MicroRNA-1 accelerates the shortening of atrial effective refractory period by regulating KCNE1 and KCNB2 expression: an atrial tachypacing rabbit model

Xiaomeng Jia, Shaohua Zheng, Xinxing Xie, Yujiao Zhang, Weizong Wang, Zhongsu Wang, Yong Zhang, Jiangrong Wang, Mei Gao, Yinglong Hou, Xiaomeng Jia, Shaohua Zheng, Xinxing Xie, Yujiao Zhang, Weizong Wang, Zhongsu Wang, Yong Zhang, Jiangrong Wang, Mei Gao, Yinglong Hou

Abstract

Background: The potential mechanisms of microRNA-1 (miR-1) in the electrical remodeling of atrial fibrillation remain unclear. The purpose of this study was to evaluate the effects of miR-1 on the atrial effective refractory period (AERP) in a right atrial tachypacing model and to elucidate the potential mechanisms.

Methods and results: QRT-PCR and western blot were used to detect the expression of the miR-1, KCNE1, and KCNB2 genes after 1-week of right atrial tachypacing in New Zealand white rabbits. The AERP was measured using a programmable multichannel stimulator, and atrial fibrillation was induced by burst stimulation in vivo. The slowly activating delayed rectifier potassium current (IKs) and AERP in atrial cells were measured by whole cell patch clamp in vitro. Right atrial tachypacing upregulated miR-1 expression and downregulated KCNE1 and KCNB2 in this study, while the AERP was decreased and the atrial IKs increased. The downregulation of KCNE1 and KCNB2 levels was greater when miR-1 was further upregulated through in vivo lentiviral infection. Electrophysiological tests indicated a shorter AERP, a great increase in the IKs and a higher atrial fibrillation inducibility. In addition, similar results were found when the levels of KCNE1 and KCNB2 were downregulated by small interfering RNA while keeping miR-1 level unaltered. Conversely, knockdown of miR-1 by anti-miR-1 inhibitor oligonucleotides alleviated the downregulation of KCNE1 and KCNB2, the shortening of AERP, and the increase in the IKs. KCNE1 and KCNB2 as the target genes for miR-1 were confirmed by luciferase activity assay.

Conclusions: These results indicate that miR-1 accelerates right atrial tachypacing-induced AERP shortening by targeting potassium channel genes, which further suggests that miR-1 plays an important role in the electrical remodeling of atrial fibrillation and exhibits significant clinical relevance as a potential therapeutic target for atrial fibrillation.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. miR-1 expression.
Figure 1. miR-1 expression.
QRT-PCR quantification of miR-1 in each group (A). Immunofluorescence analysis of atrial samples from the rabbit model of right A-TP obtained 1-week after LVs infection with miR-1-LVs (B). * P < 0.05 vs. Ctl, † P < 0.05 vs. Pacing, §P < 0.05 vs. P+miR-1, one-way ANOVA and Tukey’s post-hoc test; n = 6 independent RNA samples for each group.
Figure 2. Evaluation of KCNE1 and KCNB2…
Figure 2. Evaluation of KCNE1 and KCNB2 expression.
Western blot profiles (A) and qRT-PCR (B) of KCNE1 and KCNB2 proteins levels (33 kDa and 132 kDa, respectively) and mRNAs levels, isolated from rabbits RA. * P < 0.05 vs. Ctl, † P < 0.05 vs Pacing, §P < 0.05 vs. P+miR-1. One-way ANOVA and Tukey’s post-hoc test; n = 6 independent RNA samples for each group.
Figure 3. AERP evaluations.
Figure 3. AERP evaluations.
AERP of RA obtained from animals as described in the Materials and Methods section (A). The AERP values (y axis: ms) were obtained at 3 time points: before pacing (“a”), before infection control/recombinant LVs (“b”), and 1-week after infection (“c”). * P < 0.05 vs. “a”; † P < 0.05 vs. “b”, one-way ANOVA and Tukey’s post-hoc test; n = 6 independent rabbits for each group. AERP of atrial cells were measured by whole cell patch clamp invitro (B).* P < 0.05 vs. Ctl, † P < 0.05 vs. Pacing, §P < 0.05 vs. P+miR-1, one-way ANOVA and Tukey’s post-hoc test; n = 6 independent RNA samples for each group.
Figure 4. AF inducibility evaluation.
Figure 4. AF inducibility evaluation.
The AF inducibility in each group (A). The intracardiac ECGs (B) were obtained from the following sources: (upper ECG) sinus rhythm from the no-pacing rabbits; and (middle ECG) pacing rhythm from the rabbit with pacing; and (lower ECG) AF rhythm from the rabbit with induced AF.
Figure 5. IKs evaluations.
Figure 5. IKs evaluations.
The IKs in atrial cells was measured using whole cell patch clamp invitro. * P < 0.05 Pacing vs. Ctl, † P < 0.05 P+miR-1 vs. Pacing, § P < 0.05 P+siR-KCNE1 vs. Pacing, one-way ANOVA and Tukey’s post-hoc test; n = 6 independent RNA samples for each group.
Figure 6. Sequence alignment between miR-1 and…
Figure 6. Sequence alignment between miR-1 and putative binding sites in the 3'UTRs of KCNE1 and KCNB2.
Figure 7. Luciferase assay monitoring of the…
Figure 7. Luciferase assay monitoring of the KCNE1 / KCNB2 - miR-1 interaction.
HEK293 cells were co-transfected with miR-1- or AMO-1-plasmids and KCNE1 or KCNB2- plasmids and the mutation studies by replacing the matching site in miR-1 and the 3’UTRs of KCNE1/KCNB2 were performed as described in the Materials and Methods section. Luciferase activity, measured in the samples indicated on the “X” axis, was normalized to the activity exhibited in the control cells. * P < 0.05 vs. Ctl, † P < 0.05 vs. miR-1, one-way ANOVA and the Tukey’s post-hoc test.

