Advanced Search

Submit Manuscript

ADVANCE ONLINE PUBLICATION  |  Open Access

ORIGINAL ARTICLES

Disruption of ER ion homeostasis maintained by an ER anion channel CLCC1 contributes to ALS-like pathologies

Liang Guo1,2,3,4,† , Qionglei Mao5,6,† , Ji He7,† , Xiaoling Liu4,8 , Xuejiao Piao1,2,3,4 , Li Luo3,9 , 5,10,11,* , Hanzhi Yu2 , Qiang Song3 , Bailong Xiao1,4,8 , Dongsheng Fan7,12,* , Zhaobing Gao5,6,* , Yichang Jia1,3,4,9,*

1Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, Beijing, China
2School of Life Sciences, Tsinghua University, Beijing, China
3School of Medicine, Tsinghua University, Beijing, China
4IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China
5CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia and Medica, Chinese Academy of Sciences, Shanghai, China.
6University of Chinese Academy of Sciences, Beijing, China
7Department of Neurology, Peking University Third Hospital, Beijing, China
8School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
9Tsinghua Laboratory of Brain and Intelligence, Beijing, China
10School of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
11School of Medicine, Zhejiang University City College, Hangzhou, Zhejiang, China
12Beijing Municipal Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases, Beijing, China
These authors contributed equally: Liang Guo, Qionglei Mao, Ji He
* Correspondence: ()Dongsheng Fan(dsfan2010@aliyun.com)Zhaobing Gao(zbgao@simm.ac.cn)Yichang Jia(yichangjia@tsinghua.edu.cn)

Although anion channel activities have been demonstrated in sarcoplasmic reticulum/endoplasmic reticulum (SR/ER), their molecular identities and functions remain unclear. Here, we link rare variants of Chloride Channel CLIC Like 1 (CLCC1) to amyotrophic lateral sclerosis (ALS)-like pathologies. We demonstrate that CLCC1 is a pore-forming component of an ER anion channel and that ALS-associated mutations impair channel conductance. CLCC1 forms homomultimers and its channel activity is inhibited by luminal Ca2+ but facilitated by phosphatidylinositol 4,5-bisphosphate (PIP2). We identified conserved residues D25 and D181 in CLCC1 N-terminus responsible for Ca2+ binding and luminal Ca2+-mediated inhibition on channel open probability and K298 in CLCC1 intraluminal loop as the critical PIP2-sensing residue. CLCC1 maintains steady-state [Cl]ER and [K+]ER and ER morphology and regulates ER Ca2+ homeostasis, including internal Ca2+ release and steady-state [Ca2+]ER. ALS-associated mutant forms of CLCC1 increase steady-state [Cl]ER and impair ER Ca2+ homeostasis, and animals with the ALS-associated mutations are sensitized to stress challenge-induced protein misfolding. Phenotypic comparisons of multiple Clcc1 loss-of-function alleles, including ALS-associated mutations, reveal a CLCC1 dosage dependence in the severity of disease phenotypes in vivo. Similar to CLCC1 rare variations dominant in ALS, 10% of K298A heterozygous mice developed ALS-like symptoms, pointing to a mechanism of channelopathy dominant-negatively induced by a loss-of-function mutation. Conditional knockout of Clcc1 cell-autono

https://doi.org/10.1038/s41422-023-00798-z

FULL TEXT | PDF

Browse 210