• No results found

Electrophysiological patterning of the heart - Chapter 3: Complementary roles of Tbx3 and Nkx2-5 in the structure and function of the atrioventricular conduction system

N/A
N/A
Protected

Academic year: 2021

Share "Electrophysiological patterning of the heart - Chapter 3: Complementary roles of Tbx3 and Nkx2-5 in the structure and function of the atrioventricular conduction system"

Copied!
17
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

UvA-DARE is a service provided by the library of the University of Amsterdam (https://dare.uva.nl)

Electrophysiological patterning of the heart

Boukens, B.J.D.

Publication date 2012

Link to publication

Citation for published version (APA):

Boukens, B. J. D. (2012). Electrophysiological patterning of the heart.

General rights

It is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), other than for strictly personal, individual use, unless the work is under an open content license (like Creative Commons).

Disclaimer/Complaints regulations

If you believe that digital publication of certain material infringes any of your rights or (privacy) interests, please let the Library know, stating your reasons. In case of a legitimate complaint, the Library will make the material inaccessible and/or remove it from the website. Please Ask the Library: https://uba.uva.nl/en/contact, or a letter to: Library of the University of Amsterdam, Secretariat, Singel 425, 1012 WP Amsterdam, The Netherlands. You will be contacted as soon as possible.

(2)

Bas Boukens

Mathilda Mommersteeg

Saskia van de Velden

Corrie de Gier-de Vries

Richard Harvey

Antoon Moorman

Ruben Coronel

Vincent Christoffels

Complementary roles of Tbx3 and Nkx2-5 in

the structure and function of the adult

atrioventricular conduction system

(3)

3

Abstract

Rationale: Dysfunction of the atrioventricular (AV) node or AV bundle can cause AV block,

which requires implantation of an electronic pacemaker. Genome-wide association studies have revealed that genetic variation in multiple cardiac transcription factor loci, including NKX2-5 and

TBX3, influence the function of the conduction system. Objective: In this study we investigated

the role of Tbx3 and Nkx2-5 in the formation and the functional maintenance of the AV conduction system. Methods and results: We studied 4 groups of mice: wildtype, haploinsufficient for

Tbx3, for Nkx2-5 or for both Tbx3 and Nkx2-5. Their age was 2 months or 16 months. In these

groups, AV conduction parameters were measured, and the structure and size of the AV node and bundle were assessed using 3D reconstruction of gene expression patterns. In addition, we determined the expression pattern of proteins relevant for conduction. At 2 month of age, the AV bundle of Tbx3, Nkx2-5 and Tbx3/Nkx2-5 haploinsufficient mice was significantly smaller than that of wild type mice, whereas the size of the AV node was not different. AV delay was prolonged only in the Tbx3 haploinsufficient mutant mice. At 16 months of age, the AV bundle of Tbx3, Nkx2-5 and Tbx3/Nkx2-5 haploinsufficient mice was smaller than that of wildtype mice. The AV node size was not different between the groups but the AV delay was shorter in both

Tbx3 and Tbx3/Nkx2-5 haploinsufficient mice than in wildtype and Nkx2-5 haploinsufficient

mice. Furthermore, both Nkx2-5 and Tbx3/Nkx2-5 haploinsufficient mice developed AV block.

Conclusions: Normal levels of both Tbx3 and Nkx2-5 are required for AV bundle formation. The

(4)

3

Introduction

The cardiac conduction system enables rhythmic and coordinated activation of the heart. The development of the conduction system is guided by temporal and spatial expression of transcription factors.1 Genome-wide association studies show that variations (single

nucleotide polymorphisms; SNPs) in multiple loci containing cardiac transcription factors, including NKX2-5 and TBX3, relate to the function of the conduction system in the adult human population.2-4 These variations are likely to influence the expression level or function

of these factors, thereby modifying their action.5 On the other hand, heterozygous mutations

causing loss of function of transcription factors including NKX2-5 and TBX5 can lead to severe conduction system abnormalities like 2nd or 3rd degree AV block.6-8 Thus, the development of

the conduction system is regulated by cardiac transcription factors in a dose-dependent manner. Tbx3 is essential for conduction system development and function.9, 10 Heterozygous

mutations in TBX3 cause ulnar-mammary syndrome of congenital defects in humans. However, the consequences of heterozygous loss of function for conduction system development are unknown.11 Tbx3 has been shown to physically and functionally interact

with Nkx2-5 to regulate target gene expression in the developing cardiac conduction system.12 Heterozygous loss of Nkx2-5 results in a hypoplastic AV conduction system and AV

conduction defects in mice13 and mutations in NKX2-5 are found in patients with AV conduction

disturbances.6, 7 Despite these insights, little is known about the regulation of conduction

system homeostasis, and the roles of these factors during aging have not been explored. The AV node forms from the slowly conducting primary myocardium of the AV canal and the AV bundle forms from the initially slowly but eventually fast-conducting myocardium of the crest of the interventricular septum.14 In the AV node and AV bundle, transcription factors

including Tbx2 and Tbx3 repress the working myocardial gene program,15 whereas factors like

