hosted by
publicationslist.org
    
Henggui Zhang

henggui.zhang@manchester.ac.uk

Journal articles

2009
 
DOI   
PMID 
Oleg V Aslanidi, Philip Stewart, Mark R Boyett, Henggui Zhang (2009)  Optimal velocity and safety of discontinuous conduction through the heterogeneous Purkinje-ventricular junction.   Biophys J 97: 1. 20-39 Jul  
Abstract: Slow and discontinuous wave conduction through nonuniform junctions in cardiac tissues is generally considered unsafe and proarrythmogenic. However, the relationships between tissue structure, wave conduction velocity, and safety at such junctions are unknown. We have developed a structurally and electrophysiologically detailed model of the canine Purkinje-ventricular junction (PVJ) and varied its heterogeneity parameters to determine such relationships. We show that neither very fast nor very slow conduction is safe, and there exists an optimal velocity that provides the maximum safety factor for conduction through the junction. The resultant conduction time delay across the PVJ is a natural consequence of the electrophysiological and morphological differences between the Purkinje fiber and ventricular tissue. The delay allows the PVJ to accumulate and pass sufficient charge to excite the adjacent ventricular tissue, but is not long enough for the source-to-load mismatch at the junction to be enhanced over time. The observed relationships between the conduction velocity and safety factor can provide new insights into optimal conditions for wave propagation through nonuniform junctions between various cardiac tissues.
Notes:
 
DOI   
PMID 
Henggui Zhang, Tao Tao, Sanjay Kharche, Simon M Harrison (2009)  Modelling changes in transmural propagation and susceptibility to arrhythmia induced by volatile anaesthetics in ventricular tissue.   J Theor Biol 257: 2. 279-291 Mar  
Abstract: Volatile anaesthetics such as halothane, isoflurane and sevoflurane inhibit membrane currents contributing to the ventricular action potential. Transmural variation in the extent of current blockade induces differential effects on action potential duration (APD) in the endocardium and epicardium which may be pro-arrhythmic. Biophysical modelling techniques were used to simulate the functional impact of anaesthetic-induced blockade of membrane currents on APD and effective refractory period (ERP) in rat endocardial and epicardial cell models. Additionally, the transmural conduction of excitation waves in 1-dimensional cell arrays, the tissue's vulnerability to arrhythmogenesis and dynamic behaviour of re-entrant excitation in 2-dimensional cell arrays were studied. Simulated anaesthetic exposure reduced APD and ERP in both epicardial and endocardial cell models. The reduction in APD was greater in endocardial than epicardial cells, reducing transmural APD dispersion consistent with experimental data. However, the transmural ERP dispersion was augmented. All three anaesthetics increased the width of the tissue's vulnerable window during which a premature stimulus could induce unidirectional conduction block but only halothane reduced the critical size of ventricular substrates necessary to initiate and sustain re-entrant excitation. All three anaesthetics accelerated the rate of re-entrant excitation waves, but only halothane prolonged the lifespan of re-entry. These data illustrate in silico, that modest changes in ion channel conductance abbreviate rat ventricular APD and ERP, reduce transmural APD dispersion, but augment transmural ERP dispersion. These changes collectively enhance the propensity for arrhythmia generation and provide a substrate for re-entry circuits with a longer half life than in control conditions.
Notes:
 
DOI   
PMID 
El Harchi, McPate, Zhang, Hancox (2009)  Action potential clamp and chloroquine sensitivity of mutant Kir2.1 channels responsible for variant 3 short QT syndrome.   J Mol Cell Cardiol Mar  
Abstract: Recently identified genetic forms of short QT syndrome (SQTS) are associated with an increased risk of arrhythmia and sudden death. The SQT3 variant is associated with an amino-acid substitution (D172N) in the KCNJ2-encoded Kir2.1 K(+) channel. In this study, whole-cell action potential (AP) clamp recording from transiently transfected Chinese Hamster Ovary cells at 37 degrees C showed marked augmentation of outward Kir2.1 current through D172N channels, associated with right-ward voltage-shifts of peak repolarizing current during both ventricular and atrial AP commands. Peak outward current elicited by ventricular AP commands was inhibited by chloroquine with an IC(50) of 2.45 muM for wild-type (WT) Kir2.1, of 3.30 muM for D172N-Kir2.1 alone and of 3.11 muM for co-expressed WT and D172N (P>0.05 for all). These findings establish chloroquine as an effective inhibitor of SQT3 mutant Kir2.1 channels.
Notes:
 
DOI   
PMID 
Oleg V Aslanidi, Mark R Boyett, Halina Dobrzynski, Jue Li, Henggui Zhang (2009)  Mechanisms of transition from normal to reentrant electrical activity in a model of rabbit atrial tissue: interaction of tissue heterogeneity and anisotropy.   Biophys J 96: 3. 798-817 Feb  
Abstract: Experimental evidence suggests that regional differences in action potential (AP) morphology can provide a substrate for initiation and maintenance of reentrant arrhythmias in the right atrium (RA), but the relationships between the complex electrophysiological and anatomical organization of the RA and the genesis of reentry are unclear. In this study, a biophysically detailed three-dimensional computer model of the right atrial tissue was constructed to study the role of tissue heterogeneity and anisotropy in arrhythmogenesis. The model of Lindblad et al. for a rabbit atrial cell was modified to incorporate experimental data on regional differences in several ionic currents (primarily, I(Na), I(CaL), I(K1), I(to), and I(sus)) between the crista terminalis and pectinate muscle cells. The modified model was validated by its ability to reproduce the AP properties measured experimentally. The anatomical model of the rabbit RA (including tissue geometry and fiber orientation) was based on a recent histological reconstruction. Simulations with the resultant electrophysiologically and anatomically detailed three-dimensional model show that complex organization of the RA tissue causes breakdown of regular AP conduction patterns at high pacing rates (>11.75 Hz): as the AP in the crista terminalis cells is longer, and electrotonic coupling transverse to fibers of the crista terminalis is weak, high-frequency pacing at the border between the crista terminalis and pectinate muscles results in a unidirectional conduction block toward the crista terminalis and generation of reentry. Contributions of the tissue heterogeneity and anisotropy to reentry initiation mechanisms are quantified by measuring action potential duration (APD) gradients at the border between the crista terminalis and pectinate muscles: the APD gradients are high in areas where both heterogeneity and anisotropy are high, such that intrinsic APD differences are not diminished by electrotonic interactions. Thus, our detailed computer model reconstructs complex electrical activity in the RA, and provides new insights into the mechanisms of transition from focal atrial tachycardia into reentry.
Notes:
 
DOI   
PMID 
Wing Chiu Tong, Michele Sweeney, Carolyn J P Jones, Henggui Zhang, Stephen C O'Neill, Ian Prior, Michael J Taggart (2009)  Three-dimensional electron microscopic reconstruction of intracellular organellar arrangements in vascular smooth muscle--further evidence of nanospaces and contacts.   J Cell Mol Med 13: 5. 995-998 May  
Abstract: The sarcoplasmic reticulum (SR) of smooth muscle is crucial for appropriate regulation of Ca(2+) signalling. In visceral and vascular smooth muscles the SR is known to periodically lie in close register, within a few nanometres, to the plasma membrane. Recent work has focussed on reconstructions of the ultrastructural arrangement of this so-called peripheral SR that may be important for the genesis of phenomena such as Ca(2+) sparks. Here, we turn our attention to vascular smooth muscle and explore the 3-dimensional (3D) ultrastructural positioning of SR found deeper in the cell that is involved in the propagation of Ca(2+) waves. We use digital reconstruction and volume rendering of serial electron microscopic sections from isolated resistance arteries, pressurized in vitro to mimic cellular geometric conformations anticipated in vivo, to map SR positioning. We confirm that these central portions of SR are in close register with mitochondria and the nucleus with all three organelles tightly enveloped by a myofilament/cytoskeletal lattice. Nanospacings between the SR and individual mitochondria are visible and in three dimensions as the SR contorts to accommodate these organelles. Direct connection of the SR and nuclear membranes is confirmed. Such 3D positioning of centrally located SR further informs us of its likely role in the manifestation of spatiotemporal Ca(2+) dynamics: signal encoding may be facilitated by spatially directed release of Ca(2+) to influence several processes crucial to vascular smooth muscle and resistance artery function including myofilament activation by Ca(2+) waves, mitochondrial respiration and gene transcription.
Notes:
 
DOI   
PMID 
Stones, Billeter, Zhang, Harrison, White (2009)  The role of transient outward K(+) current in electrical remodelling induced by voluntary exercise in female rat hearts.   Basic Res Cardiol May  
Abstract: Regular exercise can lead to electrical remodelling of the heart. The cellular mechanisms associated with these changes are not well understood, and are difficult to study in human tissue but are important given that exercise is recommended to the general population. We have investigated the role played by the transient outward K(+) current (I (to)) in the changes in electrical activity seen in response to voluntary exercise training in rats. Female rats undertook 6 weeks of voluntary wheel running exercise (TRN) or were sedentary controls (SED). Monophasic action potentials (MAPs) were recorded from the surface of whole hearts. Whole cell patch clamp recordings of I (to); mRNA and protein levels of selected targets in sub-epicardial (EPI) and sub-endocardial myocardium of SED and TRN hearts were compared. In TRN rats, heart weight:body weight was significantly increased and epicardial MAPs significantly prolonged. I (to) density was reduced in TRN EPI myocytes, such that the transmural gradient of I (to) was significantly reduced (P < 0.05). Computer modelling of these changes in I (to) predicted the observed changes in action potential profile. However, transmural gradients in mRNA and protein expression for Kv4.2 or mRNA levels of the Kv4.2 regulators; KChIP2 and Irx-5 were not significantly altered by voluntary exercise. We conclude that voluntary exercise electrical remodelling is caused, at least in part, by a decrease in EPI I (to), possibly because of fewer functional channels in the membrane, which results in a fall in the transmural action potential duration gradient.
Notes:
 
DOI   
PMID 
Philip Stewart, Oleg V Aslanidi, Denis Noble, Penelope J Noble, Mark R Boyett, Henggui Zhang (2009)  Mathematical models of the electrical action potential of Purkinje fibre cells.   Philos Transact A Math Phys Eng Sci 367: 1896. 2225-2255 Jun  
Abstract: Early development of ionic models for cardiac myocytes, from the pioneering modification of the Hodgkin-Huxley giant squid axon model by Noble to the iconic DiFrancesco-Noble model integrating voltage-gated ionic currents, ion pumps and exchangers, Ca(2+) sequestration and Ca(2+)-induced Ca(2+) release, provided a general description for a mammalian Purkinje fibre (PF) and the framework for modern cardiac models. In the past two decades, development has focused on tissue-specific models with an emphasis on the sino-atrial (SA) node, atria and ventricles, while the PFs have largely been neglected. However, achieving the ultimate goal of creating a virtual human heart will require detailed models of all distinctive regions of the cardiac conduction system, including the PFs, which play an important role in conducting cardiac excitation and ensuring the synchronized timing and sequencing of ventricular contraction. In this paper, we present details of our newly developed model for the human PF cell including validation against experimental data. Ionic mechanisms underlying the heterogeneity between the PF and ventricular action potentials in humans and other species are analysed. The newly developed PF cell model adds a new member to the family of human cardiac cell models developed previously for the SA node, atrial and ventricular cells, which can be incorporated into an anatomical model of the human heart with details of its electrophysiological heterogeneity and anatomical complexity.
Notes:
2008
 
