bioRxiv preprint doi: https://doi.org/10.1101/331207; this version posted May 25, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

Cryo-EM structure of the volume-regulated anion channel

LRRC8

Go Kasuya1*, Takanori Nakane1†*, Takeshi Yokoyama2*, Yanyan Jia3, Masato

Inoue4, Kengo Watanabe4, Ryoki Nakamura1, Tomohiro Nishizawa1, Tsukasa

Kusakizako1, Akihisa Tsutsumi5, Haruaki Yanagisawa5, Naoshi Dohmae6,

Motoyuki Hattori7, Hidenori Ichijo4, Zhiqiang Yan3, Masahide Kikkawa5, Mikako

Shirouzu2, Ryuichiro Ishitani1, Osamu Nureki1‡

1Department of Biological Sciences, Graduate School of Science, The University of Tokyo, 2-11-16 Yayoi, Bunkyo-ku, Tokyo 113-0032, Japan; 2Division of Structural and Synthetic Biology, RIKEN Center for Life Science Technologies, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama-shi, Kanagawa 230-0045, Japan; 3State Key Laboratory of Medical Neurobiology, Ministry of Education Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center of Genetics and Development, Department of Physiology and Biophysics, School of Life Sciences, Fudan University, 2005 Songhu Road, Yangpu District, Shanghai 200438, China; 4Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan; 5Department of Cell Biology and Anatomy, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan; 6RIKEN, Global Research Cluster, 2-1 Hirosawa, Wako-shi, Saitama 351-0198, Japan; 7State Key Laboratory of Genetic Engineering, Collaborative Innovation Center of Genetics and Development, Department of Physiology and Biophysics, School of Life Sciences, Fudan University, 2005 Songhu Road, Yangpu District, Shanghai 200438, China. * These authors contributed equally to this work. † Present address: MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge, CB2 0QH, United Kingdom ‡Correspondence and requests for materials should be addressed to O.N. ([email protected])

— 1 — bioRxiv preprint doi: https://doi.org/10.1101/331207; this version posted May 25, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

Abstract

Maintenance of cell volume against osmotic change is crucial for proper cell functions,

such as cell proliferation and migration. The leucine-rich repeat-containing 8 (LRRC8)

are anion selective channels, and were recently identified as pore components

of the volume-regulated anion channels (VRACs), which extrude anions to decrease the

cell volume upon cell-swelling. Here, we present the human LRRC8A structure,

determined by a single-particle cryo-electron microscopy analysis. The sea

anemone-like structure represents a trimer of dimers assembly, rather than a

symmetrical hexameric assembly. The four-spanning transmembrane region has a gap

junction channel-like membrane topology, while the LRR region containing 15

leucine-rich repeats forms a long twisted arc. The channel pore is along the central axis

and constricted on the extracellular side, where the highly conserved polar and charged

residues at the tip of the extracellular helix contribute to the anion and other osmolyte

permeability. Comparing the two structural populations facilitated the identification of

both compact and relaxed conformations, suggesting that the LRR region is flexible and

mobile with rigid-body motions, which might be implicated in structural transitions

upon pore opening. Overall, our structure provides a framework for understanding the

molecular mechanisms of this unique class of ion channels.

— 2 — bioRxiv preprint doi: https://doi.org/10.1101/331207; this version posted May 25, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

Introduction

Cell volume regulation in response to changes in the extracellular osmotic pressure is a

fundamental homeostatic mechanism for all organisms1–3. Alteration of the cell volume

leads to numerous physiological processes, such as cell proliferation and migration. A

continuous decrease in the cell volume under isotonic conditions is related to the

activation of the programmed cell death pathway, and is known as apoptotic volume

decrease (AVD)4. Various proteins including membrane ion channels and transporters,

as well as cytoplasmic kinases, are essential components for cell volume regulation.

Among these proteins, the volume regulated anion channel (VRAC) is a particularly

interesting type of ion channel5. VRAC is activated by cell-swelling and releases Cl‒

ions or other osmolytes on the extracellular side and concurrently ejects water

molecules, leading to a decrease in the cell volume. This process, called regulatory

volume decrease (RVD), is observed in almost all types of eukaryotic cells.