Tbx5 activate the gene program for fast conduction in the AV bundle.6, 16, 17 Nkx2-5 plays an

important role in cardiac and conduction system gene regulation, and synergizes with both repressing and activating T-box transcription factors.12, 17 We hypothesize that a disturbed

balance in levels of these transcription factors affect the gene programs and function of the AV node and AV bundle. We addressed this hypothesis by investigating the structure and the function of the AV conduction system in young and old mice that were haploinsufficient for

Tbx3, for 5 or for both genes. Our data indicate that normal levels of Tbx3 and of

Nkx2-5 are required for the AV bundle formation and function in young and old mice, but that both factors to a large extent have distinguishable functions and act in non-redundant manners.

(5)

3

Material and Methods

Animals

The investigation conforms with the local Animal Experiments Committee of the Academic Medical Center and the Guide for the Care and Use of Laboratory Animals published by the US National Institutes of Health (NIH Publication No. 85-23, revised 1996) or the European Commission Directive 2010/63/EU. Nkx2-5-GFP and Tbx3-cre mouse lines have been described previously.18, 19 All mice had a FVB/N background.

Immunohistochemistry and in situ hybridization

In situ hybridization was performed according to a previously described method.20 Probes have

been described previously.21, 22 For immunohistochemistry, rehydration, unmasking, blocking

and washing steps were performed according to the protocol of the tetramethylrhodamide based amplification kit (Perkin Elmer). Primary antibodies used for mouse sections were: cTnI rabbit polyclonal (1:250; Hytest Ltd); Tbx3 goat polyclonal (1:500; Santa Cruz Biotechnology); Cx43 and Cx40 mouse monoclonal (1:250; US Biological); Hcn4 Goat polyclonal (1:250 Santa Cruz Biotechnology). Secondary antibodies when using amplification were: Biotinylated donkey-anti-goat (1:250; Jackson Immunology); biotinylated goat-anti-rabbit (1:250; DAKO); biotinylated goat-anti-mouse (1:250; DAKO). For visualization without the amplification step, secondary antibodies coupled to an Alexa fluorescent (1:250; Invitrogen) were used.

3D reconstruction

In short, 10 µm serial sections of the AV junction and of ventricular septum were stained for Hcn4 and Cx40 and reconstructed. Subsequently, 3D reconstruction were performed as described previously using Amira 5.2 software.23 We defined the AV node

and the AV bundle based on the expression of hyperpolarization-activated channel (Hcn4), which encodes the subunit that form the channels carrying the If current, and connexin (Cx40), which forms gap junctions with high conductance. Cx40 is expressed in the AV bundle but not in the AV node whereas Hcn4 is expressed in both structures.14

ECG

Animals were anaesthetized with 1.5% isoflurane. Electrodes were placed at the right (R) and left (L) armpit and the left groin (F). A reference electrode was placed at the right groin. ECGs were recorded (Biosemi, Amsterdam, The Netherlands, sampling rate 2,048 Hz, filtering DC 400 kHz [3dB]) for a period of 5 minutes. From these leads a standard six lead ECG was calculated as follows: I = L-R, II = F-R, III = F-L, aVR = R-(L+F)/2, aVL = L-(R+F)/2 and aVF = F-(L+R)/2.24

(6)

3

Langendorff experiments

Mice were stunned by inhalation of CO and killed by cervical dislocation. Then the heart was excised, the aorta was cannulated, mounted on a Langendorff perfusion set-up, and perfused at 37°C with Tyrode’s solution ((in mmol/L) 128 NaCl, 4.7 KCl, 1.45 CaCl2, 0.6 MgCl2, 27 NaHCO3, 0.4 NaH2PO4, and 11 glucose (pH maintained at 7.4 by equilibration with a mixture of 95% O2 and 5% CO2)). Local electrograms were acquired using a custom-built 256 channel data acquisition (Biosemi) system and analyzed using custom made software. Local electrograms were recorded from the right atrium and left ventricle during sinus rhythm and right atrial pacing at a basic cycle length of 120 ms, and twice the diastolic stimulus. The AV conduction delay was defined as the time between the start of the stimulus and the start of the first deflection on the ventricular electrogram. The refractory period of the AV node was determined by reducing the coupling interval of a premature stimulus on the right atrium (after 10 stimuli at basic cycle length 120 ms) in steps of 5 ms until activation of the ventricle failed. His potentials were measured during sinus rhythm and premature stimulation on the right atrium with a unipolar electrode that was placed on the His bundle after a small incision in the right atrium.

Statistics

Group comparisons were performed using one-way ANOVA with Bonferroni test for post hoc matched pairs. Values are given as mean ± SEM. A p-value < 0.05 was considered statistically significant.

Results

Normally sized AV node but hypoplastic AV bundle in mice haploinsufficient for Tbx3 or Nkx2-5

To study the requirement for normal levels of Tbx3 and of Nkx2-5 in the formation of the conduction system, we determined the size of the AV node and AV bundle in mice haploinsufficient for Tbx3 and/or Nkx2-5 at 2 months of age. Figure 1A shows the expression pattern of Hcn4, Cx43 and Cx40 in the AV junction of a wildtype mouse. We used Hcn4 as a marker for all AV conduction system components and Cx40 to discriminate between the AV

(7)

3

node (Cx40-negative) and the AV bundle (Cx40-positive) (Figure 1B). In order to compare the size of the AV node, bundle and bundle branches between mice haploinsufficient for Tbx3 and/or Nkx2-5 we reconstructed the expression patterns of Hcn4 and Cx40 in the AV junction and the ventricular septum. The AV node was not different in size, which contrasts previously published data in which Nkx2-5 haploinsufficiency been associated with AV node hypoplasia.13, 25 However, the AV bundle and the bundle branches were hypoplastic in mice haploinsufficient

for Tbx3 and/or Nkx2-5 (Figure 1D, n=5 for each group). The size of the AV bundle and bundle branches in mice haploinsufficient for both Tbx3 and Nkx2-5 was not different from those in mice haploinsufficient for either Tbx3 or Nkx2-5. The latter indicates that reduction of both Tbx3 and Nkx2-5 does not have an additive effect on the size of the conduction system.

Figure 1. The AV bundle and bundle branches are hypoplastic in mice haploinsufficient for Tbx3 and/

or Nkx2-5. Panel A shows Immunohistochemistry staining for Cx43, Cx40 and Hcn4 in the AV junction of a wildtype mouse. Panel B shows the transition of the AV node in the AV bundle. Not that the lower nodal cells are part of the AV bundle. Panel C shows the reconstructed expression patter of Cx40 and Hcn4 in the AV junction and the ventricular septum in wildtype mice and mice haploinsufficient for Tbx3 and/ or Nkx2-5. The bar graph in panel D shows the average size of the AV node, bundle and bundle branches. AS, atrial septum; AVN, atrioventricular node; AVB, atrioventricular bundle; VS, ventricular septum.

(8)

3

Haploinsufficiency for Tbx3 results in AV delay prolongation by an Nkx2-5-dependent mechanism

To determine the consequence of AV bundle hypoplasia observed in Tbx3, Nkx2-5 and compound heterozygotes for AV conduction, we recorded electrocardiograms. The RR, PR and QRS intervals were not different between groups (Table 1, n=6 for each group). Furthermore, the morphology of the QRS complex was not different between groups indicating that ventricular activation was not affected.

Next, we studied the properties of the AV conduction system in a Langendorff set-up to allow programmed stimulation. The refractory period of the AV node (at a basic cycle length of 120 ms) was not different across groups (Table 2). However, at each pacing interval the AV delay was larger in the Tbx3 heterozygotes than in mice from the other groups (Figure 2A). To investigate whether the increase in AV delay was the result of slower conduction in the AV node or in the ventricular conduction system, we subsequently measured HV intervals.

Figure 2. AV conduction is delayed in mice haploinsufficient for Tbx3. Panel A shows local eletrogram recordings

(9)

3

haploinsufficient for both Tbx3 and Nkx2-5, AV delay was not prolonged. This suggests that the effect of loss of one allele of Tbx3 on AV delay is attenuated by removing one allele of Nkx2-5.

Figure 3. Haploinsufficiency for Tbx3 prevents age-mediated prolongation of atrioventricular delay. The

graph in panel A shows the average conduction delay of wildtype mice and mice haploinsufficient for Tbx3 and/or Nkx2-5 at 2 and 16 months of age. Panel B shows In-situ hybridizations for Cx40 in the His bundle. The bar graph in panel C shows the average size of the atrioventricular node and ventricular conduction system in wildtype mice and mice haploinsufficient for Tbx3 and/or Nkx2-5. Panel D shows the relation between the atrioventricular delay and the size of the connection between the atrioventricular node and His bundle. AS, atrial septum; AVN, atrioventricular node; AVB, atrioventricular bundle; VS, ventricular septum

(10)