References

    1. Murphy NF, Simpson CR, Jhund PS, Stewart S, Kirkpatrick M et al. (2007) A national survey of the prevalence, incidence, primary care burden and treatment of atrial fibrillation in Scotland. Heart 93: 606-612. doi:10.1136/hrt.2006.107573. PubMed: .
    1. Burstein B, Nattel S (2008) Atrial fibrosis: mechanisms and clinical relevance in atrial fibrillation. J Am Coll Cardiol 51: 802-809. doi:10.1016/j.jacc.2007.09.064. PubMed: .
    1. Wang Z, Lu Y, Yang B (2011) MicroRNAs and atrial fibrillation: new fundamentals. Cardiovasc Res 89: 710-721. doi:10.1093/cvr/cvq350. PubMed: .
    1. Lee RC, Feinbaum RL, Ambros V (1993) The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell 75: 843-854. doi:10.1016/0092-8674(93)90529-Y. PubMed: .
    1. Lewis BP, Burge CB, Bartel DP (2005) Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets. Cell 120: 15-20. doi:10.1016/j.cell.2004.12.035. PubMed: .
    1. Zhao Y, Samal E, Srivastava D. (2005) Serum response factor regulates a muscle-specific microRNA that targets Hand2 during cardiogenesis. Nature 436(7048): 214-220.
    1. Liang Y, Ridzon D, Wong L, Chen C (2007) Characterization of microRNA expression profiles in normal human tissues. BMC Genomics 8: 166. doi:10.1186/1471-2164-8-166. PubMed: .
    1. Luo X, Zhang H, Xiao J, Wang Z (2010) Regulation of human cardiac ion channel genes by microRNAs: theoretical perspective and pathophysiological implications. Cell Physiol Biochem 25: 571-586. doi:10.1159/000315076. PubMed: .
    1. Yang B, Lin H, Xiao J, Lu Y, Luo X et al. (2007) The muscle-specific microRNA miR-1 regulates cardiac arrhythmogenic potential by targeting GJA1 and KCNJ2. Nat Med 13: 486-491. doi:10.1038/nm1569. PubMed: .
    1. Terentyev D, Belevych AE, Terentyeva R, Martin MM, Malana GE et al. (2009) miR-1 overexpression enhances Ca(2+) release and promotes cardiac arrhythmogenesis by targeting PP2A regulatory subunit B56alpha and causing CaMKII-dependent hyperphosphorylation of RyR2. Circ Res 104: 514-521. doi:10.1161/CIRCRESAHA.108.181651. PubMed: .
    1. Kamiyama N (1998) Expression of cell adhesion molecules and the appearance of adherent leukocytes on the left atrial endothelium with atrial fibrillation: rabbit experimental model. Jpn Circ J 62(11): 837-843. doi:10.1253/jcj.62.837. PubMed: .
    1. Ninio DM, Murphy KJ, Howe PR, Saint DA (2005) Dietary fish oil protects against stretch-induced vulnerability to atrial fibrillation in a rabbit model. J Cardiovasc Electrophysiol 16(11): 1189-1194. doi:10.1111/j.1540-8167.2005.50007.x. PubMed: .
    1. Yu J, Li W, Li Y, Zhao J, Wang L et al. (2011) Activation of beta(3)-adrenoceptor promotes rapid pacing-induced atrial electrical remodeling in rabbits. Cell Physiol Biochem 28: 87-96. doi:10.1159/000331717. PubMed: .
    1. Yu L, Scherlag BJ, Sha Y, Li S, Sharma T et al. (2012) Interactions between atrial electrical remodeling and autonomic remodeling: how to break the vicious cycle. Heart Rhythm 9(5): 804-809. doi:10.1016/j.hrthm.2011.12.023. PubMed: .
    1. Lu Y, Zhang Y, Wang N, Pan Z, Gao X et al. (2010) MicroRNA-328 contributes to adverse electrical remodeling in atrial fibrillation. Circulation 122: 2378-2387. doi:10.1161/CIRCULATIONAHA.110.958967. PubMed: .
    1. Gross GJ, Burke RP, Castle NA (1995) Characterisation of transient outward current in young human atrial myocytes. Cardiovasc Res 29: 112–117. doi:10.1016/S0008-6363(96)88555-5. PubMed: .
    1. Xiao L, Xiao J, Luo X, Lin H, Wang Z et al. (2008) Feedback Remodeling of Cardiac Potassium Current Expression A Novel Potential Mechanism for Control of Repolarization Reserve. Circulation 118(10): 983-992. doi:10.1161/CIRCULATIONAHA.107.758672. PubMed: .
