UB Vice President for Research and Economic Development markup
Glenna C Bett UB Profile
Obstetrics and Gynecology
Associate Professor
133 Sherman Hall
Buffalo NY 14214
716-829-2603
Export
Publication Types
Co-Authors
  • More Coauthors +
 
Authors: Leigh Korbel, Randall Rasmusson, G. Bett
Journal-article: Biophysical Journal, vol. 118, pp. 591a, 2020
Authors: Brian K. Panama, Mark W. Nowak, Brandon Franks, Leigh Korbel, Sanjot Singh, Glenna C.L. Bett, Randall L. Rasmusson
Journal-article: Journal of Pharmacological and Toxicological Methods, vol. 99, pp. 106595, 2019
Authors: Mark W. Nowak, Brian K. Panama, Sanjot Singh, Qiuming Gong, Randall L. Rasmusson, Zhengfeng Zhou, Glenna C.L. Bett
Journal-article: Journal of Pharmacological and Toxicological Methods, vol. 99, pp. 106595, 2019
Authors: Brandon Franks, Glenna C.L. Bett, Randall Rasmusson
Journal-article: Biophysical Journal, vol. 114, pp. 292a-293a, 2018
Authors: Mark W. Nowak, Brian K. Panama, Sanjot Singh, Brandon Franks, Glenna C.L. Bett, Randall L. Rasmusson
Journal-article: Biophysical Journal, vol. 114, pp. 670a, 2018
Authors: Dongrui Ma, Zhenfeng Liu, Li Jun Loh, Yongxing Zhao, Guang Li, Reginald Liew, Omedul Islam, Jianjun Wu, Ying Ying Chung, Wee Siong Teo, Chi Keong Ching, Boon Yew Tan, Daniel Chong, Kah Leng Ho, Paul Lim, Rita Yu Yin Yong, Brian K. Panama, Aaron D. Kaplan, Glenna C. L. Bett, James Ware, Connie R. Bezzina, Arie O. Verkerk, Stuart A. Cook, Randall L. Rasmusson, Heming Wei
Journal-article: Scientific Reports, vol. 8, 2018
Authors: Brian K. Panama, Mark W. Nowak, Sanjot Singh, Randall L. Rasmusson, Glenna C.L. Bett
Journal-article: Biophysical Journal, vol. 114, pp. 624a, 2018
Authors: Mark W. Nowak, Brian K. Panama, Qiuming Gong, Sanjot Singh, Randall Rasmusson, Zhengfeng Zhou, Glenna C.L. Bett
Journal-article: Biophysical Journal, vol. 114, pp. 292a, 2018
Authors: Rita Papp, Glenna C. L. Bett, Agnieszka Lis, Randall L. Rasmusson, Istv??n Baczk??, Andr??s Varr??, Guy Salama
Keywords: 17--estradiol L-type Ca2+ channel Regional heterogeneities in the heart Sex differences in arrhythmia phenotype Sex differences in the human heart Sodium-calcium exchanger
Abstract: Women have a higher risk of lethal arrhythmias than men in long QT syndrome type 2 (LQTS2), but the mechanisms remain uncertain due to the limited availability of healthy control human tissue. We have previously reported that in female rabbits, estrogen increases arrhythmia risk in drug-induced LQTS2 by upregulating L-type Ca2+ (ICa,L) and sodium-calcium exchange (INCX) currents at the base of the epicardium by a genomic mechanism. This study investigates if the effects of estrogen on rabbit ICa,L and INCX apply to human hearts. Postmortem human left ventricular tissue samples were probed with selective antibodies for regional heterogeneities of ion channel protein expression and compared to rabbit myocardium. Functionally, ICa,L and INCX were measured from female and male cardiomyocytes derived from human induced pluripotent stem cells (iPS-CMs) with the voltage-clamp technique from control and estrogen-treated iPS-CMs. In women (n=12), Cav1.2 (primary subunit of the L-type calcium channel protein 1) and NCX1 (sodium-calcium exchange protein) levels were higher at the base than apex of the epicardium (4014 and 8130%, respectively, P<0.05), but not in men (n=6) or postmenopausal women (n=6). Similarly, in cardiomyocytes derived from female human iPS-CMs, estrogen (1nM, 1-2days) increased ICa,L (31%, P<0.05) and INCX (7.5-fold, -90mV, P<0.01) and their mRNA levels (P<0.05). Moreover, in male human iPS-CMs, estrogen failed to alter ICa,L and INCX. The results show that estrogen upregulates cardiac ICa,L and INCX in women through genomic mechanisms that account for sex differences in Ca2+ handling and spatial heterogeneities of repolarization due to base-apex heterogeneities of Cav1.2 and NCX1. By analogy with rabbit studies, these effects account for human sex-difference in arrhythmia risk.
PubMedId: 28815011
Journal-article: Biology of Sex Differences, vol. 8, 2017
Authors: Mark Nowak, Sanjot Singh, Aidan Coon, Shimin Wang, Glenna Bett, Randall L. Rasmusson
Journal-article: Biophysical Journal, vol. 112, pp. 235a, 2017
Authors: Mark Nowak, Aidan Coon, Sanjot Singh, Shimin Wang, Randall Rasmusson, G. Bett
Journal-article: Biophysical Journal, vol. 112, pp. 37a, 2017
Authors: Emily R. Sekera, Heather L. Rudolph, Stephen D. Carro, Michael J. Morales, Glenna C. L. Bett, Randall L. Rasmusson, Troy D. Wood
Keywords: MALDI autism spectrum disorders biomarker mass spectrometry metabolomics solid-phase extraction
Abstract: Autism spectrum disorders (ASD) are a group of neurodevelopmental disorders lacking a clinical biomarker for diagnosis. Emerging evidence shows that intestinal microflora from ASD subjects can be distinguished from controls, suggesting metabolite differences due to the action of intestinal microbes may provide a means for identifying potential biomarkers for ASD. The aim of this study was to determine if quantitative differences in levels of stercobilin and stercobilinogen, metabolites produced by biological action of intestinal microflora, exist in the fecal matter between an ASD mouse model population and controls. Pairs of fecal samples were collected from two mouse groups, an ASD model group with Timothy syndrome 2 (TS2-NEO) and a gender-matched control group. After centrifugation, supernatant was spiked with an 18O-labeled stercobilin isotopomer and subjected to solid phase extraction for processing. Extracted samples were spotted on a stainless steel plate and subjected to matrix-assisted laser desorption and ionization mass spectrometry using dihydroxybenzoic acid as the matrix (n = 5). Peak areas for bilins and 18O-stercobilin isotopomers were determined in each fecal sample. A 40-45% depletion in stercobilin in TS2-NEO fecal samples compared with controls was observed with p < 0.05; a less dramatic depletion was observed for stercobilinogen. The results show that stercobilin depletion in feces is observed for an ASD mouse model vs. controls. This may help to explain recent observations of a less diverse microbiome in humans with ASD and may prove helpful in developing a clinical ASD biomarker.