DOI   
PMID 
Jue Li, Ian D Greener, Shin Inada, Vladimir P Nikolski, Mitsuru Yamamoto, Jules C Hancox, Henggui Zhang, Rudi Billeter, Igor R Efimov, Halina Dobrzynski, Mark R Boyett (2008)  Computer three-dimensional reconstruction of the atrioventricular node.   Circ Res 102: 8. 975-985 Apr  
Abstract: Because of its complexity, the atrioventricular node (AVN), remains 1 of the least understood regions of the heart. The aim of the study was to construct a detailed anatomic model of the AVN and relate it to AVN function. The electric activity of a rabbit AVN preparation was imaged using voltage-dependent dye. The preparation was then fixed and sectioned. Sixty-five sections at 60- to 340-microm intervals were stained for histology and immunolabeled for neurofilament (marker of nodal tissue) and connexin43 (gap junction protein). This revealed multiple structures within and around the AVN, including transitional tissue, inferior nodal extension, penetrating bundle, His bundle, atrial and ventricular muscle, central fibrous body, tendon of Todaro, and valves. A 3D anatomically detailed mathematical model (approximately 13 million element array) of the AVN and surrounding atrium and ventricle, incorporating all cell types, was constructed. Comparison of the model with electric activity recorded in experiments suggests that the inferior nodal extension forms the slow pathway, whereas the transitional tissue forms the fast pathway into the AVN. In addition, it suggests the pacemaker activity of the atrioventricular junction originates in the inferior nodal extension. Computer simulation of the propagation of the action potential through the anatomic model shows how, because of the complex structure of the AVN, reentry (slow-fast and fast-slow) can occur. In summary, a mathematical model of the anatomy of the AVN has been generated that allows AVN conduction to be explored.
Notes:
 
DOI   
PMID 
Mathieu Gautier, Henggui Zhang, Ian M Fearon (2008)  Peroxynitrite formation mediates LPC-induced augmentation of cardiac late sodium currents.   J Mol Cell Cardiol 44: 2. 241-251 Feb  
Abstract: Lysophosphatidylcholine (LPC) accumulates in the ischaemic myocardium and is arrhythmogenic. We have examined the mechanisms underlying the effects of LPC on the late cardiac Na(+) current (I(L)Na). Na(+) currents were recorded in HEK293 cells expressing Na(V)1.5 and isolated rat ventricular myocytes. LPC enhanced recombinant I(L)Na, while it reduced peak Na(+) current. Computer modeling of human ventricular myocyte action potentials predicted a marked duration prolonging effect and arrhythmogenic potential due to these effects of LPC on peak and late currents. Enhancement of recombinant I(L)Na was suppressed by the antioxidant ascorbic acid and by the NADPH oxidase inhibitor DPI. Inhibitors of the mitochondrial electron transport chain (rotenone, TTFA and myxothiazol) were without effect on LPC responses. The superoxide donor pyrogallol was without effect on I(L)Na. Enhancement of I(L)Na was abrogated by the NOS inhibitors l-NAME and 7-nitroindazole, while LPC induced an l-NAME-sensitive production of NO, measured as enhanced DAF-FM fluorescence, in both HEK293 cells and ventricular myocytes. Despite this, the NO donors SNAP and SNP caused no change in I(L)Na. However, SNAP enhanced TTX-sensitive recombinant and native I(L)Na in the presence of pyrogallol, suggesting peroxynitrite formation as a mediator of the response to LPC. In support of this, the peroxynitrite scavenger FeTPPS prevented the response of I(L)Na to LPC. Peroxynitrite formation provides a novel mechanism by which LPC regulates the late cardiac Na(+) current.
Notes:
 
DOI   
PMID 
Alan P Benson, Oleg V Aslanidi, Henggui Zhang, Arun V Holden (2008)  The canine virtual ventricular wall: a platform for dissecting pharmacological effects on propagation and arrhythmogenesis.   Prog Biophys Mol Biol 96: 1-3. 187-208 Jan/Apr  
Abstract: We have constructed computational models of canine ventricular cells and tissues, ultimately combining detailed tissue architecture and heterogeneous transmural electrophysiology. The heterogeneity is introduced by modifying the Hund-Rudy canine cell model in order to reproduce experimentally reported electrophysiological properties of endocardial, midmyocardial (M) and epicardial cells. These models are validated against experimental data for individual ionic current and action potential characteristics, and their rate dependencies. 1D and 3D heterogeneous virtual tissues are constructed, with detailed tissue architecture (anisotropy and orthotropy, due to fibre orientation and sheet structure) of the left ventricular wall wedge extracted from a diffusion tensor imaging data set. The models are used to study the effects of tissue heterogeneity and class III drugs on transmural propagation and tissue vulnerability to re-entry. We have determined relationships between the transmural dispersion of action potential duration (APD) and the vulnerable window in the 1D virtual ventricular wall, and demonstrated how changes in the transmural heterogeneity, and hence tissue vulnerability, can lead to generation of re-entry in the 3D ventricular wedge. Two class III drugs with opposite qualitative effects on transmural APD heterogeneity are considered: d-sotalol that increases transmural APD dispersion, and amiodarone that decreases it. Simulations with the 1D virtual ventricular wall show that under d-sotalol conditions the vulnerable window is substantially wider compared to amiodarone conditions, primarily in the epicardial region where unidirectional conduction block persists until the adjacent M cells are fully repolarised. Further simulations with the 3D ventricular wedge have shown that ectopic stimulation of the epicardial region results in generation of sustained re-entry under d-sotalol conditions, but not under amiodarone conditions or in control. Again, APD increase in M cells was identified as the major contributor to tissue vulnerability--re-entry was initiated primarily due to ectopic excitation propagating around the unidirectional conduction block in the M cell region. This suggests an electrophysiological mechanism for the anti- and proarrhythmic effects of the class III drugs: the relative safety of amiodarone in comparison to d-sotalol can be explained by relatively low transmural APD dispersion, and hence, a narrow vulnerable window and low probability of re-entry in the tissue.
Notes:
 
