Network


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

Hotspot


Dive into the research topics where Kathryn Houseman is active.

Publication


Featured researches published by Kathryn Houseman.


Biochemical Pharmacology | 2009

Electrophysiologic characterization of a novel hERG channel activator

Zhi Su; James T. Limberis; Andrew J. Souers; Philip R. Kym; Ann Mikhail; Kathryn Houseman; Gilbert Diaz; Xiaoqin Liu; Ruth L. Martin; Bryan F. Cox; Gary A. Gintant

Activators of the human ether-a-go-go-related gene (hERG) K(+) channel have been reported recently to enhance hERG current amplitude (five synthetic small molecules and one naturally occurring substance). Here, we characterize the effects of a novel compound A-935142 ({4-[4-(5-trifluoromethyl-1H-pyrazol-3-yl)-phenyl]-cyclohexyl}-acetic acid) on guinea-pig atrial and canine ventricular action potentials (microelectrode techniques) and hERG channels expressed in HEK-293 cells (whole-cell patch clamp techniques). A-935142 shortened cardiac action potentials and enhanced the amplitude of the hERG current in a concentration- and voltage-dependent manner. The fully activated current-voltage relationship revealed that this compound (60 microM) increased both outward and inward K(+) current as well as the slope conductance of the linear portion of the fully activated I-V relation. A-935142 significantly reduced the time constants (tau) of hERG channel activation at two example voltages (-10 mV: tau=100+/-17 ms vs. 164+/-24 ms, n=6, P<0.01; +30 mV: tau=16.7+/-1.8 ms vs. 18.9+/-1.8 ms, n=5, P<0.05) and shifted the voltage-dependence for hERG activation in the hyperpolarizing direction by 9 mV. The time course of hERG channel deactivation was slowed at multiple potentials (-120 to -70 mV). A-935142 also reduced the rate of inactivation and shifted the voltage-dependence of inactivation in the depolarizing direction by 15 mV. Recovery of hERG channel from inactivation was not affected by A-935142. In conclusion, A-935142 enhances hERG current in a complex manner by facilitation of activation, reduction of inactivation, and slowing of deactivation, and abbreviates atrial and ventricular repolarization.


Life Sciences | 2012

Characterization of A-935142, a hERG enhancer, in the presence and absence of standard hERG blockers.

Xiaoqin Liu; James T. Limberis; Zhi Su; Kathryn Houseman; Gilbert Diaz; Gary A. Gintant; Bryan F. Cox; Ruth Martin

AIMS In a previous study we found that A-935142 enhanced hERG current in a concentration-dependent manner by facilitating activation, reducing inactivation, and slowing deactivation (Su et al., 2009). A-935142 also shortened action potential duration (APD90) in canine Purkinje fibers and guinea pig atrial tissue. This study focused on the combined effects of the prototypical hERG enhancer, A-935142 and two hERG current blockers (sotalol and terfenadine). MAIN METHODS The whole-cell voltage clamp method with HEK 293 cells heterologously expressing the hERG channel (Kv 11.1) was used. KEY FINDINGS A-935142 did not compete with 3H-dofetilide binding, suggesting that A-935142 does not overlap the binding site of typical hERG blockers. In whole-cell voltage clamp studies we found: 1) 60 μM A-935142 enhanced hERG current in the presence of 150 μM sotalol (57.5±5.8%) to a similar extent as seen with A-935142 alone (55.6±5.1%); 2) 150 μM sotalol blocked hERG current in the presence of 60 μM A-935142 (43.5±1.5%) to a similar extent as that seen with sotalol alone (42.0±3.2%) and 3) during co-application, hERG current enhancement was followed by current blockade. Similar results were obtained with 60 nM terfenadine combined with A-935142. SIGNIFICANCE These results suggest that the hERG enhancer, A-935142 does not compete with these two known hERG blockers at their binding site within the hERG channel. This selective hERG current enhancement may be useful as a treatment for inherited or acquired LQTS (Casis et al., 2006).