The molecular entity of VRAC was unknown until quite recently, although extensive

experiments have been performed over the past decades, and several characteristics of

VRAC, including substrate selectivity, activation upon cell-swelling, outward

rectification, and inactivation kinetics, were functionally characterized6,7. In 2014, by

combining high-throughput RNAi screening with electrophysiological recordings, two

independent groups successfully identified the leucine-rich repeat-containing 8A

(LRRC8A) , which when mutated causes an innate immune disease,

agammaglobulinemia8, as the essential component of VRAC9,10. The LRRC8A protein

belongs to the LRRC8 family, which includes five members (LRRC8A-E) with

high amino-acid sequence similarity. All of the LRRC8 isoforms consist of about

800-850 amino acid residues11,12. Based on a sequence analysis, the N-terminal half of

— 3 — bioRxiv preprint doi: https://doi.org/10.1101/331207; this version posted May 25, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

the LRRC8 isoforms is predicted to form four transmembrane (TM) helices, while the

C-terminal half of the LRRC8 isoforms is predicted to contain up to 17 leucine-rich

repeats (LRR). A previous bioinformatics analysis suggested that the N-terminal halves

of the LRRC8 isoforms share weak homology to , , and innexin13.

Since the previous structural analyses of the channels, connexin and

, revealed that their hemichannels form a hexamer and an octamer,

respectively14–16, the LRRC8 proteins are also predicted to form hexameric or higher

TM region oligomers to create the channel pore13. Further electrophysiological analyses,

using cells expressing LRRC8 isoforms and LRRC8 isoforms embedded in lipid

bilayers, demonstrated that the heteromeric LRRC8 complex, consisting of LRRC8A

and at least one of the four other LRRC8 isoforms, is indispensable for the channel

activity in cells, while the homomeric LRRC8A protein still retains the hypotonically

induced channel activity under lipid embedded conditions10,17. This heteromeric

assembly of LRRC8A and the other LRRC8 isoforms contributes to the diversity of the

channel properties, including the gating kinetics and modulation, as well as the substrate

specificity. For example, cells expressing the LRRC8A/C heteromer are inactivated

more slowly than those expressing the LRRC8A/E heteromer10. In addition, the activity

of the LRRC8A/E heteromer is potentiated by intracellular oxidation, while the

activities of the LRRC8A/C and LRRC8A/D heteromers are inhibited by intracellular

oxidation18. Moreover, cells expressing the LRRC8A/D heteromer release uncharged

osmolytes such as taurine, and uptake anti-cancer drugs, including cisplatin, while those

expressing the LRRC8A/C/E heteromer release charged organic osmolytes19–21.

As the overall sequence of LRRC8 has little similarity to any other known classes of

ion channels, the detailed structural information of the LRRC8 protein has long been

— 4 — bioRxiv preprint doi: https://doi.org/10.1101/331207; this version posted May 25, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

awaited to elucidate the molecular architecture of LRRC8 and to clarify the molecular

mechanisms, such as substrate permeability. Here, we report the cryo-electron

microscopy (cryo-EM) structure of the homomeric human LRRC8A, at an average

resolution of 4.25 Å (local resolution of the TM region is ~3.8 Å). The present structure

provides a framework for understanding the mechanisms of this unique class of ion

channels.

— 5 — bioRxiv preprint doi: https://doi.org/10.1101/331207; this version posted May 25, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

Results

Structure determination of homomeric HsLRRC8A.

We first screened the LRRC8 proteins from various vertebrates by

Fluorescence-detection Size-Exclusion Chromatography (FSEC) and an FSEC-based

thermostability assay (FSEC-TS)22,23, and found that the human LRRC8A protein

(HsLRRC8A; the “Hs” refers to Homo sapiens), the best characterized protein among

the known LRRC8 proteins, produced symmetric and monodisperse FSEC peaks when

solubilized with digitonin (Extended Data Fig. 1a). We also confirmed the

hypotonicity-induced channel activity of the purified HsLRRC8A protein in

reconstituted liposomes (Extended Data Fig. 1b,c). Although the LRRC8 proteins

reportedly function as heteromers in cells10, the structure of the homomeric HsLRRC8A

protein may provide important insights into the basic architecture of the LRRC8

proteins, given the sequence similarity between the isoforms (Extended Data Fig. 2).

