Network


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

Hotspot


Dive into the research topics where Kate O'Driscoll is active.

Publication


Featured researches published by Kate O'Driscoll.


American Journal of Physiology-cell Physiology | 2008

Expression, localization, and functional properties of Bestrophin 3 channel isolated from mouse heart

Kate O'Driscoll; William J. Hatton; Heather R. Burkin; Normand Leblanc; Fiona C. Britton

Bestrophins are a novel family of proteins that encode calcium-activated chloride channels. In this study we establish that Bestrophin transcripts are expressed in the mouse and human heart. Native mBest3 protein expression and localization in heart was demonstrated by using a specific polyclonal mBest3 antibody. Immunostaining of isolated cardiac myocytes indicates that mBest3 is present at the membrane. Using the patch-clamp technique, we characterized the biophysical and pharmacological properties of mBest3 cloned from heart. Whole cell chloride currents were evoked in both HEK293 and COS-7 cells expressing mBest3 by elevation of intracellular calcium. mBest3 currents displayed a K(D) for Ca(2+) of approximately 175 nM. The calcium-activated chloride current was found to be time and voltage independent and displayed slight outward rectification. The anion permeability sequence of the channel was SCN(-)>I(-)>Cl(-), and the current was inhibited by niflumic acid and DIDS in the micromolar range. In addition, we generated a site-specific mutation (F80L) in the putative pore region of mBest3 that significantly altered the ion conduction and pharmacology of this channel. Our functional and mutational studies examining the biophysical properties of mBest3 indicate that it functions as a pore-forming chloride channel that is activated by physiological levels of calcium. This study reports novel findings regarding the molecular expression, tissue localization, and functional properties of mBest3 cloned from heart.


American Journal of Physiology-cell Physiology | 2015

Intracellular Ca2+ release from endoplasmic reticulum regulates slow wave currents and pacemaker activity of interstitial cells of Cajal

Mei Hong Zhu; Tae Sik Sung; Kate O'Driscoll; Sang Don Koh; Kenton M. Sanders

Interstitial cells of Cajal (ICC) provide pacemaker activity in gastrointestinal muscles that underlies segmental and peristaltic contractions. ICC generate electrical slow waves that are due to large-amplitude inward currents resulting from anoctamin 1 (ANO1) channels, which are Ca(2+)-activated Cl(-) channels. We investigated the hypothesis that the Ca(2+) responsible for the stochastic activation of ANO1 channels during spontaneous transient inward currents (STICs) and synchronized activation of ANO1 channels during slow wave currents comes from intracellular Ca(2+) stores. ICC, obtained from the small intestine of Kit(+/copGFP) mice, were studied under voltage and current clamp to determine the effects of blocking Ca(2+) uptake into stores and release of Ca(2+) via inositol 1,4,5-trisphosphate (IP3)-dependent and ryanodine-sensitive channels. Cyclocpiazonic acid, thapsigargin, 2-APB, and xestospongin C inhibited STICs and slow wave currents. Ryanodine and tetracaine also inhibited STICs and slow wave currents. Store-active compounds had no direct effects on ANO1 channels expressed in human embryonic kidney-293 cells. Under current clamp, store-active drugs caused significant depolarization of ICC and reduced spontaneous transient depolarizations (STDs). After block of ryanodine receptors with ryanodine and tetracaine, repolarization did not restore STDs. ANO1 expressed in ICC has limited access to cytoplasmic Ca(2+) concentration, suggesting that pacemaker activity depends on Ca(2+) dynamics in restricted microdomains. Our data from studies of isolated ICC differ somewhat from studies on intact muscles and suggest that release of Ca(2+) from both IP3 and ryanodine receptors is important in generating pacemaker activity in ICC.


BMC Molecular Biology | 2011

Increased complexity of Tmem16a/Anoctamin 1 transcript alternative splicing

Kate O'Driscoll; Rachel A Pipe; Fiona C. Britton

BackgroundTMEM16A (Anoctamin 1; ANO1) is an eight transmembrane protein that functions as a calcium-activated chloride channel. TMEM16A in human exhibits alternatively spliced exons (6b, 13 and 15), which confer important roles in the regulation of channel function. Mouse Tmem16a is reported to consist of 25 exons that code for a 956 amino acid protein. In this study our aim was to provide details of mouse Tmem16a genomic structure and to investigate if Tmem16a transcript undergoes alternative splicing to generate channel diversity.ResultsWe identified Tmem16a transcript variants consisting of alternative exons 6b, 10, 13, 14, 15 and 18. Our findings indicate that many of these exons are expressed in various combinations and that these splicing events are mostly conserved between mouse and human. In addition, we confirmed the expression of these exon variants in other mouse tissues. Additional splicing events were identified including a novel conserved exon 13b, tandem splice sites of exon 1 and 21 and two intron retention events.ConclusionOur results suggest that Tmem16a gene is significantly more complex than previously described. The complexity is especially evident in the region spanning exons 6 through 16 where a number of the alternative splicing events are thought to affect calcium sensitivity, voltage dependence and the kinetics of activation and deactivation of this calcium-activated chloride channel. The identification of multiple Tmem16a splice variants suggests that alternative splicing is an exquisite mechanism that operates to diversify TMEM16A channel function in both physiological and pathophysiological conditions.


British Journal of Pharmacology | 2002

Excitatory motor innervation in the canine rectoanal region: Role of changing receptor populations

Stephen Tichenor; Iain L. O. Buxton; Paul Johnson; Kate O'Driscoll; Kathleen D. Keef

Motor innervation in the canine rectoanal region was examined in isolated strips of the circular muscle layer. Contractile responses to electrical field stimulation began at lower frequencies and were more persistent in the internal anal sphincter (IAS) than in the rectum. Motor innervation to the IAS was almost exclusively sympathetic, since it was blocked by guanethidine (Guan 3 μM) while the response in the proximal rectum was approximately 50% muscarinic, and sensitive to the M3 selective antagonist 4‐diphenylacetoxy‐N‐methylpiperidine (4‐DAMP, 0.1 μM) and 50% tachykinergic, and sensitive to the neurokinin 2 (NK2) receptor antagonist GR 94800 (1 μM). From IAS to rectum there was a gradual shift in the relative contribution of intrinsic and extrinsic neural innervation. Responses to exogenously applied transmitters exhibited a similar pattern to that observed with motor innervation. Norepinephrine (NE) was most potent in the IAS and acetylcholine (ACh) and NK‐A were most potent in the proximal rectum. The responses were inhibited by prazosin, 4‐DAMP and GR 94800 respectively. A gradient in the density of adrenergic α1, muscarinic and NK2 receptors also existed from IAS to rectum as determined by measuring the binding of [3H]‐prazosin, [3H]‐quinuclidinyl benzilate ([3H]‐QNB and [3H]‐SR‐48968 to smooth muscle membranes. In summary, these data suggest that the shift in motor innervation in the rectoanal region is achieved in part by changes in receptor populations available for activation by sympathetic and enteric motor neurons.


American Journal of Physiology-cell Physiology | 2016

Influence of intracellular Ca2+ and alternative splicing on the pharmacological profile of ANO1 channels

Tae Sik Sung; Kate O'Driscoll; Haifeng Zheng; Nicholas J. Yapp; Normand Leblanc; Sang Don Koh; Kenton M. Sanders

