >A, c.1970T >C, c.2096T >C, c.2254T >A, c.2288G >C, and c.2311C >T, corresponding to p.G517E, p.G522E, p.L657P, p.I699T, p.C752S, p.C763S, and p.R771C, all of which were predicted pathogenic by in silico analysis. The results allowed us to assess the pathogenicity of each mutation and to gain novel insights into tropomyosin receptor kinase A (TRKA) downstream signaling. Each mutation was systematically analyzed for TRKA glycosylation states, intracellular and cell membrane expression patterns, nerve growth factor stimulated TRKA autophosphorylation, TRKA‐Y496 phosphorylation, PLCγ activity, and neurite outgrowth. We showed a diverse range of functional effects: one mutation appeared fully functional, another had partial activity in all assays, one mutation affected only the PLCγ pathway and four mutations were proved null in all assays. Thus, we conclude that complete abolition of TRKA kinase activity is not the only pathogenic mechanism underlying HSAN IV. By corollary, the assessment of the clinical pathogenicity of HSAN IV mutations is more complex than initially predicted and requires a multifaceted approach.


Molecular Pharmaceutics | 2015

In Silico Identification of PAP-1 Binding Sites in the Kv1.2 Potassium Channel

Christian Jorgensen; Leonardo Darré; Kenno Vanommeslaeghe; Kiyoyuki Omoto; David C. Pryde; Carmen Domene

Voltage-gated potassium channels of the Kv1 family play a crucial role in the generation and transmission of electrical signals in excitable cells affecting neuronal and cardiac activities. Small-molecule blockage of these channels has been proposed to occur via a cooperative mechanism involving two main blocking sites: the inner-pore site located below the selectivity filter, and a side-pocket cavity located between the pore and the voltage sensor. Using 0.5 μs molecular dynamics simulation trajectories complemented by docking calculations, the potential binding sites of the PAP-1 (5-(4-phenoxybutoxy)psoralen) blocker to the crystal structure of Kv1.2 channel have been studied. The presence of both mentioned blocking sites at Kv1.2 is confirmed, adding evidence in favor of a cooperative channel blockage mechanism. These observations provide insight into drug modulation that will guide further developments of Kv inhibitors.


Journal of Chemical Information and Modeling | 2017

Evaluation and Characterization of Trk Kinase Inhibitors for the Treatment of Pain: Reliable Binding Affinity Predictions from Theory and Computation

Shunzhou Wan; Agastya P. Bhati; Sarah Elizabeth Skerratt; Kiyoyuki Omoto; Veerabahu Shanmugasundaram; Sharan K. Bagal; Peter V. Coveney

Optimization of ligand binding affinity to the target protein of interest is a primary objective in small-molecule drug discovery. Until now, the prediction of binding affinities by computational methods has not been widely applied in the drug discovery process, mainly because of its lack of accuracy and reproducibility as well as the long turnaround times required to obtain results. Herein we report on a collaborative study that compares tropomyosin receptor kinase A (TrkA) binding affinity predictions using two recently formulated fast computational approaches, namely, Enhanced Sampling of Molecular dynamics with Approximation of Continuum Solvent (ESMACS) and Thermodynamic Integration with Enhanced Sampling (TIES), to experimentally derived TrkA binding affinities for a set of Pfizer pan-Trk compounds. ESMACS gives precise and reproducible results and is applicable to highly diverse sets of compounds. It also provides detailed chemical insight into the nature of ligand-protein binding. TIES can predict and thus optimize more subtle changes in binding affinities between compounds of similar structure. Individual binding affinities were calculated in a few hours, exhibiting good correlations with the experimental data of 0.79 and 0.88 from the ESMACS and TIES approaches, respectively. The speed, level of accuracy, and precision of the calculations are such that the affinity predictions can be used to rapidly explain the effects of compound modifications on TrkA binding affinity. The methods could therefore be used as tools to guide lead optimization efforts across multiple prospective structurally enabled programs in the drug discovery setting for a wide range of compounds and targets.


British Journal of Pharmacology | 2018

GI‐530159, a novel, selective, mechanosensitive two‐pore‐domain potassium (K2P) channel opener, reduces rat dorsal root ganglion neuron excitability

Alexandre J C Loucif; Pierre-Philippe Saintot; Jia Liu; Brett Antonio; Shannon G Zellmer; Katrina Yoger; Emma L. Veale; Anna Wilbrey; Kiyoyuki Omoto; Lishuang Cao; Alex Gutteridge; Neil A. Castle; Edward B. Stevens; Alistair Mathie

TREK two‐pore‐domain potassium (K2P) channels play a critical role in regulating the excitability of somatosensory nociceptive neurons and are important mediators of pain perception. An understanding of the roles of TREK channels in pain perception and, indeed, in other pathophysiological conditions, has been severely hampered by the lack of potent and/or selective activators and inhibitors. In this study, we describe a new, selective opener of TREK channels, GI‐530159.


British Journal of Pharmacology | 2017

GI-530159, a novel, selective, mechano-sensitive K2P channel opener, reduces rat dorsal root ganglion (DRG) neuron excitability.

Alexandre J C Loucif; Pierre-Philippe Saintot; Jia Liu; Brett Antonio; Shannon G Zellmer; Katrina Yoger; Emma L. Veale; Anna Wilbrey; Kiyoyuki Omoto; Lishuang Cao; Alex Gutteridge; Neil A. Castle; Edward B. Stevens; Alistair Mathie

TREK two‐pore‐domain potassium (K2P) channels play a critical role in regulating the excitability of somatosensory nociceptive neurons and are important mediators of pain perception. An understanding of the roles of TREK channels in pain perception and, indeed, in other pathophysiological conditions, has been severely hampered by the lack of potent and/or selective activators and inhibitors. In this study, we describe a new, selective opener of TREK channels, GI‐530159.

Researchain Logo
Decentralizing Knowledge