Biophysical Journal | 2011

Drug-Induced hERG Current Enhancement is Modulated by Extracellular Proton and Potassium

Zhi Su; Xiaoqin Liu; Jian Wu; Kathryn Houseman; James T. Limberis; Brian Padden; Ruth L. Martin; Bryan F. Cox; Gary A. Gintant

Acid-base disturbances and hypokalemia or hyperkalemia are frequently encountered in a variety of clinical scenarios. Extracellular proton and potassium concentrations are important modulators of hERG channel functions and biophysical properties such as deactivation and inactivation. Slowing deactivation and inactivation kinetics of hERG channel are two important components for drug-induced hERG current enhancement. This study examined if drug-induced hERG current enhancement is affected by acid-base and electrolyte aberrations at physiological temperature (37° C). Whole-cell voltage clamp technique was used to measure hERG current (step-ramp protocol: initial 1-sec depolarization step to 0 mV from −80 mV (VH) followed by a 2-sec repolarization ramp back to VH) and a typical hERG activator A-935142 was used as a tool drug. Drug-induced hERG current enhancement was potentiated by extracellular acidification (% increase for step current: 132 ± 5 vs. 58.4 ± 4.2, pH 6.8 vs. 7.4, P < 0.01; % increase for tail current: 115 ± 3 vs. 43.2 ± 3.2, P < 0.01). In contrast, alkalosis (pH 8.4) weakened the drug-induced hERG current enhancement (% increase for step: 30.3 ± 3.1 vs. 58.4 ± 4.2, pH 8.4 vs 7.4, P < 0.01; % increase for tail: 21.9 ± 2.3 vs. 43.2 ± 3.2, P < 0.01). The measured bath drug concentrations were the same at normal pH, acidosis, and alkalosis. Hypokalemia (1 mM K+) did not affect drug-induced hERG current enhancement but hyperkalemia (10 mM K+) attenuated the drug-induced hERG current enhancement (increase for step: 41.1 ± 1.6% vs. 58.4 ± 4.2%, 10 mM vs. 5 mM K+, P < 0.01; increase for tail: 31.5 ± 1.3 % vs. 43.2 ± 3.2 %, P < 0.05). These results demonstrate that hERG current enhancement by A-935142 is modulated by extracellular proton concentrations and hyperkalemia.


Biophysical Journal | 2009

Electrophysiologic Characterization of a Complex hERG Channel Activator

Zhi Su; James T. Limberis; Andrew J. Souers; Philip R. Kym; Ann Mikhail; Kathryn Houseman; Gilbert Diaz; Ruth L. Martin; Bryan F. Cox; Gary A. Gintant

Novel and specific activators of the human ether-a-go-go-related gene (hERG) K+ channel have been reported recently to enhance hERG current amplitude (4 synthetic small molecules and one naturally occurring substance). Here, we characterize the effects of a novel compound (Abbott-x) on atrial and ventricular action potentials (using microelectrode techniques) and cloned hERG channels stably expressed in HEK-293 cells (using whole cell patch clamp techniques). Abbott-x shortened cardiac action potentials and enhanced the amplitude of the hERG current in a concentration- and voltage-dependent manner. The fully activated current-voltage relationship revealed that this compound (60 uM) increased both outward and inward K+ current. The slope conductance of the linear portion of the fully activated I-V relation was increased in the presence of the compound. Abbott-x significantly reduced the time constants (τ) of hERG channel activation at two example voltages tested (-10 mV: τ =100 ± 17 vs 164 ± 24 ms, n = 6, P < 0.01; +30 mV: τ = 16.7 ± 1.8 vs 18.9 ± 1.8, n = 5, P < 0.05) and shifted the voltage-dependence for hERG activation in the hyperpolarizing direction by 9 mV (n = 7, P < 0.01). The time course of hERG channel deactivation was significantly slowed at multiple potentials tested (−120 to −70 mV). Abbott-x also significantly reduced the rate of inactivation and shifted the voltage dependence of inactivation in the depolarizing direction by 15 mV (n = 5, P < 0.05). Recovery of hERG channel from inactivation was not significantly affected by Abbott-x. In conclusion, Abbott-x enhances hERG current in a complex manner by facilitation of activation, reduction of inactivation, and slowing of deactivation, and abbreviates atrial and ventricular repolarization.