DOI   
PMID 
Henggui Zhang, Sanjay Kharche, Arun V Holden, Jules C Hancox (2008)  Repolarisation and vulnerability to re-entry in the human heart with short QT syndrome arising from KCNQ1 mutation--a simulation study.   Prog Biophys Mol Biol 96: 1-3. 112-131 Jan/Apr  
Abstract: Idiopathic short QT syndrome (SQTS) is a recently identified, genetically heterogeneous condition characterised by abbreviated QT intervals and an increased susceptibility to arrhythmia and sudden death. This simulation study identifies mechanisms by which cellular electrophysiological changes in the SQT2 (slow delayed rectifier, IKs, -linked) SQTS variant increases arrhythmia risk. The channel kinetics of the V307L mutation of the KCNQ1 subunit of the IKs channel were incorporated into human ventricular action potential (AP) models and into 1D and 2D transmural tissue simulations. Incorporating the V307L mutation into simulations reproduced defining features of the SQTS: abbreviation of the QT interval, and increases in T wave amplitude and Tpeak-Tend duration. In the single-cell model, the V307L mutation abbreviated ventricular cell AP duration at 90% repolarisation (APD90) and increased the maximal transmural voltage heterogeneity (deltaV) during APs; this resulted in augmented transmural heterogeneity of APD90 and of the effective refractory period (ERP). In the intact tissue model, the vulnerable window for unidirectional conduction block was also increased. In 2D tissue the V307L mutation facilitated and maintained reentrant excitation. Thus, in SQT2 increases in transmural heterogeneity of APD, deltaV, ERP and an increased vulnerable window for unidirectional conduction block generate an electrical substrate favourable to reentrant arrhythmia.
Notes:
 
DOI   
PMID 
Sanjay Kharche, Clifford J Garratt, Mark R Boyett, Shin Inada, Arun V Holden, Jules C Hancox, Henggui Zhang (2008)  Atrial proarrhythmia due to increased inward rectifier current (I(K1)) arising from KCNJ2 mutation--a simulation study.   Prog Biophys Mol Biol 98: 2-3. 186-197 Oct/Nov  
Abstract: Atrial fibrillation (AF) has been linked to increased inward rectifier potassium current, I(K1), either due to AF-induced electrical remodelling, or from functional changes due to the Kir2.1 V93I mutation. The aim of this simulation study was to identify at cell and tissue levels' mechanisms by which increased I(K1) facilitates and perpetuates AF. The Courtemanche et al. human atrial cell action potential (AP) model was modified to incorporate reported changes in I(K1) induced by the Kir2.1 V93I mutation in both heterozygous (Het) and homozygous (Hom) mutant forms. The modified models for wild type (WT), Het and Hom conditions were incorporated into homogeneous 1D, 2D and 3D tissue models. Restitution curves of AP duration (APD), effective refractory period (ERP) and conduction velocity (CV) were computed and both the temporal and the spatial vulnerability of atrial tissue to re-entry were measured. The lifespan and tip meandering pattern of re-entry were also characterised. For comparison, parallel simulations were performed by incorporating into the Courtmanche et al. model a linear increase in maximal I(K1) conductance. It was found that the gain-in-function of V93I 'mutant'I(K1) led to abbreviated atrial APs and flattened APD, ERP and CV restitution curves. It also hyperpolarised atrial resting membrane potential and slowed down intra-atrial conduction. V93I 'mutant'I(K1) reduced the tissue's temporal vulnerability but increased spatial vulnerability to initiate and sustain re-entry, resulting in an increased overall susceptibility of atrial tissue to arrhythmogenesis. In the 2D model, spiral waves self-terminated for WT (lifespan < 3.3 s) tissue, but persisted in Het and Hom tissues for the whole simulation period (lifespan > 10 s). The tip of the spiral wave meandered more in WT tissue than in Het and Hom tissues. Increased I(K1) due to augmented maximal conductance produced similar results to those of Het and Hom Kir2.1 V93I mutant conditions. In the 3D model the dynamic behaviour of scroll waves was stabilized by increased I(K1). In conclusion, increased I(K1) current, either by the Kir2.1 V93I mutation or by augmented maximal conductance, increases atrial susceptibility to arrhythmia by increasing the lifespan of re-entrant spiral waves and the stability of scroll waves in 3D tissue, thereby facilitating initiation and maintenance of re-entrant circuits.
Notes:
 
DOI   
PMID 
Oleg V Aslanidi, Rebecca S Dewey, Alexandra R Morgan, Mark R Boyett, Henggui Zhang (2008)  Regional differences in rabbit atrial action potential properties: mechanisms, consequences and pharmacological implications.   Conf Proc IEEE Eng Med Biol Soc 2008: 141-144  
Abstract: Regional differences in electrical action potential (AP) properties can provide a substrate for atrial arrhythmias. We quantify such differences by developing detailed AP models for the left (LA) and right (RA) rabbit atrial cells in order to study the underlying electrophysiological mechanisms, as well as their impacts on vulnerable properties of the atrial tissue. The transient outward current, Ito, is identified as the major factor contributing to the AP differences between the LA and RA cells, which suggests a potential pharmacological target for reducing heterogeneity and vulnerability of the atria.
Notes:
 