3

Tbx3 haploisufficiency prevents age-mediated AV delay prolongation

Heterozygous loss of Nkx2-5 can lead to AV delay prolongation and eventually AV block in aging mice.13 We wanted to investigate the role of Tbx3 in age-mediated AV delay prolongation

and AV block in mice. Therefore, we recorded ECGs and measured the properties of the AV node in a Langendorff set up. At 9 months we recorded ECGs to check whether AV delay had changed compared to 2 months. Although the PR interval increased somewhat compared to 2 months in all groups (compare Tables 1 and 3), the RR, QRS, PR intervals were not different

across groups at 9 months (Table 3). At 16 months, however, in explanted hearts of wildtype mice, the AV delay was significantly larger than at 2 months of age (44.8±2.3 vs 33.5±0.6, n=8 and n=6, respectively, p<0.05, Figure 3A). The refractory period of the AV node was not different between ages (Table 2). In Nkx2-5 heterozygotes, the AV delay was also larger at 16 months than at 2 months of age (40.5±2.4 vs 33.3±1.0, n=4 and n=10, respectively) but the AV delay was not different from wildtype mice at both ages. In contrast, in Tbx3 heterozygotes the AV delay was not different at 16 months of age compared to 2 months of

(11)

3

A small AV node-His connection in Tbx3 heterozygotes correlates with a short AV delay

Next, we reconstructed the AV node and the ventricular conduction system based on the expression pattern of Hcn4 and Cx40. In Figure 3B representative sections of the AV bundle of the four groups of mice are shown. Similar to the situation at 2 months of age, a hypoplastic AV bundle was observed also at 16 months of age in mice haploinsufficient for Tbx3 and/or Nkx2-5. Furthermore, the AV node was not different in size in the transgenic mice compared to in the wildtype mice, resembling what we found in the younger mice. To understand why the AV delay in aged mice was not different from young mice that missed one allele of Tbx3 we correlated the diameter of the AV bundle and the AV nodal -AV bundle transition with the AV delay within the same mouse. The diameter of the AV bundle did not correlate with the AV delay (r=0.43, p=0.25). However, the size of the transition between the AV node and the AV bundle (averaged ranking from small to large by two independent observers), indicated by the contact surface of the Cx40+/Hcn4+

myocardium with the Cx40-/Hcn4+ myocardium, significantly correlated with the AV delay

(Figure 3D). The latter suggests that both Tbx3 and Nkx2-5 haploinsufficiency affects the AV delay by affecting the structural transition between the AV node and the AV bundle.

A Tbx3-independent mechanism underlies AV conduction block in aging Nkx2-5 heterozygous mice

At 16 months, we observed 2nd or 3rd degree AV block in 3 of 8 explanted hearts (38%) from

Nkx2-5 heterozygous mice, and 3rd degree AV block in 1 of 4 hearts from mice that were

haploinsufficient for both Nkx2-5 and Tbx3. The hearts that did not have 2nd or 3rd degree AV

block had normal AV conduction (Figure 4). In hearts of wildtype (n=6) or Tbx3 haploinsufficient (n=4) mice we did not observe AV block. The percentage of hearts with AV block was not significantly different between groups. However, if we selected by haploinsufficiency for Nkx2-5, significantly more AV block occurred when compared to hearts of mice with two Nkx2-5 alleles.

Discussion

Our results show that haploinsufficiency of Tbx3, Nkx2-5 or both causes hypoplasia of the AV bundle and bundle branches by, most likely, two independent mechanisms that are, however, not additive in effect. Furthermore, in young animals, Tbx3-haploinsufficiency lead to prolonged AV delay, which can be attenuated by Nkx2-5-haploinsufficiency, whereas in old animals Tbx3-haploinsufficiency lead to a shorting of AV delay independent of

Nkx2-5 haploinsufficiency. Thus, although Tbx3 and Nkx2-Nkx2-5 interact with each other in young

mice, they seem to act in parallel in old mice. This suggests that, as expected, Tbx3 and Nkx2-5 are part of a more complex transcriptional network that regulates the formation and function of the AV conduction system. We also describe that small differences in the transition between AV node and AV bundle, independent of genotype, appear to influence

(12)

3

AV conduction. It is unclear how the transcriptional balance regulates the shaping of this structural transition, and whether other factors including like Irx326 and Tbx517, are involved.

Prolonged AV delay in Tbx3 haploinsufficient mice

The structure of the AV conduction system was similarly affected in Tbx3-, Nkx2-5- and Tbx3/

Nkx2-5-haploinsufficient mice. However, the AV conduction times were different. The latter

suggest that it is not the AV conduction structure that underlies these functional differences. In Tbx3 haploinsufficient mice, conduction in the AV node was only slowed in the isolated and denervated heart but not in the innervated heart in vivo, which is in line with previously published results of our group.9 When ectopically expressed in the heart, Tbx3 directly or

indirectly induces the expression of Cav3.1 (alpha subunit of T-type Calcium channels),19

which therefore may be downregulated in Tbx3-haploinsufficient mice. As a consequence, the expression of T-type calcium channels may be reduced in Tbx3 mutants but provide sufficient inward current for normal AV delay upon a certain degree of autonomic stimulation. When autonomic stimulation is absent, like in a Langendorff set-up, the inward calcium current may not be sufficient for normal conduction and AV delay prolongs.27 The expression of the

T-type calcium channels is also downregulated in the hearts of mice haploinsufficient for Nkx2-5.25 Therefore, the mechanism by which haploinsufficiency for Nkx2-5 counteracts

the prolonged AV delay in mice that are haploinsufficient for Tbx3 remains unclear.

Molecular mechanism by which Nkx2-5 and Tbx3 affect structure and function of the AV conduction system

Our data indicate that Tbx3 affects AV delay in a mechanism that is attenuated by a reduction in Nkx2-5. It has been shown that Tbx3 and Nkx2-5 can physically interact.12 However, to

our knowledge, a direct regulation of Nkx2-5 by Tbx3, or vice versa, has never been reported. Tbx3 may indirectly reduce Nkx2-5 expression via Prospero-Related Homeobox Protein (Prox) 1, which inhibits Nkx2-5 expression and is upregulated in Tbx3 overexpression mice.19, 25

However, the latter rather suggests an additive effect of a reduction of Tbx3 and Nkx2-5 than a counteractive effect, which is what our data indicate. Therefore, Nkx2-5 and Tbx3 most likely regulate the expression of certain genes by co-occupying the same genomic regions28 similar to

how Nkx2-5 and Tbx5 synergistically regulate the expression of inhibitor of differentiation (Id) 2 in the conduction system.17 The expression of Id2, however, is not affected in mice deficient

for either Nkx2-5 or Tbx3.9, 10 Gene expression analysis will reveal whether the expression of

(13)

3

number of mice lacking one functional Nkx2-5 allele develop AV block. In contrast to our data, other studies reported that mice haploinsufficient for Nkx2-5 have a hypoplastic AV node and prolonged PR intervals already at 2-4 months of age.13, 25 An explanation for this discrepancy

may be found in differences in genetic background. We used FVB/N mice whereas other studies were performed in C57Bl/6 or 129/SvJ mice. It has been shown that the functional consequences of a mutation can differ between mouse strains.31 Another major discrepancy is the definition

of the AV node and AV bundle, respectively. In contrast to other studies, we have used Cx40 to discriminate between the AV node (negative) and AV bundle cells (positive) in the AV junction.14

We, therefore, defined the AV node based on the presence of Hcn4 and absence of Cx40 and the AV bundle and branches on the presence of both Hcn4 and Cx40. Furthermore, instead of comparing sections we reconstructed the conduction system and compared volumes, which is more accurate. These differences most likely explain the difference in size of the AV node and bundle in Nkx2-5 haploinsufficient mice observed in our study compared to in other studies.13, 25

The role of Tbx3 in prevention of age-mediated prolongation of AV delay

Our data indicate that Tbx3 is involved in age-mediated prolongation of the AV delay. In mice, and also in human, AV conduction becomes slower during ageing whereas the conduction in the ventricular conduction system remains the same.13, 32 Not much is known about the

underlying mechanism in mice, but in rat altered collagen deposition in the AV node is thought to contribute to AV delay prolongation.33 In the atria of mice that overexpress Tbx3 the

expression of Procollagen, type VI, alpha 2 (col6a2) and Procollagen, type I, alpha 1 (col1a1) are upregulated.12 We surmise that haploinsufficiency of Tbx3 results in less collagen in the AV

node during ageing and thereby affecting AV conduction. This, however, remains to be tested. Alternatively, ion channel remodeling may be involved in AV delay prolongation during

Figure 4. Aged mice that are haploinsufficient for Nkx2-5 develop atrioventricular block. Panel A

shows local eletrogram recordings from mice haploinsufficient for Nkx2-5 with normal atrioventricular delay (upper) and atriovenricular block (lower). The bar graph in panel B shows at the right the amount atrioventricular block in wildtype mice and mice deficient for Tbx3 and/or Nkx2-5 and. At the left the amount of atrioventricular block is shown in mice that Nkx2-5 haploinsufficiency or not.

(14)

3

ageing. For the sinus node it has been reported that ageing leads to sinus node dysfunction independent of structural changes, which may be the case for the AV node as well.34

However, the role of transcription factors, like Tbx3, in maintaining the nodal gene program at older age is not known. We speculate that haploinsufficiency of Tbx3 results in a reduced nodal gene program during ageing and to faster conduction in the AV node.

Conclusion

We conclude that normal levels of Tbx3 and Nkx2-5 are required for correct formation of the AV bundle. However, in the young heart, Tbx3 and Nkx2-5 regulate AV conduction via a similar mechanism whereas in the old heart they regulate AV conduction independently.

Funding

This work was supported by grants from the Netherlands Heart Foundation (2008B062 to V.M.C. and R.C.); the European Community’s Seventh Framework Programme contract (‘CardioGeNet’ 223463 to V.M.C.).

Acknowledgements

We thank Raoul Freitas Vale Germano for his assistance.

Reference List

(1) Hatcher CJ, Basson CT. Specification of the cardiac conduction system by transcription factors. Circ

Res 2009 September 25;105(7):620-30.

(2) Holm H, Gudbjartsson DF, Arnar DO, Thorleifsson G, Thorgeirsson G, Stefansdottir H, Gudjonsson SA, Jonasdottir A, Mathiesen EB, Njolstad I, Nyrnes A, Wilsgaard T, Hald EM, Hveem K, Stoltenberg C, Lochen ML, Kong A, Thorsteinsdottir U, Stefansson K. Several common variants modulate heart rate, PR interval and QRS duration. Nat Genet 2010 January 10;42:177-22.

(3) Sotoodehnia N, Isaacs A, de Bakker PI, Dorr M, Newton-Cheh C, Nolte IM, van der HP, Muller M, Eijgelsheim M, Alonso A, Hicks AA, Padmanabhan S, Hayward C, Smith AV, Polasek O, Giovannone S, Fu J, Magnani JW, Marciante KD, Pfeufer A, Gharib SA, Teumer A, Li M, Bis JC, Rivadeneira F, Aspelund T, Kottgen A, Johnson T, Rice K, Sie MP, Wang YA, Klopp N, Fuchsberger C, Wild SH, Mateo L, I, Estrada K, Volker U, Wright AF, Asselbergs FW, Qu J, Chakravarti A, Sinner MF, Kors JA, Petersmann A, Harris TB, Soliman EZ, Munroe PB, Psaty BM, Oostra BA, Cupples LA, Perz S, de Boer RA, Uitterlinden AG, Volzke H, Spector TD, Liu FY, Boerwinkle E, Dominiczak AF, Rotter JI, van HG, Levy D, Wichmann HE, van Gilst WH, Witteman JC, Kroemer HK, Kao WH, Heckbert SR, Meitinger T, Hofman A, Campbell H, Folsom AR, van Veldhuisen DJ, Schwienbacher C, O’Donnell CJ, Volpato CB, Caulfield MJ, Connell JM, Launer L, Lu X, Franke L, Fehrmann RS, te MG, Groen HJ, Weersma RK, van den Berg LH, Wijmenga C, Ophoff RA, Navis G, Rudan I, Snieder H, Wilson JF,

(15)

3

RB, Chung MK, Barnard J, Smith JD, Van Wagoner DR, Vasan RS, Aspelund T, Eiriksdottir G, Harris TB, Launer LJ, Najjar SS, Lakatta E, Schlessinger D, Uda M, Abecasis GR, Muller-Myhsok B, Ehret GB, Boerwinkle E, Chakravarti A, Soliman EZ, Lunetta KL, Perz S, Wichmann HE, Meitinger T, Levy D, Gudnason V, Ellinor PT, Sanna S, Kaab S, Witteman JC, Alonso A, Benjamin EJ, Heckbert SR. Genome-wide association study of PR interval. Nat Genet 2010 January 10;42:153-9.

(5) van den Boogaard M, Wong LY, Tessadori F, Bakker ML, dreizehnter LK, Wakker V, Bezzina CR, ‘t Hoen PA, Bakkers J, Barnett P, Christoffels VM. Genetic variation in T-box binding element functionally affects SCN5A/SCN10A enhancer. J Clin Invest 2012 July 2;122(7):2519-30.

(6) Bruneau BG, Nemer G, Schmitt JP, Charron F, Robitaille L, Caron S, Conner DA, Gessler M, Nemer M, Seidman CE, Seidman JG. A murine model of Holt-Oram syndrome defines roles of the T-box transcription factor Tbx5 in cardiogenesis and disease. Cell 2001 September 21;106(6):709-21. (7) Schott J-J, Benson DW, Basson CT, Pease W, Silberbach GM, Moak JP, Maron BJ, Seidman CE,

Seidman JG. Congenital heart disease caused by mutations in the transcription factor NKX2-5. Science 1998;281:108-11.

(8) Basson CT, Cowley GS, Solomon SD, Weissman B, Poznanski AK, Traill TA, Seidman JG, Seidman CE. The clinical and genetic spectrum of the Holt-Oram syndrome (heart-hand syndrome). N Engl J

Med 1994 March 31;330(13):885-91.

(9) Bakker ML, Boukens BJ, Mommersteeg MTM, Brons JF, Wakker V, Moorman AFM, Christoffels VM. Transcription factor Tbx3 is required for the specification of the atrioventricular conduction system.

Circ Res 2008;102:1340-9.