Journal of Medicinal Chemistry | 1993

Discovery of a novel class of potent HIV-1 protease inhibitors containing the (R)-(hydroxyethyl)urea isostere.

Daniel P. Getman; Gary A Decrescenzo; Robert M. Heintz; Reed Kl; John J. Talley; Martin L. Bryant; Michael Clare; Kathryn Houseman; Marr Jj; Richard A. Mueller


Journal of Medicinal Chemistry | 1991

Effect of hydroxyl group configuration in hydroxyethylamine dipeptide isosteres on HIV protease inhibition. Evidence for multiple binding modes.

Daniel H. Rich; Chong Qing Sun; J. V. N. Vara Prasad; Ahammadunny Pathiasseril; Mihaly V. Toth; Garland R. Marshall; Michael Clare; Richard A. Mueller; Kathryn Houseman


Journal of Medicinal Chemistry | 1995

Inhibitors of HIV-1 Protease Containing the Novel and Potent (R)-(Hydroxyethyl)sulfonamide Isostere

Michael L. Vazquez; Martin L. Bryant; Michael Clare; Gary A Decrescenzo; Elizabeth M. Doherty; John N. Freskos; Daniel P. Getman; Kathryn Houseman; Janet Julien; Geralyn P. Kocan; Richard A. Mueller; Huey-Sheng Shieh; William C. Stallings; Roderick A. Stegeman; John J. Tilley


Journal of Medicinal Chemistry | 1992

New hydroxyethylamine HIV protease inhibitors that suppress viral replication

Daniel H. Rich; J. V. N. Vara Prasad; Chong Qing Sun; Jeremy Green; Richard Mueller; Kathryn Houseman; Debra A. MacKenzie; Miroslav Malkovsky


Journal of Medicinal Chemistry | 2007

1,4-Dihydroindeno[1,2-c ]pyrazoles with acetylenic side chains as novel and potent multitargeted receptor tyrosine kinase inhibitors with low affinity for the hERG ion channel

Jurgen Dinges; Daniel H. Albert; Lee D. Arnold; Kimba L. Ashworth; Irini Akritopoulou-Zanze; Peter F. Bousquet; Jennifer J. Bouska; George A. Cunha; Steven K. Davidsen; Gilbert Diaz; Stevan W. Djuric; Alan F. Gasiecki; Gary A. Gintant; Vijaya Gracias; Christopher M. Harris; Kathryn Houseman; Charles W. Hutchins; Eric F. Johnson; Hu Li; Patrick A. Marcotte; Ruth L. Martin; Michael R. Michaelides; Michelle Nyein; Thomas J. Sowin; Zhi Su; Paul Tapang; Zhiren Xia; Henry Q. Zhang


Journal of Pharmacological and Toxicological Methods | 2011

Ventricular rate adaptation: A novel, rapid, cellular-based in-vitro assay to identify proarrhythmic and torsadogenic compounds

Jonathon Green; Gilbert Diaz; James T. Limberis; Kathryn Houseman; Zhi Su; Ruth L. Martin; Bryan F. Cox; Stan Kantor; Gary A. Gintant

Collaboration


Dive into the Kathryn Houseman's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gilbert Diaz

TAP Pharmaceutical Products

View shared research outputs
Top Co-Authors

Avatar

Daniel H. Rich

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

J. V. N. Vara Prasad

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Chong Qing Sun

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Philip R. Kym

University of Texas at Austin

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge