Abstract

Cav1.3 L-type Ca(2+) channel is known to be highly expressed in neurons and neuroendocrine cells. However, we have previously demonstrated that the Cav1.3 channel is also expressed in atria and pacemaking cells in the heart. The significance of the tissue-specific expression of the channel is underpinned by our previous demonstration of atrial fibrillation in a Cav1.3 null mutant mouse model. Indeed, a recent study has confirmed the critical roles of Cav1.3 in the human heart (Baig, S. M., Koschak, A., Lieb, A., Gebhart, M., Dafinger, C., Nürnberg, G., Ali, A., Ahmad, I., Sinnegger-Brauns, M. J., Brandt, N., Engel, J., Mangoni, M. E., Farooq, M., Khan, H. U., Nürnberg, P., Striessnig, J., and Bolz, H. J. (2011) Nat. Neurosci. 14, 77-84). These studies suggest that detailed knowledge of Cav1.3 may have broad therapeutic ramifications in the treatment of cardiac arrhythmias. Here, we tested the hypothesis that there is a functional cross-talk between the Cav1.3 channel and a small conductance Ca(2+)-activated K(+) channel (SK2), which we have documented to be highly expressed in human and mouse atrial myocytes. Specifically, we tested the hypothesis that the C terminus of Cav1.3 may translocate to the nucleus where it functions as a transcriptional factor. Here, we reported for the first time that the C terminus of Cav1.3 translocates to the nucleus where it functions as a transcriptional regulator to modulate the function of Ca(2+)-activated K(+) channels in atrial myocytes. Nuclear translocation of the C-terminal domain of Cav1.3 is directly regulated by intracellular Ca(2+). Utilizing a Cav1.3 null mutant mouse model, we demonstrate that ablation of Cav1.3 results in a decrease in the protein expression of myosin light chain 2, which interacts and increases the membrane localization of SK2 channels.

Highlights

  • Cav1.3 Ca2ϩ channel is highly expressed in atria and pacemaking cells in the heart

  • Ablation of Cav1.3 Ca2ϩ Channels in Mouse Atrial Myocytes Results in Abnormal Surface Membrane Localization of SK2 Channels—Our previous work has demonstrated that null deletion of the Cav1.3 channel results in abnormal function of SK2 channels and prolongation of atrial repolarization and atrial arrhythmias [1, 17]

  • We further demonstrated the successful isolation of green fluorescence protein (GFP)-positive atrial myocytes in Fig. 8B using fluorescence-activated cell sorting (FACS)

Read more

Summary

Background

Cav1.3 Ca2ϩ channel is highly expressed in atria and pacemaking cells in the heart. Results: The C terminus of the Cav1.3 Ca2ϩ channel can translocate into the nucleus. The importance of the Cav1.3 isoform is underscored by the findings that Cav1.3 null mutant mice (Cav1.3Ϫ/Ϫ) show significant alteration in atrial excitability and atrial fibrillation, as well as sinoatrial and atrioventricular node dysfunction [1,2,3,4,5] These observations were surprising, yet insightful as they suggest that detailed knowledge of the different Ca2ϩ channel isoforms may have broad therapeutic ramifications in the treatment of atrial arrhythmias. Using a combination of techniques including yeast two-hybrid (Y2H) assays, biochemical analyses, immunofluorescence confocal microscopic imaging, microarray analyses, and in vivo siRNA-mediated gene silencing, we report for the first time that the C terminus of Cav1.3 translocates to the nucleus where it functions as a transcription factor to regulate the expression of SK2 channel interacting proteins

EXPERIMENTAL PROCEDURES
The abbreviations used are
RESULTS
DISCUSSION
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call