Abstract

When cells swell in hypo-osmotic solutions, chloride-selective ion channels (Clswell) activate to reduce intracellular osmolality and prevent catastrophic cell rupture. Despite intensive efforts to assign a molecular identity to the mammalian Clswell channel, it remains unknown. In an unbiased genome-wide RNA interference (RNAi) screen of Drosophila cells stably expressing an anion-sensitive fluorescent indicator, we identify Bestrophin 1 (dBest1) as the Drosophila Clswell channel. Of the 23 screen hits with mammalian homologs and predicted transmembrane domains, only RNAi specifically targeting dBest1 eliminated the Clswell current (IClswell). We further demonstrate the essential contribution of dBest1 to Drosophila IClswell with the introduction of a human Bestrophin disease-associated mutation (W94C). Overexpression of the W94C construct in Drosophila cells significantly reduced the endogenous IClswell. We confirm that exogenous expression of dBest1 alone in human embryonic kidney (HEK293) cells creates a clearly identifiable Drosophila–like IClswell. In contrast, activation of mouse Bestrophin 2 (mBest2), the closest mammalian ortholog of dBest1, is swell-insensitive. The first 64 residues of dBest1 conferred swell activation to mBest2. The chimera, however, maintains mBest2-like pore properties, strongly indicating that the Bestrophin protein forms the Clswell channel itself rather than functioning as an essential auxiliary subunit. dBest1 is an anion channel clearly responsive to swell; this activation depends upon its N-terminus.

Highlights

  • All mammalian cells express chloride channels activated by decreases in extracellular osmolality, albeit with different biophysical properties [1]

  • We confirmed that each RNA interference (RNAi) significantly reduced swelling-induced fluorescence and targeted only the mRNA from the identified gene

  • D308A is proposed to eliminate Bestrophin activation by disruption of calcium binding [39]. We introduced this mutation into dBest1 to determine if activation by calcium and cell swelling could be separated

Read more

Summary

Introduction

All mammalian cells express chloride channels activated by decreases in extracellular osmolality, albeit with different biophysical properties [1]. De-regulated constitutively active Clswell channels exacerbate several cardiac diseases, including myocardial hypertrophy and heart failure [2]. The mammalian Clswell channel- encoding gene has yet to be identified despite the wealth of proteins nominated by candidate approaches [3]. These proteins include ClC-2 [4], ClC-3 [5], P-glycoprotein [6,7], pICln [8,9], p64 [10], phospholemman [11], Best and 2 [12], TMEM16A [13], and TMEM16F [14]. The research community has yet to agree on any of these candidates as a bona fide Clswell channel

Methods
Results
Conclusion
Full Text
Paper version not known

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

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.