Network


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

Hotspot


Dive into the research topics where Brian Jow is active.

Publication


Featured researches published by Brian Jow.


Circulation Research | 1998

Inhibition of Cardiac Delayed Rectifier K+ Current by Overexpression of the Long-QT Syndrome HERG G628S Mutation in Transgenic Mice

Philip Babij; G. Roger Askew; Bart W. Nieuwenhuijsen; Chien-Min Su; Terry Roy Bridal; Brian Jow; Thomas M. Argentieri; John Kulik; Louis J. DeGennaro; Walter Spinelli; Thomas J. Colatsky

Mutations in the HERG gene are linked to the LQT2 form of the inherited long-QT syndrome. Transgenic mice were generated expressing high myocardial levels of a particularly severe form of LQT2-associated HERG mutation (G628S). Hearts from G628S mice appeared normal except for a modest enlargement seen only in females. Ventricular myocytes isolated from adult wild-type hearts consistently exhibited an inwardly rectifying E-4031-sensitive K+ current resembling the rapidly activating cardiac delayed rectifier K+ current (Ikr) in its time and voltage dependence; this current was not found in cells isolated from G628S mice. Action potential duration was significantly prolonged in single myocytes from G628S ventricle (cycle length=1 second, 26 degrees C) but not in recordings from intact ventricular strips studied at more physiological rates and temperature (200 to 400 bpm, 37 degrees C). ECG intervals, including QT duration, were unchanged, although minor aberrancies were noted in 20% (16/80) of the G628S mice studied, primarily involving the QRS complex and, more rarely, T-wave morphology. The aberrations were more commonly observed in females than males but could not be correlated with sex-based differences in action potential duration. These results establish the presence of IKr in the adult mouse ventricle and demonstrate the ability of the G628S mutation to exert a dominant negative effect on endogenous IKr in vivo, leading to the expected LQT2 phenotype of prolonged repolarization at the single cell level but not QT prolongation in the intact animal. The model may be useful in dissecting repolarization currents in the mouse heart and as a means of examining the mechanism(s) by which the G628S mutation exerts its dominant negative effect on native cardiac cells in vivo.


Journal of Pharmacology and Experimental Therapeutics | 2009

Procognitive and Neuroprotective Activity of a Novel α7 Nicotinic Acetylcholine Receptor Agonist for Treatment of Neurodegenerative and Cognitive Disorders

Renza Roncarati; Carla Scali; Thomas A. Comery; Steven M. Grauer; Suzan Aschmi; Hendrick Bothmann; Brian Jow; Dianne Kowal; Marco Gianfriddo; Cody Kelley; Ugo Zanelli; Chiara Ghiron; Simon N. Haydar; John Dunlop; Georg C. Terstappen

The α7 nicotinic acetylcholine receptor (nAChR) is a promising target for treatment of cognitive dysfunction associated with Alzheimers disease and schizophrenia. Here, we report the pharmacological properties of 5-morpholin-4-yl-pentanoic acid (4-pyridin-3-yl-phenyl)-amide [SEN12333 (WAY-317538)], a novel selective agonist of α7 nAChR. SEN12333 shows high affinity for the rat α7 receptor expressed in GH4C1 cells (Ki = 260 nM) and acts as full agonist in functional Ca2+ flux studies (EC50 = 1.6 μM). In whole-cell patch-clamp recordings, SEN12333 activated peak currents and maximal total charges similar to acetylcholine (EC50 = 12 μM). The compound did not show agonist activity at other nicotinic receptors tested and acted as a weak antagonist at α3-containing receptors. SEN12333 treatment (3 mg/kg i.p.) improved episodic memory in a novel object recognition task in rats in conditions of spontaneous forgetting as well as cognitive disruptions induced via glutamatergic [5H-dibenzo[a,d]cyclohepten-5,10-imine (dizocilpine maleate); MK-801] or cholinergic (scopolamine) mechanisms. This improvement was blocked by the α7-selective antagonist methyllycaconitine, indicating that it is mediated by α7 activation. SEN12333 also prevented a scopolamine-induced deficit in a passive avoidance task. In models targeting other cognitive domains, including attention and perceptual processing, SEN12333 normalized the apomorphine-induced deficit of prepulse inhibition. Neuroprotection of SEN12333 was demonstrated in quisqualate-lesioned animals in which treatment with SEN12333 (3 mg/kg/day i.p.) resulted in a significant protection of choline acetyltransferase-positive neurons in the lesioned hemisphere. Cumulatively, our results demonstrate that the novel α7 nAChR agonist SEN12333 has procognitive and neuroprotective properties, further demonstrating utility of α7 agonists for treatment of neurodegenerative and cognitive disorders.


