modeling atrial fibrillation using human embryonic … › bitstream › 1807 › 94524 › 1 ›...

134
MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC STEM CELLS Zachary Laksman A thesis submitted in conformity with the requirements for the Degree of Masters of Science Institute of Medical Sciences University of Toronto © Copyright by « Zachary Laksman » « 2015»

Upload: others

Post on 05-Jul-2020

0 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

MODELING ATRIAL FIBRILLATION USING

HUMAN EMBRYONIC STEM CELLS

Zachary Laksman

A thesis submitted in conformity with the requirements for the

Degree of Masters of Science

Institute of Medical Sciences

University of Toronto

© Copyright by « Zachary Laksman » « 2015»

Page 2: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

ii

Abstract

Thesis Title: Modeling Atrial Fibrillation Using Human Embryonic Stem Cells

Degree: Masters of Science, Institute of Medical Sciences, University of Toronto

Convocation: November 2015

Background: Atrial fibrillation (AF) is the most common clinical arrhythmia and is

associated with significant morbidity and mortality.

Hypothesis: “Atrial” cardiomyocytes generated from hESCs will recapitulate the key

hallmarks of human AF in a 2 dimensional tissue model, as well as demonstrate the

expected electrophysiologic changes in response to drugs.

Methods and Results: Our “atrial” cardiomyocytes were 80-90% positive for cardiac

troponin, were enriched for the atrial markers ANF and KCNJ3, and lacked ventricular

markers (MYL2 and IRX4). The “atrial” cells mimicked the human electrophysiologic

phenotype with the majority (90%) demonstrating atrial like action potentials. Optical

mapping of multicellular cell sheets was performed on rotors at baseline, after induction

of rotor formation and with the addition of commonly employed antiarrhythmic drugs.

Conclusions: We have successfully generated a human model of AF in vitro and

demonstrated its utility in screening for known, and unpredicted effects of commonly

employed anti-arrhythmics.

Page 3: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

iii

Acknowledgements

I would like to thank Dr. Peter Backx and Dr. Gordon Keller for their support and mentorship. It has been a great privilege to work with scientists of the highest caliber, and people who have earned my greatest esteem and respect. I would like to thank my beautiful daughters Kaiya, Nava, Arielle, and Samara. Thank you to all my family and friends for their help and understanding. Finally, thanks to my wife Jodi who remains the strongest person I have ever met. Thank you for continuing to believe in me and support me unconditionally. I would like to dedicate this thesis to my late father Sander Laksman.

Page 4: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

iv

Technical Contributions and Acknowledgement I acknowledge the following individuals for their contributions to my MSc project: Stephanie Protze, PhD. and Jeehoon Lee, PhD candidate Dr. Protze and Jeehoon’s work in the development and validation of the differentiation protocol used to derive the “atrial” cells under study laid the foundation upon which this project was based. They performed the qRT-PCR and aldefluor stains reported in this thesis as a part of these efforts. Wallace Yang Worked tirelessly to optimize and improve the optical mapping rig and its requisite signal processing. Dr. Mark Gagliardi, PhD Provided assistance and expert consultation in all aspects of tissue culture and directed differentiation of hPSCs Dr. Roozbeh Aschar-Sobbi, PhD Provided expert consultation on nearly all aspects of experimental cardiac electrophysiology Farzad Izaddoustdar Assisted in signal processing and technical aspects related to optical mapping

Page 5: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

v

Table of Contents

Abstract

ii

Acknowledgements iii

Technical contributions and acknowledgements iv

List of abbreviations vii

List of Tables viii

List of Figures ix

Chapter 1 Background 1 1.1 Atrial fibrillation (AF) – clinical burden 1 1.2 Normal cardiac physiology 1 1.3 AF management 3 1.3.1 Rate control vs. rhythm control of AF 3 1.3.2 AF ablation 5 1.4 Basic electrical properties and mechanisms underlying AF 7 1.5 Anti-arrhythmic drugs: dofetilide and flecainide 11 1.6 Wave theory 12 1.7 Rotors 13 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF 16 1.8 Embryonic stem cells and induced pluripotent stem cells 18 1.8.1 Directed Differentiation 20 1.8.2 Retinoic acid signaling 23 1.8.3 Molecular markers of atrial and ventricular cardiomyocytes 24 1.9 Disease modeling using hPSCs 25 1.9.1cLQTS 26 1.9.2 acquired LQTS 29

Chapter 2 Research Aims and Hypotheses 33

Chapter 3 Materials and Methods 35 3.1 Differentiation protocols 35 3.2 Flow cytometry and cell sorting 36 3.3 Molecular markers 37 3.31 qRT-PCR 37 3.3.2 Immunostaining 37 3.4 Cardiac Electrophysiology 37 3.4.1 Patch clamping 37 3.4.2 Micro Electrode Array 39 3.4.3 Optical mapping 40 3.4.3.1 Signal processing 42 3.4.4 Intracellular recordings of cell sheets 45

Page 6: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

vi

Chapter 4 Results

4.1 Differentiation protocols of “atrial” and “ventricular” cardiomyocytes 46 4.2 Molecular markers of “atrial” and “ventricular” cells 51 4.3 Single cell electrophysiology studies of “atrial” and “ventricular” cells 53 4.4 MEA studies of “atrial” and “ventricular” cell sheets 57 4.5 Optical mapping studies of “atrial” and “ventricular” cell sheets 59 4.5.1 Optical mapping the effects of antiarrhythmic drugs: flecainide and dofetilide

65

Chapter 5 Discussion 72 5.1 Molecular markers of “atrial” and “ventricular” cardiomyocytes 73 5.2 Single cell electrophysiology 73 5.3 Modeling atrial disease using hPSCs 75 5.3.1 APD restitution and heterogeneity 78 5.3.2 Conduction velocity 79 5.3.3 Effects of dofetilide on “atrial” cell sheets 80 5.3.4 Effects of flecainide on “atrial” cell sheets 81

Chapter 6 Future Directions 87 6.1 Validation of current findings 87 6.2 Study AF remodeling 90 6.3 Model other types and features of acquired AF 92 6.4 Introduce genetic variation as a determinant and model of AF 94

Chapter 7 References 99

Chapter 8 Appendix 122

Page 7: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

vii

List of abbreviations

AAD Anti-arrhythmic drug

AF Atrial fibrillation

ANF Atrial natriuretic factor

AP Action potential

APA Action potential amplitude

APD Action potential duration

APD50 Action potential duration at 50% of repolarization

APD90 Action potential duration at 90% of repolarization

CaV1.3 Alpha 1D subunit of the L-type voltage dependent calcium channel

CM Cardiomyocyte

CV Conduction velocity

Cx Connexin

DMP Diastolic membrane potential

dv/dtmax Maximum action potential upstroke velocity

EAD Early after depolarizations

EB Embryoid body

ECG Electrocardiogram

ERP Effective refractory period

ESC Embryonic stem cell

FDA U.S. Food and Drug Administration

FFT Fast fourier transform

GJ Gap junction

hPSC Human pluripotent stem cell

ICD Implantable cardioverter defibrillator

iPSC Induced pluripotent stem cell

KCNJ3 Potassium inward rectifying channel, subfamily J, member 3

LQTS Long QT syndrome

MEA Micro electrode array

MYL2 Myosin regulatory light chain-2

NCX Sodium Calcium exchanger

OAPD Optical action potential duration

PS Phase singularity

PV Pulmonary vein

qRT-PCR Real-time quantitative reverse transcription polymerase chain reaction

RA Retinoic acid

RMP Resting membrane potential

ROI Region of interest

RP Refractory period

SAN Sinoatrial node

SR Sinus rhythm

TDR Transmural dispersion of repolarization

TdP Torsades-de-pointes

Page 8: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

viii

List of Tables

Table 1: Characteristics of APs recorded from the protocol generating “atrial” cardiomyocytes compared to the protocol used to generate “ventricular” cardiomyocytes.

55

Page 9: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

ix

List of Figures

Figure 1: Flow cytometric analyses of markers of undifferentiated ES cell 46 Figure 2: Scheme of the protocol used to differentiate hESCs into “atrial” and “ventricular” cardiomyocytes

47

Figure 3: qRT-PCR results comparing relative expression of ALDH1A2 and

CYP26A1 in low induction (BMP4 3 ng/ml and Activin A 2 ng/ml) compared

to high induction (BMP4 10 ng/ml and Activin A 6 ng/ml)

48

Figure 4: Flow cytometric analyses of low induction vs. high induction with

respect to PDGFRα and Aldefluor staining

49

Figure 5: Flow cytometric analyses of important markers used in the

optimization and validation of the directed differentiation protocol (CD56,

PdgfR-α, CD90, SIRPα, cTNT).

51

Figure 6: qRT-PCR-based expression analyses of markers of atrial and

ventricular cardiomyocytes in “atrial” and “ventricular” differentiation

protocols

52

Figure 7: Fluorescent immunostaining of cTNT and the ventricular specific

marker MLC2v in “atrial” and “ventricular” differentiation protocols

53

Figure 8: Typical patch recordings of APs generated from “atrial” and

“ventricular” differentiation protocols

54

Figure 9: Showing presence of cardiomyocyte types with representative APs

and their prevalence in the “atrial” differentiation protocol

54

Figure 10: Showing presence of cardiomyocyte types with representative

APs and their prevalence in the “ventricular” differentiation protocol

55

Figure 11: Comparing the AP characteristics of cells generated from the “atrial” differentiation protocol compared to the “ventricular” differentiation protocol.

56

Figure 12: Representative patch clamp recordings of an anode break in

“atrial” cardiomyocytes

57

Figure 13: Representative MEA recordings of FPDs from “atrial” and “ventricular” cell sheets

58

Figure 14: MEA recording demonstrating pacing and capture of a cell sheet 58 Figure 15: Time sequence imaging of an “atrial” cell sheet in “SR” 60

Figure 16: Representative example of typical OAPs generated from an “atrial” cell sheet

60

Figure 17: On the left, an APD restitution curve of an “atrial” cell sheet

generated by plotting OAPD vs. rate. On the right, a recording of electrical

alternans generated in an “atrial” cell sheet during burst pacing.

61

Figure 18: Time sequence imaging of an “atrial” cell sheet in “AF” 61 Figure 19: APD maps of “SR” and “AF” of “atrial” cell sheets 62 Figure 20: Representative activation maps of “SR” and “AF” of “atrial” cell

sheets 64

Figure 21: Representative conduction velocity maps of “SR” and “AF” of “atrial” cell sheets

64

Figure 22: Representative examples of OAPDs and the effects of dofetilide and flecainide on OAP morphology in “atrial” cell sheet

65

Page 10: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

x

Figure 23: Effect of rotor induction and dofetilide on cycle length and APD in “atrial” cell sheets

66

Figure 24: Effect of rotor induction and dofetilide on conduction velocity in

“atrial” cell sheets

67

Figure 25: Effect of flecainide on cycle length and APD in “atrial” cell sheets 68 Figure 26: Effect of flecainide on conduction velocity in “atrial” cell sheets 69

Figure 27: Effect of rotor induction and dofetilide on conduction velocity in

“atrial” cell sheets as a function of the distance from the origin of the

electrical wavefront

70

Figure 28: Effect of flecainide on conduction velocity in “atrial” cell sheets as

a function of the distance from the origin of the electrical wavefront

71

Appendix Figure 1: Example of an intracellular recording of a cell sheet using

high resistance micropipettes

122

Appendix Figure 2: Serial activation maps generated after the induction of a

rotor in an “atrial” cell sheet demonstrating the initiation of two rotors after

the application of flecainide

123

Appendix Figure 3: Optical APs recorded from hiPSC derived

cardiomyocytes expressing Arclight.

124

Page 11: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

1

Chapter 1

Background

1.1 Atrial Fibrillation – clinical burden

Atrial fibrillation (AF) is the most common clinical arrhythmia, currently affecting 1-2%

of the total population 1. AF is the most common arrhythmia necessitating hospital

admission, and is a major and growing burden on the health of Canadians and our health

care system. Age is an important determinant of AF occurrence with a 25% lifetime risk

of developing AF after the age of 40. As the proportion of our populations get older,

along with a rise in a set of parallel AF risk factors, the prevalence of AF is expected to

double over the next 4 decades2. A diagnosis of AF is associated with a doubling in all-

cause mortality, a five-fold increase in stroke, an accelerated development of heart

failure, and a substantially poorer quality of life3,4. In addition to the costs of identifying

and treating cardiac risk factors leading to AF5-8, costs directly attributed to AF exceeded

$20,000/year/patient in 20109. Total costs are expected to accelerate with more

widespread use of expensive, and invasive, AF ablation ($20k/procedure)10, which can be

seen as palliation rather than a cure, touting success rates of less than 60% after 1 year

even in highly selected patient populations11.

1.2 Normal Cardiac Physiology

To begin to discuss the pathophysiology that predisposes patients to AF, we must first

review normal cardiac physiology, and the link between its conduction system and force

generating contraction. While all myocytes within the heart have the capacity to conduct

electrical impulses through gap junctions, the heart’s pump function under normal

conditions is tightly regulated by a specialized conduction system. When an electrical

Page 12: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

2

impulse (action potential) stimulates myocardium it contracts. This process, which is

fundamental to all muscle physiology, is known as excitation-contraction coupling. The

conduction system of the atria and ventricles are designed to undergo regular and orderly

depolarization/repolarization sequences in order to maximize cardiac efficiency while

minimizing arrhythmias. The sinoatrial node (SAN) acts as the primary pacemaker of the

heart, depolarizing spontaneously to determine and drive the rate of cardiac contraction.

This normal physiologic rhythm is called sinus rhythm (SR). The electrical wavefront

generated by the SAN first initiates the coordinated contraction of the right and left atria.

Atrial contraction ejects blood in the ventricles, the bottom chambers of the heart,

through the atrioventricular valves, contributing as much as 30% towards the final cardiac

output ultimately ejected by the ventricles through the semilunar valves. After atrial

depolarization, the electrical wavefront then invades the atrioventricular node, which

serves as a gateway to the specialized conduction system of the ventricles called the His-

Purkinje system. When AF is present, atria no longer beat regularly (i.e. atria "fibrillate").

AF is easily detected on electrocardiographic recordings (ECGs) as rapid, "irregularly-

irregular" QRS complexes (from ventricles) along with irregular (“random”) baseline

signals (from atria)12,13.

Within the atria, AF is characterized by the appearance of abnormal electrical re-entry

circuits which are self-sustained sequences of depolarization-repolarization cycles called

rotors or wavelets14-16. Despite cyclic patterns seen with re-entry, the electrical patterns in

AF usually have “random” signatures due to wave-breakup/conduction blocks, thus the

term "wavelets" has been proposed14. Crudely speaking, though important for AF

discussion, the propensity towards re-entry (rotors/wavelets) depends on an electrical

Page 13: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

3

property called the “atrial wavelength" (i.e. λ) which is the product of the effective

refractory period (ERP), which is ~action potential duration (APD), and the conduction

velocity (CV), which is the speed of spreading action potentials (APs) in atria. Re-entry

however does not occur without initiation by processes called “kindling” or "triggering".

Both the substrate of AF, the atrial wavelength, and the triggers of AF are potential

targets for therapy.

1.3 AF management

Although atrial beating normally makes relatively minor contributions to the heart's

overall pumping efficiency (approximately 15%), AF nevertheless has important clinical

consequences. For one, because ventricles are electrically-entrained by the atria via the

AV-node, AF leads to elevated pumping rates of the heart (typically >120 beats/min

when AF is untreated)14 which severely impairs cardiac output regulation, leading to

patient fatigue, and promoting “tachycardia induced” cardiomyopathy. The loss of

sequential atrial contractions also causes blood pooling within atrial appendages resulting

in thrombus formation, embolization and stroke, which is the major cause of morbidity

and mortality in AF patients17,18. Antithrombotic therapy is thus recommended for a

majority of patients. The decision regarding initiation of anticoagulation balances the

patient specific risks of bleeding against the treatment and patient specific benefits of

antiplatelet and/or anticoagulants 19. This decision is made independent of the choice

regarding the management of the underlying arrhythmia.

1.3.1 Rate control vs. rhythm control

There are two main strategies for the electrical management of AF, rhythm control and

rate control. Despite the intuitive attraction of restoring sinus rhythm, several trials have

Page 14: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

4

failed to show clinical benefit of this strategy over rate control. The rate control strategy

involves the use of AV nodal blocking agents, such as beta blockers, calcium channel

blockers, and digoxin, to slow impulses through the AV node while allowing

uninterrupted fibrillation of the atrial chambers. Several studies have shown non-

inferiority of rhythm control compared to rate control in terms of survival benefit 20-23,

while studies targeting a higher risk population of heart failure patients similarly did not

show clinical benefit to the rhythm control strategy 24,25. Because of this clinical trial

data, rhythm control is only indicated for patients who suffer from symptoms of AF, or

those who cannot receive adequate rate control and suffer from tachycardia induced

cardiomyopathy 19.

It is worth dissecting some of the trial data that led to our current guideline

recommendations further when considering avenues for future research and novel

therapeutics. Prior to the AFFIRM trial in 2002, a study which arguably has had the most

significant impact on the clinical management of AF, first line therapy was rhythm

control with an implicit belief that patients would benefit from fewer symptoms, better

exercise tolerance, a lower risk of stroke, eventual discontinuation of long-term

anticoagulant therapy and better survival21. In this trial of over 4000 patients over the age

of 65, and including 25% of patients with decreased left ventricular systolic function, the

rhythm control strategy was left to the discretion of the primary physician and included

anti-arrhythmic drugs (AADs) associated with increased mortality, particularly in this

type of high risk population group. The study determined that there was a trend towards

worse survival and increased hospitalizations in patients on a rhythm control compared to

a rate control strategy. Further complicating the picture, patients on rhythm control had a

Page 15: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

5

higher propensity towards discontinuing their anti-coagulation. Interestingly, 37.4% of

patients in the rhythm control group remained in AF, and 34.6% of patients in the rate

control group were in sinus rhythm, clearly demonstrating the ineffectual nature of the

era’s anti-arrhythmic strategy, and supporting the concept that this was not truly a

comparison of successful rhythm control compared to rate control. Furthermore,

crossover between groups was moderately large with 15% of rate control patients and

38% of rhythm control patients crossing over to the other treatment group 21. Later, an “

on-treatment” analysis of the AFFIRM trial showed that the presence of sinus rhythm was

in fact associated with a lower risk of death 26.

Further post-hoc analyses of rhythm vs. rate control strategies have shown a consistent

pattern of improvement in quality of life measures in patients who achieve and maintain

sinus rhythm 27,28. In the PIAF and HOT CAFE trials for example, 6-minute walk times

and maximal treadmill workloads were increased 20,29. In the Canadian Trial of Atrial

Fibrillation, quality of life measures were significantly improved at 3 months from

baseline in patients who achieved sinus rhythm 30.

1.3.2 AF ablation

From the aforementioned results, two main streams of thought have become pervasive in

the field. The first continues to target symptoms with little emphasis on the

electrocardiographic documentation of sinus rhythm or AF. The second centers on a

belief that to date, there has never been a true comparison of rate vs. rhythm control on

“hard” outcomes such as hospitalization, progression of left ventricular dysfunction, or

mortality, since we have not had a therapy that can successfully maintain a high

proportion of patients in sinus rhythm to act as a reasonable on-treatment group. One of

Page 16: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

6

the hopes for a more effective and durable therapy has been the invasive management of

catheter ablation. Remarkably, the first catheter ablation of AF was performed in 1981 by

Dr. Scheinman using high energy DC shocks to destroy the AV node as a form of rate

control 31. Since that time, the technology has rapidly evolved to include radiofrequency

energy that heats the tip of the catheter to a much higher temperature and in a

considerably more controlled fashion. The most significant advancement was

subsequently made in 1998 when Dr. Haissaguerre found that pulmonary vein (PV)

ectopy, originating from the muscular sleeves that extend from the atrial myocardium, are

frequent triggers for AF that can be successfully targeted for ablation 32. They found that

94% of the ectopic foci responsible for initiating AF originated from the PV in a group of

45 patients. Long term success rates from this procedure were 33% off AADs with an

additional 13% of patients being free of AF on AADs33. In 2001, it was Dr. Haissaguerre

again who described a novel approach to PV isolation that targeted the proximal insertion

points of the PVs to the left atrium, thus decreasing the most notable complication of PV

ectopy (PV stenosis), and providing benefit to patients who did not have inducible PV

ectopy during their electrophysiology study.

Since 2001, incremental improvements in catheter and mapping technology have

certainly improved safety and efficacy of the procedure, however the success rates remain

suboptimal. The procedure predominantly targets the triggers of AF, and to date, there

has been little convincing evidence that ablation can successfully target the underlying

substrate34. Therefore, the best results from ablation are in those with paroxysmal AF of

short duration, absence of cardiac structural disease and comorbidities, and without signs

of atrial remodeling, such as dilatation or fibrosis, as seen on MRI using late gadolinium

Page 17: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

7

enhancement 35-41. A meta-analysis of 19 studies that targeted this relatively small

population demonstrated a 68% success rate at 1 year and 60% success at maintaining

sinus rhythm at 3 years 11.

Despite the steady improvements in catheter ablation techniques, they remain expensive,

high risk procedures, which target only patients with lone paroxysmal AF, a relatively

small subpopulation of the growing population of patients with AF. Furthermore, while

AF invariably begins as isolated short-lived episodes called "paroxysmal AF", each

paroxysm accelerates atrial remodelling thereby promoting "persistent AF" (i.e. "AF-

begets-AF")42,43. As discussed previously, persistent AF is much less amenable to the

current ablation strategies, with abysmal outcomes and a lack of consensus regarding the

ability of any ablation strategy to modify the underlying substrate. For these reasons, and

the fact that there is growing evidence that maintenance of sinus rhythm is an important

consideration of outcome studies, there is renewed interest in the development of AADs

for the treatment of AF. An effective drug would not only be applicable to a wider

population, but may benefit the population of patients waiting up to 1 year for their AF

ablation in decreasing AF related remodelling which has been shown to predict worse

outcomes with ablation.

1.4 Basic electrical properties and mechanisms underlying AF

To discuss the future of AAD therapy for AF, we must first revisit the concepts alluded to

earlier, namely the basic mechanisms underlying atrial re-entry. As already mentioned,

AF generally is believed to require a vulnerable substrate that generally requires a

kindling process to initiate semi-random electrical activity featuring underlying "re-entry"

events. The fundamental appreciation of the functional determinants of AF are critical

Page 18: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

8

for understanding the mechanism of action of current AADs. Current dogma proposes

that the generation (triggers) and maintenance of continuous or fibrillatory activity in the

atria depends on the underlying properties of the atrial (and related regions like the

pulmonary veins) substrate and the balance between the determinants of refractoriness

and excitability, or the imbalance thereof. As discussed earlier, the propensity towards

re-entry in the atria depends on the atrial wavelength (i.e. λ) which is the product of the

effective refractory period (ERP), which is ~action potential duration (APD), and the

conduction velocity (CV), which is the speed of spreading APs in atria (λ = RP X CV). If

λ is small relative to the physical dimensions of atria44 then re-entry circuits are more

likely, and hypothetically the atria could accommodate a larger number of simultaneous

reentry circuits.

The action potential, a key component of the atrial wavelength, constitutes changes in the

membrane potential of cardiomyocytes. The membrane potential is established by an

unequal distribution of electrically charged ions across the sarcolemma and

predominantly by the presence of conducting ion channels in the sarcolemma. Opening

and closing of the ion channels allows ionic currents to flow across the membrane

culminating in the generation of the action potential. The direction of current depends on

the electrochemical gradient of the corresponding ions, and the conductivity of channels

that carry the current alters at different membrane potentials.

The atrial action potential is comprised of 5 phases. Depolarization (phase 0), a short-

lived hyperpolarization (phase 1), the plateau (phase 2), and repolarization (phase 3) to

resting potentials (phase 4). Activation (phase 0) is primarily driven by the depolarizing

inward Na+ current (INa). Upon reaching threshold, the rapid influx of Na+ through

Page 19: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

9

voltage-gated sodium channels causes further rapid depolarization of the membrane

(typically to +30 to +40mV), which in turn activates the transient outward potassium

current (Ito), causing very rapid, but short-lived repolarization of the membrane potential

(phase 1). Activation of L-type Ca2+ current (ICaL) follows with simultaneous activation

of a several repolarizing voltage-gated K+ channels (IK). The plateau phase (phase 2) of

the action potential and therefore the action potential duration (APD) is governed by a

delicate balance of ICaL and IK, with membrane repolarization being driven ultimately by

inactivation of L-type Ca2+ channels and progressive activation of IK (phase 3). The

resting potential (typically at -70 to -80 mV) is maintained by the inward rectifier current

IK1 and Ca2+ extrusion is carried out primarily by the electrogenic Na2+/Ca2+

exchanger (NCX) current (INCX). NCX exchanges 3Na2+ ions for each Ca2+ ion with one

net positive charge moving in the direction of sodium transport. Ion channels that are

enriched in atrial cardiomyocytes compared to other types of cardiomyocytes contribute

to the atrial specific action potential morphology. In contrast to atrial and ventricular

myocytes, SAN and AVN myocytes demonstrate slow depolarization of the resting

potential during phase 4, a property related to the relative absence of Ik1 allowing inward

currents (predominantly If) to depolarize the membrane potential 45. Slow depolarization

during phase 4 inactivates most sodium channels and thus depolarization is mainly

achieved by IcaL and T-type Ca2+ currents. INa reductions46-49 and increases in several K+

currents are linked to AF44,46,50-54. APD heterogeneity55-58 can be altered by either pacing

or vagal stimulation leading to regional differences in muscarinic-activated K+ currents,

IK,Ach16,57,59-61. It is important to note that changes in resting membrane potentials, which

Page 20: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

10

are determined by "background" K+ currents (i.e. IK1 and IK,Ach) can also contribute to AF

by modulating INa inactivation and availability.

Conduction Velocity is a measure of the spread of depolarization between electrically

coupled regions within the atria and is determined primarily by sodium current (INa)

densities/kinetics and gap junction (GJ) channel properties62-64. In atria, INa is largely

generated by Nav1.5 pore-forming α-subunits, coassembled with various β-subunits

(Nav) and other proteins65 . GJs are clusters of closely packed channels that directly

connect the cytoplasmic compartments of adjacent cells allowing the passage of ions and

small molecules 66. Each of the neighbouring cardiomyocytes contributes 1 connexon, of

which 21 members have been identified. In the atria, GJ channels are formed by both

Connexin40 (Cx-40) and Connexin-43 (Cx-43) proteins which co-localize to a

considerable extent 62,67. Cx-40 is specific for the atria, and important for atrial

conduction. Targeted deletion of Cx-40 in mice generates a phenotype of diminished

atrial conduction velocity (up to 30%) 68. Somatic mutations in the Cx-40 gene (GJA5) in

humans is associated with a heritable form of AF, thought to be related to reduced

intercellular electrical coupling 69. Taken together, changes in the expression (reductions),

distribution and phosphorylation (reductions) of GJ proteins and INa are associated with

AF46,48,49,70-74. Note the contributors to RP and CV are chamber specific and reflect the

concert of ion channels and gap junctions specific to the chamber of interest.

Since shorter wavelengths promote AF induction and maintenance, many anti-

arrhythmics target RP and thus wavelength prolongation in order to suppress AF.

Alternatively, and less important for this discussion, AADs can act by suppressing AF

Page 21: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

11

triggers. For the purpose of this study, we will discuss two anti-arrhythmics, dofetilide

and flecainide, in further depth.

1.5 Anti-arrhythmic drugs: dofetilide and flecainide

Dofetilide is a class III anti-arrhythmic which selectively inhibits the rapid component of

the time-dependent outward potassium current (Ikr) 75. Studies in dogs demonstrated

prolongation of the ERP and APD in a dose dependent manner, as well as dofetilide’s

ability to facilitate conversion of electrically induced fibrillation 76,77. Dofetilide has no

effect on the maximum rate of depolarization and does not influence conduction within

the His-Purkinje system or within the myocardium75. Data from placebo controlled trials

demonstrated the efficacy of dofetilide 78 with a 30% conversion rate to sinus rhythm, and

a 60% success rate at maintaining sinus rhythm out to 1 year, compared to 20% in the

placebo group. In patients with structural heart disease, dofetilide had no effect on

mortality, and decreased the risk of hospitalization and progression of congestive heart

failure 79,80. As predicted however, since Ikr is an important contributor to the

repolarization of ventricular cells, many patients were exposed to the pro-arrhythmic

nature of ventricular APD prolongation, which can lead to QT prolongation and put

patients at risk for torsades-de –pointes. In clinical trials, the incidence of torsades was as

high as 5% leading to a significant decrease in the clinical uptake of the drug, as well as

explicit warnings and regulations regarding its use 81,82.