(10) Frank DU, Carter KL, Thomas KR, Burr RM, Bakker ML, Coetzee WA, Tristani-Firouzi M, Bamshad MJ, Christoffels VM, Moon AM. Lethal arrhythmias in Tbx3-deficient mice reveal extreme dosage sensitivity of cardiac conduction system function and homeostasis. Proc Natl Acad Sci U S A 2011 December 27;109(3):E154-E163.

(11) Bamshad M, Lin RC, Law DJ, Watkins WS, Krakowiak PA, Moore ME, Franceschini P, Lala R, Holmes LB, Gebuhr TC, Bruneau BG, Schinzel A, Seidman JG, Seidman CE, Jorde LB. Mutations in human TBX3 alter limb, apocrine and genital development in ulnar-mammary syndrome. Nat Genet 1997 July;16(3):311-5.

(12) Hoogaars WMH, Barnett P, Rodriguez M, Clout DE, Moorman AFM, Goding CR, Christoffels VM. TBX3 and its splice variant TBX3 + exon 2a are functionally similar. Pigment Cell Melanoma Res 2008 June;21(3):379-87.

(13) Jay PY, Harris BS, Maguire CT, Buerger A, Wakimoto H, Tanaka M, Kupershmidt S, Roden DM, Schultheiss TM, O’Brien TX, Gourdie RG, Berul CI, Izumo S. Nkx2-5 mutation causes anatomic hypoplasia of the cardiac conduction system. J Clin Invest 2004 April;113(8):1130-7.

(14) Aanhaanen WT, Mommersteeg MT, Norden J, Wakker V, de Gier-de Vries C, Anderson RH, Kispert A, Moorman AF, Christoffels VM. Developmental origin, growth, and three-dimensional architecture of the atrioventricular conduction axis of the mouse heart. Circ Res 2010;107(6):728-36.

(15) Hoogaars WMH, Tessari A, Moorman AFM, de Boer PAJ, Hagoort J, Soufan AT, Campione M, Christoffels VM. The transcriptional repressor Tbx3 delineates the developing central conduction system of the heart. Cardiovasc Res 2004 June 1;62:489-99.

(16) Moskowitz IPG, Pizard A, Patel VV, Bruneau BG, Kim JB, Kupershmidt S, Roden D, Berul CI, Seidman CE, Seidman JG. The T-Box transcription factor Tbx5 is required for the patterning and maturation of the murine cardiac conduction system. Dev 2004 August;131(16):4107-16.

(17) Moskowitz IP, Kim JB, Moore ML, Wolf CM, Peterson MA, Shendure J, Nobrega MA, Yokota Y, Berul C, Izumo S, Seidman JG, Seidman CE. A molecular pathway including id2, tbx5, and nkx2-5 required

(16)

3

for cardiac conduction system development. Cell 2007 June 29;129(7):1365-76.

(18) Biben C, Weber R, Kesteven S, Stanley E, McDonald L, Elliott DA, Barnett L, Koentgen F, Robb L, Feneley M, Harvey RP. Cardiac septal and valvular dysmorphogenesis in mice heterozygous for mutations in the homeobox gene Nkx2-5. Circ Res 2000 November 10;87(10):888-95.

(19) Hoogaars WM, Engel A, Brons JF, Verkerk AO, de Lange FJ, Wong LY, Bakker ML, Clout DE, Wakker V, Barnett P, Ravesloot JH, Moorman AF, Verheijck EE, Christoffels VM. Tbx3 controls the sinoatrial node gene program and imposes pacemaker function on the atria. Genes Dev 2007 May 1;21(9):1098-112.

(20) Moorman AFM, Houweling AC, de Boer PAJ, Christoffels VM. Sensitive nonradioactive detection of mRNA in tissue sections: novel application of the whole-mount in situ hybridization protocol. J

Histochem Cytochem 2001;49:1-8.

(21) Aanhaanen WT, Brons JF, Dominguez JN, Rana MS, Norden J, Airik R, Wakker V, de Gier-de Vries C, Brown NA, Kispert A, Moorman AF, Christoffels VM. The Tbx2+ primary myocardium of the atrioventricular canal forms the atrioventricular node and the base of the left ventricle. Circ Res 2009 May 7;104(11):1267.

(22) Mommersteeg MTM, Hoogaars WMH, Prall OWJ, de Gier-de Vries C, Wiese C, Clout D.E.W., Papaioannou VE, Brown NA, Harvey RP, Moorman AFM, Christoffels VM. Molecular pathway for the localized formation of the sinoatrial node. Circ Res 2007;100:354-62.

(23) Soufan AT, Ruijter JM, van den Hoff MJB, de Boer PAJ, Hagoort J, Moorman AFM. Three-dimensional reconstruction of gene expression patterns during cardiac development. Physiol Genomics 2003;13:187-95.

(24) Boukens BJ, Hoogendijk MG, Verkerk AO, Linnenbank A, van DP, Remme CA, Fiolet JW, Opthof T, Christoffels VM, Coronel R. Early repolarization in mice causes overestimation of ventricular activation time by the QRS duration. Cardiovasc Res 2012 September 20.