PubMedId: 29147105
Journal-article: Metabolomics, vol. 13, 2017
Authors: Aaron D. Kaplan, Glenna C.L. Bett, Randall L. Rasmusson
Journal-article: Biophysical Journal, vol. 110, pp. 30a, 2016
Authors: Glenna??C.L. Bett, Aaron??D. Kaplan, Randall??L. Rasmusson
PubMedId: 26745432
Journal-article: Biophysical Journal, vol. 110, pp. 284-286, 2016
Authors: Aaron D. Kaplan, Agnieszka Lis, Carlos Li, Lei Yang, Michael J. Morales, Glenna C.L. Bett, Randall L. Rasmusson
Journal-article: Biophysical Journal, vol. 108, pp. 271a, 2015
Authors: Aaron D. Kaplan, Glenna C.L. Bett, Randall L. Rasmusson
Journal-article: Biophysical Journal, vol. 108, pp. 112a, 2015
Authors: Glenna C L Bett
Keywords: arrhythmia estrogen heart hypertrophy ion channels repolarization
Abstract: Disruption of cardiac electrical activity resulting in palpitations and syncope is often an early symptom of pregnancy. Pregnancy is a time of dramatic and dynamic physiological and hormonal changes during which numerous demands are placed on the heart. These changes result in electrical remodelling which can be detected as changes in the electrocardiogram (ECG). This gestational remodelling is a very under-researched area. There are no systematic large studies powered to determine changes in the ECG from pre-pregnancy, through gestation, and into the postpartum period. The large variability between patients and the dynamic nature of pregnancy hampers interpretation of smaller studies, but some facts are consistent. Gestational cardiac hypertrophy and a physical shift of the heart contribute to changes in the ECG. There are also electrical changes such as an increased heart rate and lengthening of the QT interval. There is an increased susceptibility to arrhythmias during pregnancy and the postpartum period. Some changes in the ECG are clearly the result of changes in ion channel expression and behaviour, but little is known about the ionic basis for this electrical remodelling. Most information comes from animal models, and implicates changes in the delayed-rectifier channels. However, it is likely that there are additional roles for sodium channels as well as changes in calcium homoeostasis. The changes in the electrical profile of the heart during pregnancy and the postpartum period have clear implications for the safety of pregnant women, but the field remains relatively undeveloped.
PubMedId: 27128800
Journal-article: Clinical science (London, England : 1979), vol. 130, 2015
Authors: Aaron D. Kaplan, Agnieszka Lis, Randall L. Rasmusson, Glenna C.L. Bett
Journal-article: Biophysical Journal, vol. 108, pp. 112a, 2015
Authors: Lu Han, Yang Li, Jason Tchao, Aaron D Kaplan, Bo Lin, You Li, Jocelyn Mich-Basso, Agnieszka Lis, Narmeen Hassan, Barry London, Glenna C L Bett, Kimimasa Tobita, Randall L Rasmusson, Lei Yang
PubMedId: 25691542
Journal-article: Cardiovascular research, vol. 106, 2015
Authors: Aaron D. Kaplan, Agnieszka Lis, Thomas R. Cimato, Emmanuel S. Tzanakakis, Qinlian Zhou, Michael J. Morales, Randall L. Rasmusson, Glenna C.L. Bett
Journal-article: Biophysical Journal, vol. 106, pp. 631a, 2014
Authors: Jonathan M Cordeiro, Tanya Zeina, Robert Goodrow, Aaron D Kaplan, Lini M Thomas, Vladislav V Nesterenko, Jacqueline A Treat, Leo Hawel, Craig Byus, Glenna C Bett, Randall L Rasmusson, Brian K Panama
Keywords: Atria Electrophysiology Potassium channel Purkinje Ventricle
Abstract: The inward rectifier potassium current, IK1, contributes to the terminal phase of repolarization of the action potential (AP), as well as the value and stability of the resting membrane potential. Regional variation in IK1 has been noted in the canine heart, but the biophysical properties have not been directly compared. We examined the properties and functional contribution of IK1 in isolated myocytes from ventricular, atrial and Purkinje tissue. APs were recorded from canine left ventricular midmyocardium, left atrial and Purkinje tissue. The terminal rate of repolarization of the AP in ventricle, but not in Purkinje, depended on changes in external K(+) ([K(+)]o). Isolated ventricular myocytes had the greatest density of IK1 while atrial myocytes had the lowest. Furthermore, the outward component of IK1 in ventricular cells exhibited a prominent outward component and steep negative slope conductance, which was also enhanced in 10 mM [K(+)]o. In contrast, both Purkinje and atrial cells exhibited little outward IK1, even in the presence of 10 mM [K(+)]o, and both cell types showed more persistent current at positive potentials. Expression of Kir2.1 in the ventricle was 76.9-fold higher than that of atria and 5.8-fold higher than that of Purkinje, whereas the expression of Kir2.2 and Kir2.3 subunits was more evenly distributed in Purkinje and atria. Finally, AP clamp data showed distinct contributions of IK1 for each cell type. IK1 and Kir2 subunit expression varies dramatically in regions of the canine heart and these regional differences in Kir2 expression likely underlie regional distinctions in IK1 characteristics, contributing to variations in repolarization in response to in [K(+)]o changes.
PubMedId: 25889894
Journal-article: Journal of molecular and cellular cardiology, vol. 84, 2014
Authors: Daniel A Gold, Aaron D Kaplan, Agnieszka Lis, Glenna C L Bett, Emily E Rosowski, Kimberly M Cirelli, Alexandre Bougdour, Saima M Sidik, Josh R Beck, Sebastian Lourido, Pascal F Egea, Peter J Bradley, Mohamed-Ali Hakimi, Randall L Rasmusson, Jeroen P J Saeij
Abstract: Toxoplasma gondii is a protozoan pathogen in the phylum Apicomplexa that resides within an intracellular parasitophorous vacuole (PV) that is selectively permeable to small molecules through unidentified mechanisms. We have identified GRA17 as a Toxoplasma-secreted protein that localizes to the parasitophorous vacuole membrane (PVM) and mediates passive transport of small molecules across the PVM. GRA17 is related to the putative Plasmodium translocon protein EXP2 and conserved across PV-residing Apicomplexa. The PVs of GRA17-deficient parasites have aberrant morphology, reduced permeability to small molecules, and structural instability. GRA17-deficient parasites proliferate slowly and are avirulent in mice. These GRA17-deficient phenotypes are rescued by complementation with Plasmodium EXP2. GRA17 functions synergistically with a related protein, GRA23. Exogenous expression of GRA17 or GRA23 alters the membrane conductance properties of Xenopus oocytes in a manner consistent with a large non-selective pore. Thus, GRA17 and GRA23 provide a molecular basis for PVM permeability and nutrient access.
PubMedId: 25974303
Journal-article: Cell host & microbe, vol. 17, 2014
Authors: Bo Lin, Yang Li, Lu Han, Aaron D Kaplan, Ying Ao, Spandan Kalra, Glenna C L Bett, Randall L Rasmusson, Chris Denning, Lei Yang
Keywords: Dilated cardiomyopathy Duchenne muscular dystrophy Induced pluripotent stem cells
Abstract: Duchenne muscular dystrophy (DMD) is caused by mutations in the dystrophin gene (DMD), and is characterized by progressive weakness in skeletal and cardiac muscles. Currently, dilated cardiomyopathy due to cardiac muscle loss is one of the major causes of lethality in late-stage DMD patients. To study the molecular mechanisms underlying dilated cardiomyopathy in DMD heart, we generated cardiomyocytes (CMs) from DMD and healthy control induced pluripotent stem cells (iPSCs). DMD iPSC-derived CMs (iPSC-CMs) displayed dystrophin deficiency, as well as the elevated levels of resting Ca(2+), mitochondrial damage and cell apoptosis. Additionally, we found an activated mitochondria-mediated signaling network underlying the enhanced apoptosis in DMD iPSC-CMs. Furthermore, when we treated DMD iPSC-CMs with the membrane sealant Poloxamer 188, it significantly decreased the resting cytosolic Ca(2+) level, repressed caspase-3 (CASP3) activation and consequently suppressed apoptosis in DMD iPSC-CMs. Taken together, using DMD patient-derived iPSC-CMs, we established an in vitro model that manifests the major phenotypes of dilated cardiomyopathy in DMD patients, and uncovered a potential new disease mechanism. Our model could be used for the mechanistic study of human muscular dystrophy, as well as future preclinical testing of novel therapeutic compounds for dilated cardiomyopathy in DMD patients.