Anoctamin-1 (ANO1) is a Ca(2+)-activated Cl(-) channel expressed in many types of cells. Splice variants of ANO1 have been shown to influence the biophysical properties of conductance. It has been suggested that several new antagonists of ANO1 with relatively high affinity and selectivity might be useful for experimental and, potentially, therapeutic purposes. We investigated the effects of intracellular Ca(2+) concentration ([Ca(2+)]i) at 100-1,000 nM, a concentration range that might be achieved in cells during physiological activation of ANO1 channels, on blockade of ANO1 channels expressed in HEK-293 cells. Whole cell and excised patch configurations of the patch-clamp technique were used to perform tests on a variety of naturally occurring splice variants of ANO1. Blockade of ANO1 currents with aminophenylthiazole (T16Ainh-A01) was highly dependent on [Ca(2+)]i Increasing [Ca(2+)]i reduced the potency of this blocker. Similar Ca(2+)-dependent effects were also observed with benzbromarone. Experiments on excised, inside-out patches showed that the diminished potency of the blockers caused by intracellular Ca(2+) might involve a competitive interaction for a common binding site or repulsion of the blocking drugs by electrostatic forces at the cytoplasmic surface of the channels. The degree of interaction between the channel blockers and [Ca(2+)]i depends on the splice variant expressed. These experiments demonstrate that the efficacy of ANO1 antagonists depends on [Ca(2+)]i, suggesting a need for caution when ANO1 blockers are used to determine the role of ANO1 in physiological functions and in their use as therapeutic agents.


American Journal of Physiology-gastrointestinal and Liver Physiology | 2016

Na+-K+-Cl− cotransporter (NKCC) maintains the chloride gradient to sustain pacemaker activity in interstitial cells of Cajal

Mei Hong Zhu; Tae Sik Sung; Masaaki Kurahashi; Lauren Okane; Kate O'Driscoll; Sang Don Koh; Kenton M. Sanders

Interstitial cells of Cajal (ICC) generate electrical slow waves by coordinated openings of ANO1 channels, a Ca2+-activated Cl- (CaCC) conductance. Efflux of Cl- during slow waves must be significant, as there is high current density during slow-wave currents and slow waves are of sufficient magnitude to depolarize the syncytium of smooth muscle cells and PDGFRα+ cells to which they are electrically coupled. We investigated how the driving force for Cl- current is maintained in ICC. We found robust expression of Slc12a2 (which encodes an Na+-K+-Cl- cotransporter, NKCC1) and immunohistochemical confirmation that NKCC1 is expressed in ICC. With the use of the gramicidin permeabilized-patch technique, which is reported to not disturb [Cl-]i, the reversal potential for spontaneous transient inward currents (ESTICs) was -10.5 mV. This value corresponds to the peak of slow waves when they are recorded directly from ICC in situ. Inhibition of NKCC1 with bumetanide shifted ESTICs to more negative potentials within a few minutes and reduced pacemaker activity. Bumetanide had no direct effects on ANO1 or CaV3.2 channels expressed in HEK293 cells or L-type Ca2+ currents. Reducing extracellular Cl- to 10 mM shifted ESTICs to positive potentials as predicted by the Nernst equation. The relatively rapid shift in ESTICs when NKCC1 was blocked suggests that significant changes in the transmembrane Cl- gradient occur during the slow-wave cycle, possibly within microdomains formed between endoplasmic reticulum and the plasma membrane in ICC. Recovery of Cl- via NKCC1 might have additional consequences on shaping the waveforms of slow waves via Na+ entry into microdomains.


Canadian Journal of Physiology and Pharmacology | 2005

Regulation of calcium-activated chloride channels in smooth muscle cells: a complex picture is emerging

Normand Leblanc; Jonathan Ledoux; Sohag N. Saleh; Amy Sanguinetti; Jeff Angermann; Kate O'Driscoll; Fiona C. Britton; Brian A. Perrino; Iain A. Greenwood


American Journal of Physiology-gastrointestinal and Liver Physiology | 2003

Spatial localization and properties of pacemaker potentials in the canine rectoanal region.

Violeta N. Mutafova-Yambolieva; Kate O'Driscoll; A. Farrelly; Sean M. Ward; Kathleen D. Keef


American Journal of Physiology-gastrointestinal and Liver Physiology | 2002

Electrical activity induced by nitric oxide in canine colonic circular muscle

Kathleen D. Keef; U. Anderson; Kate O'Driscoll; Sean M. Ward; Kenton M. Sanders


The FASEB Journal | 2010

Alternative Splicing of the murine Tmem16a Transcript in Heart

Kate O'Driscoll; William J. Hatton; Fiona C. Britton

Collaboration


Dive into the Kate O'Driscoll'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
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge