Nkx2.5/CARP signaling pathway contributes to the regulation of ion channel remodeling induced by rapid pacing in rat atrial myocytes

  • Authors:
    • Wei Wang
    • Yun Zhu
    • Jianguang Yi
    • Wei Cheng
  • View Affiliations

  • Published online on: September 6, 2016     https://doi.org/10.3892/mmr.2016.5727
  • Pages: 3848-3854
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Remodeling of atrial electrophysiology and structure is the primary feature of atrial fibrillation (AF). Evidence suggests that abnormalities in the expression levels of embryological cardiovascular development‑associated transcription factors, including Nkx2.5, are crucial for the development of AF. Rat atrial myocardial cells (AMCs) in culture dishes were placed in an electric field and stimulated. Transmission electron microscopy was used to observe the ultrastucture prior to and following rapid pacing. The action potential durations and effective refractory periods were measured. RT‑PCR and western blotting were performed to investigate the effect of rapid pacing on the expression levels of ion channel and nuclear proteins in AMCs. Nkx2.5 short interfering RNA (siRNA) transfection was performed. Through this in vitro rat AMC culture and rapid pacing model, it was demonstrated that rapid pacing shortened the action potential and downregulated the expression levels of L‑type calcium and potassium channels. Expression levels of Nkx2.5 and cardiac ankyrin repeat protein (CARP) were significantly upregulated by rapid pacing at the mRNA and protein levels. siRNA‑mediated Nkx2.5 silencing attenuated the rapid pacing‑induced downreglation of ion channel levels. These results suggest that the Nkx2.5/CARP signaling pathway contributes to the early electrical remodeling process of AF.

Introduction

Atrial fibrillation (AF) is one of the most common arrhythmias encountered in clinical practice (1). The prevalence of AF in the general population is ~1% and the risk increases with age. AF substantially increases cardiovascular morbidity and mortality. There is a 5-fold increase in the risk of stroke in patients with AF and 15–20% of strokes are caused by AF. AF is an independent risk factor for congestive heart failure and increases the mortality 2-fold. Genetic defects may be responsible for the pathogenesis of AF in a subset of patients. During AF, electrical and structural remodeling occurs continually (24). The electrophysiological and structural remodeling is crucial for the development, maintenance and recurrence of AF. Remodeling shortens the atrial wavelength of intraatrial reentry and leads to an increase in the potential number of electrical reentrant cycles, which is responsible for AF maintenance. Changes in the expression levels of ion channels, including L-type calcium (Ca) channel (LTCC) and potassium (K) channel, are important for early remodeling during AF (5). However, the pathogenic mechanisms underlying atrial structural remodeling remain to be elucidated.

Changes in atrial electrophysiology and structure, referred to as remodeling, constitute the primary features of AF occurrence, maintenance and recurrence (6,7). Changes in ion currents, including Ca2+ and K+, form the basis for early electrical remodeling in AF (8,9). Calcium entry from the extracellular space through LTCCs and the resultant intracellular Ca2+ elevation (calcium overload) was demonstrated to be crucial in the regulation of atrial frequency-dependent action potential duration (APD) and effective refractory period (ERP) (10). The transient outward K+ current mediated by the potassium channel Kv4.3 contributes to early repolarization (11).

Emerging evidence indicates that abnormalities in cardiovascular embryological development contribute to AF (12). Nkx2.5 is a critical transcription factor and its mutation is associated with AF development (1316). As a member of the NK2 family, the expression and functions of Nkx2.5 overlap with those of the GATA family during cardiovascular development (17,18). However, whether Nkx2.5 affects ion channel proteins in the context of AF remains to be elucidated. The aim of the present study was to investigate the effect of rapid pacing (rapid electrical stimulation used to simulate AF) on APD, ion channel proteins and the Nkx2.5/CARP (cardiac ankyrin repeat protein) signaling pathway.

Materials and methods

Isolation and culture of rat atrial myocardial cells (AMCs)

The present study and all experimental protocols involved were approved by the Institutional Animal Care and Use Committee of the Third Military Medical University (Chongqing, China). A total of 20 female Wistar rats (2 weeks old) were purchased from the Experimental Animal Center of the Third Military Medical University. Rats were maintained under constant temperature and humidity conditions with a 12-h light/dark cycle and ad libitum access to standard chow and water. Prior to the experiment, rats were sacrificed by CO2 inhalation and then fixed in a supine position. Following sterilization with 70% ethanol, an incision was made along the right edge of the sternum and the chest wall was removed. The heart was dissected out and washed in cold phosphate-buffered saline (PBS). The left and right atria were isolated and washed with serum-free Dulbecco's modified Eagle's medium (DMEM; Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) supplemented with penicillin and streptomycin. Under aseptic conditions, the right atrial appendage was cut into small pieces with scissors, which were then digested with 0.08% trypsin at 37°C for 5 min. The reaction was terminated by addition of DMEM containing 10% fetal bovine serum (FBS; HyClone; GE Healthcare Life Sciences, Logan, UT, USA). This solution was maintained at room temperature and the supernatant was filtered through a 100-μm mesh filter. Digestion was performed twice. Finally, suspensions of single cells were prepared by treatment of the digested product with 0.1% collagenase at 37°C for 15 min. The cells were seeded into flasks at a density of ~1×108/l, followed by incubation with DMEM containing 10% FBS at 37°C with 5% CO2. In the control group, following 72 h of routine culture, the medium was replaced with serum-free DMEM for 24 h, and then rapid pacing was performed.

Rapid pacing of AMCs

When the cell confluence reached ~80%, the culture dishes were placed in an electrical field and stimulated with 10 Hz, 1.5 V/cm using the BL-420E+ biological and functional experimental system (Chengdu Techman Software, Co., Ltd., Chengdu, China). The beating frequency of the cells was visually recorded. The survival rate of cells prior to and following the rapid pacing was assessed by 3-[4,5-dimethylthiazol-2-y1]-2,5-diphenytetrazolium bromide assay (Thermo Fisher Scientific, Inc.), according to the manufacturer's instructions, and the APD at 50 and 90% repolarization was recorded with a patch clamp at the whole cell mode.

Transmission electron microscopy

AMCs prior to and following rapid pacing were collected, transferred into Eppendorf tubes, resuspended in cold PBS and centrifuged at 200 × g for 5 min at 4°C. The supernatant was removed and cells were fixed in 2% glutaraldehyde for 2 h and post-fixed in 1% tetroxide osmium for 2 h. Following dehydration with an alcohol gradient, cells were embedded in epoxy resin 618 (Shanghai Kang Lang Biological Technology Co., Ltd., Shanghai, China). Ultrathin sections (100 nm) were prepared and contrast stained with uranyl acetate and lead citrate. Images were captured (magnification, ×6,000) using a transmission electron microscope (H7500; Hitachi, Ltd., Tokyo, Japan).

RNA interference

The short interfering RNA (siRNA) for Nkx2.5 and negative control siRNA duplexes were designed and synthesized by Shanghai GenePharma Co., Ltd. (Shanghai, China). The sequences of siRNA duplexes were as follows: Sense, 5′-UUCUCCGAACGUGUCACGUTT-3′ and anti-sense, 5′-ACGUGACACGUUCGGAGAATT-3′ for the negative control; sense, 5′-CCCUCGGGCGGAUAAGAAATT-3′ and anti-sense, 5′-UUUCUUAUCCGCCCGAGGGTC-3′ for Nkx2.5-310. In each group, 1×105 cells were seeded into 60-mm culture dishes without antibiotics. At 70% confluence, the siRNA duplexes for Nkx2.5 and negative control were added with Lipofectamine® RNAimax (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's instructions.