PubMedId: 25791035
Journal-article: Disease models & mechanisms, vol. 8, 2014
Authors: Jong J Kim, Lei Yang, Bo Lin, Xiaodong Zhu, Bin Sun, Aaron D Kaplan, Glenna C L Bett, Randall L Rasmusson, Barry London, Guy Salama
Keywords: Cellcell coupling Funny current Human myocytes from stem cells Optical mapping of calcium and action potentials Spontaneous activity Subcellular calcium waves
Abstract: The creation of cardiomyocytes derived from human induced pluripotent stem cells (hiPS-CMs) has spawned broad excitement borne out of the prospects to diagnose and treat cardiovascular diseases based on personalized medicine. A common feature of hiPS-CMs is their spontaneous contractions but the mechanism(s) remain uncertain. Intrinsic activity was investigated by the voltage-clamp technique, optical mapping of action potentials (APs) and intracellular Ca(2+) (Cai) transients (CaiT) at subcellular-resolution and pharmacological interventions. The frequency of spontaneous CaiT (sCaiT) in monolayers of hiPS-CMs was not altered by ivabradine, an inhibitor of the pacemaker current, If despite high levels of HCN transcripts (1-4). HiPS-CMs had negligible If and IK1 (inwardly-rectifying K(+)-current) and a minimum diastolic potential of -59.13.3mV (n=18). APs upstrokes were preceded by a depolarizing-foot coincident with a rise of Cai. Subcellular Cai wavelets varied in amplitude, propagated and died-off; larger Cai-waves triggered cellular sCaTs and APs. SCaiTs increased in frequency with [Ca(2+)]out (0.05-to-1.8mM), isoproterenol (1M) or caffeine (100M) (n5, p<0.05). HiPS-CMs became quiescent with ryanodine receptor stabilizers (K201=2M); tetracaine; Na-Ca exchange (NCX) inhibition (SEA0400=2M); higher [K(+)]out (58mM), and thiol-reducing agents but could still be electrically stimulated to elicit CaiTs. Cell-cell coupling of hiPS-CM in monolayers was evident from connexin-43 expression and CaiT propagation. SCaiTs from an ensemble of dispersed hiPS-CMs were out-of-phase but became synchronous through the outgrowth of inter-connecting microtubules. Automaticity in hiPS-CMs originates from a Ca(2+)-clock mechanism involving Ca(2+) cycling across the sarcoplasmic reticulum linked to NCX to trigger APs.
PubMedId: 25644533
Journal-article: Journal of molecular and cellular cardiology, vol. 81, 2014
Authors: Lu Han, Yang Li, Jason Tchao, Aaron D Kaplan, Bo Lin, You Li, Jocelyn Mich-Basso, Agnieszka Lis, Narmeen Hassan, Barry London, Glenna C L Bett, Kimimasa Tobita, Randall L Rasmusson, Lei Yang
Keywords: Cardiomyocyte Heart Hypertrophic cardiomyopathy Induced pluripotent stem cells
Abstract: Familial hypertrophic cardiomyopathy (HCM) is one the most common heart disorders, with gene mutations in the cardiac sarcomere. Studying HCM with patient-specific induced pluripotent stem-cell (iPSC)-derived cardiomyocytes (CMs) would benefit the understanding of HCM mechanism, as well as the development of personalized therapeutic strategies. To investigate the molecular mechanism underlying the abnormal CM functions in HCM, we derived iPSCs from an HCM patient with a single missense mutation (Arginine442Glycine) in the MYH7 gene. CMs were next enriched from HCM and healthy iPSCs, followed with whole transcriptome sequencing and pathway enrichment analysis. A widespread increase of genes responsible for 'Cell Proliferation' was observed in HCM iPSC-CMs when compared with control iPSC-CMs. Additionally, HCM iPSC-CMs exhibited disorganized sarcomeres and electrophysiological irregularities. Furthermore, disease phenotypes of HCM iPSC-CMs were attenuated with pharmaceutical treatments. Overall, this study explored the possible patient-specific and mutation-specific disease mechanism of HCM, and demonstrates the potential of using HCM iPSC-CMs for future development of therapeutic strategies. Additionally, the whole methodology established in this study could be utilized to study mechanisms of other human-inherited heart diseases.
PubMedId: 25209314
Journal-article: Cardiovascular research, vol. 104, 2014
Authors: Guy Salama, Glenna C L Bett
Keywords: action potential arrhythmia estradiol hormones ion channels
Abstract: Sexual dimorphism is a well-established phenomenon, but its degree varies tremendously among species. Since the early days of Einthoven's development of the three-lead galvanometer ECG, we have known there are marked differences in QT intervals of men and women. It required over a century to appreciate the profound implications of sex-based electrophysiological differences in QT interval on the panoply of sex differences with respect to arrhythmia risk, drug sensitivity, and treatment modalities. Little is known about the fundamental mechanism responsible for sex differences in electrical substrate of the human heart, in large part due to the lack of tissue availability. Animal models are an important research tool, but species differences in the sexual dimorphism of the QT interval, the ionic currents underlying the cardiac repolarization, and effects of sex steroids make it difficult to interpolate animal to human sex differences. In addition, in some species, different strains of the same animal model yield conflicting data. Each model has its strengths, such as ease of genetic manipulation in mice or size in dogs. However, many animals do not reproduce the sexual dimorphism of QT seen in humans. To match sex linked prolongation of QT interval and arrhythmogenic phenotype, the current data suggest that the rabbit may be best suited to provide insight into sex differences in humans. In the future, emerging technologies such as induced pluripotent stem cell derived cardiac myocyte systems may offer the opportunity to study sex differences in a controlled hormonal situation in the context of a sex specific human model system.
PubMedId: 24973386
Journal-article: American journal of physiology. Heart and circulatory physiology, vol. 307, 2014
Authors: Glenna Bett
PubMedId: 24901014
1: Journal of genital system & disorders, vol. 1, 2014
Authors: Hong Guo, Agnieszka Lis, Glenna C.L. Bett, Randall L. Rasmusson
Journal-article: Biophysical Journal, vol. 106, pp. 740a-741a, 2014
Authors: Vanessa M. Barnabei, Randall L. Rasmusson, Glenna C.L. Bett
PubMedId: 24440564
Journal-article: American Journal of Obstetrics and Gynecology, vol. 210, pp. 494-495, 2014
Authors: Ashish Parikh, Divyang Patel, Charles F McTiernan, Wenyu Xiang, Jamie Haney, Lei Yang, Bo Lin, Aaron D Kaplan, Glenna C L Bett, Randall L Rasmusson, Sanjeev G Shroff, David Schwartzman, Guy Salama
Keywords: INa upregulation atrial fibrillation fibrosis hypertrophy optical mapping, action potential relaxin spontaneously hypertensive rats
Abstract: Atrial fibrillation (AF) contributes significantly to morbidity and mortality in elderly and hypertensive patients and has been correlated to enhanced atrial fibrosis. Despite a lack of direct evidence that fibrosis causes AF, reversal of fibrosis is considered a plausible therapy. To evaluate the efficacy of the antifibrotic hormone relaxin (RLX) in suppressing AF in spontaneously hypertensive rats (SHR). Normotensive Wistar-Kyoto (WKY) and SHR were treated for 2 weeks with vehicle (WKY+V and SHR+V) or RLX (0.4 mg/kg per day, SHR+RLX) using implantable mini-pumps. Hearts were perfused, mapped optically to analyze action potential durations, intracellular Ca transients, and restitution kinetics, and tested for AF vulnerability. SHR hearts had slower conduction velocity (CV; P<0.01 versus WKY), steeper CV restitution kinetics, greater collagen deposition, higher levels of transcripts for transforming growth factor-, metalloproteinase-2, metalloproteinase-9, collagen I/III, and reduced connexin 43 phosphorylation (P<0.05 versus WKY). Programmed stimulation triggered sustained AF in SHR (n=5/5) and SHR+V (n=4/4), but not in WKY (n=0/5) and SHR+RLX (n=1/8; P<0.01). RLX treatment reversed the transcripts for fibrosis, flattened CV restitution kinetics, reduced action potential duration at 90% recovery to baseline, increased CV (P<0.01), and reversed atrial hypertrophy (P<0.05). Independent of antifibrotic actions, RLX (0.1 mol/L) increased Na current density, INa (2-fold in 48 hours) in human cardiomyocytes derived from inducible pluripotent stem cells (n=18/18; P<0.01). RLX treatment suppressed AF in SHR hearts by increasing CV from a combination of reversal of fibrosis and hypertrophy and by increasing INa. The study provides compelling evidence that RLX may provide a novel therapy to manage AF in humans by reversing fibrosis and hypertrophy and by modulating cardiac ionic currents.