Therefore, we set out to elucidate the structure of the homomeric HsLRRC8A. The

purified HsLRRC8A protein was vitrified on grids, and images of the grids were

recorded by electron microscopy (Extended Data Fig. 3a). In the 3D classification step,

classes #1 and #2 showed clearer densities and similar assemblies, respectively, as

compared to the other classes. Therefore, we chose these two classes for further

refinement (Extended Data Fig. 3b,c). Since these classes exhibited 3-fold rotational

symmetry, C3 symmetry was imposed in the subsequent reconstructions. In the final 3D

map reconstructed to an overall 4.25 Å resolution (local resolution of the TM region is

~3.8 Å) (Extended Data Fig. 3d,e), the densities of the six protomers of HsLRRC8A

(namely, the α to ζ subunits) were clearly observed, including the long arc-like densities

corresponding to the leucine-rich repeat (LRR) region. This density map enabled us to

— 6 — bioRxiv preprint doi: https://doi.org/10.1101/331207; this version posted May 25, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

assign the topologies of the LRR and other regions, including the extracellular,

transmembrane (TM), and intracellular regions (Fig. 1a-c, Extended Data Fig. 3b, c).

The local resolution of the core of the TM region is estimated to be ~3.8 Å, and its

density is sufficiently resolved to enable the tracing of the α-helices and the bulky

amino-acid side chains (Fig. 1a, b). The densities corresponding to most of the

extracellular and intracellular regions are also resolved well enough to trace the

secondary structures, using the structure of the previously determined nematode Innexin

6 (CeInnexin-6; the “Ce” refers to Caenorhabditis elegans) (PDB ID: 5H1Q) as a guide.

Two loops connecting the TM helices in these regions are disordered (Fig. 1d, e). In

contrast to the clear densities of the extracellular, TM and intracellular regions, the local

resolution of the LRR regions is around 5.5 Å, and only the main chain of the LRR

region is resolved in the density map (Fig. 1a, b). Therefore, we generated the

homology model of the LRR region of HsLRRC8A based on the previously determined

LRR protein structure (PDB ID: 4U08) using the Phyre2 server24, and used it to guide

the model building. The final model of HsLRRC8A, in which about 90% of the residues

were assigned, contains the residues R18-D806 and is missing parts of the extracellular

loop (T61-D91) and the intracellular loop (T176-R226 in the α, γ, and ε subunits;

T176-L232 in the β, δ, and ζ subunits).

Overall structure and subunit fold of HsLRRC8A.

The hexameric structure of HsLRRC8A adopts an upside-down sea anemone-like

shape, when viewed parallel to the membrane (Fig. 1c). The subunit structure consists

of four regions; i.e., the extracellular region (Q49-H119 and H287-L314), TM region

(W23-T48, W120-F144, I259-V286, and A315-R345), intracellular region (K145-D258

— 7 — bioRxiv preprint doi: https://doi.org/10.1101/331207; this version posted May 25, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

and R346-T412), and LRR region (L413-D806) (Fig. 1c-e), with both the N- and

C-termini residing on the cytoplasmic side. The overall structures of the six subunits are

similar, except for several points as described later, and thus we mainly discuss the

structure of the α subunit of the hexamer. The extracellular region protrudes by ~35Å

above the cell membrane, and forms the channel pore with the TM region. The LRR

region is protruded toward the intracellular side, and is twisted in a clockwise manner as

viewed from the cytoplasmic side (Fig. 1c). The TM region consists of the four TM

helices (TM1-4), which are connected by the two extracellular loops (EL1 and EL2) of

the extracellular region, and the intracellular loop (IL1) of the intracellular region (Fig.

1d, e). IL2 in the intracellular region connects the TM and LRR regions (Fig. 1d, e).