Reverse transcription-polymerase chain reaction (RT-PCR) analysis

mRNA expression levels were detected by RT-PCR. Total cellular RNA was extracted from the rat AMCs of each group using TRIzol® Reagent (Invitrogen; Thermo Fisher Scientific, Inc.) and cDNA was prepared using a Superscript II First-Strand cDNA synthesis kit (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's instructions. PCR was performed using Taq DNA Polymerase (Beijing Solarbio Science & Technology Co., Ltd., Beijing, China). The primer sequences and annealing temperatures were as follows: Forward, 5′-ATGGAGGCTGGAGCCCAGATTGA-3′ and reverse, 5′-GACATTGAGGTCCGCACCGAAGG-3′ for α1c (annealing temperature, 61.3°C); forward, 5′-GCAGCAACCTGAAATCTGAAACT-3′ and reverse, 5′-GATAAGCAATGAACCCATCTCCA-3′ for Kv4.3 (annealing temperature, 56.1°C); forward, 5′-TTGTTTCTGTCACCAGTAAC-3′ and reverse, 5′-GATGAGGAAGGAAGAGAAGC-3′ for connexin-43 (Cx43; annealing temperature, 56.3°C); forward, 5′-GTAAGCGACAGCGGCAGGAC-3′ and reverse, 5′-CGACGCCAAAGTTCACGAAG-3′ for Nkx2.5 (annealing temperature, 58.7°C); forward, 5′-GGGGTACCAGCCAACATGATG-3′ and reverse, 5′-CCCTCGAGGCCTCAGAATGTAGC-3′ for CARP (annealing temperature, 60.1°C); and forward, 5′-TGAGAGGGAAATCGTGCGTGAC-3′ and reverse, 5′-ATCTGCTGGAAGGTGGACAGTGAG-3′ for β-actin (annealing temperature, 53.9°C). The amplification process was performed for 35 cycles following an initial 45 sec denaturation at 94°C, annealed for 30 sec at the above-indicated temperatures and extended for 5 min at 72°C. PCR products were separated by agarose gel electrophoresis and stained with ethidium bromide. Band intensities were measured by densitometry and normalized to β-actin using ImageJ software version 1.5.0 (National Institutes of Health, Bethesda, MD, USA).

Western blot analysis

Rat AMCs (1.5×106) from each group were lysed with 0.5 ml radioimmunoprecipitation assay buffer (Bio-Rad Laboratories, Inc., Hercules, CA, USA) containing 5 μl phenylmethylsulfonyl fluoride. Cell lysates were centrifuged at 12,000 × g at 4°C for 30 min and the resulting supernatant (total tissue homogenate) was stored at -80°C until further analysis. Protein (15 μg) from each group was separated by 15% SDS-PAGE (200 V, 45 min) and transferred to polyvinylidene difluoride membranes. The membranes were blocked with 5% bovine serum albumin (HyClone; GE Healthcare Life Sciences) in Tris-buffered saline containing Tween 20 (TBST) for 1 h at room temperature, and then were incubated with the following primary antibodies: Rabbit anti-rat α1c (1:2,000; catalog no. AB5156; EMD Millipore, Billerica, MA, USA), rabbit anti-rat Kv4.3 (1:1,000; catalog no. AB5194; EMD Millipore), rabbit anti-rat Cx43 (1:1,000; catalog no. sc-9059; Santa Cruz Biotechnology, Inc., Dallas, TX, USA), rabbit anti-rat Nkx2.5 (1:500; catalog no. sc-14033; Santa Cruz Biotechnology, Inc.), rabbit anti-CARP (1:500; catalog no. sc-30181; Santa Cruz Biotechnology, Inc.) and rabbit anti-β-actin (1:1,000; catalog no. ab8227; Abcam, Cambridge, MA, USA). Membranes were washed three times in TBST and incubated with a horseradish peroxidase-conjugated secondary goat anti-rabbit IgG antibody (1:500; catalog no. DGSP-H-KIT-4; Beijing Dingguo Changsheng Biotechnology Co., Ltd., Beijing, China). The protein bands were visualized using SuperSensitive Enhanced Chemiluminescence solution (Beijing Dingguo Changsheng Biotechnology Co., Ltd.) and quantified using ImageJ software version 1.5.0. Band intensities were measured by densitometry and normalized to β-actin.

Statistical analysis

Statistical analysis was performed in SPSS version 17.0 (SPSS, Inc., Chicago, IL, USA). Group comparisons were performed by one-way analyses of variance, and the Student-Newman-Keuls method was used as a post-hoc test. Data are expressed as the mean ± standard deviation. P<0.05 was considered to indicate a statistically significant difference.