PubMedId: 23748429
Journal-article: Circulation research, vol. 113, 2013
Authors: Glenna Bett, Qinlian Zhou, Mimi Liu, Hong Guo, Agnieszka Lis, Randall L. Rasmusson
Journal-article: Biophysical Journal, vol. 104, pp. 126a, 2013
Authors: Glenna C L Bett, Aaron D Kaplan, Agnieszka Lis, Thomas R Cimato, Emmanuel S Tzanakakis, Qinlian Zhou, Michael J Morales, Randall L Rasmusson
Keywords: AP APD Arrhythmia DAD EAD Heart I(Ca) I(K1) I(K1,Synthetic) I(Na) I(f) I(to) Potassium channel Repolarization Stem cell TS Timothy syndrome action potential action potential duration calcium current dV/dt(max) delayed afterdepolarization early afterdepolarization h-iPSC human-induced pluripotent stem cell iPSC induced pluripotent stem cell inward rectifier potassium current maximum rate of change of voltage with time pacemaker current sodium current synthetic I(K1) transient outward potassium current
Abstract: Human-induced pluripotent stem cell (h-iPSC)-derived cardiac myocytes are a unique model in which human myocyte function and dysfunction are studied, especially those from patients with genetic disorders. They are also considered a major advance for drug safety testing. However, these cells have considerable unexplored potential limitations when applied to quantitative action potential (AP) analysis. One major factor is spontaneous activity and resulting variability and potentially anomalous behavior of AP parameters. To demonstrate the effect of using an in silico interface on electronically expressed I(K1), a major component lacking in h-iPSC-derived cardiac myocytes. An in silico interface was developed to express synthetic I(K1) in cells under whole-cell voltage clamp. Electronic I(K1) expression established a physiological resting potential, eliminated spontaneous activity, reduced spontaneous early and delayed afterdepolarizations, and decreased AP variability. The initiated APs had the classic rapid upstroke and spike and dome morphology consistent with data obtained with freshly isolated human myocytes as well as the readily recognizable repolarization attributes of ventricular and atrial cells. The application of 1 M of BayK-8644 resulted in anomalous AP shortening in h-iPSC-derived cardiac myocytes. When I(K1) was electronically expressed, BayK-8644 prolonged the AP, which is consistent with the existing results on native cardiac myocytes. The electronic expression of I(K1) is a simple and robust method to significantly improve the physiological behavior of the AP and electrical profile of h-iPSC-derived cardiac myocytes. Increased stability enables the use of this preparation for a controlled quantitative analysis of AP parameters, for example, drug responsiveness, genetic disorders, and dynamic behavior restitution profiles.
PubMedId: 24055949
Journal-article: Heart rhythm, vol. 10, 2013
Authors: Qinlian Zhou, Agnieszka Lis, G. Bett
Journal-article: Biophysical Journal, vol. 104, pp. 264a, 2013
Authors: Glenna C.L. Bett, Mateen Haroon, Beomsu Kim, Armando Arroyo, Randall L. Rasmusson, Michael J. Morales
Journal-article: Biophysical Journal, vol. 104, pp. 163a, 2013
Authors: Agniezka Lis, Thomas J. Cimato, Glenna C.L. Bett, Randall L. Rasmusson, Michael J. Morales
Journal-article: Biophysical Journal, vol. 102, pp. 674a, 2012
Authors: Sarah Crimmins, Glenna Bett, Shailini Singh, Armando Arroyo
Journal-article: American Journal of Obstetrics and Gynecology, vol. 206, pp. S271, 2012
Authors: Agnieszka Lis, Glenna C.L. Bett
Journal-article: Biophysical Journal, vol. 102, pp. 328a, 2012
Authors: Glenna CL. Bett, , Agnieszka Lis, Scott R. Wersinger, Joan S. Baizer, Michael E. Duffey, Randall L. Rasmusson, , , , ,
Journal-article: American Chinese Journal of Medicine and Science, vol. 5, pp. 135, 2012
Authors: Glenna C L Bett, Agnieszka Lis, Hong Guo, MiMi Liu, Qinlian Zhou, Randall L Rasmusson
Abstract: Several voltage-gated channels share a proline-valine-proline (proline hinge) sequence motif at the intracellular side of S6. We studied the proline hinge in Kv1.4 channels, which inactivate via two mechanisms: N- and C-type. We mutated the second proline to glycine or alanine: P558A, P558G. These mutations were studied in the presence/absence of the N-terminal to separate the effects of the interaction between the proline hinge and N- and C-type inactivation. Both S6 mutations slowed or removed N- and C-type inactivation, and altered recovery from inactivation. P558G slowed activation and N- and C-type inactivation by nearly an order of magnitude. Sensitivity to extracellular acidosis and intracellular quinidine binding remained, suggesting that transmembrane communication in N- and C-type inactivation was preserved, consistent with our previous findings of major structural rearrangements involving S6 during C-type inactivation. P558A was very disruptive: activation was slowed by more than an order of magnitude, and no inactivation was observed. These results are consistent with our hypothesis that the proline hinge and intracellular S6 movement play a significant role in inactivation and recovery. Computer modeling suggests that both P558G and P558A mutations modify early voltage-dependent steps and make a final voltage-insensitive step that is rate limiting at positive potentials.
PubMedId: 23062336
Journal-article: Biophysical journal, vol. 103, 2012
Authors: Qinlian Zhou, Glenna C L Bett, Randall L Rasmusson
Abstract: The fast component of the cardiac transient outward current, I(Ktof), is blocked by a number of drugs. The major molecular bases of I(Ktof) are Kv4.2/Kv4.3 voltage-gated potassium channels. Drugs with similar potencies but different blocking mechanisms have differing effects on action potential duration (APD). We used in silico analysis to determine the effect of I(Ktof)-blocking drugs with different blocking mechanisms on mouse ventricular myocytes. We used our existing mouse model of the action potential, and developed 4 new Markov formulations for I(Ktof), I(Ktos), I(Kur), I(Ks). We compared effects of theoretical I(Ktof)-specific channel blockers: (1) a closed state, and (2) an open channel blocker. At concentrations lower or close to IC(50), the drug which bound to the open state always had a much greater effect on APD than the drug which bound to the closed state. At concentrations much higher than IC(50), both mechanisms had similar effects at very low pacing rates. However, an open state binding drug had a greater effect on APD at faster pacing rates, particularly around 10 Hz. In summary, our data indicate that drug effects on APD are strongly dependent not only on IC(50), but also on the drug binding state.
PubMedId: 22879935
Journal-article: PloS one, vol. 7, 2012
Authors: Sarah Crimmins, Qinlian Zhou, Glenna Bett
Journal-article: American Journal of Obstetrics and Gynecology, vol. 206, pp. S111-S112, 2012
Authors: G C Bett, F Sachs
Abstract: Mechanosensitivity is a ubiquitous property of cells, and mechanosensitive ion channels (MSCs) are hypothesized to be the transducers. In the heart, MSCs are likely to account for changes in beating rate as a function of filling and for initiating stretch-induced arrhythmias (for example, following a myocardial infarction). Pharmacological agents that affect MSCs may provide a new class of antiarrhythmic drugs. 1997, Elsevier Science Inc. (Trends Cardiovasc Med 1997;7:4-8).
PubMedId: 21235857
Journal-article: Trends in cardiovascular medicine, vol. 7, 2011
Authors: Svetlana Blitshteyn, Hasti Poya, Glenna C L Bett
Abstract: Postural tachycardia syndrome (POTS), a disorder of the autonomic nervous system, predominantly occurs in women of child-bearing age. We set out to determine the clinical course and maternal and fetal outcomes in pregnant women with pre-existing POTS. Participants were asked to complete a detailed questionnaire assessing the clinical course of POTS before, during and after pregnancy, as well as complications of pregnancy, labor and delivery and fetal outcomes. Among 10 women with pre-existing POTS (pregnancy age 28 7 years, range 16-39), with a total of 17 live births, two were complicated by pre-eclampsia, 14 were normal vaginal deliveries and three were C-sections. The rate of severe vomiting or hyperemesis gravidarum in the first trimester was 59%. There were no stillbirths or congenital abnormalities. The average birth weight was 3076 733 grams, with two infants born premature. During pregnancy, POTS symptoms were either improved or stable in six of 10 women, and four of these six women utilized medications for POTS. Six months after delivery, POTS symptoms were improved in three, stable in two and worsened in five women compared to before pregnancy. POTS may have a variable clinical course in pregnancy, with 60% of women reporting either improved or stable symptoms during pregnancy, and 50% of women reporting either improved or stable symptoms 6 months after delivery. There may be a higher rate of severe vomiting in the first trimester in women with POTS than in general population.