    1. Workman AJ, Kane KA, Rankin AC (1999) Ionic basis of a differential effect of adenosine on refractoriness in rabbit AV nodal and atrial isolated myocytes. Cardiovasc Res 43(4): 974-984. doi:10.1016/S0008-6363(99)00166-2. PubMed: .
    1. Girmatsion Z, Biliczki P, Bonauer A, Wimmer-Greinecker G, Scherer M et al. (2009) Changes in microRNA-1 expression and IK1 up-regulation in human atrial fibrillation. Heart Rhythm 6: 1802-1809. doi:10.1016/j.hrthm.2009.08.035. PubMed: .
    1. Cooley N, Cowley MJ, Lin RC, Marasco S, Wong C et al. (2012) Influence of atrial fibrillation on microRNA expression profiles in left and right atria from patients with valvular heart disease. Physiol Genomics 44: 211-219. doi:10.1152/physiolgenomics.00111.2011. PubMed: .
    1. Yang B, Lu Y, Wang Z (2008) Control of cardiac excitability by microRNAs. Cardiovasc Res 79: 571-580. doi:10.1093/cvr/cvn181. PubMed: .
    1. Barhanin J, Lesage F, Guillemare E, Fink M, Lazdunski M et al. (1996) K(V)LQT1 and lsK (minK) proteins associate to form the I(Ks) cardiac potassium current. Nature 384(6604): 78-80. doi:10.1038/384078a0. PubMed: .
    1. Sanguinetti MC, Curran ME, Zou A, Shen J, Spector PS et al. (1996) Coassembly Coassembly K(V)LQT1 and minK (IsK) proteins to form cardiac IsK(Ks) potassium channel. Nature 384: 80-83. doi:10.1038/384080a0. PubMed: .
    1. Yan L, Figueroa DJ, Austin CP, Liu Y, Bugianesi RM et al. (2004) Expression of voltage-gated potassium channels in human and rhesus pancreatic islets. Diabetes 53: 597-607. doi:10.2337/diabetes.53.3.597. PubMed: .
    1. Heck A, Vogler C, Gschwind L, Ackermann S, Auschra B et al. (2011) Statistical epistasis and functional brain imaging support a role of voltage-gated potassium channels in human memory. PLOS ONE 6: e29337. doi:10.1371/journal.pone.0029337. PubMed: .
    1. Thibault K, Calvino B, Dubacq S, Roualle-de-Rouville M, Sordoillet V et al. (2012) Cortical effect of oxaliplatin associated with sustained neuropathic pain: Exacerbation of cortical activity and down-regulation of potassium channel expression in somatosensory cortex. Pain 153: 1636-1647. doi:10.1016/j.pain.2012.04.016. PubMed: .
    1. Xu C, Lu Y, Tang G, Wang R (1999) Expression of voltage-dependent K(+) channel genes in mesenteric artery smooth muscle cells. Am J Physiol 277: G1055-G1063. PubMed: .
    1. Frey BW, Lynch FT, Kinsella JM, Horowitz B, Sanders KM et al. (2000) Blocking of cloned and native delayed rectifier K channels from visceral smooth muscles by phencyclidine. Neurogastroenterol Motil 12: 509-516. doi:10.1046/j.1365-2982.2000.00225.x. PubMed: .
    1. Brahmajothi MV, Morales MJ, Liu S, Rasmusson RL, Campbell DL et al. (1996) In situ hybridization reveals extensive diversity of K+ channel mRNA in isolated ferret cardiac myocytes. Circ Res 78: 1083-1089. doi:10.1161/01.RES.78.6.1083. PubMed: .
    1. Temple J, Frias P, Rottman J, Yang T, Wu Y et al. (2005) Atrial fibrillation in KCNE1-null mice. Circ Res 97: 62-69. doi:10.1161/01.RES.0000173047.42236.88. PubMed: .
    1. Heerdt PM, Kant R, Hu Z, Kanda VA, Christini DJ et al. (2012) Transcriptomic analysis reveals atrial KCNE1 down-regulation following lung lobectomy. J Mol Cell Cardiol 53(3): 350-353. doi:10.1016/j.yjmcc.2012.05.010. PubMed: .
    1. Lim ST, Antonucci DE, Scannevin RH, Trimmer JS (2000) A novel targeting signal for proximal clustering of the Kv2.1 K+ channel in hippocampal neurons. Neuron 25(2): 385-397. doi:10.1016/S0896-6273(00)80902-2. PubMed: .
    1. Gelband CH, Warth JD, Mason HS, Zhu M, Moore JM et al. (1999) Angiotensin II type 1 receptor-mediated inhibition of K+ channel subunit kv2.2 in brain stem and hypothalamic neurons. Circ Res 84(3): 352-359. doi:10.1161/01.RES.84.3.352. PubMed: .

Source: PubMed

3
Suscribir