Results

Observation of cultured atrial myocardial cells (AMCs)

AMCs cultured for three days were heterogeneous in shape, including rod, spindle, triangular and irregular (Fig. 1A). The number of spontaneously beating cells increased following 48 h in culture. AMCs exhibited clear ultrastructural features and regular arrangement of mitochondrial cristae (Fig. 1B). At 24 h following 3-h rapid pacing, AMCs presented with a more polygonal shape, irregular myofibril arrangement and sparse myofilaments (Fig. 1C and D). At 48 and 72 h following rapid pacing, vacuolar degeneration and expanded bubbles were observed (Fig. 1E and F). The beat frequency did not alter significantly following rapid pacing (Fig. 1G); however the survival rate decreased from 48 h following rapid pacing (Fig. 1H; P<0.001). Under normal conditions, cells would proliferate for one month. Therefore, the decrease in survival rate was as a result of rapid pacing.

Electrophysiological changes in AMCs

The APD measured at 50% repolarization (APD 50) was significantly decreased from 12 h following rapid pacing (12 h, P=0.0235; 24 h, P=0.0014; 48 h, P=0.0005; Fig. 2A and B). The APD 90 was significantly decreased at 24 (P=0.056) and 48 (P=0.0021) h following rapid pacing (Fig. 2A and B). No significant differences in ERP were observed following rapid pacing (6 h, P=0.6647; 12 h, P=0.3858; 24 h, P=0.3438; 48 h, P=0.3930; Fig. 2A and C).

Effect of rapid pacing on the expression levels of ion channels and nuclear proteins in AMCs

mRNA expression levels of the LTCC protein α1c were significantly reduced at 6 h (P=0.023) following rapid pacing compared with prior to rapid pacing, and this difference increased at 12 (P=0.0053), 24 (P=0.0021) and 48 (P=0.0016) h (Fig. 3A). Western blotting correlated well with RT-PCR data, with α1c protein expression levels significantly reduced from 12 h following rapid pacing (P=0.0036; Fig. 3B). mRNA (P=0.0011) and protein (P=0.0085) expression levels of the potassium channel Kv4.3 were decreased from 12 h following rapid pacing (Fig. 3C and D). The mRNA and protein expression levels of the important gap junction protein Cx43 were not affected by rapid pacing (Fig. 3E and F). The mRNA (P=0.022) and protein (P=0.0073) expression levels of Nkx2.5, a critical cardiac transcription factor, were upregulated from 12 h following rapid pacing (Fig. 3G and H). CARP, a downstream molecule in the Nkx2.5 homeobox gene signaling pathway, exhibited a similar pattern to Nkx2.5 (mRNA, P=0.0005 and protein, P=0.0032 at 12 h; Fig. 3I and J).

Effect of Nkx2.5 inhibition on the expression levels of ion channel proteins in AMCs

As presented in Fig. 4A, transfection with Nkx2.5 siRNA inhibited the rapid pacing-induced increase in Nkx2.5 expression at the mRNA level (P=0.0089). In addition, the increase in mRNA expression levels of CARP induced by rapid pacing was inhibited by Nkx2.5 siRNA (P=0.0068; Fig. 4B and C). Protein expression levels of Nkx2.5 (P=0.046) and CARP (P=0.031) followed the same pattern (Fig. 4D and E).

Furthermore, treatment with Nkx2.5 siRNA attenuated the decrease in α1c and Kv4.3 mRNA (α1c, P= 0.028; Kv4.3, P=0.043) and protein (α1c, P=0.017; Kv4.3, P=0.019) expression levels induced by rapid pacing (Fig. 5).

Discussion

AF, the most common form of sustained cardiac arrhythmia, is characterized by uncoordinated atrial activation and chaotic electrical activity, with consequent deterioration of atrial mechanical function (19). Using the in vitro rat AMC culture and rapid pacing model, the present study demonstrated that rapid pacing shortened the APD and downregulated the expression levels of LTCC and potassium channels. Expression of Nkx2.5 and CARP were significantly upregulated by rapid pacing at the mRNA and protein levels. siRNA-mediated Nkx2.5 silencing rescued the rapid pacing-induced downreglation of ion channel expression levels, suggesting that the Nkx2.5/CARP signaling pathway contributes to the early electrical remodeling process of AF.