Flecainide is a class Ic antiarrhythmic agent, classically considered to slow CV with little

effect on ERP 83,84This in itself would not be predicted to have favourable effects on AF

as it would decrease wavelength and increase AF susceptibility. Flecainide however has

been shown in dog models to increase the atrial refractory period in a rate dependent

Page 22: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

12

fashion thus increasing wavelength at high rates 58. This has been attributed to the drug’s

effect on decreasing atrial APD accommodation during periods of increased heart rate,

possibly related to an effect on repolarizing potassium current, as well as decreasing the

heterogeneity of atrial activation 85. Flecainide has been shown in placebo controlled

trials to decrease AF recurrence once converted to sinus rhythm 86,87. Flecainide has

known pro-arrhythmic properties and has been associated with increased mortality in

patients with a history of ischemic heart disease, and left ventricular dysfunction 88-90.

Increased inducibility of ventricular arrhythmias in animal models of infarction and

ventricular dysfunction have been reproducibly observed 91. This has been attributed in

part to paradoxical amplification of flecainide induced conduction slowing in depolarized

tissue producing dispersion of conduction 91.

1.6 Wave theory

To this point, the discussion regarding AF substrate has ignored a critical concept in

arrhythmia induction and perpetuation by presuming that there is an anatomic obstacle at

the center of our reentrant circuit. To fill in this gap in our developing theorem of AF,

once again historical context will shed light on the evolving concepts. In fact, studies of

fibrillation and its mechanisms date back to the turn of the 20th century when Mayer

demonstrated sustainable circulatory activity in isolated rings of the contractile bell of the

rhyzostomous Scyphomedusa (jellyfish) and turtle ventricular muscle 92. By applying

electrical pulses at one end of the ring, Mayer induced activation wavefronts that

circulated uni-directionally around the ring. This was reproduced by Mines and Garrey

and then described as the circus movement hypothesis of reentry by Lewis in which he

described a reentrant arrhythmia as a wavefront that circulated back to its partially

Page 23: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

13

refractory tail 93 94. Wave theory continued to develop in the 1960s and 1970s, driven

predominantly by technological innovations such as high-speed cameras and the

application of computer or numerical modeling. In 1973 Alessie experimentally modeled

reentry in the absence of an anatomical obstruction thus introducing the concept of

reentry around a functional obstacle or a rotor 95. Using microelectrode recordings in an

isolated dog atrial preparation, they demonstrated sustained reentry in healthy muscle.

This led to the leading circle model of functional reentry which relied on a center of

refractoriness around which the rotating wave circulates, maintained in its state of

refractoriness through ongoing bombardment of centripetal wave fronts. In contrast to

anatomical reentry, the leading circle hypothesis proposes that there is no fully excitable

gap as the circulating wavefront must encroach on its own tail.

1.7 Rotors

Interestingly, circulating wavefronts are not unique to cardiac tissue. The concept of

rotors underpinning cardiac fibrillation stems from the field of nonequilibrium

thermodynamics. The discovery of the initial phenomenon birthing this field is attributed

to Boris Belousov who observed in the 1950s oscillations of colours in a colourless

solution of potassium bromate, cerium sulfate, malonic acid and citric acid in sulfuric

acid 96. Later, Anatoly Zhabotinsky rediscovered this reaction sequence and ultimately

disseminated the concept widely at a conference in 1968 97. The oscillatory patterns

observed in the “BZ” reaction are seen across many natural phenomena including the

growth pattern of amoeba.

Page 24: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

14

1.7.1 Physiology of rotors

A rotor is similar to the leading edge model of functional reentry with a critical

difference, the curved wavefront must meet with its wavetail at a singularity where the

tissue is not refractory 98. Rotors are hypothesized to be the drivers of cardiac fibrillation

and can be represented in 2-dimensional (2D) space as spiral waves or in 3-D space as

scroll waves 92. The wavefront represents cells that have undergone full excitation

(depolarization) and are returning to their rest state (repolarization). Gray et al

demonstrated a phase singularity (PS) using optical mapping experiments of fibrillation

in 1998, and generated phase maps which could track the spiral, its curvature, and its tip

over time and space 99. This was a dramatic step forward in the study of rotor dynamics

allowing its visualization and quantification. They demonstrated that the PS anchored the

rotor, while the spiral wave rotated around it. A rotor however, unlike the leading circle

model, allows for changes in the location of the PS dependent on subtle changes in the

rotor’s environment, or chaotic unpredictable behavior generating complex shapes.

Principal to the rotor theory is the curvature of the rotating wavefront which controls the

velocity of the impulse and the dynamics of the reentrant wavefront 100.

The leading front is curved, in contrast to the leading circle model of reentry, which gives

the activation a spiral shape. An important concept to consider is the source/sink

dynamics of a propagating wavefront. For excitation to occur, a depolarizing wave (the

source) must carry sufficient current to bring membrane potentials to threshold for firing

(activating voltage-gated sodium channels) in electrically vulnerable “downstream cells”,

which act as a sink 101 102. The geometry of the wavefront influences the source/sink ratio.

Compared to a planar wavefront, the leading edge of a convex wavefront will have

Page 25: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

15

relatively few cells driving depolarization of many cells, shifting the balance to a greater

sink than source current. This results in reduced rate of voltage rise ahead of the

wavefront, increasing the time to sodium channel activation, effectively slowing CV. The

reverse is true in concave wavefronts, where CV is higher than that of a planar wavefront.

The curvature of the spiral wave is proposed to increase progressively towards the center,

where at the tip, the curvature achieves a critical value whereby the core becomes

inaccessible to excitatory activity. This steep wavefront slows the conduction velocity to

a critical level thus forming the PS. This allows for the meandering nature of rotors, as

the core is unexcited, but is not required to be refractory and therefore can subsequently

be excited 103. Finally, the rotor is not dependent on a fixed wavelength, as is required by

anatomic reentry and the leading circle model. In fact, electrotonus is predicted to

shorten the action potentials of cells near the core 104. This final point is of particular

importance given the previous discussion of the effect of antiarrhythmics on wavelength.

In the rotor model of fibrillation, wavelength is variable and dynamic, and thus

medications are predicted to have variable effects along the curvature of the wavefront as

the complement of currents at play will vary depending on the location.

The initiation of a rotor relies on wavebreak after the interaction of an electrical impulse

or wavefront with an obstacle 105. This process can occur in a homogeneous medium

when transient heterogeneity is introduced into the system. Classically this is performed

with an S1-S2 protocol where a first wave (S1) is followed by a second wave (S2)

oriented perpendicularly to S1 106. If S2 hits S1 before it has completely repolarized, S1

acts as a barrier to S2 propagation resulting in a rotating spiral wave. Alternatively, at

high stimulation rates, electrical and or calcium alternans can be induced where action

Page 26: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

16

potential duration and or the amplitude of calcium transients demonstrate beat-to-beat

variation despite a constant stimulation frequency 107.

Alternans is a risk factor for cardiac arrhythmias including atrial fibrillation, and in some

cases can act as a prognostic tool for arrhythmia risk stratification and therapy 108 109 110.

In cardiomyocytes, beat-to-beat regulation of the membrane potential (Vm) and cytosolic

calcium are bi-directionally coupled and are dependent on the kinetics of APD restitution.

APD restitution refers to the dependence of an action potential on the preceding diastolic

interval to influence its duration. At rapid rates, APDs continue to shorten along their

restitution curves until they reach a critical point where the curve becomes so steep that

self-sustaining oscillations of APDs can occur 111 112 113 114. This underlying mechanism

has been hypothesized to be related to the time-dependent recovery of ion channels from

inactivation 115. Similarly, the calcium transient and conduction velocity have restitution

kinetics which have been identified as potential contributors to alternans and fibrillation

116.

1.7.2 Clinical relevance of rotors in AF

Although not universally accepted, rotor theory has become an important mechanistic

explanation for AF as well as a potential therapeutic target. Multiple clinical observations

have supported the existence of focal AF drivers in a subset of patients. These include the

ability of a single ablation lesion to terminate persistent AF that is resistant to

cardioversion 117, spatiotemporal stability and localized regions of stable frequencies

using dominant frequency mapping 118 119 120 and reproducible vectors of AF propagation

over time 121.The main limitation to developing a mechanistic understanding of rotors in

human AF however has been the resolution of current mapping systems, namely their

Page 27: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

17

ability to differentiate the principal components of the signal from the disorganized

activation patterns. This has required significant mathematical post-processing to

identify areas of spatiotemporal reproducibility.

Recently data from a novel mapping system employing monophasic action potential

recordings and a physiologic noise filter has led to a rotor centred ablation strategy. The

Focal Impulse and Rotor Modulation (FIRM) mapping system purports to reveal stable

electrical rotors in a majority of individuals with AF. Using direct contact gold

electrodes attached to 64 pole basket catheters, both the right and the left atria are

mapped simultaneously. Electrodes are 4-6 mm apart along each spline and separated by

4-10 mm between splines 122. The software developed to interpret the tracings advertises

its ability to examine patient specific AF electrograms based upon rate-dependent

refractoriness, or as previously described, restitution, and conduction slowing. Rotors are

identified as PS and are targeted for ablation if they are stable for minutes 123. The

rationale for localized ablation lesions has been based on the theory of ablation of

microreentrant focal atrial tachycardias, for which ablation has proven successful.

The initial clinical trial, the CONFIRM trial, demonstrated a dramatic benefit in terms of

freedom from AF compared to conventional ablation (82.4% vs. 44.9%, p = 0.001) after a

median 273 days. This benefit has been shown to persist over 3-4 years follow up and

has been replicated in a multicenter experience recently reported 124 125. While there is

certainly growing data to support rotors as drivers of AF, the community of clinicians and

scientist are still mixed with respect to their belief in the ability of focal ablation lesions

to terminate AF and prevent recurrence. The hesitation is centered around the concept

that rotors are not stable, and ablation would promote anatomical re-entry and not

Page 28: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

18

necessarily prevent recurrence of functional re-entry. There is however no published

literature to date reporting the lack of efficacy using the FIRM technology and software,

and thus critical appraisal to date has been only anecdotal. What we have certainly

learned from our growing clinical experience mapping rotors is that they can be

demonstrated using a variety of technologies and are likely important targets for therapy.

A powerful tool for the mechanistic understanding of AF and development of novel

targets would incorporate what we have learned about atrial wavelength and its response

to medications over the past century, the mathematical and clinical observations pointing

to rotors as the major drivers of AF in a model that carries the cell and tissue specific

factors already highlighted to be important in AF initiation, propagation and recurrence.

Such a tool has only recently become available with the advent and application of human

pluripotent stem cell technology. While disease modeling using human pluripotent stem

cells would not be a novel proposition, as its application and validation have

disseminated rapidly to multiple inherited and acquired diseases in multiple organ

systems including the heart, there are several unique challenges to building a model of

AF using human pluripotent stem cells. First however, one must review the source of

pluripotent stem cells, the nature of directed differentiation, and the applications of this

technology to date.

1.8 Embryonic stem cells and induced pluripotent stem cells

Embryonic stem cells (ESCs) cells are derived from totipotent cells of the developing

embryo. They are capable of unlimited proliferation while maintaining their

undifferentiated state. Human blastocyst-derived pluripotent stem cell lines were first

derived in 1998 126. Cleavage stage embryos produced by in-vitro fertilization were

Page 29: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

19

donated by individuals after informed consent. Fourteen inner cell masses were isolated

and 5 ES cell lines derived from 5 separate embryos. These cell lines had normal

karyotypes, expressed surface markers that characterize primate embryonic stem cells,

and lacked markers defining early lineages. Specifically, the cell surface markers that

characterize undifferentiated ES cells including stage-specific embryonic antigen

(SSEA)-3, SSEA-4, TRA-1-60, TRA-1-81 were expressed, while the cell lines did not

stain for SSEA-1. After 5 months in culture, these cells retained their ability to derive all

three embryonic germ layers; endoderm, mesoderm and ectoderm. When injected into

severe combined immune-deficient (SCID)-beige mice, each injected mouse formed a

teratoma that included gut epithelium (endoderm), cartilage, bone, smooth muscle and

striated muscle (mesoderm) and neural epithelium, embryonic ganglia and stratified

squamous epithelium (ectoderm). When grown in vitro to confluence, ES cells

differentiated spontaneously.

Since the introduction of human stem cell lines, a second source for pluripotent stem cells

has arisen. In 2006, Takahashi and Yamanka demonstrated that adult skin fibroblasts in

mice can be reprogrammed into ES like cells by using a retroviral vector to force the

expression of 4 transcription factors: Oct3/4, Sox2, Klf-4 and c-Myc 127. Subsequent

studies were able to recapitulate these findings using the same 4 transcription factors in

human fibroblasts and showed that these cells, human induced pluripotent stem cells

(hiPSCs), were capable of self-renewal, could remain undifferentiated in culture, and

could give rise to all somatic cell types 128. The potential to generate an unlimited number

of any of the hundreds of cell types in the human body has innumerable applications in

the fields of biomedical research and regenerative medicine. The potential however

Page 30: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

20

cannot be actualized without the ability to control and drive the directed differentiation

into the cell type required.

1.8.1 Directed differentiation

Three basic differentiation methods have been developed and applied for the

differentiation of ESCs; the formation of 3-D aggregates known as embryoid bodies

(EBs), the culture of ESCs as cell sheets, and the culture of ESCs directly on stromal

layers 129. While early protocols using fetal calf serum were difficult to reproduce and

were generally poorly optimized for the generation of a single cell lineage, several

advances including the use of serum-free media with specific inducers, and the

development of reporter ESCs to monitor progression through the developmental and

differentiation steps have advanced the field 130 131 132 133 134 135 136 137. With these new

tools, it became possible to approach the problem of directed differentiation through the

lens of developmental biology, applying insights from other model systems towards the

goal of recapitulating human developmental milestones in vitro. For the purpose of this

discussion, the focus will be primarily on the EB culture method of generating

cardiomyocytes for disease modeling and regeneration.

Understanding the steps taken by early cardiac progenitors in the developing mouse

embryo has assisted in the identification of critical signalling pathways, gene expression

profiles and surface markers that direct in vitro differentiation of hESCs. Perhaps the

most important event in embryogenesis lies in gastrulation, when uncommitted epiblast

cells migrate through a transient structure known as the primitive streak and exit as either

mesoderm or definitive endoderm. Brachyury, a T-box transcription factor, is up-

regulated as the epiblast cells enter the primitive streak, and then rapidly down regulated

Page 31: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

21

as the newly formed mesodermal cells exit the primitive streak. Specification of distinct

subpopulations is controlled both temporally and spatially suggesting different signalling

environments responsible for lineage specification. Members of the TGFβ family

including BMP4 and Nodal as well as the Wnt family are essential pathways during these

developmental steps 138 139 140. These agonists work in concert with regional expression

of inhibitors to create domains that are conducive to specifying germ layer induction 141.

Subsequent studies demonstrated that the same signalling pathways can be manipulated

in vitro to regulate primitive streak development, setting the stage for germ layer

induction 129.

The early stages of mesoderm induction can be monitored by the up regulation of fetal

liver kinase-1 (Flk-1) and PDGF receptors 142,143. The induction of early cardiac

progenitors requires initially Wnt signalling and then its subsequent inhibition in order to

specify the mesoderm, while using various concentrations of BMP4 and activin may

generate different subpopulations 144,145. Cardiomyocytes are derived from the lateral

plate mesoderm and in two waves of development of cells marked by the VEGF receptor

Flk-1 and the transcription factor Nkx2.5 146,147. Mesodermal cells migrate to the anterior

region of the embryo and organize into the cardiac crescent 148. Ultimately the cardiac

crescent fuses to become the primitive heart tube which subsequently undergoes a

complicated series of changes, or looping, resulting in heart chamber formation. The

differentiation protocols have improved tremendously over the last decade resulting in

cardiomyocyte yields of 80-90% 149.

The study of the electrophysiologic properties of ES derived cardiomyocytes has given us

significant insights into their developmental stage, and has become an important

Page 32: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

22

functional assay in the search of differentiating cells towards further cell type specificity.

While many aspects of the human pluripotent stem cell (hPSC) derived cardiomyocytes

are comparable to adult cardiomyocytes, there are several notable differences. Principally

it is believed that these differences are attributable to the immature nature of these cells,

as they beat spontaneously, have unorganized sarcomeres, and have a different

complement of ion channels compared to adults 150. In particular, the excitation-

contraction machinery appears to be immature with hPSCs lacking clear T-tubules, and

displaying disorganized sarcomeric striations and immature Ca2+ handling 151,152. Time

has been shown to be an important aspect of maturation, with cells cultured for extended

periods of time acquiring modifications in current densities and properties, increased

expression of a “more mature” host of ion channels, and significant improvements in cell

morphology 153. No studies to date however have been able to demonstrate a dramatic

impact on the relatively immature AP shape, or the Ik1 expression felt to be at least in part

responsible for the relatively depolarized resting membrane potential compared to adult

cardiomyocytes 150.

We have also learned that standard protocols154,155to generate CMs from hESCs produced

mixed populations of atrial-, pacemaker- and ventricular-like cardiomyocytes. The

majority of cells derived carry a ventricular like action potential (AP) morphology (60-

90%), with a minority of cells exhibiting atrial or nodal like AP morphologies 156. There

has been a substantial amount of evidence, both in vivo and in vitro, implicating retinoic

acid (RA) signalling as an important player in atrial specification.

Page 33: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

23

1.8.2 Retinoic acid signaling

RA signalling has been shown to regulate anterior-posterior polarisation of the heart 157.

RA treatment of mouse and chicken embryos leads to oversized atria and smaller or

missing ventricles, while the opposite is true for embryos in which RA signalling is

inhibited at critical time points 158 159. Furthermore, studies of mouse embryonic stem

cells indicated that RA signalling promotes the expression of atrial-specific genes.

Finally, exogenous treatment of RA has been shown to direct the differentiation of hESCs

into atrial like myocytes 160.

RA is synthesized from vitamin A (retinol) through a chain of 2 oxidative sequences. In

the second step, which is thought to be rate limiting, all-trans retinaldehyde is converted

to all-trans RA by any of three related aldehyde dehydrogenases, Raldh1, Raldh2, and

Raldh3. Previous studies have implicated Raldh2 as the main Raldh involved in early

cardiac development 161. Our group hypothesizes that early cardiac progenitors that

express Raldh2 are programmed to synthesize and thus respond to RA, and that atrial

precursors and not ventricular precursors would thus express Raldh2. Since RA readily

diffuses across cell membranes, ventricular progenitors would have to “protect”

themselves from RA being synthesized by neighbouring cells, and could do so by

expressing an enzyme that degrades RA. Cyp26a1, a member of the Cyp26 family of

cytochrome p450 enzymes, converts RA to metabolites that are not bioactive.

Upregulation of Cyp26a1 has been shown to attenuate RA signaling in the prospective

rostral spinal cord of zebra fish, thus limiting the downstream signaling involved in the

expression of hox genes, and demonstrating its functional role in determining the

hindbrain-spinal cord boundary 162.

Page 34: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

24

The potential therefore exists to drive cardiac differentiation to specific cell fates as

would be required for disease modeling of atrial tissue. The validation of the successful

generation of a specific cell type will require multiple modalities in order to verify the

cell’s molecular and physiologic fingerprint. The tools that are currently relied on most

heavily are the quantitative reverse transcriptase polymerase chain reaction (qRT-PCR)

targeted analysis of RNA expression, flow cytometry, immunostaining and patch

clamping. Important for the purposes of this study will be the ability to differentiate the

two major cell lineages of cardiomyocytes in our cultures, atrial like cells and ventricular

like cells.

1.8.3 Molecular markers of atrial and ventricular cardiomyocytes

Recently, comparative proteomic and transcriptomic analysis of human fetal atria and

ventricular samples has been performed identifying and validating chamber enriched and

chamber specific proteins 163. Our group has focused on the following atrial enriched

markers: atrial natriuretic factor (ANF), a potassium inward rectifying channel (KCNJ3),

Connexin-40 (Cx-40), and the alpha 1D subunit of the L-type voltage-dependent calcium

channel (CaV1.3). ANF is encoded for by the gene natriuretic peptide A (NPPA) and

belongs to a family of natriuretic peptides involved in fluid and electrolyte homeostasis.

Mutations in NPPA have been associated with familial AF. KCNJ3 or Kir3.1, associates

with other G-protein-activated potassium channels to form a heterotetrameric pore

forming complex. These multimeric G-protein gated inwardly rectifying potassium

(GIRK) channels are important in atrial electrophysiology and can be activated by

muscarinic M2 receptors. Cx-40 is encoded for by the gene GJA5. Connexins form gap

junctions in the heart through which charged ions flow between neighbouring cells thus

Page 35: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

25

facilitating action potential propagation. The two most highly expressed connexins in the

heart are Cx-40 and Cx43, however Cx-40 is expressed in atrial cardiomyocytes and not

ventricular cardiomyocytes, and has been associated with a heritable form of AF. CaV1.3

is highly expressed in atrial compared to ventricular tissue, contributes significantly to the

repolarization process in human atria, and has been linked through genome wide

association studies to AF in humans 164.

In a similar fashion, our group has utilized the absence of ventricular markers myosin

regulatory light chain-2 (MYL2) and Iroquois-class homeodomain protein 4 (IRX4) to

mark atrial cardiomyocytes and their precursors. Ventricular myosin light chain-2 (MLC-

2v) refers to the ventricular form which plays a critical role in embryonic cardiac

development and function as well as representing one of the earliest markers of

ventricular specification 165. IRX4 expression is restricted to the ventricular precursors

during the embryonic formation of the linear heart tube, and is absent from both atrial and

outflow tract precursors 166.

1.9 Disease modeling using hPSCs

The careful dissection and directed differentiation of cardiomyocyte subtypes is a

relatively new and important step forward in the field of disease modeling using hPSCs.

Prior to this advance, investigators have generated predominantly ventricular like cells

and thus focused on diseases of the ventricles. The first patient specific disease model to

be studied in depth was the long QT syndrome in 2010 when hiPSC derived

cardiomyocytes were clearly shown to have the capacity to recapitulate the clinical

disease in a dish. Since then there has been an explosion of interest and innovation

around this novel technology167 and its application in the study of innumerable cardiac

Page 36: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

26

diseases from inherited arrhythmia syndromes to cardiomyopathies168-170. Disease

modeling with hPSCs has become an important part of the armamentarium for gene

discovery, its subsequent functional analysis, and drug testing. For the purpose of this

discussion, the lessons learned in modeling congenital and acquired long QT syndrome

will be reviewed in further detail, as they will act as the building blocks upon which this

thesis was designed and carried out.

1.9.1 cLQTS

The congenital long QT syndrome (cLQTS) is a life threatening disease that represents a

leading cause of sudden cardiac death in the young 171. The electrocardiographic features

of the disease are QTc prolongation and T wave abnormalities at rest, and failure of the

QTc to shorten with exercise and epinephrine 172. The QT interval represents the

depolarization and repolarization phases of the cardiac action potential, as previously

reviewed. Decreases in repolarizing outward potassium currents or increases in

depolarizing inward sodium or calcium currents can lead to prolongation of the QT

interval, and not surprisingly then, mutations in genes encoding ion channels have been

identified as the most common pathogenic variants. Approximately 1 in 2500 healthy

live births will have an abnormally long QT interval and have congenital LQTS,

transmitted via an autosomal dominant inheritance pattern 171.

Beta-blocker therapy is the primary treatment for most patients with LQTS and offers

substantial protection from fatal cardiac events 173-176. Patients who are intolerant or

refractory to beta-blockers can be offered left cardiac sympathetic denervation 177.

Patients who have cardiac events while on beta-blockers, who have suffered a cardiac

arrest, or who are deemed sufficiently high risk, can be offered an implantable

Page 37: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

27

cardioverter defibrillator (ICD)178-181. ICD therapy however has lifelong implications, and

complications are common especially in young patients who may have the device for >20

years. In a recent review of an academic tertiary center’s outcomes for primary

prevention ICDs, 35% of patients had ICD related morbidity, and none of their LQTS

patients received appropriate shocks for LQTS related arrhythmias 182.

The majority of efforts at improving risk stratification in patients with LQTS have

focused on our rapidly improving understanding of genetics. Consequently LQTS has

become one of the best-understood and characterized monogenic genetic diseases,

serving as a model for the investigation of genotype-phenotype interactions. This

mechanistic basis and understanding of disease has afforded clinicians an improved

patient specific management strategy. This has also given researchers a unique depth to

the understanding of a monogenic disease that often carries a clear, distinct, and easily

measurable phenotype. Mutations in ion channel genes causing QT interval prolongation

do so by prolonging the action potential duration. Delayed repolarization can facilitate

early after depolarizations (EADs), a process that has been proposed to relate to L-type

calcium channel reactivation, and is dependent not only on time but also on membrane

potential 183 184. It is however widely accepted that torsades-de-pointes (TdP) is triggered

by EADs under conditions of a prolonged QT interval 185. Drugs that further prolong the

APD can aggravate the underlying QT prolongation and put patients at risk for sudden

cardiac death.

The first inherited cardiac disorder to be modeled was a patient with type-2 LQTS due to

a missense mutation in the KCNH2 gene affecting the pore-forming region of the HERG

channel 186. This mutation leads to a significant reduction of the rapid component of the

Page 38: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

28

delayed rectifier potassium current (Ikr). After an iPS line was generated and

differentiated into beating cardiomyocytes, intracellular recordings revealed APD

prolongation compared to control cells as well as a significant reduction in Ikr current. A

significant proportion (66%) of LQTS iPSC derived cardiomyocytes displayed EADs,

whereas control cells did not. The additional stress of specific Ikr blockers prolonged the

APDs further and incited an increased number and complexity of EADs. Application of

potential therapeutic agents including calcium channel blockers and pinacidil, a KATP-

channel opener, resulted in APD abbreviation and elimination of EADs.

The observations in this iPS model of LQT2 are striking, however the implications are

limited by the investigation of a tissue based arrhythmia at a single cell level. Under

normal physiologic conditions, there is dispersion across the ventricles during

repolarization 187. While the cellular basis for this continues to be debated, one theory that

has gained popularity relies on the gradient of cell types across the myocardial wall.

From epicardium to myocardium, and possibly M cells in between, ventricular

cardiomyocytes differ in terms of their repolarization properties. The ionic determinants

of the differing properties of these cell types has also been proposed to underlie the

enhanced transmural dispersion of repolarization (TDR) under conditions of prolonged

QT interval that can predispose patients to TdP and sudden cardiac death. An increase in

TDR may be essential for the development of TdP, serving as a functional reentrant

substrate for its maintenance, as well as critically facilitating the propagation of an EAD

to generate the first initiating beat 188 189 . In the absence of TDR in canine and rabbit

ventricular wedge preparation models, TdP does not develop in the setting of QT

prolongation despite frequent phase 2 EADs. Further evidence dissecting the surrogate

Page 39: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

29

outcome of QT prolongation and EADs from the clinically relevant outcome of TdP is

the observation that the incidence of TdP is not proportional to the extent of QT

prolongation in drug-induced or acquired LQTS 185. Cardiologists have long since

learned the perils of treatment paradigms based on surrogate outcomes. Perhaps the most

notable lesson has come from the Cardiac Arrhythmia Suppression Trial (CAST),

designed on the premise that suppression of ventricular ectopy after a myocardial

infarction reduces the incidence of sudden cardiac death 190. Based on a well documented

observation that ventricular ectopy was associated with sudden death after myocardial

infarction, antiarrhythmics were commonly prescribed to suppress them until the CAST

trial definitively showed that this management strategy was associated with increased

mortality. Despite the clear limitations of single cell physiology studies and surrogate

outcomes, we have gained significant insights into the utility of hPSC technology for

disease modeling and drug screening from recent studies of congenital and acquired

LQTS.

1.9.2 acquired LQTS

New drug development suffers from low phase II clinical success rates, and a daunting

price tag of 1.8 billion dollars and 12 years of development, testing, and regulatory

processes spanning the time from discovery to commercial launch 191. The need for new

technology is highlighted further by the fact that the attrition rate of new therapeutics for

any disease condition is 89% after preclinical animal testing, with 1/3 being ineffective

and an additional 1/3 having safety issues192, with cardiac toxicity being the major factor

in this attrition193. Much of this attrition rate has been attributed to the poor predictive

power of animal models. Several high profile drug withdrawals have led to mandatory

Page 40: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

30

pre-clinical screening policies to detect compounds which may prolong the QT interval

and predispose patients to TdP 194. So called acquired QTc prolongation is much more

common than congenital and typically ascribed to drugs that incidentally block the IKr

channel, but can exert this effect through any number of mechanisms, and thus prolong

the ventricular action potential duration 195,196. Current FDA mandated methods rely on

cell lines artificially expressing single cardiac ion channels, or whole animal heart and

purkinje assays 197. These screens have obvious and important limitations, with poor

sensitivity and specificity profiles for human translation, and a host of untestable

electrophysiologic toxicities that are not considered 194. Pluripotent stem cell assays have

been shown to act as efficient, reproducible screens for the efficacy of new compounds as

well as the safety profile of compounds in development 198. With the advent and

explosion of genetic engineering techniques, hPSCs carry the unique privilege of being

the only human model system that can incorporate the breadth of genetic diversity of

patient populations, while also holding the promise to personalize medical therapy and

risk stratification at the level of a single patient.

The first major effort towards the development of hPSC based drug screening was

performed at the single cell level in hiPSCs derived from healthy subjects and patients

considered to be at higher risk of developing drug induced QT prolongation. The high

risk population drew from cohorts of patients with congenital long QT syndrome, familial

hypertrophic cardiomyopathy and familial dilated cardiomyopathy 197. Cardiomyocytes

generated from high risk hiPSCs demonstrated increased susceptibility to drug induced

APD prolongation, early and delayed after depolarizations compared to control.

Subsequently, investigators from the same group generated a library of genome edited

Page 41: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

31

iPSC derived cardiomyocytes, with mutations again representing the highest risk patient

populations, and compared the effects of drugs to isogenic controls. The application of

genetically modified human cell lines demonstrated clear utility in efficient high

throughput screens for small molecules and chemical compound testing 198. Of course,

the same limitation ascribed to the LQT2 model previously described applied to single

cell drug screening assays, screening for a surrogate markers (EADs) of the actual feared

clinical outcome (TdP).

The atria and the ventricles share the same fundamental electrical properties at a single

cell level that impact the action potential profile, and at a tissue level when considering

electrical wave propagation and repolarization. Accordingly similar principles apply to

understanding arrhythmias in the two chamber types, specifically, the determinants of

wavelength, and the clinical patterns of arrhythmogenesis including fibrillation involving

reentry, conduction block, and spiral wave formation (i.e. rotors). However, there are key

differences in the physiology and pathophysiology of atria and ventricles, principally

their electrophysiologic signatures and their cell to cell communication, that can be

exploited for therapy, and tested for safety. Unfortunately, our understanding of the

mechanisms of atrial arrhythmias remains dismal and treatment approaches are grossly

ineffective and unsafe 199. One of the major barriers in the pipeline of drug development

has been the lack of appropriate models. The challenges of interspecies differences in

receptor subtypes, distribution and signaling200 are compounded by significant

differences in the ion channels and gap junctions201,202 that work in concert to dictate the

physiology and pathophysiology involved in human AF 102. The major objective of our

studies is to develop a novel platform, using human pluripotent stem cells (hPSCs), to

Page 42: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

32

study the pathophysiology of AF in order to identify novel targets for therapy and as a

platform to study the mechanisms required for proper treatment and prevention of AF.

Page 43: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

33

Chapter 2

Hypotheses and Research Aims

This study proposed to generate an in vitro model of AF using human pluripotent stem

cells. Building on the work to date from the Keller lab and published literature, we

generated cardiomyocytes from hESCs that carry the molecular and electrophysiologic

hallmarks of atrial cells. The differentiation scheme developed relies principally on

retinoic acid signaling in driving the atrial cell fate, as well as fine tuning of the BMP and

Activin signaling pathways in order to generate cells that mimic early atrial precursors.

Once validated at the single cell level, using hESC derived ventricular-like cells as

controls, we have moved forward in generating a simple tissue model in the form of a

multicellular sheet. The atrial chambers are thin compared to the ventricles, with a mean

thickness of 1-2 mm compared to ~15 mm, and thus multicellular sheets may be a

reasonable approximation of atrial tissue. This platform has the potential to facilitate

optical mapping of the electrophysiologic properties of the atria under physiologic and

pathophysiologic conditions. Thus our hypothesis is that multicellular sheets of hESC

derived atrial cardiomyocytes will serve as a reliable model of human AF and has the

potential to act as a tool to understand the mechanism of action of anti-arrhythmic drugs,

and to study and screen for novel therapeutics. Based on this hypothesis, the objectives

of this study are the following:

1. Generate atrial like cardiomyocytes from hESCs through the manipulation of Activin

and BMP signaling pathways and the addition of retinoic acid

2. Generate atrial like tissue in the form of cell sheets

3. Induce atrial fibrillation in the cell sheets

Page 44: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

34

4. Develop on optical mapping setup, and the requisite signal processing software,

capable of monitoring the electrophysiologic properties of cell sheets during sinus rhythm

and AF

6. Test the effects of AADs that are commonly used for the treatment of human AF on

the cell sheets using the acquisition setup and signal processing

Page 45: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

35

Chapter 3

Materials and Methods

3.1 Generation of cardiomyocytes from hESCs

HES3 NKX2-5egfp/w cells were maintained on irradiated mouse embryonic feeder cells in

hESC media consisting of DMEM/F12 (50:50;MEdiatech, Hedndon, VA) supplemented

with 20% knock-out serum replacement (SR), 100 μM nonessential amino acids, 2mM

glutamine, 50U/mL penicillin, 50 μg/ml streptomycin (Invitrogen Grand Island, NY), 10-

4 M Β-mercaptoethanol (Sigma, St Louis, MO), and 20 ng/mL hbFGF (R&D Systems,

Minneapolis, MN) in 6-well tissue culture plates 203. Cells were passaged to new feeders

as single-cell suspensions following dissociation with TrypLE (Life technologies).

Embryoid bodies for differentiation were generated after feeder depletion and growth of

ES cell colonies to 80% confluence. Single cell suspensions were formed after

dissociation with TrypLE. Embryoid bodies were formed by plating small aggregates at a

concentration of 500,000 cells/ml in 2 ml basic media containing StemPro34 (Invitrogen),

2mM glutamine, 4 X 10-4M monothioglycerol (MTG), 50 μg/ml ascorbic acid (Sigma),

and 0.1 ng/ml BMP-4 (R%D Systems). The aggregates were incubated at 37°C in a

hypoxic environment of 5% CO2, 5% O2, and 90% N2 on a rotator set at 70 rpm for 24

hours. Following this, the aggregates were spun in a centrifuge at 300 rpm, washed using

IMDM, and then re-suspended in the induction media.

The following concentrations of factors were used for the ventricular (control)

differentiation protocol: BMP-4, 10 ng/ml; human bFGF, 5 ng/ml; activin A, 6ng/ml;

IWP2,0.5 ng/ml; and human VEGF, 10 ng/ml. This is referred to subsequently as

10B/6A, control, or “ventricular” differentiation protocol. The factors were added with

Page 46: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

36

the following sequence: days 1–3, BMP4, bFGF and Activin A; days 3–5, VEGF and

IWP2; day5-12 VEGF, and days 12-20 only backbone media as previously described.

Media changes were performed by centrifuging the EBs at 800 rpm and washing the EBs

with IMDM. Cultures were maintained in a hypoxic 5% CO2/5% O2/90% N2

environment for the first 12 days and then transferred to a normoxic 5% CO2/air

environment.

The atrial differentiation protocol was carried out as above, with the following changes;

BMP-4 and Activin were titrated down to 3 ng/ml and 2 ng/ml respectively. On day 3,

retinoic acid (RA) was added to the culture at a concentration of 0.5 ng/ml. Other than the

addition of RA, this protocol only differed from the control protocol in terms of the

concentrations of BMP4 and Activin used at T3 employing lower concentrations. This is

referred to in text and figures as 3B/2A +RA, or “atrial” differentiation protocol.

3.2 Flow Cytometry and Cell Sorting

For cell-surface antigens, staining was carried out in PBS with 3% FCS. For intracellular

antigens, staining was carried out on cells fixed with 4% paraformaldehyde in PBS.

Staining was done in PBS with 3% FCS and 0.5% saponin (Sigma). Cells were stained at

a concentration of 2.5 × 106 cells/ml with anti-KDR- APC (R&D Systems; 1:10) and

anti-PDGFRA– PE (R&D Systems; 1:20), anti-SIRPA–PE-Cy7 (clone SE5A5;

BioLegend; 1:500), anti-CD90-APC (BD Pharmingen, 1:2000anti-CTNT (clone 13-11;

Thermo NeoMarkers; 1:400), goat anti-mouse IgG–APC(BD; 1:200). Cells assayed for

Aldefluor (STEMCELL Technologies) were prepared based on manufacturers

instructions. Incubation with the Aldefluor reagent was 45 minutes. Stained cells were

analyzed on an LSRII flow cytometer (BD Biosciences). For FACS, the cells were sorted

Page 47: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

37

at a concentration of 106 cells/ml in IMDM/5% FCS using a FACSAriaTMII (BD

Biosciences) cell sorter (SickKids-UHN Flow Cytometry Facility). Data were analyzed

using FlowJo software (Treestar).

3.3 Molecular Markers

3.3.1 Quantitative Real-Time PCR (qRT-PCR)

Total RNA was prepared with the RNAqueous-Micro Kit (Ambion) and treated with

RNase-free DNase (Ambion). RNA (500 ng to 1 μg) was reverse transcribed into cDNA

using random hexamers and Oligo(dT) with Superscript III Reverse Transcriptase

(Invitrogen). qRT-PCR was performed on a MasterCycler EP RealPlex (Eppendorf) using

QuantiFast SYBR Green PCR Kit (Qiagen). Expression levels were normalized to the

housekeeping gene TATA binding protein (TBP).

3.3.2 Immunostaining

Immunostaining was performed using the following antibodies: Polyclonal rabbit anti-

MLC2V (Synaptic systems, 1:1000) with the secondary antibody anti-rabbit rabbit IgG

Cy3 (Jackson ImmunoResearch), and mouse anti-CTNT (Thermo NeoMarkers; 1:100)

with the secondary antibody anti-mouse IgG Alexa647 (Invitrogen). DAPI (Life

Technologies, SlowFade Gold) was used to counterstain nuclei. The stained cells were

visualized using a fluorescence microscope (Leica CTR6000) and images captured using

the Leica Application Suite software.

3.4 Cardiac Electrophysiology

3.4.1 Single Cell Electrophysiology

Spin EBs resulting from control and RA-treated differentiations were dissociated at day

20 to single cells using type B collagenase and TrypLE. They were sorted based on

Page 48: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

38

DAPI-/SIRPA+/GFP+/CD90- surface markers as previously described, generating a pure

population of cardiomyocytes. Single cells were plated on matrigel-coated coverslips.

Electrophysiological measurements were performed 7-10 days after dissociation and

plating.

Spontaneous action potentials (APs) were recorded using the patch clamp technique in

the whole cell configuration 204..

APs were recorded at room temperature (22-23 °C), and

were not corrected for the calculated liquid junction potential 205.

Pipettes were pulled

from borosilicate glass (with filament 1.5 mm OD, 0.75 mm ID, Sutter Instrument

Company) using a Flaming/Brown pipette puller (model p-87, Sutter Instrument

Company) and heat polished. The resistance of these pipettes was 8-10 MΩ when filled

with recording solution. Micropipettes were positioned with a micromanipulator

(Burleigh PCS-5000 system) mounted on the stage of an inverted microscope (Olympus

IX70). Seal resistance was 2-15 GΩ. Myocytes were placed into and perfused with a bath

solution of Tyrode’s containing (in mmol/L) 140 NaCl, 2.5 KCl, 1.5 CaCl2, 1 MgCl2. 10

HEPES and 5.5 D-glucose with pH adjusted with NaOH to 7.4. Standard AP internal

solution was used to fill the pipettes which consisted of (in mM) 100 K aspartate, 10 KCl,

3 NaCl, 5 NaHCO3, 3 KHPO4, 1 MgCl2, 5 MgATP, 10 HEPES, 75 μM EGTA, with pH

adjusted to 7.2 with KOH.

Membrane potential was controlled with an Axon headstage (CV 203BU) connected to

an Axopatch 200B voltage-clamp amplifier (Axon Instruments, Foster City, CA). Data

was digitized (Axon Digidata 1320A) and acquired using Axon Clampex software

(pClamp version 8.2.0.232). Prior to cell attachment and formation of a GΩ seal,

electrode potential was adjusted to a baseline of zero current. An agar-salt bridge was

Page 49: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

39

used as the reference electrode. Rupture of the cell membrane patch was achieved using

pressure disruption by brief suction. Following rupture of the cell membrane, the

membrane capacitive transient was elicited by small depolarizing voltage pulses. Data

was analyzed using Clampfit (Molecular Devices, Sunnyvale, CA, U.S.A).

3.4.2 Micro Electrode Array (MEA) electrophysiology

MEA chips were cleaned, autoclaved, and coated with dilute matrigel overnight at 37 °C.

hESC-CMs were digested using collagenase B at a concentration of 250 U/ml in Hank’s

balanced solution (Life technologies) overnight at a concentration of 5 million cells/ml.

TrypLE was used to create single cell suspensions which were sorted based on DAPI-

/SIRPA+/NKX2-5 GFP+/CD90- surface markers as previously described, generating a

pure population of cardiomyocytes. Single cells were plated on standard 60 electrode

MEAs at a concentration of 1-2 million cells per 150 μL drop in backbone media plus 1

ng/ml ROCK inhibitor (Y-27632 Dihydrochloride Hydrate,Toronto Research Chemicals,

cat.no Y100500) generating cell sheets that were 1-1.5 cm in diameter. Extracellular

recording was performed using a MEA1060INV MEA amplifier (Multi Channel Systems,

Reutlingen, Germany). Output signals were digitized at 10 kHz by use of a PC equipped

with a MC-card data acquisition board (Multi Channel Systems). Standard measurements

were performed in IMDM supplemented with NaCl (final concentrations in mmol/L: 140

NaCl, 3.6 KCL, 1.2 CaCl2, 1 MgCl2. 10 HEPES and 5.5 D-glucose). During recordings,

temperature was kept at 37 °C. Data were recorded using Cardio2D+ (Multi Channel

Systems,) and analyzed off-line with Cardio2D (Multi Channel Systems).

Page 50: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

40

3.4.3 Optical Mapping Electrophysiology

150 μL of 100% concentrated matrigel drops were placed in the centre of 35 mm tissue

culture treated petri dishes (Falcon). The dishes were kept on ice for 30 minutes and then

the matrigel was removed leaving a thin coat of matrigel with a diameter of 1 cm. The

petri dishes were incubated at 37 °C overnight. hESC-CMs were digested using

collagenase B at a concentration of 250 U/ml in Hank’s balanced solution (Life

technologies) overnight at a concentration of 5 million cells/ml. TrypLE was used to

make single cell suspensions which were sorted based on DAPI-/SIRPA+/NKX2-5

GFP+/CD90- surface markers as previously described, generating a pure population of

cardiomyocytes. Single cells were plated on the matrigel coated petri dishes at a

concentration of 1.25 million cells per 150 μL drop in backbone media plus 1 ng/ml

ROCK inhibitor (Y-27632 Dihydrochloride Hydrate,Toronto Research Chemicals, cat.no

Y100500) generating cell sheets of 1 cm diameter. To improve the stability of the cell

sheetsand maintain confluence, various percentages of CD90+ cells generated from the

sort were spiked into the cell sheet, from 5-10%. Cell sheets were incubated under

normoxic conditions at 37 °C for 2-3 weeks prior to imaging. Once the differentiation

was optimized to generate 80-90% cardiomyocytes consistently, cells were digested into

single cells, using the aforementioned techniques, and plated directly on the matrigel as

cell sheets without sorting. This ultimately had the effect of generating confluent cell

sheets with greater consistency.

Optical mapping was performed using the voltage sensitive dye

AminoNaphthylEthenylPyridinium (Di-4-ANEPPS)(Life Technologies). Di-4-ANEPPS

is an ampiphilic compound with two hydrocarbon chains that allow it to anchor into the

Page 51: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

41

plasma membrane. When bound to the membrane, the chromophore aligns

perpendicularly to the membrane/aqueous interface and undergoes reduction in emission

intensity(~10% per 100 mV depolarization) when excited at 540 nm and monitored at

680 nm emission206. The incubation was optimal when cold Di-4-ANEPSS (on ice) was

applied to the cell sheets and then incubated for 30 minutes at 10 μM. Blebbistatin, an

inhibitor of the adenosine triphosphatases (ATPases) associated with class II myosin

isoforms in an actin-detached state, was employed to stop cardiomyocyte contraction in

an effort to avoid motion artifact. Blebbistatin has high specificity for Myosin II and has

been demonstrated previously to be an effective excitation-contraction uncoupler,

immobilizing cardiac preparations without affecting AP morphology and intracellular

calcium handling207. Blebbistatin (Sigma) was used at a concentration of 10 μM.

Imaging was performed in IMDM supplemented with NaCl (final concentrations in

mmol/L: 140 NaCl, 3.6 KCL, 1.2 CaCl2, 1 MgCl2. 10 HEPES and 5.5 D-glucose). The

Di-4-ANEPPS and blebbistatin were washed and replaced with the same IMDM

containing 2 μM Di-4-ANEPPS. The petri dish was placed on a plate warmer

constructed using copper piping sandwiched by 2 aluminum plates. The copper piping

was connected to a water bath heater and pump with rubber tubing. Sylgard was used to

coat the aluminum plate where a well had been created and sized for the 35mm petri dish.

This generated even heating across the medium in the petri dish to a temperature of 36-

37°C.

The tissue was illuminated using a mercury light source (X-Cite Exacte, Lumen

Dynamics, Mississauga, ON, Canada) with a 525 ± 50 nm band-pass filter. Fluorescent

light was collected using a 645 ± 75 nm band-pass filter. Images were captured using an

Page 52: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

42

electron multiplying charge coupled device (EMCCD) camera (Cascade 128+, Cascade

Evolve, Photometric, Tucson, AZ, U.S.A) connected to an Olympus MVX-10 upright

microscope (Center Valley, PA, U.S.A) equipped with a 0.63x c-mount adapter and a

0.38x lens relay. Frames were captured at 1408 fps, at 2x2 binning and 64x32 pixels

resolution, or 522 fps at 1X1 binning, using Image Pro Plus (Media Cybernetics,

Rockville, MD, U.S.A) software. Stimulation and sensing electrodes were constructed

from platinum wire coated with Sylgard for electrical insulation. Stimulation electrodes

were designed in a bipolar configuration in order to achieve point stimulation. These

electrodes were connected to Pulsar 6i & 6b stimulators (FHC Inc, Bowdoinham, ME,

U.S.A) for programmed stimulation. Capture threshold was determined either by direct

visualization of cell sheet contraction, or simultaneous optical mapping of signals to

confirm rate. We were able to obtain 100% capture on the majority of cell sheets using

40mV with a pulse duration of 2 ms. Burst (“on and off”) pacing at a cycle length of 50

msec for 1-2 minutes was used for AF induction. AF induction was defined by rotor

initiation in a cell sheet that had demonstrated nodal activation at baseline. Sequences of

2048 frames were captured serially to maximize data acquisition while minimizing

photobleaching.

3.4.3.1 Signal Processing and Data

Optical action potential duration (OAPD) measurements were performed using ImageJ,

an open source image processing program designed for scientific multidimensional

images. Images were analyzed using minimal processing as defined as an averaging of

the image sequences which were then appended, and the unprocessed images were

subtracted from the processed images. This processing, in addition to removing the

Page 53: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

43

background, enhanced the pixel intensity changes associated with depolarization to aid in

analysis. In other words, for every pixel, let I(x,y) be the input. Let O1 be the first stage

of output. Then O1(x,y) = Iave(x,y) - I(x,y) + 10000, where Iave(x,y) is the time average of

pixel(x,y) and I(x,y) is the intensity of the individual pixel.

Optical APs were generated by measuring stacks over regions of interest (ROIs) using

both raw data and minimal processing as previously defined. When recordings had

suitable signal to noise ratios, 1 pixel was used to generate optical APs. When the signal

to noise ratio was less favourable, ROIs were selected, up to an area of 10 pixels, over a

region of the cell sheet that was in a single phase, meaning the activation wavefront

would cross the ROI at the same time in each pixel included.

In order to generate activation maps and conduction velocity maps, recordings were

analyzed using Scroll software (courtesy of Sergey Mironov). Processed images were

subjected to temporal and spatial filtering to reduce high frequency noise. The activation

time (the time of the first derivative of the flurorescence signal corresponding to the

steepest segment of the optical AP) was defined for each pixel first, followed by

generation of the activation map and the isochronal activation map. The activation time

delay between two adjacent neighbours in the vertical, or horizontal components divided

by the distance between the two neighbours was used to calculate the vector gradient for

each pixel. The resultant local gradient vector was then reciprocated to obtain the

conduction velocity vector magnitude, and inversed to gain conduction velocity vector

direction. The resultant vector velocity map was then used to guide ROI analysis of

conduction velocity. The cell sheet was divided into tertiles based on their distance from

the focus of electrical generation and propagation in the cell sheet (either nodal or the

Page 54: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

44

centre of a rotor, identified as the phase singularity). ROIs were taken across each tertile,

and then combined to generate a mean and standard deviation of conduction velocity

across the cell sheet.

Generating APD maps: Advanced Signal Processing

Due to the low S/N ratios, further processing was required. The first filtering was spatial.

Either of the following were used: 7x7 kernel averaging or bandwidth filtering using a 2D

FFT. The 2D FFT implementation was that given by ImageJ. The bandwidth used was 8 -

64 pixels. Let O2 be the 2nd output stage.

O2(t) = spatial filtering(O1(t))

A temporal filtering was also required. Pixel-by-pixel, this was done by taking the FFT of

O2. Zeroing the coefficients associated with higher frequencies k as well as the negative

frequencies, and then reconstructing the signal with an IFFT. The k = Cutoff frequency

was chosen to be as high as possible that will enable a good estimation of the action

potential duration. The cutoff frequency ranged from 5Hz to 58Hz. In all cases, this

meant including the fundamental frequency and several of its harmonics to many of its

harmonics. Let O3 be the third output stage.

O3(x,y) = IFFT(0 to cutoff frequency of O2(x,y))

Depending on the amount and type of filtering the baseline of O3 may have a low

frequency component. A filtering is performed by using the rolling ball algorithm to

establish the baseline. In this variation of the rolling ball algorithm, the height of the ball

is scaled to the signal swing O3 as the ball is moved temporally beneath the signal. The

radius of the ball is tweaked between 128 to 256 time points to establish a baseline that

was manually checked for accuracy.

Page 55: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

45

O4(x,y) = rolling ball baseline filtering(O3(x,y))

The APD map was generated from O4. The measurement was taken from the beginning

of the upstroke (moving away from the baseline) to the first occurrence of the data point

less than half of the peak value.

3.4.4 Intracellular Recordings of Cell Sheets

Micropipettes were pulled from borosilicate glass (thin walled pipettes measuring 1.2mm

outer diameter, 0.75 mm internal diameter, and thick walled pipettes measuring 1.2 mm

outer diameter and 0.4 mm internal diameter, WPI, Sarasota, FL, U.S.A) using a

Flaming/Brown pipette puller (model p-87, Sutter Instrument Company, Novato, CA,

U.S.A). Pipettes were filled with 3M KCl solution and the resistance was ~30 MΩ for

thin walled pipettes and 80-100 MΩ for thick walled pipettes. High resistance

microelectrodes were required so as to minimize cellular damage, and minimize K+

leakage. Pipettes were pulled with wispy, long shanks that allowed for flexibility of the

pipette when impaled in thin tissue. A vacuum chamber was used after filling to remove

bubbles. Pipettes were positioned using course manipulators (Newport, Irvine, CA,

U.S.A) and recordings were acquired at 1000 Hz using an Axopatch 200B amplifier with

a MiniDigi 1A digitizer and Axoscope acquisition software (Molecular Devices,

Sunnyvale, CA, U.S.A). Impalement of tissue was performed by first positioning the

microelectrode on the surface of the tissue which was evident by a drop in the potential

recording.

Page 56: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

46

Chapter 4

Results

4.1 Generation of cardiomyocytes from hESCs

We employed the HES3 line of hESCs which had sequences encoding enhanced GFP

(eGFP) introduced into the NKX2-5 locus by homologous recombination (NKX2-5egfp/w)

which has previously demonstrated its utility and reliability in faithfully reporting

endogenous Nkx2.5 expression 208. The homeobox gene Nkx2-5 is the earliest known

marker of vertebrate heart development, expressed in early cardiac progenitor cells and

through adulthood209. This line was chosen to facilitate quantification of cardiac

differentiation and purification of cardiomyocytes when performing single cell analyses.

This human ES cell line expressed cell surface makers that characterize undifferentiated

human cells including stage-specific antigen (SSEA)-3, SSEA-4, TRA-I-60, TRA1-81.

These cells did not stain strongly for SSEA-1 (s 1) 126.

Figure 1: Flow cytometric analyses of the frequencies of cell surface markers used routinely to identify undifferentiated hESCs; stage specific embryonic antigen 3 and 4 (SSEA3, SSEA4) and two human EC cell antigens Tra-1-60 and Tra-181 also used to mark undifferentiated hESCs. SSEA1 is expressed on differentiated hESCs. Flow cytometry performed after 5 days of hESC culture on MEFs at the time point when cells are dissociated for the T0 of differentiation. Here we show, based on gates set on the unstained sample on the far left, high frequencies of makers of undifferentiated hESCs and a low frequency of a marker of differentiated hESCs.

Page 57: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

47

To generate cardiomyocytes from hESCs, we employed the embryoid body staged

differentiation protocol that involved the formation of a primitive streak like population

defined by T (BRACHYURY) expression (days2-4), the induction and specification of

cardiac mesoderm (MESP1; days 3 and 4), and the expansion of cardiovascular lineages

154. This protocol uses the combination of Activin and BMP at the primitive streak and

mesoderm stage, and then the addition of the WNT inhibitor IWP2 as well as retinoic

acid (RA) to the developing cardiomyocytes (Figure 2). As previously mentioned, RA

signaling is crucial for atrial chamber development in vivo, and its activation has been

shown to effectively drive atrial cardiomyocyte differentiation in mouse and human ESCs

161 158 210 160.

Figure 2: Scheme of the protocol used to differentiate hESCs towards the cardiomyocyte lineage highlighting the three main stages of development: 1) mesoderm induction, 2) cardiovascular specification and 3) maturation. Protocols for the generation of “ventricular” and “atrial” cardiomyocytes differed in terms of the concentration of Activin and BMP4 used (high vs. low respectively) and whether or not retinoic acid (RA) was added to the cultures at T3. RA is added to the low Activin and BMP4 induction protocol in order to generate “atrial” cardiomyocytes as discussed in the text.

Our group has gone on to show that the manipulation of the BMP and Activin signaling

pathways can generate two distinct cardiac precursors as defined by their expression of

Page 58: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

48

either Raldh2 (“atrial” precursosrs), or upregulation of Cyp26a1 (“ventricular”

progenitors) in a temporal pattern consistent with developmental cues (Figure 3). The

first protocol uses lower levels of BMP4 and Activin during mesoderm induction

(3B/2A), while the second uses higher concentrations (10B/6A) as discussed in the

Methods section. The 3B/2A protocol displayed a higher relative expression of RALDH2,

the main Raldh involved in early cardiac development. The 10B/6A protocol produced a

population of cells that upregulated , Cyp26a1, the cytochrome p450 enzyme that

degrades RA.

Figure 3: qRT-PCR results comparing the relative expression of ALDH1A2 and CYP26A1 over time in a protocol incorporating low levels of BMP4 and Activin (3B/2A) and a protocol with higher BMP4 and Activin (10B/6A). This highlights the different cell types generated through the manipulation of the strength of signaling in these pathways. While both protocols were previously demonstrated to make cardiomyocytes at high frequencies, the 3B/2A protocol generated a population of cells with significantly higher expression of ALDH1A2 (the gene encoding RALDH2) than the 10B/6A protocol. In parallel, the 10B/6A protocol generated a population of cells with significantly higher CYP26A1 (the cytochrome p450 enzyme that degrades RA). These graphs highlight the importance of the careful manipulation of the BMP4 and Activin pathways in directing differentiation of a cell population poised to generate and employ RA (3B/2A), in contrast to a cell population that upregulates an enzyme to protect itself from RA exposure (10B/6A). Unpublished data reproduced with permission from Dr. Stephanie Protze and Jeehoon Lee. To validate the function of the two systems, our group employed a commercially

available assay known as Aldefluor 211. Briefly, cells are incubated with a fluorescent

aldefluor substrate that when oxidized by an aldehyde dehydrogenase can no longer

Page 59: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

49

efflux from a cell and becomes effectively trapped. The amount of fluorescence exhibited

reflects the activity of aldehyde dehydrogenase in the cell. The addition of 4-

diethylaminobenzaldehyde (DEAB), which is a potent inhibitor of Aldh enzymes,

prevents the enzymatic oxidation of this product thereby providing a negative control for

fluorescence. Our group demonstrated that the activity of aldehyde dehydrogenases was

restricted to a population of cardiac precursors exposed to lower concentrations of BMP4

and Activin, suggesting that this population of cells not only expresses Raldh2, but that

the enzyme is functional in these cells and not in the “ventricular” precursors (Figure 4).

Figure 4: Flow cytometric analyses plotting the frequencies of aldefluor positive cells against PDGFRα positivity (a marker of cardiac progenitor cells). Using the aldefluor assay, cells that express high levels of aldehyde dehydrogenase will fluoresce, in this study used as a potential functional marker of RALDH2 activity. DEAB, a potent inhibitor of Aldh enzymes, serves as a negative control. Once again, two protocols are compared incorporating either low levels of BMP4 and Activin (3B/2A) or higher BMP4 and Activin (10B/6A). While both protocols generate a high frequency of PDGFRα+ cells (an early marker of cardiomyocyte differentiation efficiency), only the 3B/2A protocol generates a high frequency of aldefluor+ cells. This functional assay reiterates the importance of precise manipulation of the BMP4 and Activin signaling pathways in patternings cells that have the machinery necessary to generate and thus utilize RA. Unpublished data reproduced with the permission of Jeehoon Lee and Dr. Stephanie Protze.

Page 60: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

50

In order to monitor and optimize the efficiency of the differentiation protocol, several

surface makers were monitored at key commitment stages. The induction of cardiac

mesoderm was monitored by the temporal expression of CD56, KDR (Flk-1) as well as

PdgfR-α. CD56/NCAM marks the differentiation of early mesodermal progenitors from

hESCs. These mesodermal progenitors however are multipotent, giving rise to many

mesodermal lineages including cardiomyocytes 212. KDR/Flk-1 has demonstrated its

utility in marking the induction of cardiac mesoderm in mouse and human ESCs,

however it is also expressed in different mesoderm populations 142. PdgfR-α is co-

expressed with Flk-1 in the cardiac mesoderm at embryonic stages and is found on

cardiac progenitor cells in the cardiac crescent 213 143. Optimizing the induction steps to

generate greater than 60% Flk-1+ PdgfR-α + cells generates cultures of highly enriched

cardiomyocytes 155. The earliest time point at which we monitor mesodermal induction is

at day 3 or day 4, looking for the emergence of a double-positive population (>80%) of

CD56+ and Pdgfr-α+ cells. The generation of cardiomyocytes was quantified using flow

cytometry by staining of cardiac troponin T (cTNT) and the surface marker SIRPA,

which has previously been shown to uniquely mark the cardiomyocyte lineage in hPSC

differentiation cultures (Figure 5) 214.

Page 61: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

51

Figure 5: Flow cytometric analyses plotting the frequencies of several important markers used in the optimization and validation of the differential protocols employed. The plot on the left demonstrates an optimal profile on day 4 with a high frequency of CD56+ and PdfR-α+ cells. The middle plot is representative of a good differentiation protocol which generates a high frequency of SIRPα+ cells. The frequency of CD90+ cells is also routinely assessed, and employed to optimize purification of cardiomyocytes, to quantify the frequency of CD90+ cells in the cell sheet culture, or to sort out CD90+ cells and then incorporate them into the cell sheet at controlled frequencies. The final plot on the right demonstrates an efficient differentiation protocol generating 91.9% cTNT+ cells.

4.2 Molecular markers of “atrial” and “ventricular” cardiomyocytes

Differentiation cultures were analyzed for atrial and ventricular specific markers with and

without the addition of RA using qRT-PCR (Figure 6). Cultures that were exposed to RA

compared to cultures that were not exposed to RA were enriched in the atrial specific

markers ANF (148.3 ± 9.4 vs. 16.4 ± 3.9, p < 0.0001, n = 4), KCNJ3 (0.58 ± 0.16 vs.

0.046 ± 0.01, p = 0.01, n = 4), Cx-40 (1.6 ± 0.4 vs. 0.019 ± 0.01, p <0.01, n = 5), and

CaCNA1d (13.0 ± 1.5 vs. 5.6 ± 1.0, p <0.01, n=5). Cultures that were exposed to RA

expressed significantly less ventricular specific markers compared to cultures that were

not exposed to RA including MLC2V (0.63 ± 0.3, p <0.01, n =5), and IRX4 (0.037 ±

0.02 vs. 0.46 ± 0.1, p <0.01, n =5).

Page 62: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

52

Figure 6: qRT-PCR-based expression analyses of a control protocol (10B/6A) generating “ventricular” cardiomyocytes and the RA protocol (3B/2A+RA) incorporating lower levels of BMP4 and Activin as well as RA. Cell populations were analyzed at T20 of the differentiation protocol. The RA protocol was able to generate cells that are enriched in atrial specific markers (NPPA, KCNJ3, and GJA5) and lack ventricular markers (MYL, IRX4). Values are shown relative the housekeeping gene TBP. Error bars represent standard deviation of the mean from the values of independent experiments (N≥4); *P≤0.05, **P≤0.01, ***P≤0.001 as analyzed by Student's T-test. Unpublished data reproduced with the permission of Jeehoon Lee and Dr. Stephanie Protze.

Immunostaining confirmed the qRT-PCR findings and demonstrated that RA treated cells

expressed cTNT, but did not express MLC2v, whereas control cells expressed both

MLC2v and cTNT (Figure 7).

Page 63: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

53

Figure 7: Fluorescent immunostaining for the presence of MLC2v and cTNT in day 20 populations. DAPI staining shows cell nuclei. On the left is the control (10B/6A) protocol generating cells that express both cTNT and the ventricular specific marker MLC2v. On the right, the atrial protocol (3B/2A+RA), that includes the addition of retinoic acid, generating cells that express cTNT but do not express MLC2v. Unpublished data reproduced with the permission of Jeehoon Lee and Dr. Stephanie Protze.

4.3 Single Cell Electrophysiology

Cardiomyocytes derived from hESCs that received RA during their differentiation

protocol (“atrial” cardiomyocytes) had predominantly atrial action potential (AP)

morphologies, whereas cardiomyocytes that were not exposed to RA (“ventricular” or

control cardiomyocytes) had predominantly ventricular action potential morphologies

(Figure 8). The “atrial” differentiation protocol, that included the addition of RA,

generated 90% atrial like APs, 5% nodal like APs and 5% ventricular like APs (n = 20

cells) (Figure 9). The control, or “ventricular” differentiation protocol, that did not

include the addition of RA, generated 85% ventricular like APs, 10% atrial like APs and

5% nodal like APs (n = 20 cells) (Figure 10). The electrophysiologic characteristics of

the recorded APs can be seen in Table 1.

Page 64: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

54

Figure 8: Single cell patch recordings demonstrating typical APs of RA (3B/2A+RA) compare to the control protocol (10B/6A without RA). Cells are carried through to day 20, to allow for maturation and the genesis of contracting cardiomyocytes, where they were then plated as single cells and patched in Tyrode’s solution to record single cardiomyocyte APs. RA treated cells demonstrate atrial AP morphologies whereas control cells that were not exposed to RA demonstrate ventricular AP morphologies.

Figure 9: Showing the presence of cardiomyocyte types with representative AP profiles and their prevalence (n = 20 cells) in the atrial directed differentiation protocol (3B/2A + RA) used to generate atrial cardiomyocytes from hESCs. Cells are carried through to day 20, to allow for maturation and the genesis of contracting cardiomyocytes, where they were then plated as single cells and patched in Tyrode’s solution to record single CM APs. The majority of APs carried an atrial phenotype (90%).

Page 65: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

55

Figure 10: Showing the presence of cardiomyocyte types with representative AP profiles and their prevalence (n = 20 cells) in the standard directed differentiation protocol (control, 10B/6A) used to generate cardiomyocytes from hESCs. Cells are carried through to day 20, to allow for maturation and the genesis of contracting cardiomyocytes, where they were then plated as single cells and patched in Tyrode’s solution to record single CM APs. The majority of APs carried a ventricular phenotype (85%).

N (cells)

dv/dtmax

(V/s)

DMP (mV)

APA (mV) APD50

(ms) APD90

(ms)

Control “Ventricular”

20 54 ± 3 -53 ± 1 101 ± 2 479 ± 59 547.2 ± 59

RA treated “Atrial”

20 44 ± 5 -50 ± 1 78 ± 3** 33 ± 6** 187 ± 13**

Table 1: Characteristics of APs recorded from the protocol generating atrial cardiomyocytes (3B/2A + RA) compared to the protocol used to generate primarily ventricular cardiomyocytes (10B/6A). The majority of cells demonstrated features of electrophysiologic immaturity with more positive membrane potentials, and lower upstroke velocities and action potential amplitudes. APA, action potential amplitude; APD50, action potential duration at 50% of repolarization; APD90, action potential duration at 90% of repolarization; DMP, diastolic membrane potential; dv/dtmax, maximum action potential upstroke velocity; N, cell number; **P≤0.0001 as analyzed by Student's T-test.

Comparing the cardiomyocytes generated from the “atrial” differentiation protocol to the

cardiomyocytes generated from the “ventricular” or control protocol, as would be

expected, the “atrial” cardiomyocytes had a shorter APD50 (33 ± 6 ms vs. 479 ± 59, p <

0.0001) (Figure 11) There was no significant difference between differentiation protocols

with respect to the maximum upstroke velocity, or the maximum diastolic potential. Of

Page 66: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

56

note, both of theses values are considerably more positive than that expected of adult

tissue. In addition, “atrial” cells had a lower action potential amplitude (78 ± 3 mV vs.

101 ± 2 mV, p <0.0001), and a dramatically shorter APD90 (187 ± 13 ms vs. 547 ± 59

ms, p <0.0001).

Figure 11: Comparing the AP characteristics of cells generated from the “Ventricular” (10B/6A) differentiation protocol compared to the “Atrial” (3B/2A + RA) differentiation protocol. n = 60 APs representing 20 cells (3APs/cell). Error bars represent 95% standard deviation from the mean. **P≤0.0001 as analyzed by Student's T-test.

There was a significant correlation between the action potential amplitude and the

maximum diastolic potential (R2 = 0.373, p<0.001), which was also seen when

considering the maximum upstroke velocity and the maximum diastolic potential (R2 =

0.176, p = 0.001). Using a technique called an anode break, negative current was injected

into the cells in order to achieve a more negative resting membrane potential. This was

done repeatedly until a resting membrane potential of -70 mV was achieved, and then the

current was extinguished allowing the cell to fire a spontaneous action potential. Using

this technique, a higher action potential amplitude and maximum upstroke velocity were

achieved (Figure 12). Finally, “atrial” cardiomyocytes had a much faster spontaneous

beating rate, as demonstrated by a shorter cycle length, compared to the “ventricular”

cells (680 ± 5 ms vs. 1670 ± 12 ms, p < 0.0001).

Page 67: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

57

Figure 12: Patch clamp recording in Tyrode’s solution of a cardiomyocyte generated from the atrial protocol (3B/2A + RA) during an anode break. The injection of negative current into the cell drove the resting membrane potential to a more negative value and then released. The subsequent action potential amplitude is clearly higher than the subsequent APs starting from a less negative resting membrane potential in the image on the left. On the right, the same cell with multiple time sequences overlayed to demonstrate the reproducibility of this maneuver in achieving higher action potential amplitudes.

4.4 MEA Electrophysiology

“Atrial” and “ventricular” cell sheets were plated on MEA chips as described. External

field potentials were successfully recorded from the MEAs of both atrial and ventricular

cell sheets (Figure 13). These are akin to an electrocardiogram measurement on patients

and have clearly demonstrable depolarization and repolarization phases facilitating

measurements of field potential durations (FPDs) which are directly linked to the action

potential duration. As expected from the single cell analyses, the FPDs of “atrial” cell

sheetss were much shorter than that of “ventricular” cell sheets. Pacing was performed

through the Cardio2D system in a unipolar configuration. Capture was reliably

demonstrated by direct visualization of the cell sheet, the demonstration of capture at the

tail end of the pacing artefact, and through routine electrophysiologic principles including

Page 68: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

58

the perturbation of cycle length as demonstrated by the pacing cycle length, the

spontaneous beating rate, and the post-pacing interval (Figure 14).

Figure 13: MEA recording of a “ventricular” cell sheet on the left generated from the 10B/6A differentiation protocol compared to an “atrial” cell sheet on the right generated from the 3B/2A + RA protocol. Cell sheets were allowed to beat spontaneously. FPDs recorded and labeled for both cell sheets. The “atrial” cell sheet displays a shorter FPD as would be predicted by the single cell data in which atrial cells had significantly faster repolarization times. FPD = field potential duration.

Figure 14: MEA recording of “atrial” cell sheet (generated from the 3B/2A + RA differentiation protocol). Capture is demonstrated at the pacing rate and thus pacing has successfully overdriven (either suppressed in the case of automatic activity or accelerated in the setting of triggered activity) the spontaneous pacemaker. The post pacing cycle length (CL), commonly referred to as the post pacing interval, is the time required for the spontaneous activity to resume, which then returns to its spontaneous rate. The clear demonstration of these three intervals, in addition to the capture signal distinct from the pacing artefact, in this tracing strongly supports our ability to pace and capture the cell sheets at this pacing rate. Fibrillatory activity, appreciated by direct visualization of the cell sheets, was achieved in

“atrial” (n=3), but not “ventricular” (n=3) cell sheets despite aggressive burst pacing

Page 69: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

59

protocols down to the shortest cycle length of 50 ms. Similarly, 2 point stimulation using

the S1-S2 protocol previously described was not able to induce fibrillatory activity in the

ventricular cell sheets despite numerous and aggressive pacing protocols and field

generating configurations (data not shown). Unfortunately, fibrillatory activity in the

atrial cell sheets reduced the signal to noise ratio on the recorded field potentials, and

signals were no longer distinguishable. The inability to monitor and thus map AF on the

MEA system available led us to abandon this methodology in favour of optical mapping.

4.5 Optical mapping electrophysiology

The optical mapping configuration was optimized such that activation wavefronts could

be visualized with minimal processing. Advanced processing techniques were utilized for

noise reduction and signal augmentation, but were not used for analyses. Fifty percent of

“atrial” cell sheets had a propensity to develop spontaneous fibrillatory activity within the

first week of plating, with the prevalence of fibrillatory activity decreasing over time. At

the time of optical mapping, 1 in 4 cell sheets would continue to exhibit fibrillatory

activity. This occurred for the most part in cell sheets that had areas of non-confluence,

where “holes” in the cell sheets acted as anchors for re-entry (not shown). Non-confluent

cell sheets were discarded.

In confluent cell sheets, spontaneous fibrillatory activity at the time of optical mapping

was infrequent. “Atrial” cell sheets demonstrated spontaneous beating rates of 78 ± 14

bpm, with the majority developing spontaneous activity typically at the edges of the cell

sheets where a favourable source-to-sink relationship exists215 for cardiomyocyte clusters

with the fastest intrinsic firing rate (Figure 15). Once optimized, optical action potentials

(OAPs) could be recorded in a majority of cell sheets (Figure 16).

Page 70: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

60

Figure 15: A series of sequential still images taken from the optical recording of an atrial cell sheet (3B/2A + RA) of 1 cm diameter using 10 μM Di-4-ANEPPS and 10 μM blebbistatin and captured on an EMCCD. Electrical activity appears to originate from the top right corner of the cell sheet and propagate more rapidly along the edge of the cell sheet than towards its centre. This generates an activation wave front that ultimately circumnavigates the cell sheet and then moves relatively uniformly towards its centre, where the greatest sink to source mismatch occurs. Ultimately the wavefront extinguishes at the centre of the cell sheet, and a new wavefront is generated from the same origin and propagates in the same manner.

Figure 16: Typical optically recorded APs of an “atrial” cell sheet (3B/2A + RA) in “AF” using 10 μM Di-4-ANEPPS and 10 μM blebbistatin and captured on an EMCCD. The OAPs carry the typical morphology of atrial APs. The amplitude relates to the amount of fluorescent change of the voltage sensitive dye that occurs during a depolarization and repolarization event. The bipolar electrodes were able to pace and generate point stimulation which propagated

uniformly through the cell sheet. Increase of the pacing rate resulted in shortening of the

“atrial” APD (Figure 17). Burst on and off pacing at a cycle length of 50 ms for 1-2

minutes consistently generated “AF” as demonstrated by an increase in beating rate, the

development of continuous electrical activity in the cell sheet, and the generation of

Page 71: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

61

rotors (Figure 18). On one occasion, electrical alternans was recorded during rapid pacing

(cycle length of 50 ms), and was followed by rotor initiation (Figure 17).

Figure 17: On the left, optical action potential duration (OAPD) measurements on the Y axis plotted as a function of rate on the X axis demonstrating the APD restitution curve of a typical “atrial” cell sheet. At faster rates, the APD restitution curve becomes increasingly steep. On the right, electrical alternans is induced in the same cell sheet at a pacing rate of 1200 bpm demonstrating cyclic, beat-to-beat variations in AP amplitude and action potential duration at a constant stimulation frequency. The cell sheet cannot be captured 1:1 at this pacing rate. After this recording, “AF” was recorded in the cell sheet upon cessation of pacing.

Figure 18: A series of sequential still images taken from the optical recording of an “atrial” cell sheet (3B/2A + RA) of 1 cm diameter using 10 μM Di-4-ANEPPS and 10 μM blebbistatin and captured on an EMCCD. A rotor continuously generates an electrical wavefront, anchored by the phase singularity. Once initiated, the rotors were extremely stable, persisting in culture for as long as 8

weeks. When comparing the APD maps of a cell sheet in sinus rhythm compared to a cell

sheet which had been induced into rotor formation and remained persistently so

chronically (for 6 weeks), there is a clear difference in the APD heterogeneity (Figure

Page 72: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

62

19). This heterogeneity is one manifestation of the consequences of electrical remodelling

taking place in the cell sheet after rotor induction that can be identified both acutely and

chronically in this model.

Figure 19: Action potential duration (APD) maps generated from the “atrial” cell sheets using DI4-ANEPPS and an EMCCD. The figure legend represents APDs in msec. The image on the left was generated from a cell sheet prior to arrhythmia induction, while the APD map on the right represents a stable rotor that was induced with burst pacing. Note the increased APD heterogeneity in the APD map of the rotor compared to the APD map on the left. This heterogeneity is one manifestation of the nearly immediate electrical remodelling that is taking place in the cell sheet that can be identified both acutely and chronically in our system. As discussed previously, the inability of the excitation wavefront to depolarize the PS of

an ongoing rotor, and the tendency of the wavefront to curve around the PS create the

basis for rotor initiation and maintenance216. It has been previously demonstrated that the

wavefront curvature is highest around the tip resulting in slowing of the conduction

velocity near the PS, or centre of the rotor. Further from the centre, the curvature is

reduced and conduction velocity increases. Several mapping techniques have evolved to

assist in the understanding and analysis of rotor dynamics. Phase mapping was first able

to identify the PS by giving it an arbitrary phase, and its surrounding element a

continuous progression of phases equal to ±2π 99. More recently, the Hillbert transform

Page 73: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

63

has facilitated the computing of the instantaneous phase 217. Another important

developments was the analysis of the time-dependent behaviour of rotors using a Fast

Fourier Transform Analysis and generating a frequency map of the signals of interest.

Selecting the maximum frequency in the Fourier spectrum allowed for the construction of

dominant frequency maps 218.

Cardiac sheets, predominantly using rat neonatal rat and embryonic chick ventricular

cardiomyocytes, have demonstrated their utility in generating and mapping 2-D rotors

and re-entry 219 220. In these cases, owing to the stability of rotors in this model,

activation maps were able to capture the same data content generated by phase maps and

DF maps, required of more complex and three dimensional structures. We successfully

generated activation maps (Figure 20) and conduction velocity maps (Figure 21) at both

baseline, when a single nodal source acts as a pacemaker (henceforth referred to as “sinus

rhythm” or “SR”) and after rotor induction (henceforth referred to as “AF”). Activation

maps appropriately identified the PS where depolarization and repolarization wavefronts

collided, and captured the property of increasing curvature as the excitation wavefront

propagated away from the PS. Conduction velocity maps were able to demonstrate the

inverse relationship between conduction velocity and distance from the PS.

Page 74: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

64

Figure 20: Activation maps generated from recordings of “atrial” cell sheets using DI-4ANEPPS and an EMCCD. Optical APs are obtained on each pixel of the EMCCD and the accrued points are assigned to an isochronal color scale based on their respective activation times. On the left, a cell sheet before the induction of an arrhythmia. The cell sheet spontaneously generated a concentric ring of triggered activity which starts on the periphery of the culture and propagates uniformly to the centre (see Figure 15). This is likely a result of the aforementioned source sink relationship which generates edge effects in the culture. On the right, an activation map recorded after the induction of a rotor using burst pacing (see Figure 18). Note the increase in frequency after the induction of continuous rotor activity in the dish simulating the induction of a major driver of clinical AF. Isochronal lines are recorded in ms and labeled on the map.

Figure 21: Conduction Velocity (CV) maps generated from the activation maps in Figure 20. Again, the map on the left demonstrating baseline activity before rotor induction with uniform progression of electrical signal from the periphery of the dish to the centre. The arrows represent the vector of the electrical propagation at each point, and the size of the arrows represents the magnitude of the velocity. On the right, a CV map generated after rotor induction. Note the smaller magnitude of the arrows at the core of the rotor. This facilitates rotor stability and in fact anchors the rotor around an area of a phase singularity. Here, the convex curvature reaches a critical value preventing electrical activity to invade the core and thus maintaining the unexcitable obstacle around which the re-entrant rhythm circulates.

Page 75: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

65

4.5.1 Optical mapping the effects of AADs

To test the appropriateness and applicability of our model system to the tasks of drug

screening and drug discovery, we first sought out to demonstrate the known effects of

commonly used AADs. Indeed in our preliminary studies we observed predictable

effects of flecainide and dofetilide on the OAP morphology (Figure 22). The major and

important limitation to these studies was the rapid photobleaching of voltage sensitive

dyes thus limiting the exposure time and therefore the dose range of each drug trialed.

Figure 22: Optical action potentials (OAPs) from cell sheets of “atrial” cardiomyocytes derived from hESCs are recorded using the voltage sensitive dye DI4-ANEPPS on an EMCCD. Representative OAPs from an “atrial” cell sheet after induction of a rotor using burst pacing are demonstrated in Figure 5A. In Figure 5B, the baseline recording (in black) is compared to a second recording after the addition of 1 μM dofetilide (in blue), a specific IKr blocker. There is prolongation of the OAP as predicted by the effects of dofetilide on the major repolarizing K+ current in these cells. Figure 5C again demonstrates another “atrial” cell sheet in which a rotor is induced and OAPs are

recorded (in black), and a second OAP recorded after the addition of 10 µM flecainide (in

red). Slowing of the upstroke of the cardiac action potential can be appreciated, as would be predicted by the sodium channel blocking properties of the drug. Interestingly, there is also OAP prolongation, which may be related to the additional K+ blocking effects of flecainide that are predicted to extend the atrial wavelength and induce rotor termination. Dofetilide is a class III anti-arrhythmic which selectively inhibits the rapid component of

the time-dependent outward potassium current (Ikr) thus causing APD prolongation

without influencing the rate of depolarization or conduction velocity 75. As predicted

however, dofetilide is not specific to the atria and thus causes APD prolongation and

Page 76: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

66

resultant QT prolongation in the ventricles, predisposing patient to torsades-de-pointes

and sudden death. Cardiomyocytes derived from hESCs and hiPSCs have demonstrated

exquisite sensitivity to Ikr blockade exceeding that of animal tissue assays 221. Atrial

cardiomyocytes derived from hPSCs have not yet been tested for their response to Ikr

blockade, and so we chose to test dofetilide as our first AAD screen on our model.

Importantly, we chose to apply AADs after induction of “AF” to generate a clinically

relevant tissue model that would have greater translational meaning and potential.

“AF” was induced in rotors during “SR” with burst pacing as described. The induction of

“AF” in these cultures did not have an effect on the mean OAPDs across the cell sheet

(Figure 23). As would be predicted, dofetilide at a concentration of 1 μM dramatically

prolonged the OAPD of “atrial” cardiomyocytes during “AF”. When comparing cell

sheets during “AF” and then after exposure of dofetilide, there was a significant

prolongation of the OAPD (529 ± 137 vs. 809 ± 432, p = 0.02) (n =3) without a

significant effect on cycle length (687 ± 170 ms vs. 809 ± 432 ms, ns)(n=3).

Figure 23: Plotting the sequential effects of rotor induction and 1 μM dofetilide on the optically mapped APDs and cycle length of “atrial” cell sheets. The induction of a rotor decreased the cycle length (increased the rate) without a significant effect on the APD.

Page 77: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

67

Dofetilide significantly prolonged the APD without a significant effect on cycle length. Error bars represent 95% standard deviation generated from three independent experiments (N=3); *P≤0.05 as analyzed by Student's t-test. Dofetilide alone was not able to convert “AF” to “SR” acutely (within 4 hours). The

introduction of overdrive pacing (pacing at a cycle length exceeding that of the ongoing

rotor) to the cultures that were exposed to dofetilide however was able to convert “AF” to

“SR” as demonstrated by the return of a nodal pacemaker driving the rate of the system,

and the elimination of reentry and rotor dynamics (n=2). Interestingly, on one occasion,

overdrive pacing extinguished one rotor immediately transitioning to another rotor with a

different PS that was geographically distinct from the first. This second rotor was then

eliminated by overdrive pacing leading to “SR”. Dofetilide did not have an effect on

conduction velocity across the cell sheets (n = 3)(Figure 24).

Figure 24: Demonstrating the effects of rotor induction, 100 nM dofetilide and 1 μM dofetilide on conduction velocity in “atrial” cell sheets. When considering the conduction velocity across the cell sheet, dofetilide does not impact on conduction velocity. Error bars represent 95% standard deviation generated from three independent experiments (N=3). The next anti-arrhythmic trialed was flecainide, a class Ic AAD which blocks the Nav1.5

sodium channel in the heart, slowing the rapid upstroke component of the cardiac AP.

Page 78: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

68

Flecainide has been shown to reduce excitability and slow conduction velocity.

Flecainide also has effects on the repolarizing potassium currents which are proposed to

increase wavelength thus explaining the clinical observation of decreased AF

susceptibility for patients on this drug. Addition of flecainide to cell sheets in “AF”

appeared to have an effect on the cycle length on two cell sheets trialed (840 ± 396 ms vs

1766 ± 278 ms), with less of an impact appreciable on OAPD prolongation in both cell

sheets trialed (523 ± 119 vs to 789 ± 160 ms) (Figure 25).

Figure 25: Plotting the sequential effects of 5 μM flecainide and 10 μM flecainide on the optically mapped APDs and cycle length of a single “atrial” cell sheet after the induction of a rotor. Flecainide appears to slow cycle length in a dose dependent manner without exerting a significant independent effect on action potential duration. Also as expected, the addition of flecainide appeared to decrease conduction velocity

across the cell sheets in “AF” (3.5 ± 2.4 cm/s vs 2.0 ± 1.8 cm/s)(n=2). The addition of

flecainide did not convert the cell sheets from “AF” to “SR” (n=3) (Figure 26).

Page 79: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

69

Figure 26: Demonstrating the effects of 5 μM flecainide and 10 μM flecainide on the conduction velocity of an “atrial” cell sheet after the induction of a rotor. Flecainide appears to have a dose dependent effect on conduction velocity as seen on both cell sheets trialed. A mean and 95% standard deviation are represented for all conduction velocities recorded from the conduction velocity map of one cell sheet. To further dissect the effects of AADs on our human model of SR and AF, we analyzed

the conduction velocity maps in tertiles according to the geographic distance from the

focal source, either the nodal pacemaker in the setting of “SR” or the PS in the setting of

“AF”. This was based on our observation that our model had recapitulated the principles

components of curvature and conduction velocity essential to rotor initiation and

maintenance, and our hypothesis that AADs may have a differential effect depending on

the location on the rotor. The induction of a rotor indeed had a dramatic effect on slowing

conduction velocity at the source of the electrical wavefront (3.8 ± 2.1 cm/s vs. 0.5 ± 0.2,

p = 0.04)(n=3) with a trend towards the inverse effect on the outer tertile of the cell sheet

Page 80: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

70

that did not reach statistical significance. Dofetlide, as would be predicted, did not have

an effect on conduction velocity at any site along the rotor (Figure 27). Flecainide

however appeared to have a differential effect on conduction velocity slowing that was

dependent on the distance from the PS, unexpectedly slowing the CV disproportionately

at the site of the broadest curvature on the rotor (from 5.2 ± 4.9 cm/s to 2.2 ± 2.0 cm/s)

(n=2) (Figure 28).

Figure 27: Demonstrating the effect of dofetilide on conduction velocity as a function of distance from the origin of the electrical wavefront. The cell sheet was divided into thirds according to the distance from the origin. At baseline, the cell sheet has a spontaneous pacemaker which drives the cell sheet in “SR”. The origin is therefore at the site of the earliest activation in this case. After “AF” induction, the origin is defined as the phase singularity (PS), or centre of the rotor. Regions of interest are drawn over these regions of the conduction velocity maps generating means and standard deviations. There is a significant drop in the conduction velocity near the source after rotor induction (p = 0.04) which persists during the sequential uptitration of dofetilide, from 100 nM to 1 μM. Dofetilide does not appear to have any effect on conduction velocity, regardless of the distance from the PS. Error bars represent 95% standard deviation generated from three independent experiments (N=3).

Page 81: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

71

Figure 28: Demonstrating the effect of flecainide on conduction velocity as a function of distance from the origin of the electrical wavefront. Two independent “atrial” cell sheets are displayed. The cell sheets were divided into thirds according to the distance from the origin. Displaying only data obtained after the induction of a rotor with the centre of the rotor defined as the phase singularity (PS). Regions of interest are drawn over these regions of the conduction velocity maps generating means and standard deviations. After rotor induction, as before, the conduction velocity is slowest at the centre of the rotor and increases with increasing distance from the core. The addition of flecainide appears to have a dose dependent effect on slowing conduction velocity at the middle and outer third of the cell sheet, yet does not appear to exert any influence on the rotor’s core.

Page 82: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

72

Chapter 5

Discussion

Atrial fibrillation is an important contributor to the morbidity and mortality of Canadians,

and is a major and growing burden on our health care system and our economy1-9. While

the pace of discovery and innovation in the understanding and treatment of AF has

accelerated rapidly over the last two decades, the initial description of the disease nearly a

century ago has withstood the test of time and remained remarkably accurate despite the

relatively primitive tools of the age. A series of critical reviews in 1924 outlined the

clinical principles of AF, and the general hypotheses regarding AF initiation and

maintenance that continue to be hotly debated today 222 223. Perhaps stemming from this

confusion regarding the mechanistic basis of the disease, clinicians have limited tools

with which to target and treat the substrate that drives recurrence and facilitates the

remodeling process involved in the disease’s propensity for self-promotion. Rotor theory

has become a pre-eminent component of AF mechanistic study and treatment owing to

technologic innovations that have facilitated a greater appreciation of its importance, and

a more detailed understanding of its components. Over a parallel, yet only recently

overlapping time course, there has been a rapid advancement of hPSC technology in the

generation of pure lineages of well characterized cardiomyocytes in vitro through the

application of the fundamentals of developmental biology.

A major step forward in the application of hPSC derived cardiomyocytes to disease

modeling has been the careful dissection of embryonic cues which drive cell specific

fates in the embryo, and mimicking them in vitro to differentiate hPSCs into pure

lineages of cardiomyocyte subpopulations. While the ability of hPSC derived

Page 83: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

73

cardiomyocytes to model electrical cardiac disorders at a single cell level, predominantly

in ventricular like cells, has been exploited in a number of disease phenotypes and drug

screens, to our knowledge this is the first study to examine atrial tissue in the setting of a

tissue based arrhythmia. This has important implications for the study of AF and the

search for novel therapies, as well as significant potential to translate our growing

understanding of the genetic contributors of AF directly to the application of personalized

medicine.

5.1 Molecular markers of “atrial” and “ventricular” cardiomyocytes

Our group has developed a differentiation protocol that has generated an enriched

population of atrial like cells through the manipulation of BMP and Activin signaling,

and the addition of retinoic acid. The cells have undergone thorough molecular

characterization which has demonstrated their similarities to atrial cardiomyocytes and

their distinct signature compared to adult ventricular cardiomyocytes, and ventricular like

cardiomyocytes generated from hESCs. Specifically, the atrial like cell population is

enriched with the atrial markers ANF, KCNJ3, Cx40 and Cav1.3, whereas the ventricular

like cell population is enriched in MLC-2v and IRX4. Functional analyses of these cells

through patch clamping has demonstrated their clear and distinct electrophysiologic

properties which again mimic atrial cardiomyocytes.

5.2 Single cell electrophysiology

We have shown that the atrial like cell population consists of an overwhelming majority

of cells displaying an atrial AP morphology (90%), whereas the ventricular like cell

population equally exhibits a majority of cells that display a ventricular AP morphology

(85%). These cell populations importantly differ in their APD50 profiles, with atrial like

Page 84: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

74

cells displaying dramatically shorter APD50s. As previously mentioned, the single cell

electrophysiologic properties recorded suggest that these cells are immature compared to

their adult counterparts. This finding is not unexpected given the fact that there are

known differences between adult and fetal cardiomyocytes in terms of their complement

of ion channels and the resultant AP profile224.

Adult ventricular and atrial cardiomyocytes exhibit a resting phase (phase 4) in which the

resting membrane potential (RMP) does not change. This is primarily due to the

rectifying potassium current (Ik1) which stabilized the RMP at ~-85mV, which is the

reversal potential of K+. In hPSC derived cardiomyocytes, the RMP is much less

negative, a property which has been attributed to the lower expression of Ik1156,225. We,

and others, have indirectly shown that the relatively depolarized state of the hPSC

cardiomyocytes leads to a decrease in the functional availability of Na+ channels by

driving the membrane potential to more negative values and demonstrating an increase

the action potential amplitude and upstroke velocity 65,156. An additional explanation for

the relatively low upstroke velocity and action potential amplitude seen in hPSC derived

cardiomyocytes compared to published values for adult cardiomyocytes may be that cells

were patched at room temperature since the principal component of these measures is

sodium current. Sodium channels are temperature dependent, and thus cells analyzed at

37 °C may demonstrate more sodium current, and thus higher values for upstroke

velocity and action potential amplitude. Cardiomyocytes derived from hPSCs also

demonstrate large pacemaker currents (funny current, If), which is very low in adult

cardiomyocytes, causing diastolic depolarization and spontaneous contraction of hPSCs

in culture as seen in our study.

Page 85: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

75

There are many other features of immaturity not directly addressed in our study including

morphologic parameters, sarcomeric organization, calcium handling and excitation-

contraction coupling, and metabolism224. There have been many proposed methods to

promote maturation of hPSC derived cardiomyocytes including increasing time in

culture, electrical stimulation, application of mechanical strain, chemically inducing

maturation, and the promotion of maturation by incorporating other cells types or

extracellular substrates into culture or engineered 3-D structures. In spite of all of these

efforts, many of the features of maturation have only been seen in isolation, and certain

features of the adult phenotype have never been reproduced in culture such as the present

of T-tubules.

5.3 Modeling atrial diseases using hPSCs

Despite the known limitations related to the immaturity of hPSC derived cardiomyocytes,

a fully mature state is not felt to be a necessary pre-requisite to the use of these cells to

model disease. The FDA in fact has recently proposed a directive that all new drugs

should be tested for their effects on all ion channels in human cardiomyocytes, and have

suggested that hPSC derived cardiomyocytes were an ideal model and may be

appropriate for this purpose224. While the weight of evidence to support this notion in

ventricular like hPSC derived cardiomyocytes has already been reviewed in the

background section, there has been a recent study demonstrating the utility of atrial like

cells in drug screening and drug discovery. By employing the same HES3 NKX2-5egfp/w

cell line as we have employed for our studies, Devalla et. al, generated atrial like cells

through the manipulation of BMP4, Activin and Wnt signaling, as well as the addition of

RA 226. Similar to our findings, they showed that the expression of atrial specific ion

Page 86: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

76

channel genes was confined to their atrial like cells compared to their ventricular like

controls. Using patch clamping, they went on to show that in response to multiple atrial

specific ion channel blockers; vernakalant and Kv1.5 blocker XEN-D0101, hESC derived

atrial but not ventricular like cells demonstrated AP prolongation at the single cell level.

Additionally, and as would be expected of atrial cells, a novel Kir3.1/3.4 blocker restored

the AP shortening caused by carbachol in the hESC derived atrial cardiomyocytes. Taken

together, these results convincingly demonstrate that the cell line employed in our study,

when differentiated in cardiomyocytes using a very similar protocol to the one our group

has independently developed, generates a cell type that is appropriate, at least at the

single cell level, for the pre-clinical testing of atrial specific antiarrhythmics. We have

taken this concept a step further by demonstrating reliable and predictable drug responses

of atrial tissue in the setting of a clinically relevant arrhythmia model.

The novelty and strength of our study is the tissue based model of disease which

encompasses many aspects of clinical arrhythmology that cannot be studied at the single

cell level. In particular, we have focused on the determinants of the atrial wavelength,

conduction velocity and action potential duration, as well as the fundamental properties

of rotor dynamics. Optical mapping is a well known method to monitor and analyze the

propagation of cell excitation in whole cardiac tissues, and thus we set out to employ this

tool to study the functional properties of our multicellular network of “atrial” cells at

baseline and with the induction of complex phenomena such as reentry and spiral wave

propagation.

In our first efforts to validate our model, we have shown that “atrial” cell sheets can

demonstrate properties of electrical wavefront activation and propagation which can be

Page 87: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

77

captured on our optical mapping setup. We have successfully designed bipolar electrodes

capable of capturing the cell sheets reliably, and generated point source stimulation with

which to control and test our cell sheets. We have gone further to demonstrate the normal

physiologic property of APD restitution in our “atrial” cell sheets, and taken advantage of

this principle to generate “AF” by driving the cell sheets to the steepest part of their APD

restitution curves to facilitate self-sustaining oscillations. We have for the first time,

observed electrical alternans at rapid pacing rates preceding the onset of “AF” in human

“atrial” tissue. Electrical alternans is a recognized clinical risk factor for cardiac

arrhythmias including AF.

“AF” in our model system was defined by spiral wave activity that carried several

signatures of rotor dynamics including the ability to self-sustain a PS, the generation of a

curved wavefront with its maximal curvature and slowest conduction velocity centered on

the PS, the ability to overdrive the system with the addition of AADs that extend

wavelength, the spontaneous initiation of a distinct PS after overdrive pacing of the

original PS, and the ability of overdrive pacing and AADs to convert the reentrant

arrhythmia. We have successfully optically mapped the induced arrhythmia to generate

activation maps, measure optical APs in the setting of an arrhythmia, and generated

vector and conduction velocity maps giving us the unique ability to measure local

conduction velocity.

Page 88: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

78

5.3.1 APD restitution and heterogeneity

We have shown that OAPs across a cell sheet in “AF” display greater heterogeneity than

OAPs in “SR” (Figure 18). Dispersion of APDs in the atria occur under normal

physiologic conditions in humans, both in newborns and adults, and has been

documented in a number of other animal species 42,227 228. The APD and effective

refractory period shorten progressively with increased distance form the SAN, a finding

which stems from the initial observation that the atrial AP shape changes as a function of

distance and independent from the excitation sequence. This has been proposed to

protect the atria from anatomic reentry because of the resistance to unidirectional block in

the setting of an ectopic focus, and ascribed to changes in regional ionic properties 229 230.

In computer modeling studies, APD heterogeneity has indeed demonstrated a protective

effect on the development of anatomic reentry, but in contrast, did not afford protection

from functional reentry as would be seen in rotor formation 231. Wavebreak was in fact

more notable in areas of increased APD gradient leading way to stable rotor formation.

It has been proposed that such gradients contribute to the establishment of acute AF in the

structurally normal heart 232. In human AF, the atrium can achieve exceedingly high

frequencies, and thus extreme shortening of the APD would be required for APD

maintenance. Mechanisms that increased APD heterogeneity and regional abbreviation in

areas that achieve the highest frequencies, such as rotors driving AF, would lead to

increased stability of microreentrant sources by reducing interaction between the front of

the wavefront and its tail, and allowing for rapid curling of the wavefront around the PS

12 218. The APD maps generated in “SR” and “AF” indeed demonstrate a significant

difference in APD heterogeneity (ΔAPD 38 in SR vs. ΔAPD 191 in AF). At the core,

Page 89: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

79

there is a clear boundary where the wavefront, with a very short APD, meets the wavetail

with a longer APD. It is not surprising however that we did not observe a dramatic

change in the minimum APD observed because the AF model did not induce the dramatic

increases in frequency that are observed in humans (up to 16-18 Hz), likely relating to the

conduction velocity as the rate limiting component.

5.3.2 Conduction velocity

The conduction velocities observed in our model system are much slower than that seen

in cardiac tissue in vivo. There are several possible explanations for this finding, which

has been recapitulated by many other groups using hPSCs in a cell sheet format 224. As

discussed, the RMP of the cells is less negative than that seen in vivo, leading to reduced

availability of sodium channels which are a major contributor to the propagation velocity.

This was demonstrated in our single cell patch clamping studies, and has also been

observed in micropipette recordings of our cell sheets (Appendix Figure 1).

Another major contributor to conduction velocity is the density and composition of gap

junctions. Conduction velocity however is determined by the structural organization of

gap junctions as well which co-localize with sodium channels at the intercalated disks.

Intercalated disks are found on the shortest edge of two neighbouring cardiomyocytes

thus facilitating more rapid conduction of electrical signals in the longitudinal direction

233. In cell sheets of hPSCs, gap junctions are in contrast found on all sides of the

membrane 234. A recent study has however demonstrated that over time and up to one

month in culture, hPSC derived cardiomyocytes display increasingly developed

intercalated disks, including attachment zones and gap junctions, compared to at baseline

235. This increasing degree of ultrastructural organization was associated with increasing

Page 90: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

80

conduction velocity in the cell sheets over time. We hypothesize that this is the

underlying mechanism for our observation that spontaneous rotors were less prevalent in

our “atrial” cell sheets over time. Finally, the non-cardiomyocyte population in our cell

sheet cultures could significantly impact on conduction velocity.

Most important for the applicability of our model to serve as a drug screen is the response

of our system to AADs with known mechanisms of action. The first AAD that we chose

to test in our model system was Dofetilide, a class III anti-arrhythmic that selectively

inhibits the rapid component of the time-dependent outward potassium current (Ikr) 75.

Dofetilide is a potent AAD which has demonstrated clear utility in the management of

AF 78, however its use has been restricted by its lack of chamber specificity, causing

significant APD prolongation of the ventricles thus putting patients at risk of sudden

cardiac death. As previously mentioned, hPSC derived cardiomyocytes have previously

been shown to be more sensitive to the effects of Ikr blockade than other animal models,

implicating Ikr as a principal component of hPSC cardiomyocyte repolarization221. Our

study is the first to demonstrate the impact of dofetilide on APD prolongation in hESC

derived atrial like cardiomyocytes, the first to demonstrate OAPD prolongation in a tissue

model of hPSC derived cardiomyocytes, and the first to interrogate the effects of

dofetilide on an ongoing rotor in an hESC derived tissue model of AF. As we would

expect, dofetilide had no effect on conduction velocity over the cell sheets, and did not

disturb conduction velocity at any site of the ongoing spiral wave.

5.3.3 Effects of dofetilide on “atrial” cell sheets

Dofetilide alone did not convert “AF” in our dish to “SR”. Previous studies have

suggested that atrial wavelength plays a critical role in the antifibrillatory action of AADs

Page 91: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

81

as wavelength prolongation was associated with atrial refractory period, and mapping of

AF on AADs showed a reduction in the number of wavelets observed 58. Other groups

have proposed that the antifibrillatory effects are indirectly associated with wavelength

prolongation, and directly linked to prolongation of the excitable gap during periods of

AF236. According to the multiple-wavelet hypothesis, the stability of AF is determined by

the number of wavelets. Widening of the excitable gap would lower the chance that

fibrillation waves encounter areas of partially refractory tissue and thus slowing, and

fractionation of wavelets will occur less frequently. Furthermore, widening of the

excitable gap would promote fusion, and on balance, the generation and extinction of

fibrillatory waves would be expected to be reduced. We have demonstrated our ability to

overdrive pace ongoing rotors easily when treated with dofetilide, which was not the case

prior to the application of the drug. This indirectly points to dofetilide’s action on

extending the excitable gap, and supports the notion that fusion and extinction of rotors

are important components of the conversion of AF to SR. Further in support of this

theory are the mechanisms of action of flecainide, the second antiarrhythmic trialed in

our model.

5.3.4 Effects of flecainide on “atrial” cell sheets

We chose to study flecainide as it is a commonly used AAD for the treatment of AF.

Flecainide is a class I antiarrhythmic agent known to depress the maximum upstroke

velocity of APs in atrial and ventricular tissue relating to a block of the sodium current

(INa) 237. Flecainide has been shown in a number of models to decrease conduction

velocity significantly in the setting of AF without a significant effect on ERP 238. During

functional reentry, the electrical wavefront is circulating around an area of functional

Page 92: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

82

conduction block, often making sharp turns at pivot points. This would predict that the

wavefront would be forced to increase its curvature and thus set up a source-sink

mismatch resulting in conduction delay at the pivot points. This mismatch could

theoretically be preferentially aggravated by flecainide’s effects on slowing of conduction

velocity, and has been proposed as a mechanism explaining flecainide’s clinical efficacy.

Since AF relies upon the random reentry effects of wavelets entering an area previously

activated by another wavelet, drugs that increase the size of the functional circuit, and

decrease the number of wavelets available, would in effect increase the excitable gap.

The dogma that flecainide has no effect on ERP has been questioned, and may in fact be

specific to the AF model in question. Flecainide has been shown to terminate AF in the

vagotonic model of canine AF by causing tachycardia dependent increases in atrial

refractoriness and wavelength 58. This has been attributed to the drug’s effect on

decreasing atrial APD accommodation during periods of increased heart rate, possibly

related to an effect on repolarizing potassium current, as well as decreasing the

heterogeneity of atrial activation 85. Rate-dependent sodium channel block however could

also explain the observed prolongation in refractoriness independently of changes in

repolarization by depressing atrial excitability. Complicating the issue further is the

observation that flecainide has shown differential effects on potassium currents in animal

models and humans. As an example, the Kv1.5-based human Ikur has been proposed as a

potentially interesting target for novel therapies of AF, with numerous compounds now

undergoing clinical testing. The human channel is however resistant to flecainide,

whereas the dog counterpart has been shown to be quite sensitive, highlighting the

importance of using human tissue for drug screening 239.

Page 93: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

83

We have shown that flecainide appears to have an effect on conduction velocity slowing

in our model of AF. We also noted flecainide’s effect on cycle length without a clear

independent effect of APD prolongation. While this data is clearly preliminary owing to

the limited number of repeated experiments performed, we plan on further testing to

determine the reproducibility of our findings. The following discussion is based on our

preliminary observations which will form the basis for hypotheses that have yet to be

tested.

As previously discussed, the known clinically efficacy of class Ic drugs is seemingly

paradoxic in light of our understanding of wavelength dependent reentry when

considering only the effects of conduction velocity. Anything that slows conduction

velocity without altering refractoriness would be expected to decrease the wavelength and

thus increase the propensity of tissue to develop reentrant arrhythmias. We have in fact

clearly demonstrated this principal in our model with our unique ability to optically map

rotors, and their response to drug effects. On one occasion, the addition of flecainide to a

stable rotor induced by burst pacing had a dramatic effect on conduction velocity slowing

that was not accompanied by a substantial increase in APD. This slowed the cycle

length of the rotor and facilitated the initiation of two separate rotors in an “atrial” cell

sheet (Appendix Figure 2). This finding, although predictable, would argue against the

notion that flecainide decreases the number of wavelets in AF and thus would require an

alternate explanation for its observed anti-fibrillatory effects. We propose an alternate

mechanism, based on early preliminary findings that relate to the regional variation of

drug effects along the curvature of an ongoing rotor.

Page 94: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

84

Flecainide has been shown to produce rate-dependent reductions in atrial conduction

velocity as well as reducing the heterogeneity of wavelengths across the atria 85. We have

observed regional variation in the effects of conduction velocity slowing with the

addition of flecainide to “AF” in our model. There was an effect on conduction velocity

slowing at points along the spiral wave that had the broadest curvature, where the

conduction velocity was greatest at baseline, while there was seemingly no effect on

conduction velocity at the centre of the spiral where the wavefront curled tightly around

the PS, corresponding to the area with the slowest conduction velocity at baseline.

Flecainide is known to cause potent voltage-and frequency-dependent inhibition of

cardiac Na+ channels. Voltage gated sodium channels can exist in three distinct states;

deactivated (closed), activated (open) or inactivated (closed). With the membrane at its

resting potential, the Na+ channels are in their deactivated states. Activation gates

subsequently open in response to an electrical current, thus allowing Na+ ions to flow

into the cell and causing the membrane potential to increase and thus depolarize. At a

specific membrane potential, the Na+ channels become inactivated until the membrane

potential becomes negative enough and the cell returns to its deactivated state, ready to

participate in the subsequent depolarization event.

Flecainide has a strong preference for binding to the activated states of the sodium

channel (ie open and inactivated) and its effect is enhanced by rapid repetitive

depolarizations, and increases over the range of voltages where the channels activate 240.

This provides one potential explanation for our preliminary observation in that the centre

of the rotor may be in a relatively deactivated state, while cells along the broader

Page 95: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

85

curvature that demonstrate a more rapid conduction velocity before treatment with

flecainide, are in relatively activated states and thus preferentially effected by flecainide.

This would certainly be supported by the observation that conduction velocity in the rotor

is lowest at its center and increases as a function of distance from the centre if the

observed conduction velocity reflects the active and inactivated states of the sodium

channel. It however directly contradicts the observations made by Allessie in a commonly

used and referenced goat model of AF. Allessie described a preferential effect of class I

drugs at areas where the rotor curvature was steepest, preferentially depressing

conduction velocity of wavelets a their pivot points 236.

An alternative explanation for our preliminary finding is that it is in fact calcium currents

and not sodium currents that predominate and drive the wavelets surrounding the rotor’s

core in our model. It has been well established that patients who have had prolonged

durations of AF show less of a response to both Na+ and K+ blocking agents 241. This has

been a proposed consequence of changes in ion channel function during atrial

remodelling in response to tachycardia and AF, a phenomenon known as “AF begets

AF”. In light of our observation, it is interesting to reflect on the fact that T-type Ca+

channel blockers appear to suppress atrial remodelling whereas L-type Ca+ channels

blockers are ineffective. In addition, Bepridil which acts on both L-type and T-type Ca+

channels has an unusual ability to convert long-standing AF242. It is possible therefore

that this hypothesis holds true in human AF. Our enthusiasm for this observation is

tempered by questions relating to the fundamental translational capacity of our model,

specifically when interrogating ion channels that have a signature in our model that is

quite distinct from the adult phenotype.

Page 96: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

86

A feature of electrophysiologic immaturity, not previously discussed in this work, is the

expression of T-type calcium channels. These channels help to confer automaticity in

differentiating cardiomyocytes 243. In embryonic and neonatal cardiomyocytes, T-type

calcium channels have been observed at higher densities and their expression nearly

disappears in the working myocardium of the adult heart. Although difficult to quantify

in terms of current because of the lack of specific blockers, greater than 50% of hiPSC

derived cardiomyocytes display functional ICa,T244. If we are therefore able to go on to

conclusively demonstrate that the regional variation in flecainide’s effect on our model

system is dependent on the function of T-type Ca+ channels near the rotor, we would

have to go on to demonstrate the applicability of this finding to the human system. This

is of course a limitation of all model systems, emphasized here to contrast cell types,

highlight limitations, and provide a cautionary note on the translation of this novel model.

Page 97: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

87

Chapter 6

Future Directions

The immediate path forward is clear, and our experience to date has taught us important

lessons regarding the limitations related to our current model system. Our future

directions are divided into three main themes: 1) the validation of findings to date 2) the

continued interrogation, refinement and improvement of our model system to study atrial

remodeling 3) adapting our model to answer new fundamental and clinically relevant

questions related to the identification, prevention and treatment of AF.

6.1 Validation of current findings

The principle outcomes under study have been the components of the atrial wavelength,

namely the action potential duration and the conduction velocity. In order to validate our

findings, we have incorporated micropipette recordings into our optical mapping setup.

We have successfully recorded intracellular potentials using high resistance

micropipettes, and will use these to quantify the effects of AADs on the AP morphology

and APD measurements as a standard with which to judge the accuracy of our OAP

recordings. Using this technique we have also begun to validate our conduction velocity

measurements, and generate restitution curves for conduction velocity as a function of

rate, using our stimulation probes and micropipette recordings. By measuring the time

taken from stimulation on one end of the cell sheet to the depolarization of a cell on the

opposite side of the cell sheet, and measuring the distance travelled, we have begun to

generate accurate measurements of conduction velocity while simultaneously optical

mapping to ensure point source stimulation and uniform unidirectional propagation.

Finally, we are generating “ventricular” cell sheets in order to contrast our findings with a

Page 98: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

88

distinct cell type which should display differences in the determinants of wavelength at

baseline as well as different responses to AADs. We have already observed a significant

decrease in the propensity of “ventricular” cell sheets to generate rotors using our MEA

protocol, and expect to demonstrate the same findings in our optically mapped cell sheets.

To further validate our findings, we plan on testing AADs that have atrial specific effects

and demonstrating the predictable nature of responses in our “atrial” cell sheets compared

to “ventricular” cell sheets. This would include more recently proposed agents for the

treatment of AF: Tertiapin-Q, an IKAch blocking agent, and Ca-activated K+ channels

blockers. In addition, we propose to analyze the effects of vernakalant, which

preferentially blocks atrial-specific Kv1.5-based IKur in many models including atrial like

hESC derived cardiomyocytes. Vernakalant is used exclusively for cardioversion with

high efficacy, and thus is of interest to be tested in our model that has not yet

demonstrated purely pharmacologic cardioversion 245-247

In order to further interrogate our model, we plan on examining the structural

components of our cell sheets in greater detail. Our first step will be the identification

and quantification of Cx-40, a gap junction that is selectively expressed in atria. Another

principle component of conduction velocity and its heterogeneity across cardiac tissue

that we have not yet addressed is anisotropy, which will be interrogated by analyzing the

gross arrangement of cells in culture through immunostaining and/or electron

microscopy. Once quantified, it would be ideal to incorporate these structural measures,

as well as the ionic determinants of wave propagation, into a mathematical model that

could complement findings from the in vitro model as well as help direct future

experimentation. There are 5 models of human atrial electrophysiology that have been

Page 99: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

89

published248. Our in vitro model could help to evaluate and refine these models.

Collectively these tools could serve to provide an in vitro and in silico assessment of

known and novel therapeutics.

The major limitation of our study has been our reliance on currently available voltage

sensitive dyes. We have trialed 3 such dyes (Di-4-ANEPPS, Di-8-ANEPPS, and RH237)

in our model system, and ultimately chose to optimize and utilize Di-4-ANEPPS as it

displayed the best signal to noise (S:N) ratio on our optical mapping setup. These dyes all

have the limitation of generating poor S:N ratios in the cell sheet format, and rapidly

photobleaching thus limiting the recording time available when using a high intensity

light source. For future studies we will use hESCs expressing Arclight249, with better (~2-

fold) S:N, and virtually no photobleaching compared to Di-4-ANNEPS249. ArcLight is a

novel genetically encoded voltage probe, developed by fusing the voltage sensor domain

of the Ciona intestinalis voltage-sensitive phosphatase to a super ecliptic pHlurion

carrying the point mutation A227D 250. Arclight has already demonstrated its utility in

measuring APs noninvasively using optical mapping of hESC derived cardiomyocytes

which quantitatively and qualitatively track with patch clamp recordings 249. Arclight is

typically introduced into cells using transient transfection or lentiviral transduction. We

have already acquired an hiPSC line that stably expresses Arclight from our collaborator.

This line has successfully been differentiated using the EB protocol previously described.

The derivative cardiomyocytes generated OAPs with excellent S:N that do not exhibit

perceptible signs of photobleaching (Appendix Figure 3). Arclight has a relatively

sluggish response time compared to Di-4-ANNEPS which will significantly affect

estimates of AP upstroke (underestimated by ~30%), however will impact immeasurably

Page 100: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

90

on key mapping results (activation/phase, CV and APD). We will overcome this

limitation by recording APs using micropipettes, as previously described, which will be

used to "calibrate" optical data.

Extracellular field recordings (ALA multi-electrode array or MEA system) will be used

to: a) monitor electrical activity thereby (crudely) validating our activation/CV plots, b)

assess the P-wave and local electrogram “fractionation”, which links to arrhythmia

susceptibility251-253, and 3) induce and terminate arrhythmias with appropriate pacing.

This last point will be a necessary component of future studies, removing the necessity of

introducing an unsterile pacing electrode into the tissue culture medium and thus

preserving it for repeated studies. The incorporation of a stable and sterile pacing

platform as well as the utility of Arclight in repeated measures over time, will allow us to

interrogate a question that remains fundamental to the understanding and management of

AF and has been alluded to repeatedly in this document, that is “AF begets AF”.

6.2 Study AF remodeling

Regardless of its ultimate cause, AF invariably begins as isolated short-lived episodes

called "paroxysmal AF". Each paroxysm accelerates atrial remodeling thereby promoting

"persistent AF"42,43. The functional, structural, electrical and biochemical changes in atria

leading to increased AF vulnerability, providing the substrate for AF maintenance, and

making cardioversion increasingly difficult, are called remodeling14,15,254. Although some

clinical predictors of the transition from paroxysmal to persistent AF have been

identified, the molecular, biochemical and electrophysiologic mechanisms for this

remodeling remain largely unknown255,256. We propose to utilize our novel model of AF

to study the molecular, cellular and electrical changes underlying AF remodeling. As AF-

Page 101: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

91

begets-AF, we hypothesize that rotor induction will drive time-dependent remodeling.

Indeed, APD and CV heterogeneity have been shown to develop in our cell sheets with

rotors. Moreover, we observe spontaneous rotors in a small subset of our cell sheets

which implies the presence of triggers. Thus, our cultures display dynamic changes in

wavelength (APD and CV) as well as triggers, the two key components of the AF

syndrome, which is unlike mouse/goat/dog/sheep animal models in which triggering is

externally induced.

To look at the effects of AF on remodelling in our “atrial” cardiomyocyte cultures, we

propose to make measurements at 3 time points after the induction of stable rotors: after 1

day, representing early clinical AF, after 7 days, representing the clinical time point used

to diagnose persistent AF, and after 30 days, representing chronic persistent AF. Control

cultures will be maintained in regular “sinus rhythm” (“SR”) for equivalent periods. We

will also study the effects of reverse remodelling; if possible, “SR” will be restored in the

chronic persistent “AF” group and then studied 1 week later along with a time matched

control. Since AF is ultimately an electrical phenomenon, we will use our optical

mapping rig to interrogate the electrical remodelling process as previously described. Of

course with the ability to make repeated optical mapping recordings over time, multiple

timepoints can be chosen in each cell sheet until they reach a “terminal” endpoint, such as

enzymatic digestion for single cell analysis. In addition we will be able to interrogate

other important measures of electrical remodelling that require exposure times that

exceed the capabilities of Di-4-ANEPPS because of the photobleaching and toxicity

previously described.

Page 102: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

92

Using the Arclight line we will be able to routinely measure the effective refractory

period (ERP) with programmed external stimulation, as is routinely done in human

subjects. This is achieved using trains of 8 field pulses at 500 msec (S1) followed by

variable shorter cycle pulses (S2) until capture does not occur. S1 is also varied (400,

300, 200) allowing ERP to be determined at multiple cycle lengths. We expect that the

longer cell sheets are kept in "AF", the easier it will be to generate rotors, and these

differences will correlate systematically with differences in (inter-related) rotor/electrical

properties such as257-260: shorter APD, shorter ERP, faster rotor frequencies, greater rotor

curvatures, slower CV, and more complex activation patterns or wave breaks261. Resting

membrane potentials may become more negative262 (more background IK1). These

hypotheses will be tested by performing single cell cardiac electrophysiology studies

using patch clamping in order to dissect the observed alterations caused by electrical

remodelling into the functions and interactions of single channels and currents. To

complement these single cell studies, RNA will be isolated for targeted qRT-PCR

analysis. A future consideration could be the inclusion of RNA seq analysis.

To this point we have focused on the rapid pacing model of AF. We propose however to

test multiple acquired models of AF in the future using the aforementioned study design.

We currently are focused on three potential acquired stressors that we propose to include

in future studies; elevated parasympathetic activity, stretch, and fibrosis.

6.3 Model other types and features of acquired AF

Previous studies have established the involvement of elevated parasympathetic activity

and related IK,ACH activity in AF which has resulted in efforts to develop blockers of these

channels263. We will look for changes in the expression pattern of IK,ACH, GIRK1 and

Page 103: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

93

GRK4, M2- muscarinic receptors, and RGS regulator proteins, as well as constitutive

activity of GRK-related channels264. We will also perform studies and analyses in the

presence of low levels of the stable muscarinic agonist, carbachol. We predict that low

levels of carbachol will be associated with electrophysiologic markers of maturation, and

that higher levels of carbachol will facilitate wavebreak and multiple wavelets

degenerating into chaotic activity.

Elevated venous/atrial pressure and atrial stretch are major factors in AF strongly

stimulating cardiac remodeling via signaling pathways leading to increased oxidative

stress 18,265-2676,265,266,267. Atrial stretch shortens atrial ERP through increased IKAch, Icl, and

Ik1268,269as well as ICa,L inactivation270 and reduced gap junctions271. We plan on

investigating the effects of cyclical stretch on hESC derived cardiomyocytes and looking

at measures of maturation and remodeling.

Finally, since many acquired forms of AF demonstrate increased interstitial atrial fibrosis,

which correlates inversely with treatment success272-275, we will combine cardiac

fibroblasts with our atrial CMs. Our initial experiments either used a cell sorting strategy

to purify the population to contain only cardiomyocytes, or incorporated ill defined

CD90+ population. We propose to repeat the same studies in cultures containing various

percentages of cardiac fibroblasts generated from hESCs using a published protocol276. It

has already been established that CD90+ cells improve maturation of cardiac gene

expression patterns, and various structural/functional measures277. In addition, fibroblasts

secrete endothelin-1 which promotes the generation and persistence of myofibroblasts,

and induce the expression of a wide variety of ECM components, including collagen type

I.

Page 104: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

94

In contrast to other fibroblast sources, generating cardiac fibroblasts from hESCs allows

for the production of pure unlimited numbers of well characterized cardiac fibroblasts

that do not proliferate uncontrollably in our backbone culture medium. These are novel

studies with uncertain outcomes; nevertheless, we do expect that the cell sheets in which

“AF” has been induced will have increased accumulation of fibroblasts and connective

tissue since AF induces atrial fibroblasts (myofibroblasts) to proliferate and deposit

extracellular matrix278-281. This is expected to alter tissue architecture and myocyte-to-

myocyte electrical coupling as well as to create additional capacitive loads on CMs282.

We anticipate that the end result will be collective interference with electrical conduction

and increased propensity to arrhythmia initiation and maintenance. All of these properties

can be readily determined using the approaches detailed already. This novel model

system could be a powerful platform for developing new comprehensive therapies for

treating AF that specifically target fibrosis283,284.

6.4 Introduce genetic variation as a determinant and model of AF

Building on the lessons learned from our model of acquired AF, we propose to study

heritable forms of AF using hiPSCs and/or genomic editing. We have learned from the

Framingham studies that 58% of AF patients have no obvious cardiac disease or risk

factors285. AF incidence shows a strong heritability, and a genetic predisposition has been

shown to contribute to AF risk. Recently numerous AF associated mutations, candidate

genes and risk loci have been identified, yet few functional studies have deciphered their

mechanisms of action or validated their role in disease. We propose to employ our model

of human “atrial” cardiomyocytes derived from hESCs to study the underlying

mechanism for AF vulnerability in these patients with the potential to identify novel

Page 105: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

95

therapies. As a first test of our hypothesis, we will use the CRISPR/Cas9 system to

introduce genetic changes into a background of normal hESCs. As a test case, we propose

to study the effects of GATA6 (heterozygous and homozygous) mutations in our system

286. Autosomal dominant mutations in GATA6 have been identified in cases of loneAF287.

GATA6 was chosen for several reasons; firstly, it has recently been linked to AF in rare

families with mendelian inheritance patterns and strong penetrance and expressivity.

Second, it is a transcription factor expressed in the early cardiac mesoderm 288. Third,

although GATA6 has been implicated in normal gap junction expression during periods

of stress or injury in the atria289, GATA6 is likely (by virtue of being a transcriptional

factor) to affect the expression of many genes and could provide some interesting clues

into multiple functional genes impacting on AF, which could yield greater insights into

AF mechanisms more broadly. To complement this data set, we propose to differentiate

pure populations of nodal, atrial, and ventricular cell types from the genomically edited

hESCs in order to study the effects of these mutations on the different cardiac lineages.

By studying the impact of various drug treatments, we can determine whether and how

the atrial remodeling and AF induced by GATA6 mutations relate to the remodeling

induced by AF itself. If the mechanisms are quite different, these studies could be used to

identify personalized optimal drug therapies for these patients carrying these mutations,

as well as numerous other patients who may have as yet unidentified mutations. Most

importantly, this type of investigation has the potential to generate and test new

hypotheses regarding propensity to and mechanism of disease that may be potential

preventative and/or therapeutic targets.

Page 106: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

96

Other gene mutations will also be studied such as the TBX5 gene which causes Holt-

Oram syndrome and has been linked to lone AF 290,291. TBX5 likely lies upstream of

ANF and Cx-40, and may contribute to Nav1.5 expression 290,291. TBX5 mutations have

been associated with sinus node disease, AV node disease as well as QT prolongation 292.

Most recently, TBX5 has been identified as a novel locus for AF in a population of over

7000 European and Japanese patients with lone AF293. Although informative, the

analysis of isolated mutations in the backbone of an otherwise genetically normal stem

cell line will require complementary studies using hiPSCs. The strength in the hiPSC

model lies in the fact that in encompasses a host of unknown complementary and

confounding genetic variations which may play an indispensible role in the expression of

the disease experienced by the patients.

The final future direction to be discussed is the incorporation of bioengineering into our

AF model, that will use engineered heart tissue for analyses. Although the 2D geometry

of atrial cell sheets mimic in many respects that of atrial tissue, which is very thin and

unlikely to support 3D rotors (scroll waves), there are unwanted edge effects which

impact on source-sink matching, and have no parallels in human atria. Our atrial cell

sheets also show characteristics consistent with immaturity such as spontaneous beating.

Although it is conceivable that our cell sheets will mature with extended time in culture,

we have found limitations to time dependent maturation, consistent with other similar

studies153,244,294. In fact, studies of heart development show us that cardiac cells require

many environmental cues to drive adult patterning. Other efforts to promote maturity

have focused on electrical stimulation, mechanical strain, biochemical agents, co-

cultures, 3D culture, extracellular substrates, and genetic manipulations224. We plan to

Page 107: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

97

study the impact of geometry and immaturity simultaneously using co-cultures of cardiac

fibroblasts (as previously described), as well as alternative sources of human cardiac

fibroblasts and CD90+ cells mixed with cardiomyocytes and incorporated into 3D

biowires295. Biowires can be mechanically loaded, electrically stimulated, and exposed to

selected chemical inducers of maturation. We will study biowires in detail using the same

approaches previously outlined including arrhythmia induction (i.e. complete electrical

datasets, beating rates, various membrane currents). We know that atrial cell sheets

support sustained rotors much more easily than ventricular cell sheets, and expect that

sustained rotors will be readily induced in atrial biowires. We will screen using markers

of structural maturation such as sarcomeric banding and myofilament density on

immunofluorescence and electron microscopy, physiologic maturation by examining the

amplitude of calcium transients and the shape and resting membrane potentials of

optically mapped and micropipette recorded action potentials, and finally looking at

effects on cell cycle activity as well as monitoring mRNA and protein levels 296 297 296.

Numerous studies have shown that cell responses to drugs in 3D culture better predict in

vivo tissue functionality compared to 2D high throughput screens. On the other hand,

high throughput drug screening technologies relying on single cells or looking at single

biochemical or gene expression assays have been enormously successful in testing and

developing novel compounds cheaply and quickly. Most recently there has been a

movement towards tissue based assays, or organ-on-chip platforms for high throughput

assays to improve the translational relevance of these screens by approximating more

closely the disease or the organ of interest. We would propose that our model is well

Page 108: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

98

suited for the application of high throughput screening technologies once we can generate

tissue efficiently and reproducibly that has predictable performance parameters.

The strength in our model is the extreme nature of control which can ultimately be

exerted over countless variables, from the genetic make-up of the cell to the cell’s

environment. The challenge henceforth will be to understand each of these variables

under a defined set of extreme conditions such that the model’s behavior can be

understood and evaluated. A further challenge will be the scalability of the readout

analysis which must be interpreted in quantifiable patterns, and with the assistance of

computer learning, rather than the current form of qualitative observations and semi-

quantitative recordings that are hypothesis generating. The experiments described in our

future directions will define the most reliable measures, and help to focus our attention on

the outputs with the greatest signal. Once identified, and before a major investment into

scalability, these variables must be strictly correlated with important clinical endpoints to

ensure that our model has the capacity to exert a positive impact on patients for whom

these studies have been devised.

Page 109: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

99

References

1 Andrade, J., Khairy, P., Dobrev, D. & Nattel, S. The clinical profile and

pathophysiology of atrial fibrillation: relationships among clinical features,

epidemiology, and mechanisms. Circulation research 114, 1453-1468,

doi:10.1161/CIRCRESAHA.114.303211 (2014).

2 Lloyd-Jones, D. M. et al. Lifetime risk for development of atrial fibrillation: the

Framingham Heart Study. Circulation 110, 1042-1046,

doi:10.1161/01.CIR.0000140263.20897.42 (2004).

3 Weiss, J. N. et al. The dynamics of cardiac fibrillation. Circulation 112, 1232-

1240, doi:10.1161/CIRCULATIONAHA.104.529545 (2005).

4 McManus, D. D., Rienstra, M. & Benjamin, E. J. An update on the prognosis of

patients with atrial fibrillation. Circulation 126, e143-146,

doi:10.1161/CIRCULATIONAHA.112.129759 (2012).

5 Heeringa, J. et al. Subclinical atherosclerosis and risk of atrial fibrillation: the

rotterdam study. Archives of internal medicine 167, 382-387, doi:167/4/382

[pii]

10.1001/archinte.167.4.382 (2007).

6 Mathew, S. T., Patel, J. & Joseph, S. Atrial fibrillation: mechanistic insights and

treatment options. European journal of internal medicine 20, 672-681,

doi:S0953-6205(09)00140-X [pii]

10.1016/j.ejim.2009.07.011 (2009).

7 Movahed, M. R., Hashemzadeh, M. & Jamal, M. M. Diabetes mellitus is a strong,

independent risk for atrial fibrillation and flutter in addition to other

cardiovascular disease. Int J Cardiol 105, 315-318, doi:S0167-

5273(05)00523-1 [pii]

10.1016/j.ijcard.2005.02.050 (2005).

8 Nattel, S. Therapeutic implications of atrial fibrillation mechanisms: can

mechanistic insights be used to improve AF management? Cardiovascular

research 54, 347-360 (2002).

9 Wodchis, W. P., Bhatia, R. S., Leblanc, K., Meshkat, N. & Morra, D. A review of

the cost of atrial fibrillation. Value in health : the journal of the International

Society for Pharmacoeconomics and Outcomes Research 15, 240-248,

doi:10.1016/j.jval.2011.09.009 (2012).

10 Khaykin, Y. & Shamiss, Y. Cost of atrial fibrillation: invasive vs non-invasive

management in 2012. Current cardiology reviews 8, 368-373 (2012).

11 Ganesan, A. N. et al. Long-term outcomes of catheter ablation of atrial

fibrillation: a systematic review and meta-analysis. Journal of the American

Heart Association 2, e004549, doi:10.1161/JAHA.112.004549 (2013).

12 Mandapati, R., Skanes, A., Chen, J., Berenfeld, O. & Jalife, J. Stable

microreentrant sources as a mechanism of atrial fibrillation in the isolated

sheep heart. Circulation 101, 194-199 (2000).

13 Sarmast, F. et al. Cholinergic atrial fibrillation: I(K,ACh) gradients determine

unequal left/right atrial frequencies and rotor dynamics. Cardiovascular

research 59, 863-873 (2003).

Page 110: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

100

14 Nattel, S., Li, D. & Yue, L. Basic mechanisms of atrial fibrillation--very new

insights into very old ideas. Annu Rev Physiol 62, 51-77 (2000).

15 Schotten, U. et al. Electrical and contractile remodeling during the first days

of atrial fibrillation go hand in hand. Circulation 107, 1433-1439 (2003).

16 Shinagawa, K., Shi, Y. F., Tardif, J. C., Leung, T. K. & Nattel, S. Dynamic nature

of atrial fibrillation substrate during development and reversal of heart

failure in dogs. Circulation 105, 2672-2678 (2002).

17 Farrar, M. W., Bogart, D. B., Chapman, S. S. & Rigden, L. B. Atrial fibrillation in

athletes. Mo Med 103, 297-301 (2006).

18 Nattel, S. & Opie, L. H. Controversies in atrial fibrillation. Lancet 367, 262-272

(2006).

19 January, C. T. et al. 2014 AHA/ACC/HRS guideline for the management of

patients with atrial fibrillation: a report of the American College of

Cardiology/American Heart Association Task Force on Practice Guidelines

and the Heart Rhythm Society. Journal of the American College of Cardiology

64, e1-76, doi:10.1016/j.jacc.2014.03.022 (2014).

20 Hohnloser, S. H., Kuck, K. H. & Lilienthal, J. Rhythm or rate control in atrial

fibrillation--Pharmacological Intervention in Atrial Fibrillation (PIAF): a

randomised trial. Lancet 356, 1789-1794 (2000).

21 Wyse, D. G. et al. A comparison of rate control and rhythm control in patients

with atrial fibrillation. The New England journal of medicine 347, 1825-1833,

doi:10.1056/NEJMoa021328 (2002).

22 Van Gelder, I. C. et al. A comparison of rate control and rhythm control in

patients with recurrent persistent atrial fibrillation. The New England journal

of medicine 347, 1834-1840, doi:10.1056/NEJMoa021375 (2002).

23 Carlsson, J. et al. Randomized trial of rate-control versus rhythm-control in

persistent atrial fibrillation: the Strategies of Treatment of Atrial Fibrillation

(STAF) study. Journal of the American College of Cardiology 41, 1690-1696

(2003).

24 Caldeira, D., David, C. & Sampaio, C. Rate vs rhythm control in patients with

atrial fibrillation and heart failure: a systematic review and meta-analysis of

randomised controlled trials. European journal of internal medicine 22, 448-

455, doi:10.1016/j.ejim.2011.05.001 (2011).

25 Roy, D. et al. Rhythm control versus rate control for atrial fibrillation and

heart failure. The New England journal of medicine 358, 2667-2677,

doi:10.1056/NEJMoa0708789 (2008).

26 Corley, S. D. et al. Relationships between sinus rhythm, treatment, and

survival in the Atrial Fibrillation Follow-Up Investigation of Rhythm

Management (AFFIRM) Study. Circulation 109, 1509-1513,

doi:10.1161/01.CIR.0000121736.16643.11 (2004).

27 Singh, S. N. et al. Quality of life and exercise performance in patients in sinus

rhythm versus persistent atrial fibrillation: a Veterans Affairs Cooperative

Studies Program Substudy. Journal of the American College of Cardiology 48,

721-730, doi:10.1016/j.jacc.2006.03.051 (2006).

28 Guedon-Moreau, L. et al. Impact of the control of symptomatic paroxysmal

atrial fibrillation on health-related quality of life. Europace : European pacing,

Page 111: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

101

arrhythmias, and cardiac electrophysiology : journal of the working groups on

cardiac pacing, arrhythmias, and cardiac cellular electrophysiology of the

European Society of Cardiology 12, 634-642, doi:10.1093/europace/euq007

(2010).

29 Opolski, G. et al. Rate control vs rhythm control in patients with nonvalvular

persistent atrial fibrillation: the results of the Polish How to Treat Chronic

Atrial Fibrillation (HOT CAFE) Study. Chest 126, 476-486,

doi:10.1378/chest.126.2.476 (2004).

30 Dorian, P. & Mangat, I. Quality of life variables in the selection of rate versus

rhythm control in patients with atrial fibrillation: observations from the

Canadian Trial of Atrial Fibrillation. Cardiac electrophysiology review 7, 276-

279, doi:10.1023/B:CEPR.0000012395.33292.cd (2003).

31 Nishida, K. & Nattel, S. Atrial fibrillation compendium: historical context and

detailed translational perspective on an important clinical problem.

Circulation research 114, 1447-1452, doi:10.1161/CIRCRESAHA.114.303466

(2014).

32 Haissaguerre, M. et al. Spontaneous initiation of atrial fibrillation by ectopic

beats originating in the pulmonary veins. The New England journal of

medicine 339, 659-666, doi:10.1056/NEJM199809033391003 (1998).

33 Scheinman, M. M. & Morady, F. Nonpharmacological approaches to atrial

fibrillation. Circulation 103, 2120-2125 (2001).

34 Nishida, K., Datino, T., Macle, L. & Nattel, S. Atrial fibrillation ablation:

translating basic mechanistic insights to the patient. Journal of the American

College of Cardiology 64, 823-831, doi:10.1016/j.jacc.2014.06.1172 (2014).

35 Chang, S. L. et al. Biatrial substrate properties in patients with atrial

fibrillation. Journal of cardiovascular electrophysiology 18, 1134-1139,

doi:10.1111/j.1540-8167.2007.00941.x (2007).

36 Pappone, C. et al. Mortality, morbidity, and quality of life after circumferential

pulmonary vein ablation for atrial fibrillation: outcomes from a controlled

nonrandomized long-term study. Journal of the American College of

Cardiology 42, 185-197 (2003).

37 Gaita, F. et al. Long-term clinical results of 2 different ablation strategies in

patients with paroxysmal and persistent atrial fibrillation. Circulation.

Arrhythmia and electrophysiology 1, 269-275,

doi:10.1161/CIRCEP.108.774885 (2008).

38 Bhargava, M. et al. Impact of type of atrial fibrillation and repeat catheter

ablation on long-term freedom from atrial fibrillation: results from a

multicenter study. Heart rhythm : the official journal of the Heart Rhythm

Society 6, 1403-1412, doi:10.1016/j.hrthm.2009.06.014 (2009).

39 Tzou, W. S. et al. Long-term outcome after successful catheter ablation of

atrial fibrillation. Circulation. Arrhythmia and electrophysiology 3, 237-242,

doi:10.1161/CIRCEP.109.923771 (2010).

40 Ouyang, F. et al. Long-term results of catheter ablation in paroxysmal atrial

fibrillation: lessons from a 5-year follow-up. Circulation 122, 2368-2377,

doi:10.1161/CIRCULATIONAHA.110.946806 (2010).

Page 112: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

102

41 Medi, C. et al. Pulmonary vein antral isolation for paroxysmal atrial

fibrillation: results from long-term follow-up. Journal of cardiovascular

electrophysiology 22, 137-141, doi:10.1111/j.1540-8167.2010.01885.x

(2011).

42 Wijffels, M. C., Kirchhof, C. J., Dorland, R. & Allessie, M. A. Atrial fibrillation

begets atrial fibrillation. A study in awake chronically instrumented goats.

Circulation 92, 1954-1968 (1995).

43 Anne, W. et al. Self-terminating AF depends on electrical remodeling while

persistent AF depends on additional structural changes in a rapid atrially

paced sheep model. Journal of molecular and cellular cardiology 43, 148-158

(2007).

44 Nishida, K., Michael, G., Dobrev, D. & Nattel, S. Animal models for atrial

fibrillation: clinical insights and scientific opportunities. Europace : European

pacing, arrhythmias, and cardiac electrophysiology : journal of the working

groups on cardiac pacing, arrhythmias, and cardiac cellular electrophysiology

of the European Society of Cardiology 12, 160-172, doi:eup328 [pii]

10.1093/europace/eup328 (2010).

45 Amin, A. S., Tan, H. L. & Wilde, A. A. Cardiac ion channels in health and

disease. Heart rhythm : the official journal of the Heart Rhythm Society 7, 117-

126, doi:10.1016/j.hrthm.2009.08.005 (2010).

46 Tsai, C. T., Lai, L. P., Hwang, J. J., Lin, J. L. & Chiang, F. T. Molecular genetics of

atrial fibrillation. Journal of the American College of Cardiology 52, 241-250,

doi:S0735-1097(08)01200-X [pii]

10.1016/j.jacc.2008.02.072 (2008).

47 Choi, G. et al. Spectrum and frequency of cardiac channel defects in

swimming-triggered arrhythmia syndromes. Circulation 110, 2119-2124,

doi:01.CIR.0000144471.98080.CA [pii]

10.1161/01.CIR.0000144471.98080.CA (2004).

48 Watanabe, H. et al. Mutations in sodium channel beta1- and beta2-subunits

associated with atrial fibrillation. Circulation. Arrhythmia and

electrophysiology 2, 268-275, doi:CIRCEP.108.779181 [pii]

10.1161/CIRCEP.108.779181 (2009).

49 Francis, J. & Antzelevitch, C. Atrial fibrillation and Brugada syndrome. Journal

of the American College of Cardiology 51, 1149-1153, doi:S0735-

1097(08)00174-5 [pii]

10.1016/j.jacc.2007.10.062 (2008).

50 Chen, Y. H. et al. KCNQ1 gain-of-function mutation in familial atrial

fibrillation. Science 299, 251-254 (2003).

51 Dobrev, D. et al. The G protein-gated potassium current I(K,ACh) is

constitutively active in patients with chronic atrial fibrillation. Circulation

112, 3697-3706 (2005).

52 Li, J., McLerie, M. & Lopatin, A. N. Transgenic upregulation of IK1 in the

mouse heart leads to multiple abnormalities of cardiac excitability. American

journal of physiology. Heart and circulatory physiology 287, H2790-2802

(2004).

Page 113: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

103

53 Roberts, J. D. & Gollob, M. H. Impact of genetic discoveries on the

classification of lone atrial fibrillation. Journal of the American College of

Cardiology 55, 705-712, doi:S0735-1097(09)03989-8 [pii]

10.1016/j.jacc.2009.12.005 [doi].

54 Temple, J. et al. Atrial fibrillation in KCNE1-null mice. Circulation research 97,

62-69 (2005).

55 Fareh, S., Villemaire, C. & Nattel, S. Importance of refractoriness

heterogeneity in the enhanced vulnerability to atrial fibrillation induction

caused by tachycardia-induced atrial electrical remodeling. Circulation 98,

2202-2209 (1998).

56 Lukas A & Antzelevitch C. Phase 2 reentry as a mechanism of initiation of

circus movement reentry in canine epicardium exposed to simulated

ischemia. Cardiovascular research 32, 593-603 (1996).

57 Katsouras, G. et al. Differences in atrial fibrillation properties under vagal

nerve stimulation versus atrial tachycardia remodeling. Heart rhythm : the

official journal of the Heart Rhythm Society 6, 1465-1472 (2009).

58 Wang, Z., Page, P. & Nattel, S. Mechanism of flecainide's antiarrhythmic action

in experimental atrial fibrillation. Circulation research 71, 271-287 (1992).

59 Kneller, J. et al. Cholinergic atrial fibrillation in a computer model of a two-

dimensional sheet of canine atrial cells with realistic ionic properties.

Circulation research 90, E73-87 (2002).

60 Korantzopoulos, P., Kolettis, T. M., Goudevenos, J. A. & Siogas, K. Errors and

pitfalls in the non-invasive management of atrial fibrillation. Int J Cardiol

104, 125-130, doi:S0167-5273(05)00210-X [pii]

10.1016/j.ijcard.2004.11.014 (2005).

61 Po, S. S. et al. Rapid and stable re-entry within the pulmonary vein as a

mechanism initiating paroxysmal atrial fibrillation. Journal of the American

College of Cardiology 45, 1871-1877 (2005).

62 Desplantez, T., Dupont, E., Severs, N. J. & Weingart, R. Gap junction channels

and cardiac impulse propagation. The Journal of membrane biology 218, 13-

28, doi:10.1007/s00232-007-9046-8 (2007).

63 Henriquez, A. P. et al. Influence of dynamic gap junction resistance on

impulse propagation in ventricular myocardium: a computer simulation

study. Biophysical journal 81, 2112-2121, doi:S0006-3495(01)75859-6 [pii]

10.1016/S0006-3495(01)75859-6 (2001).

64 Hinch, R. An analytical study of the physiology and pathology of the

propagation of cardiac action potentials. Prog Biophys Mol Biol 78, 45-81,

doi:S0079610702000068 [pii] (2002).

65 Amin, A. S., Asghari-Roodsari, A. & Tan, H. L. Cardiac sodium channelopathies.

Pflugers Archiv : European journal of physiology 460, 223-237,

doi:10.1007/s00424-009-0761-0 (2010).

66 van der Velden, H. M. & Jongsma, H. J. Cardiac gap junctions and connexins:

their role in atrial fibrillation and potential as therapeutic targets.

Cardiovascular research 54, 270-279 (2002).

Page 114: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

104

67 van der Velden, H. M. et al. Gap junctional remodeling in relation to

stabilization of atrial fibrillation in the goat. Cardiovascular research 46, 476-

486 (2000).

68 Verheule, S. et al. Cardiac conduction abnormalities in mice lacking the gap

junction protein connexin40. Journal of cardiovascular electrophysiology 10,

1380-1389 (1999).

69 Gollob, M. H. et al. Somatic mutations in the connexin 40 gene (GJA5) in atrial

fibrillation. The New England journal of medicine 354, 2677-2688,

doi:10.1056/NEJMoa052800 (2006).