(25) Risebro CA, Petchey LK, Smart N, Gomes J, Clark J, Vieira JM, Yanni J, Dobrzynski H, Davidson S, Zuberi Z, Tinker A, Shui B, Tallini YI, Kotlikoff MI, Miquerol L, Schwartz RJ, Riley PR. Epistatic Rescue of Nkx2.5 Adult Cardiac Conduction Disease Phenotypes by Prospero-Related Homeobox Protein 1 and HDAC3. Circ Res 2012 July 6;111(2):e19-e31.

(26) Zhang SS, Kim KH, Rosen A, Smyth JW, Sakuma R, Delgado-Olguin P, Davis M, Chi NC, Puviindran V, Gaborit N, Sukonnik T, Wylie JN, Brand-Arzamendi K, Farman GP, Kim J, Rose RA, Marsden PA, Zhu Y, Zhou YQ, Miquerol L, Henkelman RM, Stainier DY, Shaw RM, Hui CC, Bruneau BG, Backx PH. Iroquois homeobox gene 3 establishes fast conduction in the cardiac His-Purkinje network. Proc

Natl Acad Sci U S A 2011 August 16;108(33):13576-81.

(27) Mangoni ME, Traboulsie A, Leoni AL, Couette B, Marger L, Le QK, Kupfer E, Cohen-Solal A, Vilar J, Shin HS, Escande D, Charpentier F, Nargeot J, Lory P. Bradycardia and slowing of the atrioventricular conduction in mice lacking CaV3.1/{alpha}1G T-Type Calcium Channels. Circ Res 2006 2006 May 11;98(11):1422-30.

(28) van den Boogaard M, Wong LYE, Tessadori F, Bakker ML, dreizehnter LK, Wakker V, Bezzina CR, ‘t Hoen PAC, Barnett P, Christoffels VM. T-box targets scn5a/10a are functionally affected by a genetic variation. J Clin Invest 2012.

(17)

3

September 19.

(31) Remme CA, Scicluna BP, Verkerk AO, Amin AS, van BS, Beekman L, Deneer VH, Chevalier C, Oyama F, Miyazaki H, Nukina N, Wilders R, Escande D, Houlgatte R, Wilde AA, Tan HL, Veldkamp MW, de Bakker JM, Bezzina CR. Genetically determined differences in sodium current characteristics modulate conduction disease severity in mice with cardiac sodium channelopathy. Circ Res 2009 June 5;104(11):1283-92.

(32) Fleg JL, Das DN, Wright J, Lakatta EG. Age-associated changes in the components of atrioventricular conduction in apparently healthy volunteers. J Gerontol 1990 May;45(3):M95-100.

(33) Schmidlin O, Bharati S, Lev M, Schwartz JB. Effects of physiological aging on cardiac

electrophysiology in perfused Fischer 344 rat hearts. Am J Physiol 1992 January;262(1 Pt 2):H97-105. (34) Froese A, Breher SS, Waldeyer C, Schindler RF, Nikolaev VO, Rinne S, Wischmeyer E, Schlueter J,

Becher J, Simrick S, Vauti F, Kuhtz J, Meister P, Kreissl S, Torlopp A, Liebig SK, Laakmann S, Muller TD, Neumann J, Stieber J, Ludwig A, Maier SK, Decher N, Arnold HH, Kirchhof P, Fabritz L, Brand T. Popeye domain containing proteins are essential for stress-mediated modulation of cardiac pacemaking in mice. J Clin Invest 2012 March 1;122(3):1119-30.

Referenties

GERELATEERDE DOCUMENTEN

Light in strongly scattering semiconductors - diffuse transport and Anderson localization.. Gomez

The direction of prop- agationn of light is randomized in an average distance given by the transport mean freee path L The photonic strength of the system is defined by the inverse

Naast de totale transmissie, hebben wij ook de totale reflectiee gemeten, oftewel de fractie van het licht dat gereflecteerd wordt door het monster,, en de coherente

Autonomic modulation is essential for proper functioning of the heart and contributes to the prognosis of patients with heart failure and congenital heart disease. Early

In chapter 6, the development of the sinus venosus myocardium and sinoatrial node after inhibition of the RHOA- ROCK pathway is studied in chicken embryos.. The effect of inhibition

In human, clinical arrhythmias are often related to sinus venosus related structures in the atria, such as the crista terminalis (embryonic right venous valve), the myocardium of

The CCS initiates and coordinates electrical activation of the myocardium, which is essential for normal functioning of the heart. Rhythmic contraction of the PHT

As mentioned, it was shown that in the region of the developing AVN, different cell populations (i.e. relative high expression of HCN4 and ISL1 in the myocardial continuity