PubMedId: 22185354
Journal-article: The journal of maternal-fetal & neonatal medicine : the official journal of the European Association of Perinatal Medicine, the Federation of Asia and Oceania Perinatal Societies, the International Society of Perinatal Obstetricians, vol. 25, 2011
Authors: Patrick L Bader, Mehrdad Faizi, Leo H Kim, Scott F Owen, Michael R Tadross, Ronald W Alfa, Glenna C L Bett, Richard W Tsien, Randall L Rasmusson, Mehrdad Shamloo
Abstract: Autism and autism spectrum disorder (ASD) typically arise from a mixture of environmental influences and multiple genetic alterations. In some rare cases, such as Timothy syndrome (TS), a specific mutation in a single gene can be sufficient to generate autism or ASD in most patients, potentially offering insights into the etiology of autism in general. Both variants of TS (the milder TS1 and the more severe TS2) arise from missense mutations in alternatively spliced exons that cause the same G406R replacement in the Ca(V)1.2 L-type calcium channel. We generated a TS2-like mouse but found that heterozygous (and homozygous) animals were not viable. However, heterozygous TS2 mice that were allowed to keep an inverted neomycin cassette (TS2-neo) survived through adulthood. We attribute the survival to lowering of expression of the G406R L-type channel via transcriptional interference, blunting deleterious effects of mutant L-type channel overactivity, and addressed potential effects of altered gene dosage by studying Ca(V)1.2 knockout heterozygotes. Here we present a thorough behavioral phenotyping of the TS2-neo mouse, capitalizing on this unique opportunity to use the TS mutation to model ASD in mice. Along with normal general health, activity, and anxiety level, TS2-neo mice showed markedly restricted, repetitive, and perseverative behavior, altered social behavior, altered ultrasonic vocalization, and enhanced tone-cued and contextual memory following fear conditioning. Our results suggest that when TS mutant channels are expressed at levels low enough to avoid fatality, they are sufficient to cause multiple, distinct behavioral abnormalities, in line with the core aspects of ASD.
PubMedId: 21878566
Journal-article: Proceedings of the National Academy of Sciences of the United States of America, vol. 108, 2011
Authors: Glenna C L Bett, Qinlian Zhou, Randall L Rasmusson
Abstract: HERG (Kv11.1, KCNH2) is a voltage-gated potassium channel with unique gating characteristics. HERG has fast voltage-dependent inactivation, relatively slow deactivation, and fast recovery from inactivation. This combination of gating kinetics makes study of HERG difficult without using mathematical models. Several HERG models have been developed, with fundamentally different organization. HERG is the molecular basis of I(Kr), which plays a critical role in repolarization. We programmed and compared five distinct HERG models. HERG gating cannot be adequately replicated using Hodgkin-Huxley type formulation. Using Markov models, a five-state model is required with three closed, one open, and one inactivated state, and a voltage-independent step between some of the closed states. A fundamental difference between models is the presence/absence of a transition directly from the proximal closed state to the inactivated state. The only models that effectively reproduce HERG data have no direct closed-inactivated transition, or have a closed-inactivated transition that is effectively zero compared to the closed-open transition, rendering the closed-inactivation transition superfluous. Our single-channel model demonstrates that channels can inactivate without conducting with a flickering or bursting open-state. The various models have qualitative and quantitative differences that are critical to accurate predictions of HERG behavior during repolarization, tachycardia, and premature depolarizations.
PubMedId: 21806931
Journal-article: Biophysical journal, vol. 101, 2011
Authors: Glenna C.L. Bett, MiMi Liu, Randall L. Rasmusson
Journal-article: Biophysical Journal, vol. 100, pp. 426a, 2011
Authors: QinLian Zhou, Glenna C.L. Bett
Journal-article: Biophysical Journal, vol. 100, pp. 426a, 2011
Authors: Michael E. Duffey, Glenna C. L. Bett
PubMedId: 22086250
Journal-article: The Journal of Physiology, vol. 589, pp. 5345-5346, 2011
Authors: Armando Arroyo, Beom Su Kim, Amanda Biehl, John Yeh, Glenna C L Bett
Abstract: Regular and timely electrical activity of gonadotrophin-releasing hormone (GnRH) neurons accompanies the pulsatile release of GnRH that plays a central role in regulating fertility. Although transient outward A-type currents (I(A)) have been electrophysiologically identified in GnRH neurons, the molecular identity of the channels that underlie these currents are unknown. Several families of voltage-gated potassium channels can underlie I(A). However, the biophysical properties of I(A) described in previous electrophysiological studies are strongly characteristic of members of the Kv4 family of voltage-gated channels. We, therefore, sought to determine the presence of Kv4 channels in GnRH neurons. We used reverse transcriptase-polymerase chain reaction (RT-PCR) and Western blot analysis to determine whether Kv4 messenger RNA (mRNA) and protein are present in the rat medial preoptic area (MPOA) and median eminence (ME). We used double-label immunohistochemistry to determine whether Kv4 colocalized with GnRH cell bodies in the MPOA and GnRH axons in the ME. Kv4.3 channels co-localized with GnRH in the MPOA but not in the ME. Neither Kv4.2 nor Kv4.1 co-localized with GnRH in either the MPOA or the ME. The electrical activity of GnRH neurons changes dramatically during the estrous cycle. We, therefore, studied the change in Kv4.3 expression in GnRH neurons during the estrous cycle. In the estrus phase, 58.05% of GnRH neurons expressed Kv4.3 compared to 74.48% in diestrus-proestrus rats (P < .05). Our data suggest that Kv4.3 is the major molecular component of I(A) in GnRH neurons, and furthermore that the expression of Kv4.3 changes significantly during the rat estrous cycle.
PubMedId: 20861393
Journal-article: Reproductive sciences (Thousand Oaks, Calif.), vol. 18, 2010
Authors: Glenna C L Bett, Isidore Dinga-Madou, Qinlian Zhou, Vladimir E Bondarenko, Randall L Rasmusson
Abstract: Kv1.4 channels are Shaker-related voltage-gated potassium channels with two distinct inactivation mechanisms. Fast N-type inactivation operates by a ball-and-chain mechanism. Slower C-type inactivation is not so well defined, but involves intracellular and extracellular conformational changes of the channel. We studied the interaction between inactivation mechanisms using two-electrode voltage-clamp of Kv1.4 and Kv1.4N (amino acids 2-146 deleted to remove N-type inactivation) heterologously expressed in Xenopus oocytes. We manipulated C-type inactivation by introducing a lysine-tyrosine point mutation (K532Y, equivalent to Shaker T449Y) that diminishes C-type inactivation. We used experimental data to develop a comprehensive computer model of Kv1.4 channels to determine the interaction between activation and N- and C-type inactivation mechanisms needed to replicate the experimental data. C-type inactivation began at lower voltage preactivated states, whereas N-type inactivation was coupled directly to the open state. A model with distinct N- and C-type inactivated states was not able to reproduce experimental data, and direct transitions between N- and C-type inactivated states were required, i.e., there is coupling between N- and C-type inactivated states. C-type inactivation is the rate-limiting step determining recovery from inactivation, so understanding C-type inactivation, and how it is coupled to N-type inactivation, is critical in understanding how channels act to repetitive stimulation.