DOI   
PMID 
Sanjay Kharche, Henggui Zhang (2008)  Simulating the effects of atrial fibrillation induced electrical remodeling: a comprehensive simulation study.   Conf Proc IEEE Eng Med Biol Soc 2008: 593-596  
Abstract: Mechanisms underlying atrial fibrillation (AF) are poorly understood. In this study, we computationally evaluated the functional roles of AF induced electrical remodeling (AFER) on atrial electrical excitations. Experimental data of AFER on human atrial myocytes were incorporated into a biophysically detailed model of human atrial cells to simulate the effects of AFER at cellular and tissue levels. Our results show that AFER dramatically abbreviated atrial action potential duration (APD90) and effective refractory period that were quantitatively consistent with experimental data. A typical feature of loss in rate dependent accommodation of APD90 was observed. AFER slowed down atrial conduction velocity, but facilitated atrial conduction at high excitation rates. AFER increased tissue's spatial vulnerability for initiation and maintenance of AF remarkably. The overall susceptibility of human atrium to arrhythmia was increased. Most importantly AFER increased the stability of reentrant waves in 2D and 3D models prolonging their lifespan. While reentrant excitation waves self-terminated under Control conditions, the same became persistent or degenerated into multiple wavelets leading to spatio-temporal chaos under AFER conditions with accelerated re-entrant excitation rates. There was an increase in dominant frequency. In conclusion, our simulations substantiated a link between AFER and persistence of AF, providing mechanistic insights towards better understanding of "AF begets AF".
Notes:
 
DOI   
PMID 
Laurent Sallé, Sanjay Kharche, Henggui Zhang, Fabien Brette (2008)  Mechanisms underlying adaptation of action potential duration by pacing rate in rat myocytes.   Prog Biophys Mol Biol 96: 1-3. 305-320 Jan/Apr  
Abstract: Heart rate is an essential determinant of cardiac performance. In rat ventricular myocytes, a sudden increase in rate yields to a prolongation of the action potential duration (APD). The mechanism underlying this prolongation is controversial: it has been proposed that the longer APD is due to either: (1) a decrease in K+ currents only or (2) an increase in Ca2+ current only. The aim of this study was to quantitatively investigate the contribution of Ca2+ and K+ currents in the adaptation of APD to pacing rate. Simulation using a mathematical model of ventricular rat cardiac cell model [Pandit, S.V., Clark, R.B., Giles, W.R., Demir, S.S., 2001. A mathematical model of action potential heterogeneity in adult rat left ventricular myocytes. Biophys. J. 81, 3029-3051] predicted a role in the prolongation of APD for K+ currents only. In patch clamp experiments, increasing the pacing rate leads to a significant increase in APD in both control and detubulated myocytes, although it was more marked in control than detubulated myocytes. Supporting the model prediction, we observed that increasing stimulation frequency leads to a decrease in K+ currents in voltage clamped rat ventricular myocytes (square and action potential waveforms), and to a similar extent in both cell types. We have also observed that frequency-dependent facilitation of Ca2+ current occurred in control cells but not in detubulated cells (square and action potential waveforms). From these experiments, we calculated that the relative contribution of Ca2+ and K+ currents to the longer APD following an increase in pacing rate is approximately 65% and approximately 35%, respectively. Therefore, in contrast to the model prediction, Ca2+ current has a significant role in the adaptation of APD to pacing rate. Finally, we have introduced a simplistic modification to the Pandit's model to account for the frequency-dependent facilitation of Ca2+ current.
Notes:
2007
 
DOI   
PMID 
John J Borg, Jules C Hancox, Henggui Zhang, Christopher Ian Spencer, Hongyu Li, Roland Z Kozlowski (2007)  Differential pharmacology of the cardiac anionic background current I(AB).   Eur J Pharmacol 569: 3. 163-170 Aug  
Abstract: A novel anionic background conductance (I(AB)) in cardiac ventricular myocytes has recently been identified but at present there is comparatively little information on its pharmacological modulation. This study investigated the effects of on I(AB) of four pyrethroid agents tefluthrin (a selective activator of this current), tetramethrin, fenpropathrin and alpha-cypermethrin in addition to other well known chloride channel modulators (chlorotoxin, gadolinium and picrotoxin). Guinea-pig ventricular myocytes were isolated using an enzymatic and mechanical dispersion procedure and all electrophysiological measurements were made using the whole-cell patch-clamp technique. In contrast to other anion conductances (stretch- or volume-regulated chloride current (I(Cl,vol)), a cAMP-dependent Cl(-) current (I(Cl,cAMP))) I(AB) was augmented by tefluthrin, fenpropathrin, alpha-cypermethrin (but not tetramethrin). I(AB) was insensitive to chlorotoxin, gadolinium and picrotoxin. Thus, I(AB) exhibits a distinct pharmacological profile from other known cardiac anion conductances.
Notes:
 
DOI   
PMID 
Ming Lei, Henggui Zhang, Andrew A Grace, Christopher L-H Huang (2007)  SCN5A and sinoatrial node pacemaker function.   Cardiovasc Res 74: 3. 356-365 Jun  
Abstract: The SCN5A gene encodes specific voltage-dependent Na+ channels abundant in cardiac muscle that open and close at specific stages of cardiac activity in response to voltage change, thereby controlling the magnitude and timecourse of voltage-dependent Na+ currents (iNa) in cardiac muscle cells. Although iNa has been recorded from sinoatrial (SA) node pacemaker cells, its precise role in SA node pacemaker function remains uncertain. This review summarizes recent findings bearing upon: (i) Sinus node dysfunction resulting from genetic mutations in SCN5A; (ii) Sinus node function in the murine cardiac model with targeted disruptions of the SCN5A gene; (iii) Experimental and computational evaluations of the functional roles of iNa in SA node pacemaker function. Taken together, these new observations suggest strong correlations between SCN5A-encoded Na+ channel and SA node pacemaker function.
Notes:
 
DOI   
PMID 
Michael John Taggart, Andrew Blanks, Sanjay Kharche, Arun Holden, Bin Wang, Henggui Zhang (2007)  Towards understanding the myometrial physiome: approaches for the construction of a virtual physiological uterus.   BMC Pregnancy Childbirth 7 Suppl 1: 06  
Abstract: Premature labour (PTL) is the single most significant factor contributing to neonatal morbidity in Europe with enormous attendant healthcare and social costs. Consequently, it remains a major challenge to alleviate the cause and impact of this condition. Our ability to improve the diagnosis and treatment of women most at risk of PTL is, however, actually hampered by an incomplete understanding of the ways in which the functions of the uterine myocyte are integrated to effect an appropriate biological response at the multicellular whole organ system. The level of organization required to co-ordinate labouring uterine contractile effort in time and space can be considered immense. There is a multitude of what might be considered mini-systems involved, each with their own regulatory feedback cycles, yet they each, in turn, will influence the behaviour of a related system. These include, but are not exclusive to, gestational-dependent regulation of transcription, translation, post-translational modifications, intracellular signaling dynamics, cell morphology, intercellular communication and tissue level morphology. We propose that in order to comprehend how these mini-systems integrate to facilitate uterine contraction during labour (preterm or term) we must, in concert with biological experimentation, construct detailed mathematical descriptions of our findings. This serves three purposes: firstly, providing a quantitative description of series of complex observations; secondly, proferring a database platform that informs further testable experimentation; thirdly, advancing towards the establishment of a virtual physiological uterus and in silico clinical diagnosis and treatment of PTL.
Notes:
2006
 
DOI   
PMID 
Ling Xia, Yu Zhang, Henggui Zhang, Qing Wei, Feng Liu, Stuart Crozier (2006)  Simulation of Brugada syndrome using cellular and three-dimensional whole-heart modeling approaches.   Physiol Meas 27: 11. 1125-1142 Nov  
Abstract: Brugada syndrome (BS) is a genetic disease identified by an abnormal electrocardiogram (ECG) (mainly abnormal ECGs associated with right bundle branch block and ST-elevation in right precordial leads). BS can lead to increased risk of sudden cardiac death. Experimental studies on human ventricular myocardium with BS have been limited due to difficulties in obtaining data. Thus, the use of computer simulation is an important alternative. Most previous BS simulations were based on animal heart cell models. However, due to species differences, the use of human heart cell models, especially a model with three-dimensional whole-heart anatomical structure, is needed. In this study, we developed a model of the human ventricular action potential (AP) based on refining the ten Tusscher et al (2004 Am. J. Physiol. Heart Circ. Physiol. 286 H1573-89) model to incorporate newly available experimental data of some major ionic currents of human ventricular myocytes. These modified channels include the L-type calcium current (I(CaL)), fast sodium current (I(Na)), transient outward potassium current (I(to)), rapidly and slowly delayed rectifier potassium currents (I(Kr) and I(Ks)) and inward rectifier potassium current (I(Ki)). Transmural heterogeneity of APs for epicardial, endocardial and mid-myocardial (M) cells was simulated by varying the maximum conductance of I(Ks) and I(to). The modified AP models were then used to simulate the effects of BS on cellular AP and body surface potentials using a three-dimensional dynamic heart-torso model. Our main findings are as follows. (1) BS has little effect on the AP of endocardial or mid-myocardial cells, but has a large impact on the AP of epicardial cells. (2) A likely region of BS with abnormal cell AP is near the right ventricular outflow track, and the resulting ST-segment elevation is located in the median precordium area. These simulation results are consistent with experimental findings reported in the literature. The model can reproduce a variety of electrophysiological behaviors and provides a good basis for understanding the genesis of abnormal ECG under the condition of BS disease.
Notes:
 
DOI   
PMID 
Gunnar Seemann, Christine Höper, Frank B Sachse, Olaf Dössel, Arun V Holden, Henggui Zhang (2006)  Heterogeneous three-dimensional anatomical and electrophysiological model of human atria.   Philos Transact A Math Phys Eng Sci 364: 1843. 1465-1481 Jun  
Abstract: Investigating the mechanisms underlying the genesis and conduction of electrical excitation in the atria at physiological and pathological states is of great importance. To provide knowledge concerning the mechanisms of excitation, we constructed a biophysical detailed and anatomically accurate computer model of human atria that incorporates both structural and electrophysiological heterogeneities.The three-dimensional geometry was extracted from the visible female dataset. The sinoatrial node (SAN) and atrium, including crista terminalis (CT), pectinate muscles (PM), appendages (APG) and Bachmann's bundle (BB) were segmented in this work. Fibre orientation in CT, PM and BB was set to local longitudinal direction.Descriptions for all used cell types were based on modifications of the Courtemanche et al. model of a human atrial cell. Maximum conductances of Ito, IKr and ICa,L were modified for PM, CT, APG and atrioventricular ring to reproduce measured action potentials (AP). Pacemaker activity in the human SAN was reproduced by removing IK1, but including If, ICa,T, and gradients of channel conductances as described in previous studies for heterogeneous rabbit SAN.Anisotropic conduction was computed with a monodomain model using the finite element method. The transversal to longitudinal ratio of conductivity for PM, CT and BB was 1:9. Atrial working myocardium (AWM) was set to be isotropic.Simulation of atrial electrophysiology showed initiation of APs in the SAN centre. The excitation spread afterwards to the periphery near to the region of the CT and preferentially towards the atrioventricular region. The excitation extends over the right atrium along PM. Both CT and PM activated the right AWM. Earliest activation of the left atrium was through BB and excitation spread over to the APG. The conduction velocities were 0.6ms-1 for AWM, 1.2ms-1 for CT, 1.6ms-1 for PM and 1.1ms-1 for BB at a rate of 63bpm.The simulations revealed that bundles form dominant pathways for atrial conduction. The preferential conduction towards CT and along PM is comparable with clinical mapping. Repolarization is more homogeneous than excitation due to the heterogeneous distribution of electrophysiological properties and hence the action potential duration.
Notes:
2005
 
DOI   
PMID 
Henggui Zhang, Clifford J Garratt, Jiujiang Zhu, Arun V Holden (2005)  Role of up-regulation of IK1 in action potential shortening associated with atrial fibrillation in humans.   Cardiovasc Res 66: 3. 493-502 Jun  
Abstract: OBJECTIVES: Although previous studies in dogs have indicated a minimal role for changes in I(K1) in the shortening of action potential duration (APD) associated with atrial fibrillation (AF), in humans, there is evidence for significant AF-induced up-regulation of this current. In this computer model study, we investigated the relative contributions of the remodeling of I(K1), L-type calcium current, and other remodeled ionic channel currents to AF-induced APD reduction in human atrium. METHODS: Two computer models of electrical activity of human atrial cell were modified by incorporating experimental data of AF-induced changes in human atrial ionic channel conductance and kinetics reported by Bosch et al. (I(CaL), I(to), I(K1), and I(Na)) (AF-1) and Workman et al. (I(CaL), I(to), and I(K1)) (AF-2). The roles and relative importance of individually remodeled ion channels in the APD reduction in human atrium were evaluated by the removal and exclusive methods, in which remodeling of specific currents was omitted, or considered in isolation, in the two models. RESULTS: When tested together, previously reported AF-induced changes in sarcolemmal ion currents result in marked shortening of atrial APD(90). With the AF-1 remodeled parameters, there is a 62% reduction in APD(90) for the Nygren et al. model, and a 68% reduction for the Courtemanche et al. model, which are comparable to experimental results of 60% reduction seen in humans. When tested individually, AF-1-induced changes in I(CaL), I(K1), or I(to) alone result in APD(90) reduction of 20%, 64%, and -10%, respectively, for the Nygren et al. model, and 27%, 40%, and 11.6%, respectively, for the Courtemanche et al. model. With the AF-2 remodeled parameters, there is a 47% reduction in APD(90) for the Nygren et al. model and a 49% reduction for the Courtemanche et al. model, which are also comparable to experimental results of 45% reduction. When tested individually, AF-2-induced changes in I(CaL) or I(K1) alone result in APD(90) reduction of 20% and 40%, respectively, for the Nygren et al. model, and 14% and 21%, respectively, for the Courtemanche et al. model. CONCLUSION: Previously reported changes in L-type Ca(2+) current are insufficient to account for the observed reduction in atrial APD associated with persistent AF. Up-regulation of I(K1) has a greater influence on atrial APD in the human model.
Notes:
 
DOI   
PMID 
Mark R Boyett, Jue Li, Shin Inada, Halina Dobrzynski, Jurgen E Schneider, Arun V Holden, Henggui Zhang (2005)  Imaging the heart: computer 3-dimensional anatomic models of the heart.   J Electrocardiol 38: 4 Suppl. 113-120 Oct  
Abstract: Since the 1960s, models of the action potential in various cardiac cell types have been developed, and since the 1990s, 3-dimensional anatomic (or geometric) models of various cardiac structures have been developed. We are approaching the time when, for one species, we should have a complete set of action potential and anatomic models for the various cardiac tissues and then we will have realized the aim of constructing a "virtual heart" with accurate anatomy and electrophysiology. However, already the two types of model are beginning to be used in tandem to reconstruct the activation sequence of the heart both during sinus rhythm and arrhythmias.
Notes:
 
DOI   
PMID 
Ming Lei, Catharine Goddard, Jie Liu, Anne-Laure Léoni, Anne Royer, Simon S-M Fung, Guosheng Xiao, Aiqun Ma, Henggui Zhang, Flavien Charpentier, Jamie I Vandenberg, William H Colledge, Andrew A Grace, Christopher L-H Huang (2005)  Sinus node dysfunction following targeted disruption of the murine cardiac sodium channel gene Scn5a.   J Physiol 567: Pt 2. 387-400 Sep  
Abstract: We have examined sino-atrial node (SAN) function in hearts from adult mice with heterozygous targeted disruption of the Scn5a gene to clarify the role of Scn5a-encoded cardiac Na+ channels in normal SAN function and the mechanism(s) by which reduced Na+ channel function might cause sinus node dysfunction. Scn5a+/- mice showed depressed heart rates and occasional sino-atrial (SA) block. Their isolated peripheral SAN pacemaker cells showed a reduced Na+ channel expression and slowed intrinsic pacemaker rates. Wild-type (WT) and Scn5a+/- SAN preparations exhibited similar activation patterns but with significantly slower SA conduction and frequent sino-atrial conduction block in Scn5a+/- SAN preparations. Furthermore, isolated WT and Scn5a+/- SAN cells demonstrated differing correlations between cycle length, maximum upstroke velocity and action potential amplitude, and cell size. Small myocytes showed similar, but large myocytes reduced pacemaker rates, implicating the larger peripheral SAN cells in the reduced pacemaker rate that was observed in Scn5a+/- myocytes. These findings were successfully reproduced in a model that implicated i(Na) directly in action potential propagation through the SAN and from SAN to atria, and in modifying heart rate through a coupling of SAN and atrial cells. Functional alterations in the SAN following heterozygous-targeted disruption of Scn5a thus closely resemble those observed in clinical sinus node dysfunction. The findings accordingly provide a basis for understanding of the role of cardiac-type Na+ channels in normal SAN function and the pathophysiology of sinus node dysfunction and suggest new potential targets for its clinical management.
Notes:
2004
 