EL1 possesses one α-helix (EL1H) and one β-strand (EL1β), while EL2 possesses two

β-strands (EL2β1 and EL2β2) (Fig. 1d, e). IL1 possesses three α-helices (IL1H1–3),

while IL2 possesses four α-helices (IL2H1–4) (Fig. 1d, e). EL1 and IL1 contain

disordered regions (T61–D91 and T176–R226), suggesting that these loops have

conformational flexibility (Fig. 1d, e, Extended Data Fig. 2). While a previous

bioinformatics analysis suggested that LRRC8 contains a 17 LRR repeat12, the present

structure revealed that the LRR region consists of the leucine-rich repeat N-terminal

helix (LRRNT), fifteen leucine-rich repeats (LRR1–15), and the following C-terminal

helices (LRRCT) (Fig. 1d, e, Extended Data Fig. 2).

Differences in structures and interactions among HsLRRC8A subunits.

With its global C3 symmetry, the hexameric HsLRRC8A structure exhibits the “trimer

of dimers” architecture; in other words, the structures of the α, γ, and ε subunits, and

those of the β, δ, and ζ subunits, are identical (Fig. 1c, 2a). In contrast, a distinctive

— 8 — bioRxiv preprint doi: https://doi.org/10.1101/331207; this version posted May 25, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

structural difference is observed between the α and β subunits. The LRR region is

rotated by ~22° relative to the extracellular, TM, and intracellular regions (Fig. 2b). No

large structural difference is observed in each region, as their structures superimpose

well with RMSD values less than 1 Å (0.39 Å, 0.40 Å, 0.98 Å, and 0.47 Å for the

extracellular, TM, intracellular, and LRR regions, respectively), suggesting their

structural rigidity (Fig. 2b, Extended Data Fig. 4). Therefore, this structural difference

between the subunits can be ascribed to the rotation of the IL2H4 helix in the IL2 loop,

which connects the intracellular and LRR regions as a hinge (Fig. 2c).

As a result of this structural difference between the subunits, several variations are

observed in the interfaces between the subunits; e.g., the α subunit tightly interacts with

the β subunit, while it loosely interacts with the ζ subunit (Fig. 2a). Notably, in the

“tight” interaction, there are extensive contacts between the adjacent subunits from the

extracellular region to the LRR region, whereas in the “loose” interaction, there are

almost no contacts between the adjacent subunits in the lower part of the TM region, the

intracellular region, and the upper part of the LRR region (Fig. 2a). At the TM region,

the subunit interface is highly hydrophobic and mainly formed by the TM2 helix from

one subunit, and the TM1 and TM4 helices from the other adjacent subunit (Extended

Data Fig. 5a, b). There is a “lateral crevice” open to the lipid bilayer across the central

channel pore, and it is wider in the loose interface than in the tight interface (Extended

Data Fig. 5a, b). At the intracellular region, the subunit interface is highly hydrophilic

and formed by the IL1H1 and IL1H3 helices from one subunit, and the IL1H2, IL2H2

and IL2H3 helices from the other adjacent subunit (Extended Data Fig. 5c, d). In this

region, extensive interactions are formed in the tight interface, while few interactions

are observed in the loose interface (Extended Data Fig. 5c, d). At the LRR region,

— 9 — bioRxiv preprint doi: https://doi.org/10.1101/331207; this version posted May 25, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

extensive interactions throughout all of the LRR repeats are observed in the tight

interface, while in the loose interface, fewer interactions are formed between the

C-terminal LRRCT helices from one subunit and the middle of the LRR repeats from

the other adjacent subunit (Extended Data Fig. 5e, f). Notably, a heterozygous LRRC8A

truncation mutant, in which the C-terminal 91 amino acids are replaced by 35 amino

acids encoded in the intron sequences, has been reported to suppress the LRRC8A

function, which causes an innate immune disease8,9. This disease-related truncation

might abrogate the interactions at this loose interface, thereby affecting the proper

formation of the hexameric channel structure. In contrast, the interactions in the tight

and loose interfaces of the extracellular region are almost the same (Extended Data Fig.