PubMedId: 21190652
Journal-article: Biophysical journal, vol. 100, 2010
Authors: Qinlian Zhou, Glenna C L Bett
Abstract: Human ether--go-go-related gene (HERG, Kv11.1, KCNH2) voltage-gated K(+) channels dominate cardiac action potential repolarization. In addition, HERG channels play a role in neuronal and smooth cell excitability as well as cancer pathology. Extracellular pH (pH(o)) is modified during myocardial ischemia, inflammation, and respiratory alkalosis, so understanding the response of HERG channels to changes in pH is of clinical significance. The relationship between pH(o) and HERG channel gating appears complex. Acidification has previously been reported to speed, slow, or have no effect on activation. We therefore undertook comprehensive analysis of the effect of pH(o) on HERG channel activation. HERG channels have unique and complex activation gating characteristics with both voltage-sensitive and voltage-insensitive steps in the activation pathway. Acidosis decreased the activation rate, suppressed peak current, and altered the sigmoidicity of gating near threshold potentials. At positive voltages, where the voltage-insensitive transition is rate limiting, pH(o) modified the voltage-insensitive step with a pK(a) similar to that of histidine. Hill coefficient analysis was incompatible with a coefficient of 1 but was well described by a Hill coefficient of 4. We derived a pH(o)-sensitive term for a five-state Markov model of HERG channel gating. This model demonstrates the mechanism of pH(o) sensitivity in HERG channel activation. Our experimental data and mathematical model demonstrate that the pH(o) sensitivity of HERG channel activation is dominated by the pH(o) sensitivity of the voltage-insensitive step, in a fashion that is compatible with the presence of at least one proton-binding site on each subunit of the channel tetramer.
PubMedId: 20363888
Journal-article: American journal of physiology. Heart and circulatory physiology, vol. 298, 2010
Authors: Derek L. Beahm, Ki-Hyuk Yoo, Glenna C.L. Bett, John M. Canty, James A. Fallavollita, Randall L. Rasmusson
Journal-article: Biophysical Journal, vol. 98, pp. 412a-413a, 2010
Authors: Glenna C.L. Bett, Mimi Liu, Randall L. Rasmusson
Journal-article: Biophysical Journal, vol. 96, pp. 665a, 2009
Authors: Glenna C.L. Bett, Vladimir E. Bondarenko, Randall L. Rasmusson
Journal-article: Biophysical Journal, vol. 96, pp. 665a-666a, 2009
Authors: Mandeep K Brar, Douglas W Laube, Glenna C L Bett
Abstract: The purpose of this study was to determine whether regular quantitative feedback improved medical student performance on the National Board of Medical Examiners Obstetrics and Gynecology subject test. We examined the effect of including regular quantitative feedback (in the form of biweekly quizzes) in the obstetrics and gynecology clerkship on student performance on the National Board of Medical Examiners Obstetrics and Gynecology examination at the University at Buffalo. Students who completed the clerkship format including the regular feedback scored significantly higher than students who completed the clerkship without feedback (70.3 +/- 7.1 vs 68.2 +/- 8.6; P < .005; mean +/- SD; t test). The number of students failing the examination was significantly reduced from 6.39% to 0.47% (chi2 test; P < .001). Introduction of regular quantitative feedback significantly improved student performance on the National Board of Medical Examiners test, and resulted in a significant decrease in the number of students failing the course.
PubMedId: 17980197
Journal-article: American journal of obstetrics and gynecology, vol. 197, 2007
Authors: Glenna C L Bett, Randall L Rasmusson
Abstract: Reconciling ion channel alpha-subunit expression with native ionic currents and their pharmacological sensitivity in target organs has proved difficult. In native tissue, many K(+) channel alpha-subunits co-assemble with ancillary subunits, which can profoundly affect physiological parameters including gating kinetics and pharmacological interactions. In this review, we examine the link between voltage-gated potassium ion channel pharmacology and the biophysics of ancillary subunits. We propose that ancillary subunits can modify the interaction between pore blockers and ion channels by three distinct mechanisms: changes in (1) binding site accessibility; (2) orientation of pore-lining residues; (3) the ability of the channel to undergo post-binding conformational changes. Each of these subunit-induced changes has implications for gating, drug affinity and use dependence of their respective channel complexes. A single subunit may modulate its associated alpha-subunit by more than one of these mechanisms. Voltage-gated potassium channels are the site of action of many therapeutic drugs. In addition, potassium channels interact with drugs whose primary target is another channel, e.g. the calcium channel blocker nifedipine, the sodium channel blocker quinidine, etc. Even when K(+) channel block is the intended mode of action, block of related channels in non-target organs, e.g. the heart, can result in major and potentially lethal side-effects. Understanding factors that determine specificity, use dependence and other properties of K(+) channel drug binding are therefore of vital clinical importance. Ancillary subunits play a key role in determining these properties in native tissue, and so understanding channel-subunit interactions is vital to understanding clinical pharmacology.
PubMedId: 18096604
Journal-article: The Journal of physiology, vol. 586, 2007
Authors: Glenna C L Bett, Michael J Morales, Derek L Beahm, Michael E Duffey, Randall L Rasmusson
Abstract: KCNQ1 (Kv7.1 or KvLQT1) encodes the alpha-subunit of a voltage-gated potassium channel found in tissues including heart, brain, epithelia and smooth muscle. Tissue-specific characteristics of KCNQ1 current are diverse, due to modification by ancillary subunits. In heart, KCNQ1 associates with KCNE1 (MinK), producing a slowly activating voltage-dependent channel. In epithelia, KCNQ1 co-assembles with KCNE3 (Mirp2) producing a constitutively open channel. Chromanol 293B is a selective KCNQ1 blocker. We studied drug binding and frequency dependence of 293B on KCNQ1 and ancillary subunits expressed in Xenopus oocytes. Ancillary subunits altered 293B potency up to 100-fold (IC(50) for KCNQ1 = 65.4 +/- 1.7 microm; KCNQ1/KCNE1 = 15.1 +/- 3.3 microm; KCNQ1/KCNE3 = 0.54 +/- 0.18 microm). Block of KCNQ1 and KCNQ1/KCNE3 was time independent, but 293B altered KCNQ1/KCNE1 activation. We therefore studied frequency-dependent block of KCNQ1/KCNE1. Repetitive rapid stimulation increased KCNQ1/KCNE1 current biphasically, and 293B abolished the slow component. KCNQ1/KCNE3[V72T] activates slowly with a KCNQ1/KCNE1-like phenotype, but retains the high affinity binding of KCNQ1/KCNE3, demonstrating that subunit-mediated changes in gating can be dissociated from subunit-mediated changes in affinity. This study demonstrates the KCNQ1 pharmacology is significantly altered by ancillary subunits. The response of KCNQ1 to specific blockers will therefore be critically dependent on the electrical stimulation pattern of the target organ. Furthermore, the dissociation between gating and overall affinity suggests that mutations in ancillary subunits can potentially strongly alter drug sensitivity without obvious functional changes in gating behaviour, giving rise to unexpected side-effects such as a predisposition to acquired long QT syndrome.
PubMedId: 16887873
Journal-article: The Journal of physiology, vol. 576, 2006
Authors: Shimin Wang, Vladimir E Bondarenko, Yu-jie Qu, Glenna C L Bett, Michael J Morales, Randall L Rasmusson, Harold C Strauss
Abstract: Kv4.3 inactivation is a complex multiexponential process, which can occur from both closed and open states. The fast component of inactivation is modulated by the N-terminus, but the mechanisms mediating the other components of inactivation are controversial. We studied inactivation of Kv4.3 expressed in Xenopus laevis oocytes, using the two-electrode voltage-clamp technique. Inactivation during 2000 ms pulses at potentials positive to the activation threshold was described by three exponents (46 +/- 3, 152 +/- 13, and 930 +/- 50 ms at +50 mV, n = 7) whereas closed-state inactivation (at potentials below threshold) was described by two exponents (1079 +/- 119 and 3719 +/- 307 ms at -40 mV, n = 9). The fast component of open-state inactivation was dominant at potentials positive to -20 mV. Negative to -30 mV, the intermediate and slow components dominated inactivation. Inactivation properties were dependent on pulse duration. Recovery from inactivation was strongly dependent on voltage and pulse duration. We developed an 11-state Markov model of Kv4.3 gating that incorporated a direct transition from the open-inactivated state to the closed-inactivated state. Simulations with this model reproduced open- and closed-state inactivation, isochronal inactivation relationships, and reopening currents. Our data suggest that inactivation can proceed primarily from the open state and that multiple inactivation components can be identified.