In the current study, the APD of rat AMCs was significantly reduced 12 h subsequent to rapid pacing, while no effect on the ERP was observed at any time point. In AF patients, shortened APD results in decreased wavelength of reentry circuits and atrial electrical remodeling, thus facilitating the maintenance of AF and inhibiting the natural termination of AF (20,21). The lack of an effect on ERP may be due to differences in pacing rate, electric field strength and varying cell sources. The causes of APD shortening include: i) Increased outward K+ currents; ii) decreased inward Ca2+ current; and iii) a combination of the above two factors (22). L-type Ca2+ current is activated by membrane depolarization and contributes to the formation of the action potential plateau phase (23). As in the ventricular muscle cells, the transient outward K+ current (Ito) is the basis of early rapid repolarization of atrial action potential, while Kv4.3 is the major pore-forming subunit of Ito channels (24). Atrial rapid pacing, as occurs in atrial fibrillation, may lead to a decrease in the density of functional ion channels (Na+, Ca2+ and K+) (25); however, no effect was observed on the intrinsic properties of single ion channels.

A calcium channel current (ICa) is essential for action potential and excitation-contraction coupling of myocardial cells (26). The voltage-dependent calcium channels are typically divided into L- and T-type channels, and Ca2+ influx mediated by LTCCs is an important factor regulating human atrial frequency-dependent APD. The present study revealed that the expression level of α1c at 6 h subsequent to rapid pacing was significantly reduced compared with prior to pacing, becoming stable at 24 h. The reduction in LTCC currents is critical for the shortening of the action potential cycle, and decreased calcium influx is harmful to atrial mechanical contraction. The expression level of Kv4.3 was significantly reduced from 12 h subsequent to rapid pacing. These results are largely in accordance with other experimental models of atrial fibrillation, and may reflect an attempt to prevent the shortening of APD and ERP; however, the underlying mechanism requires further study.

Gap junctions between cardiac cells provide connections and a low-resistance pathway interconnecting cardiomyocytes (27). These coordinate myocardial action potential and synchronous contraction. The gap junction Cx proteins present in heart cells include Cx40 and Cx43 (28). A change in the structure and density of Cx may result in changes in conductivity anisotropy and conduction velocity of atrial myocytes, ideal conditions for reentrant arrhythmia (29). In the present study, no significant changes in Cx43 were observed, which may be due to various factors: The pacing duration may not have been long enough; changes in Cx43 may be the result of long-term AF; or changes in the distribution of Cx43 may be of greater importance than its expression levels. Further investigations are required to elucidate the role of gap junction proteins in atrial electrical remodeling.

Anomalies in embryological cardiovascular development contribute to the initiation of AF (30,31). Various transcription factors, including Nkx2.5, were identified as essential in cardiovascular genesis (32,33). Gutierrez-Roelens et al (34) first identified an Nkx2.5 mutation suggested to be associated with the atrial fibrillation phenotype. Homeobox gene Nkx2.5, also referred to as cardiac-specific homeobox gene, belongs to the NK-2 homeobox family. Nkx2.5 is crucial for myocardial cell differentiation and heart tube formation, and is involved in the atrioventricular separation and conduction system (35). As a downstream mediator of Nkx2.5, CARP contributes to the maintenance of complete sarcomere structure and function, and is involved in the regulation of intracellular calcium (36). The present study demonstrated that the expression levels of Nkx2.5 and CARP were significantly elevated during the early phase following fast pacing, indicating that Nkx2.5 is important in undifferentiated cells and in differentiated cardiomyocytes. As the Nkx2.5/CARP signaling pathway is a critical regulator of cell development, cell communication and intracellular calcium, it was hypothesized that the Nkx2.5/CARP signaling pathway may be critical for ion channel remodeling in the early stages of atrial fibrillation.