5g, h). The interactions are formed by the extensive contacts between the EL1H helix

and the three-stranded β-sheet (EL1β, EL2β1, and EL2β2) from the other adjacent

subunit (Extended Data Fig. 5g, h).

Overall, these structural comparisons between the subunits suggest that the flexibility

around the IL2H4 helix, which connects the intracellular and LRR regions, causes the

structural differences between the subunits, which may enable the formation of the

“trimer of dimers” architecture of the hexameric channel, with the wide crevice in the

loose interface.

Channel pore

HsLRRC8A has a channel pore along the central axis, perpendicular to the membrane.

The channel pore is mainly composed of the EL1H helix in the extracellular region and

the TM1 helix in the TM region, as well as the IL1H1 and IL1H3 helices in the

intracellular region (Fig. 3a). The diameter of the pore is about ~ 25 Å on the

— 10 — bioRxiv preprint doi: https://doi.org/10.1101/331207; this version posted May 25, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

intracellular side, and it widens to about 50 Å at the lower part of the TM region, and

then narrows to less than 10 Å at the upper part of the extracellular region (Fig. 3b-e).

Therefore, the channel pore formed by the hexameric LRRC8A is accessible from both

the extracellular and intracellular sides, and thus the present structure of LRRC8A

probably represents the open conformation. The residues lining the channel pore are

mainly hydrophilic, suggesting that the pore is filled with water molecules in the present

structure. A previous experiment revealed that the T44C mutation alters the I– versus

Cl– anion selectivity of HsLRRC8A, and the MTSES (2-sulfonatoethyl

methanethiosulfonate) modification of the T44C mutant significantly reduced the

channel activity, suggesting that T44 is involved in the pore formation9. Consistently, in

our HsLRRC8A structure, T44 is located on TM1 and involved in the formation of the

solvent-accessible surface (Fig. 3b, d). The most-constricted site of the pore has a

diameter of about 7.6 Å, which is wide enough to permeate a hydrated Cl– ion. This

constriction site is mainly formed by the side chains of the positively charged R103

residues (Fig. 3b, c). In addition, a negatively charged residue, D102, is located next to

R103, and is positioned just above R103 like a “lid” for the constriction site. These

structural features suggest that the D102 and R103 residues play an important role in the

ion permeation (Fig. 3b, c). Furthermore, a recent electrophysiological analysis

demonstrated that three residues, corresponding to K98, Y99, and D100 in the LRRC8A

isoform, are involved in the voltage dependence and kinetics of the LRRC8 isoforms25.

Single and combination mutants of these residues reportedly affected the rate and

voltage dependence of inactivation, the kinetics of inactivation, and the ion selectivity25.

Consistently, these three residues are located in the loop preceding the EL1H helix and

are exposed to the extracellular entrance of the channel pore in the present HsLRRC8A

— 11 — bioRxiv preprint doi: https://doi.org/10.1101/331207; this version posted May 25, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

structure (Fig. 4a), suggesting their importance for the ion permeation.

Based on the present HsLRRC8A structure, as well as the previous functional analysis,

we hypothesized that the N-terminal tip of the EL1H helix (D102-Y106) plays a critical

role in the ion permeation, as it resides on the extracellular side of the channel pore and

constitutes its most-constricted site (Fig. 4a,b). To corroborate this notion, we generated

a series of single alanine mutants of residues D102 to Y106 in HsLRRC8A and

performed cell-based measurements of the VRAC activity, using assays originally

developed upon the discovery of the LRRC8A protein as a VRAC component9,10. First,

by a confocal microscopic analysis, we confirmed that the signals derived from these

mutants are observed at the cell membrane (Extended Data Fig. 6a). In addition, the

FSEC analysis of these mutants produced symmetric and monodisperse peaks, similar

to those of the wild-type channel, confirming the proper structural integrities of these

mutant channels (Extended Data Fig. 6b). We transfected the halide-sensitive YFP

mutant26 with or without HsLRRC8A wild-type (WT) into HEK293A cells, in which

the endogenous LRRC8A gene is knocked out, and verified the assay system:

HsLRRC8A WT could rescue the hypotonicity-induced iodide quenching response in

the LRRC8A KO cells (Fig. 4c, d). We then evaluated each of the HsLRRC8A mutants

within the range of WT expression levels (Extended Data Fig. 6c), and found that the

D102A, Q105A and Y106A mutants decreased the quenching speed as compared to the

wild-type channel (Fig. 4c, d, Extended Data Fig. 6d). Even though the reduction of the

quenching speed cannot exactly explain how the mutations affect the channel properties,

such as the rate of ion permeation and the channel open probability, the results support

our hypothesis that the N-terminal tip of the EL1H helix is important for the ion

permeation. Interestingly, the amino-acid sequences in this region of the other LRRC8

— 12 — bioRxiv preprint doi: https://doi.org/10.1101/331207; this version posted May 25, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

isoforms deviate from that of LRRC8A (Fig. 4b). These differences in the amino acid

sequences in this region may alter the properties of this constriction site in the channel

pore, thereby modulating the substrate specificity of the heteromeric LRRC8 hexamers

containing different isoforms20,21.

Putative structural change of LRRC8

During the 3D classification, in addition to classes #1 and #2 used for the 4.25 Å

reconstruction, we found that class #7 also showed clearer LRR densities than the other

classes (Extended Data Fig. 3b). Interestingly, class #7 exhibited the distorted

arrangement of the LRR densities, as compared to those in classes #1 and #2, and thus it

may represent another structural population of LRRC8A. Therefore, we performed the

3D reconstruction of class #7 without imposing any symmetry. The overall resolution of

the resulting map was 9.17 Å, and we could assign the extracellular, TM, intracellular,

and LRR regions. We then constructed a model of class #7, based on the higher

resolution structure.

In this model constructed from class #7, the LRR region is substantially deviated and

more loosely packed as compared to that in the high-resolution structure from classes #1

and #2 (Fig. 5a, b). Accordingly, we refer to this class #7 structure as the “relaxed form”

(Fig. 5a), and the higher-resolution structure from classes #1 and #2 as the “compact

form” (Fig. 5b). In the relaxed form, the LRR regions are further tilted and expanded

away from the central axis, with their width elongated by ~20 Å and height shortened

by ~8 Å, as compared with the compact form (Fig. 5a, b). The C3 symmetry observed in

the compact form is absent and the arrangement of the LRR regions is completely

asymmetric. In contrast, the arrangement of the secondary structure elements in the

— 13 — bioRxiv preprint doi: https://doi.org/10.1101/331207; this version posted May 25, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

extracellular, TM, and intracellular regions is quite similar to that observed in the

compact form. The structures of the LRR regions in both the relaxed and compact forms

were also similar, including their curvatures. Thus, the overall structural difference

between the relaxed and compact forms can be explained by the rigid-body motions of

the LRR and the other regions, in which the IL2H4 helix in the IL2 loop serves as a

hinge. Interestingly, a similar structural difference is also observed between the α and β

subunits in the compact form (Fig. 2, Extended Data Fig. 4). These structural

differences suggest the structural flexibility around the IL2H4 helix in the IL2 loop,

which might be involved in the important structural transition for the channel function

(Fig. 5c).

Structural comparison with gap junction channels

Consistent with the previous bioinformatics analysis13, the extracellular, TM, and

intracellular regions of HsLRRC8A share several structural features with those of the

CeInnexin-6 and human connexin 26 (HsConnexin-26) gap junction channels (Extended

Data Fig. 7)14,15, although they have low sequence identity (~15% with CeInnexin-6;

~18% with HsConnexin-26). In the TM region, they share the same topology of four

TM helices, in which TM1 is the innermost helix. In the extracellular region, they

commonly possess conserved disulfide bonds between the EL1 and EL2 loops, and

common secondary structure elements, including helix EL1H, as well as the β-sheet

bridging the EL1 and EL2 loops (Extended Data Fig. 7a-c). In the intracellular region,

helices IL1H1 and IL1H3 in the IL1 loop are also conserved in CeInnexin-6 (Extended