PubMedId: 16100281
Journal-article: Biophysical journal, vol. 89, 2005
Authors: Glenna C L Bett, Michael J Morales, Harold C Strauss, Randall L Rasmusson
Abstract: Rapidly activating Kv4 voltage-gated ion channels are found in heart, brain, and diverse other tissues including colon and uterus. Kv4.3 can co-assemble with KChIP ancillary subunits, which modify kinetic behavior. We examined the affinity and use dependence of nifedipine block on Kv4.3 and its modulation by KChIP2b. Nifedipine (150 microM) reduced peak Kv4.3 current approximately 50%, but Kv4.3/KChIP2b current only approximately 27%. Nifedipine produced a very rapid component of open channel block in both Kv4.3 and Kv4.3/KChIP2b. However, recovery from the blocked/inactivated state was strongly sensitive to KChIP2b. Kv4.3 Thalf,recovery was slowed significantly by nifedipine (120.0+/-12.4 ms vs. 213.1+/-18.2 ms), whereas KChIP2b eliminated nifedipine's effect on recovery: Kv4.3/KChIP2b Thalf,recovery was 45.3+/-7.2 ms (control) and 47.8+/-8.2 ms (nifedipine). Consequently, Kv4.3 exhibited use-dependent nifedipine block in response to a series of depolarizing pulses which was abolished by KChIP2b. KChIPs alter drug affinity and use dependence of Kv4.3.
PubMedId: 16414350
Journal-article: Biochemical and biophysical research communications, vol. 340, 2005
Authors: Armando Arroyo, Beomsu Kim, Randall L Rasmusson, Glenna Bett, John Yeh
Abstract: The current research was conducted to determine whether hyperpolarization-activated cyclic nucleotide-gated (HCN1-4) channels are expressed in gonadotropin-releasing hormone (GnRH) neurons in the female rat hypothalamus and immortalized GnRH neurons (GT1-7 cells). Double-label fluorescence immunohistochemistry was used to colocalize HCN1-4 channels and GnRH in GnRH neurons in the female rat hypothalamus. Reverse transcriptase-polymerase chain reaction (RT-PCR), Western blotting, and immunocytochemistry were used to analyze HCN channel gene expression in GT1-7 cells. Double-label fluorescence immunohistochemistry showed that 43% of hypothalamic GnRH neurons immunostained for HCN2 and 90% of GnRH neurons immunostained for HCN3. RT-PCR and Western blot showed expression of all four HCN channel subunits in GT1-7 cells. Double-label immunocytochemistry showed cytoplasmic immunostaining of HCN2 and HCN3 in GT1-7 cells. This study demonstrates for the first time that HCN channels are expressed in GnRH neurons in the rat hypothalamus and GT1-7 cells. Our research supports the hypothesis that HCN channels may be involved in electrical bursting activity and pulsatile GnRH secretion in endogenous GnRH neurons and GT1-7 cells.
PubMedId: 16879992
Journal-article: Journal of the Society for Gynecologic Investigation, vol. 13, 2005
Authors: Vladimir E Bondarenko, Gyula P Szigeti, Glenna C L Bett, Song-Jung Kim, Randall L Rasmusson
Abstract: We have developed a mathematical model of the mouse ventricular myocyte action potential (AP) from voltage-clamp data of the underlying currents and Ca2+ transients. Wherever possible, we used Markov models to represent the molecular structure and function of ion channels. The model includes detailed intracellular Ca2+ dynamics, with simulations of localized events such as sarcoplasmic Ca2+ release into a small intracellular volume bounded by the sarcolemma and sarcoplasmic reticulum. Transporter-mediated Ca2+ fluxes from the bulk cytosol are closely matched to the experimentally reported values and predict stimulation rate-dependent changes in Ca2+ transients. Our model reproduces the properties of cardiac myocytes from two different regions of the heart: the apex and the septum. The septum has a relatively prolonged AP, which reflects a relatively small contribution from the rapid transient outward K+ current in the septum. The attribution of putative molecular bases for several of the component currents enables our mouse model to be used to simulate the behavior of genetically modified transgenic mice.
PubMedId: 15142845
Journal-article: American journal of physiology. Heart and circulatory physiology, vol. 287, 2004
Authors: Glenna C L Bett, Randall L Rasmusson
Abstract: HERG (Human ether--go-go-related gene) potassium channels are crucial for cardiac action potential repolarization. HERG channels are also found in neuronal and tumor cells. The effect of pHo on HERG is of clinical significance because of changes in pH during myocardial ischemia, inflammation, and respiratory alkalosis. We present evidence for the presence of multiple proton binding sites in HERG. Extracellular protons bind rapidly and reversibly to affect both activation and deactivation. However, these effects occur in two distinct pHo ranges. The deactivation rate has a pKa of 6.76 +/- 0.02 compared to pKa of 5.50 +/- 0.02 for changes in current suppression, which suggests the presence of at least two proton binding sites on HERG with functionally distinct properties.
PubMedId: 14716075
Journal-article: Cell biochemistry and biophysics, vol. 39, 2004
Authors: Glenna C L Bett, Randall L Rasmusson
Abstract: C-type inactivation is present in many voltage-gated potassium channels and is probably related to 'slow' inactivation in calcium and sodium channels. The mechanisms underlying C-type inactivation are unclear, but it is sensitive to mutations on both the extra- and intracellular sides of the channel. We used an N-terminal deleted channel with a valine to alanine point mutation at the intracellular side of S6 (fKv1.4[V561A]DeltaN). This construct alters recovery from inactivation and inverts the relationship between C-type inactivation and [K(+)](o). We used this inverted relationship to examine C-type inactivation and coupling mechanisms between N- and C-type inactivation. The valine to alanine mutation reduces the channel's affinity for both quinidine and the N-terminal domain. However, binding of the N-terminal or quinidine restores normal recovery from inactivation. This suggests that coupling between N- and C-type inactivation is dominated by allosteric mechanisms. The permeation mechanism, driven by a reduction in permeant [K(+)](o) following pore block (which would retard C-type inactivation), contributes minimally to coupling in these channels. We propose that the cytoplasmic half of S6 forms part of the N-terminal binding site, as previously predicted from X-ray crystallography studies in the distantly related KcsA channel. Binding of the N-terminal domain or a positively charged lipophilic compound such as quinidine interacts with the hydrophobic moieties on S6 in the bound state. This binding can orientate S6 into a conformation which resembles the normal C-type inactivated state. This is the probable mechanism by which drug or N-terminal binding increases the rate of C-type inactivation via an allosteric mechanism.
PubMedId: 14608006
Journal-article: The Journal of physiology, vol. 556, 2003
Authors: Vladimir E Bondarenko, Glenna C L Bett, Randall L Rasmusson
Abstract: We have developed a model of Ca(2+) handling in ferret ventricular myocytes. This model includes a novel L-type Ca(2+) channel, detailed intracellular Ca(2+) movements, and graded Ca(2+)-induced Ca(2+) release (CICR). The model successfully reproduces data from voltage-clamp experiments, including voltage- and time-dependent changes in intracellular Ca(2+) concentration ([Ca(2+)](i)), L-type Ca(2+) channel current (I(CaL)) inactivation and recovery kinetics, and Ca(2+) sparks. The development of graded CICR is critically dependent on spatial heterogeneity and the physical arrangement of calcium channels in opposition to ryanodine-sensitive release channels. The model contains spatially distinct subsystems representing the subsarcolemmal regions where the junctional sarcoplasmic reticulum (SR) abuts the T-tubular membrane and where the L-type Ca(2+) channels and SR ryanodine receptors (RyRs) are localized. There are eight different types of subsystems in our model, with between one and eight L-type Ca(2+) channels distributed binomially. This model exhibits graded CICR and provides a quantitative description of Ca(2+) dynamics not requiring Monte-Carlo simulations. Activation of RyRs and release of Ca(2+) from the SR depend critically on Ca(2+) entry through L-type Ca(2+) channels. In turn, Ca(2+) channel inactivation is critically dependent on the release of stored intracellular Ca(2+). Inactivation of I(CaL) depends on both transmembrane voltage and local [Ca(2+)](i) near the channel, which results in distinctive inactivation properties. The molecular mechanisms underlying many I(CaL) gating properties are unclear, but [Ca(2+)](i) dynamics clearly play a fundamental role.