DOI   
PMID 
Henggui Zhang, Jules C Hancox (2004)  In silico study of action potential and QT interval shortening due to loss of inactivation of the cardiac rapid delayed rectifier potassium current.   Biochem Biophys Res Commun 322: 2. 693-699 Sep  
Abstract: The rapid delayed rectifier K(+) current, I(Kr), plays a key role in repolarisation of cardiac ventricular action potentials (APs). In recent years, a novel clinical condition denoted the short QT syndrome (SQTS) has been identified and, very recently, gain in function mutations in the gene encoding the pore-forming sub-unit of the I(Kr) channel have been proposed to underlie SQTS in some patients. Here, computer simulations were used to investigate the effects of the selective loss of voltage-dependent inactivation of I(Kr) upon ventricular APs and on the QT interval of the electrocardiogram. I(Kr) and inactivation-deficient I(Kr) were incorporated into Luo-Rudy ventricular AP models. Inactivation-deficient I(Kr) produced AP shortening that was heterogeneous between endocardial, mid-myocardial, and epicardial ventricular cell models, irrespective of whether heterogeneity between these sub-regions was incorporated of slow delayed rectifier K(+) current (I(Ks)) alone, or of I(Ks) together with that of transient outward K(+) current. The selective loss of rectification of I(Kr) did not augment transmural dispersion of AP repolarisation, as AP shortening was greater in mid-myocardial than in endo- or epicardial cell models. Simulated conduction through a 1 D transmural ventricular strand was altered by incorporation of inactivation-deficient I(Kr) and the reconstructed QT interval was shortened. Collectively, these results substantiate the notion that selective loss of I(Kr) inactivation produces a gain in I(Kr) function that causes QT interval shortening.
Notes:
2002
 
PMID 
Henggui Zhang, Arun V Holden, Mark R Boyett (2002)  Sustained inward current and pacemaker activity of mammalian sinoatrial node.   J Cardiovasc Electrophysiol 13: 8. 809-812 Aug  
Abstract: INTRODUCTION: A novel sustained inward Na+ current i(sv), which sensitive to Ca2+-antagonists and potentiated by beta-adrenergic stimulation, has been described in pacemaker cells of rabbit, guinea pig, and rat sinoatrial node, as well as rabbit AV node. Although i(st) has been suggested to be an important pacemaker current, this has never been tested experimentally because of the lack of a specific blocker. In this study, we address the role of i(st) in the pacemaker activity of the sinoatrial node cell using computer models. METHODS AND RESULTS: The newly developed models of Zhang et al. for peripheral and central rabbit sinoatrial node cells and models of Noble and Noble, Demir et al., Wilders et al., and Dokos et al. for typical rabbit sinoatrial node cells were modified to incorporate equations for i(st). The conductance g(st) was chosen to give a current density-voltage relationship consistent with experimental data. In the models of Zhang et al. (periphery), Noble and Noble, and Dokos et al., in which i(st) was smaller or about the same amplitude as other inward currents, i(st) increased the pacemaking rate by 0.6%, 2.2%, and 0.8%, respectively. In the models of Zhang et al. (center), Demir et al., and Wilders et al., in which i(st) was larger than some other inward ionic currents, i(st) increased the pacemaking rate by 7%, 20%, and 14%, respectively. CONCLUSION: i(st) has the potential to be a regulator of pacemaker activity, although its importance will depend on the amplitude of i(st) relative to the amplitude of other inward currents involved in pacemaker activity.
Notes:
 
PMID 
Henggui Zhang, Arun V Holden, Denis Noble, Mark R Boyett (2002)  Analysis of the chronotropic effect of acetylcholine on sinoatrial node cells.   J Cardiovasc Electrophysiol 13: 5. 465-474 May  
Abstract: INTRODUCTION: The ionic basis underlying the negative chronotropic effect of acetylcholine (ACh) on sinoatrial (SA) node cells is unresolved and controversial. In the present study, mathematical modeling was used to address this issue. METHODS AND RESULTS: The known concentration-dependent effects of ACh on iK,ACh, iCa,L, and i(f) were introduced into models of rabbit central and peripheral SA node cells. In the central and peripheral models, 9 x 10(-8) and 14 x 10(-8) M ACh, respectively, caused a 50% decrease in pacemaking rate, whereas in rabbit SA node to approximately 7.4 x 10(-8) M ACh caused such a decrease. In the models, iK,ACh was primarily responsible for the decrease and actions of ACh on iCa,L or i(f) alone caused a negligible effect. Although the inhibition of i(f) did not directly contribute to the chronotropic effect, it was indirectly important, because it minimized the opposition by i(f ) to the decrease of rate caused by activation of iK,ACh. The central model was more sensitive to ACh than the peripheral model. CONCLUSION: The chronotropic effect of ACh is principally the result of activation of iK,ACh, and inhibition of iCa,L plays little or no role. Inhibition of i(f) and possible inhibition of ib,Na play an important facilitative role by reducing the ability of i(f) and ib,Na to curtail the chronotropic effect caused by activation of iK,ACh.
Notes:
Powered by publicationslist.org.