Data Fig. 7a, b). In contrast, HsLRRC8A possesses several distinct structural features as

compared to the gap junction channels. First, the subunit interfaces of HsConnexin-26

— 14 — bioRxiv preprint doi: https://doi.org/10.1101/331207; this version posted May 25, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

and CeInnexin-6 are tightly packed and the large crevice is not observed (Extended

Data Fig. 7d-f)14,15, suggesting that the crevice of LRRC8 is involved in its specific

function. Second, the HsConnexin-26 and CeInnexin-6 structures revealed that their

short N-terminal extension (~15 residues) forms a short α helix (NTH), which is

involved in the channel pore formation (Extended Data Fig. 7g-i). This NTH was

speculated to be important for the ion permeation and gating in the gap junction

channels14,15. HsLRRC8A also possesses a short extension at its N terminus; however,

its density was not observed in the present cryo-EM structure. The amino-acid sequence

of the N-terminal extension of HsLRRC8A is quite different from those of both

HsConnexin-26 and CeInnexin-6, and is not predicted to form a helix, unlike the cases

of HsConnexin-26 and CeInnexin-6 (Extended Data Fig. 7j). Thus, the N-terminal

extension of LRRC8 would not form a helix and is likely to play a different role from

those of the gap junction channels. Third, in the TM region, while TM1 in HsLRRC8A

is straight and faces toward the intracellular side, those in the gap junction channels are

bent at the middle, and consequently the N-termini of the gap junction channels face

toward the extracellular side to form the channel pore (Extended Data Fig. 7b, c).

Fourth, in the intracellular region, the IL1 and IL2 loops of HsLRRC8A (144 residues

in IL1; 67 residues in IL2) are longer than those of CeInnexin-6 (54 residues in IL1; 59

residues in IL2) (Extended Data Fig. 7a, b). As a result, in HsLRRC8A, the long linker

between the IL1H2 and IL1H3 helices in IL1, as well as the IL2H4 helix in IL2, directly

interacts with the LRR region. Particularly, the IL2H4 helix, which connects the

intracellular and LRR regions, only exists in the LRRC8 isoforms. Fifth, in the

extracellular region, EL1H in HsLRRC8A (14 residues) is longer than those in the gap

junction channels (10 residues), and the N-terminal tip of the EL1H helix in

— 15 — bioRxiv preprint doi: https://doi.org/10.1101/331207; this version posted May 25, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

HsLRRC8A forms the channel pore with the TM1 helix (Extended Data Fig. 7a-c, g-i).

In addition, the sizes and structures of the EL1 and EL2 loops of the gap junction

channels are different from those of HsLRRC8A (Extended Data Fig. 7a-c). Indeed, the

extracellular regions of HsConnexin-26 and CeInnexin-6 were shown to interact with

each other to form the gap junction channel, in which the EL1 and EL2 loops provide

the interaction interface14,15. Therefore, these structural differences in the subunit

interfaces, as well as the extracellular, TM, and intracellular regions, contribute to the

functional diversity between the volume-regulated channels and the gap junction

channels.

Discussion

In this study, we present the homomeric human LRRC8A cryo-EM structure, the first

structure of a member of the LRRC8 protein family responsible for the VRAC function.

The overall structure, reconstructed at 4.25 Å resolution (~3.8 Å in the TM region),

exhibited a hexameric assembly with a trimer of dimers, rather than a symmetrical

hexamer, revealing the architecture of this unique class of ion channels (Fig. 1) The

channel pore observed along the central axis is open toward both the extra- and

intracellular sides, suggesting that the present structure represents the channel-open

form (Fig. 3).