In the present study, although Nkx2.5-siRNA transfected AMCs inhibited the downregulation of α1c and Kv4.3 expression levels induced by rapid pacing, this downregulation was not completely reversed, suggesting that ion channel remodeling is regulated by multiple factors.

In conclusion, the results of the present study demonstrated that rapid pacing may shorten APD and induce the downregulation of the LTCC protein, α1c and potassium channel, Kv4.3, resembling the electrophysiological properties of atrial fibrillation. The Nkx2.5/CARP signaling pathway was upregulated by rapid pacing, while Nkx2.5 siRNA-mediated gene silencing inhibited the rapid pacing-induced ion channel downregulation. These results indicate that the Nkx2.5/CARP signaling pathway may be involved in the early channel remodeling process during rapid pacing. These findings may have implications for the early detection of AF, and suggest potential targets for prophylaxis.

Acknowledgments

The present study was supported by the National Natural Science Foundation of China (grant no. 30600252).

References

1 

Wang XH, Huang CX, Wang Q, Li RG, Xu YJ, Liu X, Fang WY and Yang YQ: A novel GATA5 loss-of-function mutation underlies lone atrial fibrillation. Int J Mol Med. 31:43–50. 2013.

2 

Zhou YM, Zheng PX, Yang YQ, Ge ZM and Kang WQ: A novel PITX2c loss-of-function mutation underlies lone atrial fibrillation. Int J Mol Med. 32:827–834. 2013.PubMed/NCBI

3 

Yoon N, Cho JG, Kim KH, Park KH, Sim DS, Yoon HJ, Hong YJ, Park HW, Kim JH, Ahn Y, et al: Beneficial effects of an angiotensin-II receptor blocker on structural atrial reverse-remodeling in a rat model of ischemic heart failure. Exp Ther Med. 5:1009–1016. 2013.PubMed/NCBI

4 

Qiu XB, Xu YJ, Li RG, Xu L, Liu X, Fang WY, Yang YQ and Qu XK: PITX2C loss-of-function mutations responsible for idiopathic atrial fibrillation. Clinics (Sao Paulo). 69:15–22. 2014. View Article : Google Scholar

5 

Gan TY, Qiao W, Xu GJ, Zhou XH, Tang BP, Song JG, Li YD, Zhang J, Li FP, Mao T and Jiang T: Aging-associated changes in L-type calcium channels in the left atria of dogs. Exp Ther Med. 6:919–924. 2013.PubMed/NCBI

6 

Xu GJ, Gan TY, Tang BP, Chen ZH, Mahemuti A, Jiang T, Song JG, Guo X, Li YD, Zhou XH, et al: Alterations in the expression of atrial calpains in electrical and structural remodeling during aging and atrial fibrillation. Mol Med Rep. 8:1343–1352. 2013.PubMed/NCBI

7 

Fu G, Cao Y, Lu J, Li J, Liu L, Wang H, Su F and Zheng Q: Programmed cell death-1 deficiency results in atrial remodeling in C57BL/6 mice. Int J Mol Med. 31:423–429. 2013.

8 

Heijman J and Dobrev D: Systems approaches to post-operative atrial fibrillation-do they help us to better understand the ionic basis of the arrhythmogenic substrate? J Mol Cell Cardio. 53:320–322. 2012. View Article : Google Scholar

9 

Nattel S and Dobrev D: The multidimensional role of calcium in atrial fibrillation pathophysiology: Mechanistic insights and therapeutic opportunities. Eur Heart J. 33:1870–1877. 2012. View Article : Google Scholar : PubMed/NCBI

10 

Ren Y, Zhang M, Zhang T and Huang R: Effect of ouabain on myocardial remodeling in rats. Exp Ther Med. 6:65–70. 2013.PubMed/NCBI

11 

Huo R, Sheng Y, Guo WT and Dong DL: The potential role of Kv4.3 K+ channel in heart hypertrophy. Channels (Austin). 8:203–209. 2014. View Article : Google Scholar