Journal of Pharmacology and Experimental Therapeutics | 2009

Old and New Pharmacology: Positive Allosteric Modulation of the α7 Nicotinic Acetylcholine Receptor by the 5-Hydroxytryptamine2B/C Receptor Antagonist SB-206553 (3,5-Dihydro-5-methyl-N-3-pyridinylbenzo[1,2-b:4,5-b′]di pyrrole-1(2H)-carboxamide)

John Dunlop; Tim Lock; Brian Jow; Fabrizio Sitzia; Steven M. Grauer; Flora Jow; Angela Kramer; Mark R. Bowlby; Andrew D. Randall; Dianne Kowal; Adam M. Gilbert; Thomas A. Comery; James LaRocque; Veronica Soloveva; Jon T. Brown; Renza Roncarati

The α7 nicotinic acetylcholine receptor (nAChR) has been implicated in Alzheimers disease and schizophrenia, leading to efforts targeted toward discovering agonists and positive allosteric modulators (PAMs) of this receptor. In a Ca2+ flux fluorometric imaging plate reader assay, SB-206553 (3,5-dihydro-5-methyl -N-3-pyridinylbenzo [1, 2-b:4,5 -b′]-di pyrrole-1(2H)-carboxamide), a compound known as a 5-hydroxytryptamine2B/2C receptor antagonist, produced an 8-fold potentiation of the evoked calcium signal in the presence of an EC20 concentration of nicotine and a corresponding EC50 of 1.5 μM for potentiation of EC20 nicotine responses in GH4C1 cells expressing the α7 receptor. SB-206553 was devoid of direct α7 receptor agonist activity and selective against other nicotinic receptors. Confirmation of the PAM activity of SB-206553 on the α7 nAChR was obtained in patch-clamp electrophysiological experiments in GH4C1 cells, where it failed to evoke any detectable currents when applied alone, yet dramatically potentiated the currents evoked by an EC20 (17 μM) and EC100 (124 μM) of acetylcholine (ACh). Native nicotinic receptors in CA1 stratum radiatum interneurons of rat hippocampal slices could also be activated by ACh (200 μM), an effect that was entirely blocked by the α7-selective antagonist methyllycaconitine (MLA). These ACh currents were potentiated by SB-206553, which increased the area of the current response significantly, resulting in a 40-fold enhancement at 100 μM. In behavioral experiments in rats, SB-206553 reversed an MK-801 (dizocilpine maleate)-induced deficit in the prepulse inhibition of acoustic startle response, an effect attenuated in the presence of MLA. This latter observation provides further evidence in support of the potential therapeutic utility of α7 nAChR PAMs in schizophrenia.


Assay and Drug Development Technologies | 2008

Functional Properties of α7 Nicotinic Acetylcholine Receptors Co-expressed with RIC-3 in a Stable Recombinant CHO-K1 Cell Line

Renza Roncarati; Tamara Seredenina; Brian Jow; Flora Jow; Silvia Papini; Angela Kramer; Hendrick Bothmann; John Dunlop; Georg C. Terstappen

Heterologous functional expression of alpha7 nicotinic acetylcholine receptors (nAChRs) is difficult to achieve in mammalian cell lines, and the reasons have been associated with a lack of expression of the putative chaperone factor RIC-3. Here, we describe the generation and functional and pharmacological characterization of a Chinese hamster ovary (CHO)-K1 cell line co-expressing the human alpha7 nAChR and RIC-3. Stable recombinant cells expressing alpha7 nAChR on the plasma membrane were selected by binding of fluorochrome-conjugated alpha-bungarotoxin and fluorescence-activated cell sorting. The presence of functional alpha7 channels was demonstrated by whole cell patch clamp recordings. Nicotine and acetylcholine induced rapid desensitizing currents with 50% effective concentration values of 14 and 37 microM, respectively, with agonist-evoked currents detected in approximately 75% of the cell population. Surprisingly, when tested in a FLIPR (Molecular Devices, Sunnyvale, CA) Ca(2+) assay, activation of alpha7 nAChRs was measured only when nicotinic agonists were applied either in the presence of the positive allosteric modulator (PAM) PNU-120596 or after pretreatment of cells with the tyrosine kinase inhibitor genistein. No Ca(2+) influx was measured upon addition of agonists alone or together with allosteric potentiators such as 5-hydroxyindole that predominantly increase the apparent peak amplitude without robustly affecting the current desensitization rate, as exemplified by PNU-120596. These results show that functional alpha7 nAChRs can stably be expressed in the non-neuronal CHO-K1 cell line. This recombinant cell system is useful for characterization of alpha7 nAChRs and to study the mechanism of action of chemical modulators, in particular the detection of PAMs capable of slowing receptor desensitization kinetics.


Journal of Cardiovascular Pharmacology | 2002

Sphingosine modulates myocyte electrophysiology, induces negative inotropy, and decreases survival after myocardial ischemia

Gregory S. Friedrichs; Robert E. Swillo; Brian Jow; Terry Roy Bridal; Randy Numann; Linda Warner; Loran Marie Killar; Kurex Sidek

Contractility studies in isolated feline myocytes have demonstrated that sphingosine, a metabolite stimulated by tumor necrosis factor (TNF) binding, decreases intracellular calcium release and depresses inotropic activity. This study investigated the electrophysiologic effects of sphingosine in isolated cat myocytes as well as the cardiodynamic consequence of TNF, sphingosine, and its metabolic precursors in vivo. In cat myocytes, sphingosine markedly decreased action potential duration, lowered action potential plateau, and inhibited L-type calcium current (I Ca-L ). After administration of TNF, sphingomyelin, C2-ceramide, or sphingosine, only C2-ceramide and sphingosine depressed cardiac function in normal rats. Negative inotropic effects of C2-ceramide were attenuated by N-oleoylethanolamine (NOE), a ceramidase inhibitor that blocks sphingosine formation. Rats pretreated with NOE before undergoing 30 min of acute regional myocardial ischemia followed by 150 min of reperfusion exhibited improved survival. Most deaths could be attributed to acute pump failure accompanied by bradycardia. Myocardial infarct size and peak serum TNF were not different between NOE- and vehicle-treated groups (3,908 ± 1097 pg/ml and 3,027 ± 846 pg/ml, respectively). These results indicate that sphingosine exerts direct inhibitory effects on the action potential and I Ca-L in isolated feline myocytes, consistent with previously reported sphingosine activity on I Ca-L in isolated rat myocytes. The in vivo study suggests that reducing sphingosine production with N-oleoylethanolamine attenuates cardiodepression and can improve overall survival after ischemic injury. Clearly, agents that modulate sphingosine production limit cardiodepression and may provide a therapeutic benefit in clinical conditions of myocardial inflammatory injury.


European Journal of Pharmacology | 1999

The effects of ZD6169 on the ATP-dependent K+ current (IKATP) in isolated cat ventricular myocytes

Brian Jow; Randy Numann

The effect of the K(ATP) channel opener ZD6169 [(S)-N-(4-benzoyl-phenyl)-3,3, 3-trifluoro-2-hydroxy-2-methyl-propionamide] currently under development for the treatment of urinary incontinence was explored in acutely isolated adult feline ventricular myocytes. ZD6169 activated a current over a wide range of concentrations (0.1-100 microM) that is completely blocked by 10 microM glyburide thereby identifying it as I(K(ATP)). The maximum activation of K(ATP) current was observed at 10 microM; higher concentrations decreased current activation. In contrast, the standard K(ATP) channel opener cromakalim showed a more usual concentration-response relationship, with increasing current for increased concentrations and no signs of saturation or reversal. The bell-shaped dose-response relationship for ZD6169 activation of I(K(ATP)) has also been seen in bladder myocytes, albeit at a lower concentration, and it has been proposed to contribute to the reported lack of in vivo cardiovascular side effects. We compared the effects of ZD6169 to cromakalim and showed that both compounds dramatically shorten cardiac myocyte action potential duration and that ZD6169 does so in spite of the bell-shaped concentration-response relationship for activation of K(ATP) current.


Biochemical Pharmacology | 2007

In vitro screening strategies for nicotinic receptor ligands.

John Dunlop; Renza Roncarati; Brian Jow; Hendrick Bothmann; Tim Lock; Dianne Kowal; Mark R. Bowlby; Georg C. Terstappen


Molecular Pharmacology | 2005

Characterization of Novel Aryl-Ether, Biaryl, and Fluorene Aspartic Acid and Diaminopropionic Acid Analogs as Potent Inhibitors of the High-Affinity Glutamate Transporter EAAT2

John Dunlop; Beal McIlvain; Tikva Carrick; Brian Jow; Qiang Lu; Dianne Kowal; Stephen Lin; Alexander Alexei Greenfield; Cristina Grosanu; Kristi Fan; Robert E. Petroski; John T. Williams; Alan C. Foster; John A. Butera


Archive | 2001

Method of treating or inhibiting cellular injury or cell death

Gregory S. Friedrichs; Roberto Edward Swillo; Brian Jow; Terry Roy Bridal; Randal Edward Numann; Linda Warner; Loran Marie Killar


Bioorganic & Medicinal Chemistry Letters | 2005

Synthesis and biological activities of aryl-ether-, biaryl-, and fluorene-aspartic acid and diaminopropionic acid analogs as potent inhibitors of the high-affinity glutamate transporter EAAT-2.

Alexander Alexei Greenfield; Cristina Grosanu; John Dunlop; Beal McIlvain; Tikva Carrick; Brian Jow; Qiang Lu; Dianne Kowal; John P. Williams; John A. Butera

Collaboration


Dive into the Brian Jow'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