70 Burstein, B. et al. Changes in connexin expression and the atrial fibrillation

substrate in congestive heart failure. Circulation research 105, 1213-1222,

doi:CIRCRESAHA.108.183400 [pii]

10.1161/CIRCRESAHA.108.183400 (2009).

71 Chaldoupi, S. M., Loh, P., Hauer, R. N., de Bakker, J. M. & van Rijen, H. V. The

role of connexin40 in atrial fibrillation. Cardiovascular research 84, 15-23,

doi:cvp203 [pii]

10.1093/cvr/cvp203 (2009).

72 Liu, X. et al. Decreased connexin 43 and increased fibrosis in atrial regions

susceptible to complex fractionated atrial electrograms. Cardiology 114, 22-

29, doi:000210398 [pii]

10.1159/000210398 (2009).

73 Rossman, E. I. et al. The gap junction modifier, GAP-134 [(2S,4R)-1-(2-

aminoacetyl)-4-benzamido-pyrrolidine-2-carboxylic acid], improves

conduction and reduces atrial fibrillation/flutter in the canine sterile

pericarditis model. The Journal of pharmacology and experimental

therapeutics 329, 1127-1133, doi:jpet.108.150102 [pii]

10.1124/jpet.108.150102 (2009).

74 Li, J. Y. et al. Atrial gap junctions, NF-kappaB and fibrosis in patients

undergoing coronary artery bypass surgery: the relationship with

postoperative atrial fibrillation. Cardiology 112, 81-88, doi:000141012 [pii]

10.1159/000141012 (2009).

75 Rasmussen, H. S., Allen, M. J., Blackburn, K. J., Butrous, G. S. & Dalrymple, H. W.

Dofetilide, a novel class III antiarrhythmic agent. Journal of cardiovascular

pharmacology 20 Suppl 2, S96-105 (1992).

76 Gwilt, M. et al. UK-68,798: a novel, potent and highly selective class III

antiarrhythmic agent which blocks potassium channels in cardiac cells. The

Journal of pharmacology and experimental therapeutics 256, 318-324 (1991).

77 Knilans, T. K., Lathrop, D. A., Nanasi, P. P., Schwartz, A. & Varro, A. Rate and

concentration-dependent effects of UK-68,798, a potent new class III

antiarrhythmic, on canine Purkinje fibre action potential duration and Vmax.

British journal of pharmacology 103, 1568-1572 (1991).

78 Singh, S. et al. Efficacy and safety of oral dofetilide in converting to and

maintaining sinus rhythm in patients with chronic atrial fibrillation or atrial

flutter: the symptomatic atrial fibrillation investigative research on dofetilide

(SAFIRE-D) study. Circulation 102, 2385-2390 (2000).

Page 115: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

105

79 Kober, L. et al. Effect of dofetilide in patients with recent myocardial

infarction and left-ventricular dysfunction: a randomised trial. Lancet 356,

2052-2058 (2000).

80 Torp-Pedersen, C. et al. Dofetilide in patients with congestive heart failure

and left ventricular dysfunction. Danish Investigations of Arrhythmia and

Mortality on Dofetilide Study Group. The New England journal of medicine

341, 857-865, doi:10.1056/NEJM199909163411201 (1999).

81 Fuster, V. et al. 2011 ACCF/AHA/HRS focused updates incorporated into the

ACC/AHA/ESC 2006 Guidelines for the management of patients with atrial

fibrillation: a report of the American College of Cardiology

Foundation/American Heart Association Task Force on Practice Guidelines

developed in partnership with the European Society of Cardiology and in

collaboration with the European Heart Rhythm Association and the Heart

Rhythm Society. Journal of the American College of Cardiology 57, e101-198,

doi:10.1016/j.jacc.2010.09.013 (2011).

82 Sanoski, C. A. Atrial fibrillation and managed care: current approaches and

future directions for long-term therapy. Introduction. Journal of managed

care pharmacy : JMCP 15, S3 (2009).

83 Campbell, T. J. Kinetics of onset of rate-dependent effects of Class I

antiarrhythmic drugs are important in determining their effects on

refractoriness in guinea-pig ventricle, and provide a theoretical basis for

their subclassification. Cardiovascular research 17, 344-352 (1983).

84 Harrison, D. C. Antiarrhythmic drug classification: new science and practical

applications. The American journal of cardiology 56, 185-187 (1985).

85 Wang, Z., Feng, J. & Nattel, S. Idiopathic atrial fibrillation in dogs:

electrophysiologic determinants and mechanisms of antiarrhythmic action of

flecainide. Journal of the American College of Cardiology 26, 277-286 (1995).

86 Anderson, J. L. et al. Prevention of symptomatic recurrences of paroxysmal

atrial fibrillation in patients initially tolerating antiarrhythmic therapy. A

multicenter, double-blind, crossover study of flecainide and placebo with

transtelephonic monitoring. Flecainide Supraventricular Tachycardia Study

Group. Circulation 80, 1557-1570 (1989).

87 Pietersen, A. H. & Hellemann, H. Usefulness of flecainide for prevention of

paroxysmal atrial fibrillation and flutter. Danish-Norwegian Flecainide

Multicenter Study Group. The American journal of cardiology 67, 713-717

(1991).

88 Akiyama, T., Pawitan, Y., Greenberg, H., Kuo, C. S. & Reynolds-Haertle, R. A.

Increased risk of death and cardiac arrest from encainide and flecainide in

patients after non-Q-wave acute myocardial infarction in the Cardiac

Arrhythmia Suppression Trial. CAST Investigators. The American journal of

cardiology 68, 1551-1555 (1991).

89 Nappi, J. M. & Anderson, J. L. Flecainide: a new prototype antiarrhythmic

agent. Pharmacotherapy 5, 209-221 (1985).

90 Estes, N. A., 3rd, Garan, H. & Ruskin, J. N. Electrophysiologic properties of

flecainide acetate. The American journal of cardiology 53, 26B-29B (1984).

Page 116: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

106

91 Packer, D. L., Munger, T. M., Johnson, S. B. & Cragun, K. T. Mechanism of lethal

proarrhythmia observed in the Cardiac Arrhythmia Suppression Trial: role of

adrenergic modulation of drug binding. Pacing and clinical electrophysiology :

PACE 20, 455-467 (1997).

92 Pandit, S. V. & Jalife, J. Rotors and the dynamics of cardiac fibrillation.

Circulation research 112, 849-862, doi:10.1161/CIRCRESAHA.111.300158

(2013).

93 Mines, G. R. On dynamic equilibrium in the heart. The Journal of physiology

46, 349-383 (1913).

94 Lewis, T. Oliver-Sharpey Lectures ON THE NATURE OF FLUTTER AND

FIBRILLATION OF THE AURICLE. British medical journal 1, 590-593 (1921).

95 Allessie, M. A., Bonke, F. I. & Schopman, F. J. Circus movement in rabbit atrial

muscle as a mechanism of trachycardia. Circulation research 33, 54-62

(1973).

96 Garanin, O. A., Popov, N. V., Efimov, S. V., Udovikov, A. I. & Grigor'eva, G. V.

[The dynamics of the species composition of nidiculous animals in different

types of nests of the little suslik]. Parazitologiia 29, 37-42 (1995).

97 Zhabotinsky, A. M. A history of chemical oscillations and waves. Chaos 1, 379-

386, doi:10.1063/1.165848 (1991).

98 Vaquero, M., Calvo, D. & Jalife, J. Cardiac fibrillation: from ion channels to

rotors in the human heart. Heart rhythm : the official journal of the Heart

Rhythm Society 5, 872-879, doi:10.1016/j.hrthm.2008.02.034 (2008).

99 Gray, R. A., Pertsov, A. M. & Jalife, J. Spatial and temporal organization during

cardiac fibrillation. Nature 392, 75-78, doi:10.1038/32164 (1998).

100 Fast, V. G. & Kleber, A. G. Role of wavefront curvature in propagation of

cardiac impulse. Cardiovascular research 33, 258-271 (1997).

101 Panfilov, A. V. & Pertsov, A. M. [Mechanism of spiral waves initiation in active

media, connected with critical curvature phenomenon]. Biofizika 27, 886-889

(1982).

102 Schotten, U., Verheule, S., Kirchhof, P. & Goette, A. Pathophysiological

mechanisms of atrial fibrillation: a translational appraisal. Physiological

reviews 91, 265-325, doi:10.1152/physrev.00031.2009 (2011).

103 Gray, R. A. et al. Mechanisms of cardiac fibrillation. Science 270, 1222-1223;

author reply 1224-1225 (1995).

104 Jalife, J. Inward rectifier potassium channels control rotor frequency in

ventricular fibrillation. Heart rhythm : the official journal of the Heart Rhythm

Society 6, S44-48, doi:10.1016/j.hrthm.2009.07.019 (2009).

105 Jalife, J. & Pandit, S. V. Ionic mechanisms of wavebreak in fibrillation. Heart

rhythm : the official journal of the Heart Rhythm Society 2, 660-663,

doi:10.1016/j.hrthm.2005.03.016 (2005).

106 Pertsov, A. M., Davidenko, J. M., Salomonsz, R., Baxter, W. T. & Jalife, J. Spiral

waves of excitation underlie reentrant activity in isolated cardiac muscle.

Circulation research 72, 631-650 (1993).

107 Wohlfart, B. Analysis of mechanical alternans in rabbit papillary muscle. Acta

physiologica Scandinavica 115, 405-414, doi:10.1111/j.1748-

1716.1982.tb07098.x (1982).

Page 117: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

107

108 Walker, M. L. & Rosenbaum, D. S. Repolarization alternans: implications for

the mechanism and prevention of sudden cardiac death. Cardiovascular

research 57, 599-614 (2003).

109 Pitt, G. S., Dun, W. & Boyden, P. A. Remodeled cardiac calcium channels.

Journal of molecular and cellular cardiology 41, 373-388,

doi:10.1016/j.yjmcc.2006.06.071 (2006).

110 Verrier, R. L. & Nieminen, T. T-wave alternans as a therapeutic marker for

antiarrhythmic agents. Journal of cardiovascular pharmacology 55, 544-554,

doi:10.1097/FJC.0b013e3181d6b781 (2010).

111 Nolasco, J. B. & Dahlen, R. W. A graphic method for the study of alternation in

cardiac action potentials. Journal of applied physiology 25, 191-196 (1968).

112 Tolkacheva, E. G., Anumonwo, J. M. & Jalife, J. Action potential duration

restitution portraits of mammalian ventricular myocytes: role of calcium

current. Biophysical journal 91, 2735-2745,

doi:10.1529/biophysj.106.083865 (2006).

113 Tolkacheva, E. G., Romeo, M. M., Guerraty, M. & Gauthier, D. J. Condition for

alternans and its control in a two-dimensional mapping model of paced

cardiac dynamics. Physical review. E, Statistical, nonlinear, and soft matter

physics 69, 031904 (2004).

114 Watanabe, M. A. & Koller, M. L. Mathematical analysis of dynamics of cardiac

memory and accommodation: theory and experiment. American journal of

physiology. Heart and circulatory physiology 282, H1534-1547,

doi:10.1152/ajpheart.00351.2001 (2002).

115 Gelzer, A. R. et al. Dynamic mechanism for initiation of ventricular fibrillation

in vivo. Circulation 118, 1123-1129,

doi:10.1161/CIRCULATIONAHA.107.738013 (2008).

116 Kanaporis, G. & Blatter, L. A. The mechanisms of calcium cycling and action

potential dynamics in cardiac alternans. Circulation research 116, 846-856,

doi:10.1161/CIRCRESAHA.116.305404 (2015).

117 Herweg, B., Kowalski, M. & Steinberg, J. S. Termination of persistent atrial

fibrillation resistant to cardioversion by a single radiofrequency application.

Pacing and clinical electrophysiology : PACE 26, 1420-1423 (2003).

118 Lazar, S., Dixit, S., Marchlinski, F. E., Callans, D. J. & Gerstenfeld, E. P. Presence

of left-to-right atrial frequency gradient in paroxysmal but not persistent

atrial fibrillation in humans. Circulation 110, 3181-3186,

doi:10.1161/01.CIR.0000147279.91094.5E (2004).

119 Wu, T. J. et al. Simultaneous biatrial computerized mapping during

permanent atrial fibrillation in patients with organic heart disease. Journal of

cardiovascular electrophysiology 13, 571-577 (2002).

120 Sahadevan, J. et al. Epicardial mapping of chronic atrial fibrillation in

patients: preliminary observations. Circulation 110, 3293-3299,

doi:10.1161/01.CIR.0000147781.02738.13 (2004).

121 Sanders, P. et al. Spectral analysis identifies sites of high-frequency activity

maintaining atrial fibrillation in humans. Circulation 112, 789-797,

doi:10.1161/CIRCULATIONAHA.104.517011 (2005).

Page 118: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

108

122 Schricker, A. A., Lalani, G. G., Krummen, D. E. & Narayan, S. M. Rotors as

drivers of atrial fibrillation and targets for ablation. Current cardiology

reports 16, 509, doi:10.1007/s11886-014-0509-0 (2014).

123 Ganesan, A. N. et al. Bipolar electrogram shannon entropy at sites of

rotational activation: implications for ablation of atrial fibrillation.

Circulation. Arrhythmia and electrophysiology 6, 48-57,

doi:10.1161/CIRCEP.112.976654 (2013).

124 Narayan, S. M. et al. Ablation of rotor and focal sources reduces late

recurrence of atrial fibrillation compared with trigger ablation alone:

extended follow-up of the CONFIRM trial (Conventional Ablation for Atrial

Fibrillation With or Without Focal Impulse and Rotor Modulation). Journal of

the American College of Cardiology 63, 1761-1768,

doi:10.1016/j.jacc.2014.02.543 (2014).

125 Miller, J. M. et al. Initial independent outcomes from focal impulse and rotor

modulation ablation for atrial fibrillation: multicenter FIRM registry. Journal

of cardiovascular electrophysiology 25, 921-929, doi:10.1111/jce.12474

(2014).

126 Thomson, J. A. et al. Embryonic stem cell lines derived from human

blastocysts. Science 282, 1145-1147 (1998).

127 Takahashi, K. & Yamanaka, S. Induction of pluripotent stem cells from mouse

embryonic and adult fibroblast cultures by defined factors. Cell 126, 663-

676, doi:10.1016/j.cell.2006.07.024 (2006).

128 Takahashi, K., Okita, K., Nakagawa, M. & Yamanaka, S. Induction of

pluripotent stem cells from fibroblast cultures. Nature protocols 2, 3081-

3089, doi:10.1038/nprot.2007.418 (2007).

129 Murry, C. E. & Keller, G. Differentiation of embryonic stem cells to clinically

relevant populations: lessons from embryonic development. Cell 132, 661-

680, doi:10.1016/j.cell.2008.02.008 (2008).

130 Ng, E. S. et al. The primitive streak gene Mixl1 is required for efficient

haematopoiesis and BMP4-induced ventral mesoderm patterning in

differentiating ES cells. Development 132, 873-884, doi:10.1242/dev.01657

(2005).

131 Kubo, A. et al. Development of definitive endoderm from embryonic stem

cells in culture. Development 131, 1651-1662, doi:10.1242/dev.01044

(2004).

132 Wiles, M. V. & Johansson, B. M. Embryonic stem cell development in a

chemically defined medium. Experimental cell research 247, 241-248,

doi:10.1006/excr.1998.4353 (1999).

133 Yasunaga, M. et al. Induction and monitoring of definitive and visceral

endoderm differentiation of mouse ES cells. Nature biotechnology 23, 1542-

1550, doi:10.1038/nbt1167 (2005).

134 Fehling, H. J. et al. Tracking mesoderm induction and its specification to the

hemangioblast during embryonic stem cell differentiation. Development 130,

4217-4227 (2003).

135 Gadue, P., Huber, T. L., Paddison, P. J. & Keller, G. M. Wnt and TGF-beta

signaling are required for the induction of an in vitro model of primitive

Page 119: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

109

streak formation using embryonic stem cells. Proceedings of the National

Academy of Sciences of the United States of America 103, 16806-16811,

doi:10.1073/pnas.0603916103 (2006).

136 Tada, S. et al. Characterization of mesendoderm: a diverging point of the

definitive endoderm and mesoderm in embryonic stem cell differentiation

culture. Development 132, 4363-4374, doi:10.1242/dev.02005 (2005).

137 Ying, Q. L., Stavridis, M., Griffiths, D., Li, M. & Smith, A. Conversion of

embryonic stem cells into neuroectodermal precursors in adherent

monoculture. Nature biotechnology 21, 183-186, doi:10.1038/nbt780 (2003).

138 Hogan, B. L. Bone morphogenetic proteins in development. Current opinion in

genetics & development 6, 432-438 (1996).

139 Conlon, F. L. et al. A primary requirement for nodal in the formation and

maintenance of the primitive streak in the mouse. Development 120, 1919-

1928 (1994).

140 Schier, A. F. Nodal signaling in vertebrate development. Annual review of cell

and developmental biology 19, 589-621,

doi:10.1146/annurev.cellbio.19.041603.094522 (2003).

141 Gadue, P., Huber, T. L., Nostro, M. C., Kattman, S. & Keller, G. M. Germ layer

induction from embryonic stem cells. Experimental hematology 33, 955-964,

doi:10.1016/j.exphem.2005.06.009 (2005).

142 Ema, M., Takahashi, S. & Rossant, J. Deletion of the selection cassette, but not

cis-acting elements, in targeted Flk1-lacZ allele reveals Flk1 expression in

multipotent mesodermal progenitors. Blood 107, 111-117,

doi:10.1182/blood-2005-05-1970 (2006).

143 Kataoka, H. et al. Expressions of PDGF receptor alpha, c-Kit and Flk1 genes

clustering in mouse chromosome 5 define distinct subsets of nascent

mesodermal cells. Development, growth & differentiation 39, 729-740 (1997).

144 Ueno, S. et al. Biphasic role for Wnt/beta-catenin signaling in cardiac

specification in zebrafish and embryonic stem cells. Proceedings of the

National Academy of Sciences of the United States of America 104, 9685-9690,

doi:10.1073/pnas.0702859104 (2007).

145 Era, T. et al. Multiple mesoderm subsets give rise to endothelial cells,

whereas hematopoietic cells are differentiated only from a restricted subset

in embryonic stem cell differentiation culture. Stem cells 26, 401-411,

doi:10.1634/stemcells.2006-0809 (2008).

146 Moretti, A. et al. Multipotent embryonic isl1+ progenitor cells lead to cardiac,

smooth muscle, and endothelial cell diversification. Cell 127, 1151-1165,

doi:10.1016/j.cell.2006.10.029 (2006).

147 Wu, S. M. et al. Developmental origin of a bipotential myocardial and smooth

muscle cell precursor in the mammalian heart. Cell 127, 1137-1150,

doi:10.1016/j.cell.2006.10.028 (2006).

148 DeRuiter, M. C., Poelmann, R. E., VanderPlas-de Vries, I., Mentink, M. M. &

Gittenberger-de Groot, A. C. The development of the myocardium and

endocardium in mouse embryos. Fusion of two heart tubes? Anatomy and

embryology 185, 461-473 (1992).

Page 120: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

110

149 Lian, X. et al. Directed cardiomyocyte differentiation from human pluripotent

stem cells by modulating Wnt/beta-catenin signaling under fully defined

conditions. Nature protocols 8, 162-175, doi:10.1038/nprot.2012.150 (2013).

150 van den Heuvel, N. H., van Veen, T. A., Lim, B. & Jonsson, M. K. Lessons from

the heart: mirroring electrophysiological characteristics during cardiac

development to in vitro differentiation of stem cell derived cardiomyocytes.

Journal of molecular and cellular cardiology 67, 12-25,

doi:10.1016/j.yjmcc.2013.12.011 (2014).

151 Satin, J. et al. Calcium handling in human embryonic stem cell-derived

cardiomyocytes. Stem cells 26, 1961-1972, doi:10.1634/stemcells.2007-0591

(2008).

152 Itzhaki, I. et al. Calcium handling in human induced pluripotent stem cell

derived cardiomyocytes. PloS one 6, e18037,

doi:10.1371/journal.pone.0018037 (2011).

153 Lundy, S. D., Zhu, W. Z., Regnier, M. & Laflamme, M. A. Structural and

functional maturation of cardiomyocytes derived from human pluripotent

stem cells. Stem cells and development 22, 1991-2002,

doi:10.1089/scd.2012.0490 (2013).

154 Yang, L. et al. Human cardiovascular progenitor cells develop from a KDR+

embryonic-stem-cell-derived population. Nature 453, 524-528,

doi:10.1038/nature06894 (2008).

155 Kattman, S. J. et al. Stage-specific optimization of activin/nodal and BMP

signaling promotes cardiac differentiation of mouse and human pluripotent

stem cell lines. Cell stem cell 8, 228-240, doi:10.1016/j.stem.2010.12.008

(2011).

156 Ma, J. et al. High purity human-induced pluripotent stem cell-derived

cardiomyocytes: electrophysiological properties of action potentials and

ionic currents. American journal of physiology. Heart and circulatory

physiology 301, H2006-2017, doi:10.1152/ajpheart.00694.2011 (2011).

157 Xavier-Neto, J. et al. Retinoid signaling and cardiac anteroposterior

segmentation. Genesis 31, 97-104 (2001).

158 Hochgreb, T. et al. A caudorostral wave of RALDH2 conveys anteroposterior

information to the cardiac field. Development 130, 5363-5374,

doi:10.1242/dev.00750 (2003).

159 Xavier-Neto, J. et al. A retinoic acid-inducible transgenic marker of sino-atrial

development in the mouse heart. Development 126, 2677-2687 (1999).

160 Zhang, Q. et al. Direct differentiation of atrial and ventricular myocytes from

human embryonic stem cells by alternating retinoid signals. Cell research 21,

579-587, doi:10.1038/cr.2010.163 (2011).

161 Niederreither, K. et al. Embryonic retinoic acid synthesis is essential for heart

morphogenesis in the mouse. Development 128, 1019-1031 (2001).

162 Emoto, Y., Wada, H., Okamoto, H., Kudo, A. & Imai, Y. Retinoic acid-

metabolizing enzyme Cyp26a1 is essential for determining territories of

hindbrain and spinal cord in zebrafish. Developmental biology 278, 415-427,

doi:10.1016/j.ydbio.2004.11.023 (2005).

Page 121: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

111

163 Lu, Z. Q., Sinha, A., Sharma, P., Kislinger, T. & Gramolini, A. O. Proteomic

analysis of human fetal atria and ventricle. Journal of proteome research 13,

5869-5878, doi:10.1021/pr5007685 (2014).

164 Ellinor, P. T. et al. Common variants in KCNN3 are associated with lone atrial

fibrillation. Nature genetics 42, 240-244, doi:10.1038/ng.537 (2010).

165 O'Brien, T. X., Lee, K. J. & Chien, K. R. Positional specification of ventricular

myosin light chain 2 expression in the primitive murine heart tube.

Proceedings of the National Academy of Sciences of the United States of

America 90, 5157-5161 (1993).

166 Bruneau, B. G. et al. Cardiac expression of the ventricle-specific homeobox

gene Irx4 is modulated by Nkx2-5 and dHand. Developmental biology 217,

266-277, doi:10.1006/dbio.1999.9548 (2000).

167 Moretti, A. et al. Patient-specific induced pluripotent stem-cell models for

long-QT syndrome. The New England journal of medicine 363, 1397-1409,

doi:10.1056/NEJMoa0908679 (2010).

168 Lan, F. et al. Abnormal calcium handling properties underlie familial

hypertrophic cardiomyopathy pathology in patient-specific induced

pluripotent stem cells. Cell stem cell 12, 101-113,

doi:10.1016/j.stem.2012.10.010 (2013).

169 Sun, N. et al. Patient-specific induced pluripotent stem cells as a model for

familial dilated cardiomyopathy. Science translational medicine 4, 130ra147,

doi:10.1126/scitranslmed.3003552 (2012).

170 Kim, C. et al. Studying arrhythmogenic right ventricular dysplasia with

patient-specific iPSCs. Nature 494, 105-110, doi:10.1038/nature11799

(2013).

171 Schwartz, P. J., Crotti, L. & Insolia, R. Long-QT syndrome: from genetics to

management. Circulation. Arrhythmia and electrophysiology 5, 868-877,

doi:10.1161/CIRCEP.111.962019 (2012).

172 Ackerman, M. J. et al. HRS/EHRA expert consensus statement on the state of

genetic testing for the channelopathies and cardiomyopathies: this document

was developed as a partnership between the Heart Rhythm Society (HRS)

and the European Heart Rhythm Association (EHRA). Europace : European

pacing, arrhythmias, and cardiac electrophysiology : journal of the working

groups on cardiac pacing, arrhythmias, and cardiac cellular electrophysiology

of the European Society of Cardiology 13, 1077-1109,

doi:10.1093/europace/eur245 (2011).

173 Villain, E. et al. Low incidence of cardiac events with beta-blocking therapy in

children with long QT syndrome. European heart journal 25, 1405-1411,

doi:10.1016/j.ehj.2004.06.016 (2004).

174 Monnig, G. et al. Implantable cardioverter-defibrillator therapy in patients

with congenital long-QT syndrome: a long-term follow-up. Heart rhythm : the

official journal of the Heart Rhythm Society 2, 497-504,

doi:10.1016/j.hrthm.2005.02.008 (2005).

175 Schwartz, P. J. et al. Who are the long-QT syndrome patients who receive an

implantable cardioverter-defibrillator and what happens to them?: data from

the European Long-QT Syndrome Implantable Cardioverter-Defibrillator

Page 122: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

112

(LQTS ICD) Registry. Circulation 122, 1272-1282,

doi:10.1161/CIRCULATIONAHA.110.950147 (2010).

176 Horner, J. M. et al. Implantable cardioverter defibrillator therapy for

congenital long QT syndrome: a single-center experience. Heart rhythm : the

official journal of the Heart Rhythm Society 7, 1616-1622,

doi:10.1016/j.hrthm.2010.08.023 (2010).

177 Schwartz, P. J. et al. Left cardiac sympathetic denervation in the management

of high-risk patients affected by the long-QT syndrome. Circulation 109,

1826-1833, doi:10.1161/01.CIR.0000125523.14403.1E

01.CIR.0000125523.14403.1E [pii] (2004).

178 Ackerman, M. J. et al. HRS/EHRA expert consensus statement on the state of

genetic testing for the channelopathies and cardiomyopathies: this document

was developed as a partnership between the Heart Rhythm Society (HRS)

and the European Heart Rhythm Association (EHRA). Europace 13, 1077-

1109, doi:eur245 [pii]

10.1093/europace/eur245 (2011).

179 Nunn, L. M. & Lambiase, P. D. Genetics and cardiovascular disease--causes

and prevention of unexpected sudden adult death: the role of the SADS clinic.

Heart 97, 1122-1127, doi:10.1136/hrt.2010.218511

hrt.2010.218511 [pii] (2011).

180 Zipes, D. P. et al. ACC/AHA/ESC 2006 guidelines for management of patients

with ventricular arrhythmias and the prevention of sudden cardiac death: a

report of the American College of Cardiology/American Heart Association

Task Force and the European Society of Cardiology Committee for Practice

Guidelines (Writing Committee to Develop Guidelines for Management of

Patients With Ventricular Arrhythmias and the Prevention of Sudden Cardiac

Death). J Am Coll Cardiol 48, e247-346 (2006).

181 Tang, A. S. et al. Canadian Cardiovascular Society/Canadian Heart Rhythm

Society position paper on implantable cardioverter defibrillator use in

Canada. Can J Cardiol 21 Suppl A, 11-18 (2005).

182 Olde Nordkamp, L. R. et al. The ICD for primary prevention in patients with

inherited cardiac diseases: indications, use, and outcome: a comparison with

secondary prevention. Circulation. Arrhythmia and electrophysiology 6, 91-

100, doi:10.1161/CIRCEP.112.975268 (2013).

183 Viswanathan, P. C. & Rudy, Y. Pause induced early afterdepolarizations in the

long QT syndrome: a simulation study. Cardiovascular research 42, 530-542

(1999).

184 Clancy, C. E. & Rudy, Y. Linking a genetic defect to its cellular phenotype in a

cardiac arrhythmia. Nature 400, 566-569, doi:10.1038/23034 (1999).

185 Lankipalli, R. S., Zhu, T., Guo, D. & Yan, G. X. Mechanisms underlying

arrhythmogenesis in long QT syndrome. Journal of electrocardiology 38, 69-

73, doi:10.1016/j.jelectrocard.2005.06.008 (2005).

186 Itzhaki, I. et al. Modelling the long QT syndrome with induced pluripotent

stem cells. Nature 471, 225-229, doi:10.1038/nature09747 (2011).

Page 123: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

113

187 Yan, G. X. & Martin, J. Electrocardiographic T wave: a symbol of transmural

dispersion of repolarization in the ventricles. Journal of cardiovascular

electrophysiology 14, 639-640 (2003).