12 

Yue L, Xie J and Nattel S: Molecular determinants of cardiac fibroblast electrical function and therapeutic implications for atrial fibrillation. Cardiovasc Res. 89:744–753. 2011. View Article : Google Scholar :

13 

Huang RT, Xue S, Xu YJ, Zhou M and Yang YQ: A novel NKX2.5 loss-of-function mutation responsible for familial atrial fibrillation. Int J Mol Med. 31:1119–1126. 2013.PubMed/NCBI

14 

Yuan F, Qiu XB, Li RG, Qu XK, Wang J, Xu YJ, Liu X, Fang WY, Yang YQ and Liao DN: A novel NKX2-5 loss-of-function mutation predisposes to familial dilated cardiomyopathy and arrhythmias. Int J Mol Med. 35:478–486. 2015.

15 

Wang J, Zhang DF, Sun YM, Li RG, Qiu XB, Qu XK, Liu X, Fang WY and Yang YQ: NKX2-6 mutation predisposes to familial atrial fibrillation. Int J Mol Med. 34:1581–1590. 2014.PubMed/NCBI

16 

Yu H, Xu JH, Song HM, Zhao L, Xu WJ, Wang J, Li RG, Xu L, Jiang WF, Qiu XB, et al: Mutational spectrum of the NKX2-5 gene in patients with lone atrial fibrillation. Int J Med Sci. 11:554–563. 2014. View Article : Google Scholar : PubMed/NCBI

17 

Zhang Y, Rath N, Hannenhalli S, Wang Z, Cappola T, Kimura S, Atochina-Vasserman E, Lu MM, Beers MF and Morrisey EE: GATA and Nkx factors synergistically regulate tissue-specific gene expression and development in vivo. Development. 134:189–198. 2007. View Article : Google Scholar

18 

Wang J, Zhang DF, Sun YM and Yang YQ: A novel PITX2c loss-of-function mutation associated with familial atrial fibrillation. Eur J Med Genet. 57:25–31. 2014. View Article : Google Scholar

19 

Shi HF, Yang JF, Wang Q, Li RG, Xu YJ, Qu XK, Fang WY, Liu X and Yang YQ: Prevalence and spectrum of GJA5 mutations associated with lone atrial fibrillation. Mol Med Rep. 7:767–774. 2013.PubMed/NCBI

20 

Iwasaki YK, Nishida K, Kato T and Nattel S: Atrial fibrillation pathophysiology: Implications for management. Circulation. 124:2264–2274. 2011. View Article : Google Scholar : PubMed/NCBI

21 

Lee HL, Chang PC, Chou CC, Wo HT, Chu Y, Wen MS, Yeh SJ and Wu D: Blunted proarrhythmic effect of nicorandil in a Langendorff-perfused phase-2 myocardial infarction rabbit model. Pacing Clin Electrophysiol. 36:142–151. 2013. View Article : Google Scholar

22 

Shaw RM and Rudy Y: Electrophysiologic effects of acute myocardial ischemia: A theoretical study of altered cell excitability and action potential duration. Cardiovasc Res. 35:256–272. 1997. View Article : Google Scholar : PubMed/NCBI

23 

Wang X, Wang X, Gu Y, Wang T and Huang C: Wenxin Keli attenuates ischemia-induced ventricular arrhythmias in rats: Involvement of L-type calcium and transient outward potassium currents. Mol Med Rep. 7:519–524. 2013.

24 

Zhang H, Wu S, Huang C and Li X: Long-term treatment of spontaneously hypertensive rats with losartan and molecular basis of modulating Ito of ventricular myocytes. Mol Med Rep. 9:1959–1967. 2014.PubMed/NCBI

25 

Nattel S, Burstein B and Dobrev D: Atrial remodeling and atrial fibrillation: Mechanisms and implications. Circ Arrhythm Electrophysio. 1:62–73. 2008. View Article : Google Scholar

26 

Shi S, Liu T, Li Y, Qin M, Tang Y, Shen JY, Liang J, Yang B and Huang C: Chronic N-methyl-D-aspartate receptor activation induces cardiac electrical remodeling and increases susceptibility to ventricular arrhythmias. Pacing Clin Electrophysiol. 37:1367–1377. 2014. View Article : Google Scholar : PubMed/NCBI