PubMedId: 14630639
Journal-article: American journal of physiology. Heart and circulatory physiology, vol. 286, 2003
Authors: Shimin Wang, Michael J. Morales, Yu-Jie Qu, Glenna C. L. Bett, Harold C. Strauss, Randall L. Rasmusson
Journal-article: The Journal of Physiology, vol. 546, pp. 387-401, 2003
Authors: XueJun Jiang, Glenna C L Bett, XiaoYan Li, Vladimir E Bondarenko, Randall L Rasmusson
Abstract: Channels are water-filled membrane-spanning proteins, which undergo conformational changes as they gate, i.e. open or close. These conformational changes affect both the shape of the channel and the volume of the water-filled pore. We measured the changes in pore volume associated with activation, deactivation, C-type inactivation and recovery in an N-terminal-deleted mutant of the Kv1.4 K+ channel (Kv1.4DeltaN) expressed in Xenopus oocytes. We used giant-patch and cut-open oocyte voltage clamp techniques and applied solutes which are too large to enter the pore mouth to exert osmotic pressure and thus favour smaller pore volume conformations. Applied intracellular osmotic pressure (300 mM sucrose) sped inactivation (time constants (tauinactivation): control, 0.66 +/- 0.09 s; hyperosmotic solution, 0.29 +/- 0.04 s; n = 5, P < 0.01), sped deactivation (taudeactivation: control, 18.8 +/- 0.94 ms; hyperosmotic solution, 8.01 +/- 1.92 ms; n = 5, P < 0.01), and slowed activation (tauactivation: control, 1.04 +/- 0.05 ms; hyperosmotic solution, 1.96 +/- 0.31 ms; n = 5, P < 0.01). These effects were reversible and solute independent. We estimated the pore volume change on inactivation to be about 4500 A3. Osmotic pressure had no effect when applied extracellularly. These data suggest that the intracellular side of the pore closes during C-type inactivation and the volume change is similar to that associated with activation or deactivation. This is also similar to the pore volume estimated from the crystal structure of KcsA and MthK K+ channels. Intracellular osmotic pressure also strongly inhibited re-opening currents associated with recovery from inactivation, which is consistent with a physical similarity between the C-type inactivated and resting closed state.
PubMedId: 12730347
Journal-article: The Journal of physiology, vol. 549, 2003
Authors: Shimin Wang, Michael J Morales, Yu-Jie Qu, Glenna C L Bett, Harold C Strauss, Randall L Rasmusson
PubMedId: 12527726
Journal Article: The Journal of physiology, vol. 546, 2003
Authors: Glenna C L Bett, Shuiping Dai, Donald L Campbell
PubMedId: 12096055
Journal Article: The Journal of physiology, vol. 542, 2002
Authors: Glenna C. L. Bett, Shuiping Dai, Donald L. Campbell
Journal-article: The Journal of Physiology, vol. 542, pp. 107-117, 2002
Authors: Xiaoyan Li, Glenna C L Bett, Xuejun Jiang, Vladimir E Bondarenko, Michael J Morales, Randall L Rasmusson
Abstract: Kv1.4 encodes a slowly recovering transient outward current (I(to)), which inactivates by a fast N-type (intracellular ball and chain) mechanism but has slow recovery due to C-type inactivation. C-type inactivation of the NH(2)-terminal deletion mutant (fKv1.4DeltaN) was inhibited by 98 mM extracellular K(+) concentration ([K(+)](o)), whereas N-type was unaffected. In 98 mM [K(+)](o), removal of intracellular K(+) concentration ([K(+)](i)) speeded C-type inactivation but had no effect on N-type inactivation, suggesting that C-type inactivation is sensitive to K(+) binding to intracellular sites. C-type inactivation is thought to involve closure of the extracellular pore mouth. However, a valine to alanine mutation on the intracellular side of S6 (V561A) of fKv1.4DeltaN alters recovery and results in anomalous speeding of C-type inactivation with increasing [K(+)](o). Extracellular pH (pH(o)) modulated both N- and C-type inactivation through an S5-H5 linker histidine (H508) with acidosis speeding both N- and C-type inactivation. Mutation of an extracellular lysine to a tyrosine (K532Y) slowed C-type inactivation and inhibited the pH dependence of both N- and C-type inactivation. These results suggest that mutations, [K(+)], and pH modulate inactivation through membrane-spanning mechanisms involving S6.
PubMedId: 12388308
Journal-article: American journal of physiology. Heart and circulatory physiology, vol. 284, 2002
Authors: Glenna Bett, Randall Rasmusson
Book-chapter: Quantitative Cardiac Electrophysiology, pp. 1-60, 2002
Authors: Tao Zeng, Glenna C. L. Bett, Frederick Sachs
Journal-article: American Journal of Physiology-Heart and Circulatory Physiology, vol. 278, pp. H548-H557, 2000
Authors: G.C.L. Bett, F. Sachs
Journal-article: The Journal of Membrane Biology, vol. 173, pp. 237, 2000
Authors: G C Bett, F Sachs
PubMedId: 10667920
Journal Article: The Journal of membrane biology, vol. 173, 2000
Authors: G.C.L. Bett, F. Sachs
Journal-article: Journal of Membrane Biology, vol. 173, pp. 255-263, 2000
Authors: J I Vandenberg, G C Bett, T Powell
Abstract: The purpose of this investigation was to determine to what extent the swelling-activated Cl- current (ICl,swell) contributes to swelling-induced changes in the resting membrane potential and action potential duration (APD) in ventricular myocytes. Action potentials were recorded from guinea pig ventricular myocytes using conventional whole cell recording techniques. Cell swelling caused initial lengthening followed by a variable shortening of APD. In 59% of cells this secondary APD shortening had a 4,4'-diisothiocyanostilbene-2,2'-disulfonic acid (DIDS)-sensitive component, consistent with a contribution from ICl,swell. Furthermore, DIDS partially antagonized the depolarization of the resting membrane potential that occurred during cell swelling. We have modeled the ICl,swell using the Oxsoft Heart computer program. Action potential changes predicted by the model agree well with the observed DIDS-sensitive component of the change in the action potential during cell swelling. We conclude that activation of ICl,swell contributes to shortening of APD and depolarization of the resting membrane potential during cell swelling in cardiac myocytes.
PubMedId: 9277351
Journal-article: The American journal of physiology, vol. 273, 1997
Authors: D Noble, G Bett
PubMedId: 8275516
Journal Article: Cardiovascular research, vol. 27, 1993
Authors: D. Noble, G. Bett
Journal-article: Cardiovascular Research, vol. 27, pp. 1701-1712, 1993
Authors: G Bett, D Noble, S Noble, Y Earm, W K Ho, I S So
PubMedId: 1529788
Journal Article: Advances in experimental medicine and biology, vol. 311, 1992
Authors: Glenna Bett, Denis Noble, Susan Noble, Yung Earm, Won-Kyung Ho, In-Suk So
Book-chapter: Excitation-Contraction Coupling in Skeletal, Cardiac, and Smooth Muscle, pp. 453-454, 1992
Authors: D Noble, S J Noble, G C Bett, Y E Earm, W K Ho, I K So
PubMedId: 1785860
Journal Article: Annals of the New York Academy of Sciences, vol. 639, 1991
Authors: D. NOBLE, S. J. NOBLE, G. C. L. BETT, Y. E. EARM, W. K. HO, I. K. SO
Journal-article: Annals of the New York Academy of Sciences, vol. 639, pp. 334-353, 1991
Presented by CUBS © 2024