188 Yan, G. X. et al. Phase 2 early afterdepolarization as a trigger of polymorphic

ventricular tachycardia in acquired long-QT syndrome : direct evidence from

intracellular recordings in the intact left ventricular wall. Circulation 103,

2851-2856 (2001).

189 Yan, G. X. et al. Ventricular hypertrophy amplifies transmural repolarization

dispersion and induces early afterdepolarization. American journal of

physiology. Heart and circulatory physiology 281, H1968-1975 (2001).

190 Echt, D. S. et al. Mortality and morbidity in patients receiving encainide,

flecainide, or placebo. The Cardiac Arrhythmia Suppression Trial. The New

England journal of medicine 324, 781-788,

doi:10.1056/NEJM199103213241201 (1991).

191 Paul, S. M. et al. How to improve R&D productivity: the pharmaceutical

industry's grand challenge. Nature reviews. Drug discovery 9, 203-214,

doi:10.1038/nrd3078 (2010).

192 Kola, I. & Landis, J. Can the pharmaceutical industry reduce attrition rates?

Nature reviews. Drug discovery 3, 711-715, doi:10.1038/nrd1470 (2004).

193 MacDonald, J. S. & Robertson, R. T. Toxicity testing in the 21st century: a view

from the pharmaceutical industry. Toxicological sciences : an official journal

of the Society of Toxicology 110, 40-46, doi:10.1093/toxsci/kfp088 (2009).

194 Mordwinkin, N. M., Lee, A. S. & Wu, J. C. Patient-specific stem cells and

cardiovascular drug discovery. Jama 310, 2039-2040,

doi:10.1001/jama.2013.282409 (2013).

195 Walker, B. D. et al. Congenital and acquired long QT syndromes. The Canadian

journal of cardiology 19, 76-87 (2003).

196 Chevalier, P. et al. Non-invasive testing of acquired long QT syndrome:

evidence for multiple arrhythmogenic substrates. Cardiovascular research 50,

386-398 (2001).

197 Liang, P. et al. Drug screening using a library of human induced pluripotent

stem cell-derived cardiomyocytes reveals disease-specific patterns of

cardiotoxicity. Circulation 127, 1677-1691,

doi:10.1161/CIRCULATIONAHA.113.001883 (2013).

198 Wang, Y. et al. Genome editing of isogenic human induced pluripotent stem

cells recapitulates long QT phenotype for drug testing. Journal of the

American College of Cardiology 64, 451-459, doi:10.1016/j.jacc.2014.04.057

(2014).

199 Lafuente-Lafuente, C., Longas-Tejero, M. A., Bergmann, J. F. & Belmin, J.

Antiarrhythmics for maintaining sinus rhythm after cardioversion of atrial

fibrillation. The Cochrane database of systematic reviews 5, CD005049,

doi:10.1002/14651858.CD005049.pub3 (2012).

200 Brito-Martins, M., Harding, S. E. & Ali, N. N. beta(1)- and beta(2)-

adrenoceptor responses in cardiomyocytes derived from human embryonic

stem cells: comparison with failing and non-failing adult human heart. British

journal of pharmacology 153, 751-759, doi:10.1038/sj.bjp.0707619 (2008).

Page 124: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

114

201 Lu, H. R., Marien, R., Saels, A. & De Clerck, F. Species plays an important role

in drug-induced prolongation of action potential duration and early

afterdepolarizations in isolated Purkinje fibers. Journal of cardiovascular

electrophysiology 12, 93-102 (2001).

202 Lian, Q., Chow, Y., Esteban, M. A., Pei, D. & Tse, H. F. Future perspective of

induced pluripotent stem cells for diagnosis, drug screening and treatment of

human diseases. Thrombosis and haemostasis 104, 39-44, doi:10.1160/TH10-

05-0269 (2010).

203 Kennedy, M., D'Souza, S. L., Lynch-Kattman, M., Schwantz, S. & Keller, G.

Development of the hemangioblast defines the onset of hematopoiesis in

human ES cell differentiation cultures. Blood 109, 2679-2687,

doi:10.1182/blood-2006-09-047704 (2007).

204 Hamill, O. P., Marty, A., Neher, E., Sakmann, B. & Sigworth, F. J. Improved

patch-clamp techniques for high-resolution current recording from cells and

cell-free membrane patches. Pflugers Archiv : European journal of physiology

391, 85-100 (1981).

205 Barry, P. H. & Lynch, J. W. Liquid junction potentials and small cell effects in

patch-clamp analysis. The Journal of membrane biology 121, 101-117 (1991).

206 Fluhler, E., Burnham, V. G. & Loew, L. M. Spectra, membrane binding, and

potentiometric responses of new charge shift probes. Biochemistry 24, 5749-

5755 (1985).

207 Fedorov, V. V. et al. Application of blebbistatin as an excitation-contraction

uncoupler for electrophysiologic study of rat and rabbit hearts. Heart rhythm

: the official journal of the Heart Rhythm Society 4, 619-626,

doi:10.1016/j.hrthm.2006.12.047 (2007).

208 Elliott, D. A. et al. NKX2-5(eGFP/w) hESCs for isolation of human cardiac

progenitors and cardiomyocytes. Nature methods 8, 1037-1040,

doi:10.1038/nmeth.1740 (2011).

209 Schwartz, R. J. & Olson, E. N. Building the heart piece by piece: modularity of

cis-elements regulating Nkx2-5 transcription. Development 126, 4187-4192

(1999).

210 Gassanov, N. et al. Retinoid acid-induced effects on atrial and pacemaker cell

differentiation and expression of cardiac ion channels. Differentiation;

research in biological diversity 76, 971-980, doi:10.1111/j.1432-

0436.2008.00283.x (2008).

211 Storms, R. W. et al. Isolation of primitive human hematopoietic progenitors

on the basis of aldehyde dehydrogenase activity. Proceedings of the National

Academy of Sciences of the United States of America 96, 9118-9123 (1999).

212 Evseenko, D. et al. Mapping the first stages of mesoderm commitment during

differentiation of human embryonic stem cells. Proceedings of the National

Academy of Sciences of the United States of America 107, 13742-13747,

doi:10.1073/pnas.1002077107 (2010).

213 Prall, O. W. et al. An Nkx2-5/Bmp2/Smad1 negative feedback loop controls

heart progenitor specification and proliferation. Cell 128, 947-959,

doi:10.1016/j.cell.2007.01.042 (2007).

Page 125: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

115

214 Dubois, N. C. et al. SIRPA is a specific cell-surface marker for isolating

cardiomyocytes derived from human pluripotent stem cells. Nature

biotechnology 29, 1011-1018, doi:10.1038/nbt.2005 (2011).

215 Xie, Y., Sato, D., Garfinkel, A., Qu, Z. & Weiss, J. N. So little source, so much sink:

requirements for afterdepolarizations to propagate in tissue. Biophysical

journal 99, 1408-1415, doi:10.1016/j.bpj.2010.06.042 (2010).

216 Cabo, C. et al. Vortex shedding as a precursor of turbulent electrical activity in

cardiac muscle. Biophysical journal 70, 1105-1111, doi:10.1016/S0006-

3495(96)79691-1 (1996).

217 Bray, M. A. & Wikswo, J. P. Considerations in phase plane analysis for

nonstationary reentrant cardiac behavior. Physical review. E, Statistical,

nonlinear, and soft matter physics 65, 051902 (2002).

218 Mansour, M. et al. Left-to-right gradient of atrial frequencies during acute

atrial fibrillation in the isolated sheep heart. Circulation 103, 2631-2636

(2001).

219 Gonzalez, H., Nagai, Y., Bub, G., Glass, L. & Shrier, A. Reentrant waves in a ring

of embryonic chick ventricular cells imaged with a Ca2+ sensitive dye. Bio

Systems 71, 71-80 (2003).

220 Iravanian, S. et al. Functional reentry in cultured monolayers of neonatal rat

cardiac cells. American journal of physiology. Heart and circulatory physiology

285, H449-456, doi:10.1152/ajpheart.00896.2002 (2003).

221 Gibson, J. K., Yue, Y., Bronson, J., Palmer, C. & Numann, R. Human stem cell-

derived cardiomyocytes detect drug-mediated changes in action potentials

and ion currents. Journal of pharmacological and toxicological methods 70,

255-267, doi:10.1016/j.vascn.2014.09.005 (2014).

222 Hewlett, A. W. Some Clinical Aspects of Auricular Fibrillation: A Critical

Review. California and western medicine 22, 479-483 (1924).

223 Janse, M. J. & Rosen, M. R. History of arrhythmias. Handbook of experimental

pharmacology, 1-39 (2006).

224 Veerman, C. C. et al. Immaturity of Human Stem-Cell-Derived Cardiomyocytes

in Culture: Fatal Flaw or Soluble Problem? Stem cells and development,

doi:10.1089/scd.2014.0533 (2015).

225 Magyar, J. et al. Effects of endothelin-1 on calcium and potassium currents in

undiseased human ventricular myocytes. Pflugers Archiv : European journal

of physiology 441, 144-149 (2000).

226 Devalla, H. D. et al. Atrial-like cardiomyocytes from human pluripotent stem

cells are a robust preclinical model for assessing atrial-selective

pharmacology. EMBO molecular medicine 7, 394-410,

doi:10.15252/emmm.201404757 (2015).

227 Spach, M. S., Dolber, P. C. & Anderson, P. A. Multiple regional differences in

cellular properties that regulate repolarization and contraction in the right

atrium of adult and newborn dogs. Circulation research 65, 1594-1611

(1989).

228 Nygren, A., Lomax, A. E. & Giles, W. R. Heterogeneity of action potential

durations in isolated mouse left and right atria recorded using voltage-

Page 126: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

116

sensitive dye mapping. American journal of physiology. Heart and circulatory

physiology 287, H2634-2643, doi:10.1152/ajpheart.00380.2004 (2004).

229 Feng, J., Yue, L., Wang, Z. & Nattel, S. Ionic mechanisms of regional action

potential heterogeneity in the canine right atrium. Circulation research 83,

541-551 (1998).

230 Li, D., Zhang, L., Kneller, J. & Nattel, S. Potential ionic mechanism for

repolarization differences between canine right and left atrium. Circulation

research 88, 1168-1175 (2001).

231 Ridler, M. E., Lee, M., McQueen, D., Peskin, C. & Vigmond, E. Arrhythmogenic

consequences of action potential duration gradients in the atria. The

Canadian journal of cardiology 27, 112-119, doi:10.1016/j.cjca.2010.12.002

(2011).

232 Jalife, J., Berenfeld, O. & Mansour, M. Mother rotors and fibrillatory

conduction: a mechanism of atrial fibrillation. Cardiovascular research 54,

204-216 (2002).

233 Jansen, J. A., van Veen, T. A., de Bakker, J. M. & van Rijen, H. V. Cardiac

connexins and impulse propagation. Journal of molecular and cellular

cardiology 48, 76-82, doi:10.1016/j.yjmcc.2009.08.018 (2010).

234 Vreeker, A. et al. Assembly of the cardiac intercalated disk during pre- and

postnatal development of the human heart. PloS one 9, e94722,

doi:10.1371/journal.pone.0094722 (2014).

235 Kadota, S. et al. Development of a reentrant arrhythmia model in human

pluripotent stem cell-derived cardiac cell sheets. European heart journal 34,

1147-1156, doi:10.1093/eurheartj/ehs418 (2013).

236 Wijffels, M. C., Dorland, R., Mast, F. & Allessie, M. A. Widening of the excitable

gap during pharmacological cardioversion of atrial fibrillation in the goat:

effects of cibenzoline, hydroquinidine, flecainide, and d-sotalol. Circulation

102, 260-267 (2000).

237 Hiraoka, M., Sunami, A., Fan, Z. & Sawanobori, T. Multiple ionic mechanisms

of early afterdepolarizations in isolated ventricular myocytes from guinea-

pig hearts. Annals of the New York Academy of Sciences 644, 33-47 (1992).

238 Yamamoto, W. et al. Effects of the selective KACh channel blocker NTC-801

on atrial fibrillation in a canine model of atrial tachypacing: comparison with

class Ic and III drugs. Journal of cardiovascular pharmacology 63, 421-427,

doi:10.1097/FJC.0000000000000065 (2014).

239 Herrera, D. et al. A single residue in the S6 transmembrane domain governs

the differential flecainide sensitivity of voltage-gated potassium channels.

Molecular pharmacology 68, 305-316, doi:10.1124/mol.104.009506 (2005).

240 Ramos, E. & O'Leary M, E. State-dependent trapping of flecainide in the

cardiac sodium channel. The Journal of physiology 560, 37-49,

doi:10.1113/jphysiol.2004.065003 (2004).

241 Nattel, S., Burstein, B. & Dobrev, D. Atrial remodeling and atrial fibrillation:

mechanisms and implications. Circulation. Arrhythmia and electrophysiology

1, 62-73, doi:10.1161/CIRCEP.107.754564 (2008).

Page 127: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

117

242 Fujiki, A. & Inoue, H. Pharmacological cardioversion of long-lasting atrial

fibrillation. Circulation journal : official journal of the Japanese Circulation

Society 71 Suppl A, A69-74 (2007).

243 Yanagi, K. et al. Hyperpolarization-activated cyclic nucleotide-gated channels

and T-type calcium channels confer automaticity of embryonic stem cell-

derived cardiomyocytes. Stem cells 25, 2712-2719,

doi:10.1634/stemcells.2006-0388 (2007).

244 Ivashchenko, C. Y. et al. Human-induced pluripotent stem cell-derived

cardiomyocytes exhibit temporal changes in phenotype. American journal of

physiology. Heart and circulatory physiology 305, H913-922,

doi:10.1152/ajpheart.00819.2012 (2013).

245 Conde, D. & Baranchuk, A. Vernakalant for the conversion of atrial

fibrillation: the new kid on the block? Annals of noninvasive electrocardiology

: the official journal of the International Society for Holter and Noninvasive

Electrocardiology, Inc 19, 299-302, doi:10.1111/anec.12164 (2014).

246 Guerra, F., Matassini, M. V., Scappini, L., Urbinati, A. & Capucci, A. Intravenous

vernakalant for the rapid conversion of recent onset atrial fibrillation:

systematic review and meta-analysis. Expert review of cardiovascular therapy

12, 1067-1075, doi:10.1586/14779072.2014.943662 (2014).

247 Savelieva, I., Graydon, R. & Camm, A. J. Pharmacological cardioversion of

atrial fibrillation with vernakalant: evidence in support of the ESC Guidelines.

Europace : European pacing, arrhythmias, and cardiac electrophysiology :

journal of the working groups on cardiac pacing, arrhythmias, and cardiac

cellular electrophysiology of the European Society of Cardiology 16, 162-173,

doi:10.1093/europace/eut274 (2014).

248 Wilhelms, M. et al. Benchmarking electrophysiological models of human

atrial myocytes. Frontiers in physiology 3, 487,

doi:10.3389/fphys.2012.00487 (2012).

249 Leyton-Mange, J. S. et al. Rapid cellular phenotyping of human pluripotent

stem cell-derived cardiomyocytes using a genetically encoded fluorescent

voltage sensor. Stem cell reports 2, 163-170,

doi:10.1016/j.stemcr.2014.01.003 (2014).

250 Cao, G. et al. Genetically targeted optical electrophysiology in intact neural

circuits. Cell 154, 904-913, doi:10.1016/j.cell.2013.07.027 (2013).

251 Haley, C. L. et al. Validation of a novel algorithm for quantification of the

percentage of signal fractionation in atrial fibrillation. Europace : European

pacing, arrhythmias, and cardiac electrophysiology : journal of the working

groups on cardiac pacing, arrhythmias, and cardiac cellular electrophysiology

of the European Society of Cardiology 15, 447-452,

doi:10.1093/europace/eus361 (2013).

252 Lau, D. H. et al. Stability of complex fractionated atrial electrograms: a

systematic review. Journal of cardiovascular electrophysiology 23, 980-987,

doi:10.1111/j.1540-8167.2012.02335.x (2012).

253 Berenfeld, O. & Jalife, J. Complex fractionated atrial electrograms: is this the

beast to tame in atrial fibrillation? Circulation. Arrhythmia and

electrophysiology 4, 426-428, doi:10.1161/CIRCEP.111.964841 (2011).

Page 128: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

118

254 Van Gelder, I. C. et al. Chronic atrial fibrillation. Success of serial

cardioversion therapy and safety of oral anticoagulation. Archives of internal

medicine 156, 2585-2592 (1996).

255 Corradi, D. Atrial fibrillation from the pathologist's perspective.

Cardiovascular pathology : the official journal of the Society for Cardiovascular

Pathology 23, 71-84, doi:10.1016/j.carpath.2013.12.001 (2014).

256 Pinho-Gomes, A. C., Reilly, S., Brandes, R. P. & Casadei, B. Targeting

inflammation and oxidative stress in atrial fibrillation: role of 3-hydroxy-3-

methylglutaryl-coenzyme a reductase inhibition with statins. Antioxidants &

redox signaling 20, 1268-1285, doi:10.1089/ars.2013.5542 (2014).

257 Jalife, J. & Kaur, K. Atrial remodeling, fibrosis, and atrial fibrillation. Trends in

cardiovascular medicine, doi:10.1016/j.tcm.2014.12.015 (2014).

258 Atienza, F., Martins, R. P. & Jalife, J. Translational research in atrial

fibrillation: a quest for mechanistically based diagnosis and therapy.

Circulation. Arrhythmia and electrophysiology 5, 1207-1215,

doi:10.1161/CIRCEP.111.970335 (2012).

259 Berenfeld, O. & Jalife, J. Mechanisms of atrial fibrillation: rotors, ionic

determinants, and excitation frequency. Cardiology clinics 32, 495-506,

doi:10.1016/j.ccl.2014.07.001 (2014).

260 Baher, A. et al. Short-term cardiac memory and mother rotor fibrillation.

American journal of physiology. Heart and circulatory physiology 292, H180-

189, doi:10.1152/ajpheart.00944.2005 (2007).

261 Grandi, E. et al. Human atrial action potential and Ca2+ model: sinus rhythm

and chronic atrial fibrillation. Circulation research 109, 1055-1066,

doi:10.1161/CIRCRESAHA.111.253955 (2011).

262 Qi, X. Y. et al. Fibroblast inward-rectifier potassium current upregulation in

profibrillatory atrial remodeling. Circulation research 116, 836-845,

doi:10.1161/CIRCRESAHA.116.305326 (2015).

263 Chen, P. S., Chen, L. S., Fishbein, M. C., Lin, S. F. & Nattel, S. Role of the

autonomic nervous system in atrial fibrillation: pathophysiology and therapy.

Circulation research 114, 1500-1515, doi:10.1161/CIRCRESAHA.114.303772

(2014).

264 Voigt, N., Makary, S., Nattel, S. & Dobrev, D. Voltage-clamp-based methods for

the detection of constitutively active acetylcholine-gated I(K,ACh) channels in

the diseased heart. Methods in enzymology 484, 653-675, doi:10.1016/B978-

0-12-381298-8.00032-0 (2010).

265 Shiroshita-Takeshita, A., Brundel, B. J., Lavoie, J. & Nattel, S. Prednisone

prevents atrial fibrillation promotion by atrial tachycardia remodeling in

dogs. Cardiovascular research 69, 865-875 (2006).

266 Cagli, K. E. et al. Plasma levels of tumor necrosis factor-alpha and its

receptors in patients with mitral stenosis and sinus rhythm undergoing

percutaneous balloon valvuloplasty. Heart Vessels 25, 131-137,

doi:10.1007/s00380-009-1175-9 (2010).

267 Li, J. et al. Role of inflammation and oxidative stress in atrial fibrillation.

Heart rhythm : the official journal of the Heart Rhythm Society 7, 438-444,

doi:S1547-5271(09)01380-0 [pii]

Page 129: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

119

10.1016/j.hrthm.2009.12.009 (2010).

268 Qi, J. et al. Effects of potassium channel blockers on changes in refractoriness

of atrial cardiomyocytes induced by stretch. Experimental biology and

medicine 234, 779-784, doi:10.3181/0902-RM-45 (2009).

269 Kalifa, J. et al. Anti-arrhythmic effects of I (Na), I (Kr), and combined I (Kr)-I

(CaL) blockade in an experimental model of acute stretch-related atrial

fibrillation. Cardiovascular drugs and therapy / sponsored by the International

Society of Cardiovascular Pharmacotherapy 21, 47-53, doi:10.1007/s10557-

007-6001-y (2007).

270 Dobrev, D., Carlsson, L. & Nattel, S. Novel molecular targets for atrial

fibrillation therapy. Nature reviews. Drug discovery 11, 275-291,

doi:10.1038/nrd3682 (2012).

271 Ueda, N., Yamamoto, M., Honjo, H., Kodama, I. & Kamiya, K. The role of gap

junctions in stretch-induced atrial fibrillation. Cardiovascular research 104,

364-370, doi:10.1093/cvr/cvu202 (2014).

272 Boldt, A. et al. Fibrosis in left atrial tissue of patients with atrial fibrillation

with and without underlying mitral valve disease. Heart 90, 400-405 (2004).

273 Frustaci, A. et al. Histological substrate of atrial biopsies in patients with lone

atrial fibrillation. Circulation 96, 1180-1184 (1997).

274 Kostin, S. et al. Structural correlate of atrial fibrillation in human patients.

Cardiovascular research 54, 361-379 (2002).

275 Woods, C. E. & Olgin, J. Atrial fibrillation therapy now and in the future: drugs,

biologicals, and ablation. Circulation research 114, 1532-1546,

doi:10.1161/CIRCRESAHA.114.302362 (2014).

276 Witty, A. D. et al. Generation of the epicardial lineage from human pluripotent

stem cells. Nature biotechnology 32, 1026-1035, doi:10.1038/nbt.3002

(2014).

277 Thavandiran, N. et al. Design and formulation of functional pluripotent stem

cell-derived cardiac microtissues. Proceedings of the National Academy of

Sciences of the United States of America 110, E4698-4707,

doi:10.1073/pnas.1311120110 (2013).

278 Qi, X. Y. et al. Fibroblast Inward-Rectifier Potassium Current Upregulation in

Profibrillatory Atrial Remodeling. Circulation research,

doi:10.1161/CIRCRESAHA.116.305326 (2015).

279 Suarez, G. & Meyerrose, G. Heart failure and galectin 3. Annals of translational

medicine 2, 86, doi:10.3978/j.issn.2305-5839.2014.09.10 (2014).

280 Pellman, J., Lyon, R. C. & Sheikh, F. Extracellular matrix remodeling in atrial

fibrosis: mechanisms and implications in atrial fibrillation. Journal of

molecular and cellular cardiology 48, 461-467,

doi:10.1016/j.yjmcc.2009.09.001 (2010).

281 Lin, C. S. & Pan, C. H. Regulatory mechanisms of atrial fibrotic remodeling in

atrial fibrillation. Cell Mol Life Sci 65, 1489-1508 (2008).

282 de Bakker, J. M. et al. Slow conduction in the infarcted human heart. 'Zigzag'

course of activation. Circulation 88, 915-926 (1993).

283 Hanna, N., Cardin, S., Leung, T. K. & Nattel, S. Differences in atrial versus

ventricular remodeling in dogs with ventricular tachypacing-induced

Page 130: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

120

congestive heart failure. Cardiovascular research 63, 236-244,

doi:10.1016/j.cardiores.2004.03.026 (2004).

284 Nakajima, H. et al. Atrial but not ventricular fibrosis in mice expressing a

mutant transforming growth factor-beta(1) transgene in the heart.

Circulation research 86, 571-579 (2000).

285 Tucker, N. R. & Ellinor, P. T. Emerging directions in the genetics of atrial

fibrillation. Circulation research 114, 1469-1482,

doi:10.1161/CIRCRESAHA.114.302225 (2014).

286 Gonzalez, F. et al. An iCRISPR platform for rapid, multiplexable, and inducible

genome editing in human pluripotent stem cells. Cell stem cell 15, 215-226,

doi:10.1016/j.stem.2014.05.018 (2014).

287 Yang, Y. Q. et al. Prevalence and spectrum of GATA6 mutations associated

with familial atrial fibrillation. International journal of cardiology 155, 494-

496, doi:10.1016/j.ijcard.2011.12.091 (2012).

288 Zhao, R. et al. Loss of both GATA4 and GATA6 blocks cardiac myocyte

differentiation and results in acardia in mice. Developmental biology 317,

614-619, doi:10.1016/j.ydbio.2008.03.013 (2008).

289 Remond, M. C. et al. GATA6 reporter gene reveals myocardial phenotypic

heterogeneity that is related to variations in gap junction coupling. American

journal of physiology. Heart and circulatory physiology 301, H1952-1964,

doi:10.1152/ajpheart.00635.2011 (2011).

290 Arnolds, D. E. et al. TBX5 drives Scn5a expression to regulate cardiac

conduction system function. The Journal of clinical investigation 122, 2509-

2518, doi:10.1172/JCI62617 (2012).

291 Postma, A. V. et al. A gain-of-function TBX5 mutation is associated with

atypical Holt-Oram syndrome and paroxysmal atrial fibrillation. Circulation

research 102, 1433-1442, doi:10.1161/CIRCRESAHA.107.168294 (2008).

292 Zang, X. et al. SNP rs3825214 in TBX5 is associated with lone atrial

fibrillation in Chinese Han population. PloS one 8, e64966,

doi:10.1371/journal.pone.0064966 (2013).

293 Sinner, M. F. et al. Integrating genetic, transcriptional, and functional analyses

to identify 5 novel genes for atrial fibrillation. Circulation 130, 1225-1235,

doi:10.1161/CIRCULATIONAHA.114.009892 (2014).

294 Sartiani, L. et al. Developmental changes in cardiomyocytes differentiated

from human embryonic stem cells: a molecular and electrophysiological

approach. Stem cells 25, 1136-1144, doi:10.1634/stemcells.2006-0466

(2007).

295 Nunes, S. S. et al. Biowire: a platform for maturation of human pluripotent

stem cell-derived cardiomyocytes. Nature methods 10, 781-787,

doi:10.1038/nmeth.2524 (2013).

296 Zhang, M. et al. Universal Cardiac Induction of Human Pluripotent Stem Cells

in 2D and 3D formats - Implications for In-Vitro Maturation. Stem cells,

doi:10.1002/stem.1964 (2015).

297 Yang, X. et al. Tri-iodo-l-thyronine promotes the maturation of human

cardiomyocytes-derived from induced pluripotent stem cells. Journal of

Page 131: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

121

molecular and cellular cardiology 72, 296-304,

doi:10.1016/j.yjmcc.2014.04.005 (2014).

Page 132: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

122

Appendix

Appendix Figure 1: Intracellular recordings of a “ventricular” cell sheet performed with a

high resistance (~30 MΩ) thin walled glass micropipette. Sharp spikes represent pacing

artifact. The cell sheet was paced from the edge of one side of the cell sheet, and a cell

on the opposite side was impaled for this recording. The conduction velocity was

calculated to be ~4 cm/s for this recording. The resting membrane potential, upstroke

velocity and action potential amplitude mimic those recorded from single cell patch

clamping studies. In particular, the resting membrane potential sits at ~-55 mV,

essentially the same as seen in single cell studies.

Page 133: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

123

Appendix Figure 2: Activation maps generated from recordings of an “atrial” cell sheet

and the sequential uptitration of flecainide dosing using DI-4ANEPPS and an EMCCD.

Activation maps demonstrate the dose dependent effects of flecainide on the cycle length

and the number of excitation wavelets in the cell sheet. Initially a stable rotor is induced

on the left. After the addition of 5 μM flecainide, the cycle length is substantially

reduced and the curvature of the rotor is decreased without affecting the location of the

phase singularity (PS)(labeled as a white star on figure). After the addition of 10 μM

flecainide, two distinct rotors appear, each rotating around their own PS independently

and causing wave collision along sites of wavefront interaction. Wave collision did not

generate more wavelets in this cell sheet, and instead the activation wavefront

extinguished at the site of collision.

Page 134: MODELING ATRIAL FIBRILLATION USING HUMAN EMBRYONIC … › bitstream › 1807 › 94524 › 1 › Laks… · 1.7.1 Physiology of rotors 14 1.7.2 Clinical relevance of rotors in AF

124

Appendix Figure 3: Optical Action Potentials (OAPs) recorded using the Arclight hiPSC cell line differentiated into cardiomyocytes and recorded on an EMCCD. Cells contain a genetically encoded voltage sensor attached to eGFP which reports a change in membrane voltage as changes in fluorescence. Note the 20% change in fluorescent signal intensity (Y axis) demarcating depolarization of the hiPSC derived cardiomyocytes. Note also the absence of photobleaching over 20 seconds of recording time, overcoming a major limitation of the voltage sensitive dyes. This was repeated on many occasions on the same day and on sequential days for up to 1 week with preservation of signal.