27 

Zhang Q, Deng C, Rao F, Modi RM, Zhu J, Liu X, Mai L, Tan H, Yu X, Lin Q, et al: Silencing of desmoplakin decreases connexin43/Nav1.5 expression and sodium current in HL-1 cardiomyocytes. Mol Med Rep. 8:780–786. 2013.PubMed/NCBI

28 

Yan Y, Huang J, Ding F, Mei J, Zhu J, Liu H and Sun K: Aquaporin 1 plays an important role in myocardial edema caused by cardiopulmonary bypass surgery in goat. Int J Mol Med. 31:637–643. 2013.PubMed/NCBI

29 

Valderrábano M: Influence of anisotropic conduction properties in the propagation of the cardiac action potential. Prog Biophys MolBiol. 94:144–168. 2007. View Article : Google Scholar

30 

Mommersteeg MT, Christoffels VM, Anderson RH and Moorman AF: Atrial fibrillation: A developmental point of view. Heart Rhythm. 6:1818–1824. 2009. View Article : Google Scholar : PubMed/NCBI

31 

Hong K and Xiong Q: Genetic basis of atrial fibrillation. Curr Opin Cardiol. 29:220–226. 2014. View Article : Google Scholar : PubMed/NCBI

32 

Searcy RD, Vincent EB, Liberatore CM and Yutzey KE: A GATA-dependent nkx-2.5 regulatory element activates early cardiac gene expression in transgenic mice. Development. 125:4461–4470. 1998.PubMed/NCBI

33 

Mahida S: Transcription factors and atrial fibrillation. Cardiovasc Res. 101:194–202. 2014. View Article : Google Scholar

34 

Gutierrez-Roelens I, De Roy L, Ovaert C, Sluysmans T, Devriendt K, Brunner HG and Vikkula M: A novel CSX/NKX2-5 mutation causes autosomal-dominant AV block: Are atrial fibrillation and syncopes part of the phenotype? Eur J Hum Genet. 14:1313–1316. 2006. View Article : Google Scholar : PubMed/NCBI

35 

Moskowitz IP, Kim JB, Moore ML, Wolf CM, Peterson MA, Shendure J, Nobrega MA, Yokota Y, Berul C, Izumo S, et al: A molecular pathway including Id2, Tbx5, and Nkx2-5 required for cardiac conduction system development. Cell. 129:1365–1376. 2007. View Article : Google Scholar : PubMed/NCBI

36 

Chen B, Zhong L, Roush SF, Pentassuglia L, Peng X, Samaras S, Davidson JM, Sawyer DB and Lim CC: Disruption of a GATA4/Ankrd1 signaling axis in cardiomyocytes leads to sarcomere disarray: Implications for anthracycline cardiomyopathy. PLoS One. 7:e357432012. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October-2016
Volume 14 Issue 4

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wang W, Zhu Y, Yi J and Cheng W: Nkx2.5/CARP signaling pathway contributes to the regulation of ion channel remodeling induced by rapid pacing in rat atrial myocytes. Mol Med Rep 14: 3848-3854, 2016
APA
Wang, W., Zhu, Y., Yi, J., & Cheng, W. (2016). Nkx2.5/CARP signaling pathway contributes to the regulation of ion channel remodeling induced by rapid pacing in rat atrial myocytes. Molecular Medicine Reports, 14, 3848-3854. https://doi.org/10.3892/mmr.2016.5727
MLA
Wang, W., Zhu, Y., Yi, J., Cheng, W."Nkx2.5/CARP signaling pathway contributes to the regulation of ion channel remodeling induced by rapid pacing in rat atrial myocytes". Molecular Medicine Reports 14.4 (2016): 3848-3854.
Chicago
Wang, W., Zhu, Y., Yi, J., Cheng, W."Nkx2.5/CARP signaling pathway contributes to the regulation of ion channel remodeling induced by rapid pacing in rat atrial myocytes". Molecular Medicine Reports 14, no. 4 (2016): 3848-3854. https://doi.org/10.3892/mmr.2016.5727