Although LRRC8A is an essential component of the VRAC activity, overexpression of

the LRRC8A gene in the quintuple-knockout LRRC8−/− cells or quadruple-knockout

LRRC8(B/C/D/E)−/− cells did not rescue the VRAC activity, suggesting that the

homomer of LRRC8A does not form the active VRAC pore in vivo. Nevertheless,

interestingly, the homomer of LRRC8A reconstituted into lipid droplet bilayers still

— 16 — bioRxiv preprint doi: https://doi.org/10.1101/331207; this version posted May 25, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

retains VRAC characteristics, including hypotonicity-induced currents and sensitivity to

a specific VRAC inhibitor, DCPIB

(4-(2-butyl-6,7-dichlor-2-cyclopentyl-indan-1-on-5-yl) oxybutyric acid)17. We also

confirmed that the homomeric HsLRRC8A protein used in this study has

hypotonicity-induced channel activity upon osmotic change, using the reconstituted

liposomes (Extended Data Fig. 1b,c). Furthermore, a previous biochemical experiment

showed that the overall assembly, including the oligomeric state, of the LRRC8A

homomer is similar to those of the LRRC8A-containing heteromers. The present

structure of the homomeric HsLRRC8A provides important structural insights into the

LRRC8A-containing heteromers. To visualize the conserved and diversified sites

among the LRRC8 gene family members, we calculated the conservation score using

the program ConSurf27, and mapped the score on the molecular surface of HsLRRC8A

(Extended Data Fig. 8a). The result showed that the subunit interface and the channel

pore in the TM, extracellular, and intracellular regions, except for the constriction site,

are highly conserved among the LRRC8 gene family members. In contrast, the

membrane side of the TM region, the overall LRR region, and the constriction site of

the pore exhibited relatively low conservation scores. Next, based on the present

structure of HsLRRC8A, we constructed a model structure of the HsLRRC8 heteromer

containing HsLRRC8D (54% sequence identity with HsLLRC8A; Extended Data Fig.

8b). Although the stoichiometry of the heteromeric LRRC8 is still unknown, this

(LRRC8A)5·LRRC8D heteromer may be one of the oligomeric states present in the

cellular context17. In this model structure, no large steric clashes are observed at the

subunit interfaces (Extended Data Fig. 8b), suggesting that the formation of the

hexamer, as well as the crevice between the subunits, is plausible in the

— 17 — bioRxiv preprint doi: https://doi.org/10.1101/331207; this version posted May 25, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

LRRC8A-containing heteromers. In contrast, the structure of the pore constriction site,

which is mainly formed by the helix EL1H, is different from that of the homomeric

LRRC8A (Extended Data Fig. 8b). These results again underscore the notion that the

sequence diversity in the pore constriction site is important for the functional diversity

of the LRRC8A-containing heteromers.

In the subunit interface of the TM region, we observed the lateral crevice formed

between the subunits, which is opened toward the membrane side (Fig. 2a). Although it

is currently unknown whether the activity of LRRC8 proteins is influenced by the lipid

environment, it is possible that the LRRC8 proteins might directly interact with the lipid

molecules through these crevices, in a similar manner to other channels, such as the

mechanosensitive two-pore domain K+ channels28. A recent electrophysiological

analysis using purified LRRC8 proteins demonstrated that low ionic strength alone is

sufficient for their activation, and it was speculated that the sensor for the ionic strength

exists in the IL1 loop, as well as in LRRCT17. Indeed, the long linker between the

IL1H2 and IL1H3 helices, which is largely disordered in the present structure, contains

several charged residues that are conserved among LRRC8 isoforms (Extended Data

Fig. 2). Although the question of whether the ionic strength directly modulates the

channel activities of the LRRC8 proteins remains to be answered, these observations

lead us to speculate that the interaction of the IL1 loop with ions affects its

conformation, thereby changing the arrangement of the LRR and other regions, as

observed in the present relaxed form (Fig. 5a, c), to regulate the channel activity. To

understand the gating mechanism of the LRRC8 protein depending on the cellular

volume change, the structural analysis of the closed form of LRRC8 is required.

— 18 — bioRxiv preprint doi: https://doi.org/10.1101/331207; this version posted May 25, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

Methods

Protein expression, purification and sample preparation for cyro-EM

The human LRRC8A WT protein (NCBI Reference sequence number: NP_062540.2)

was cloned from human brain cDNA (ZYAGEN) into the pEG BacMam vector, with an

C-terminal GFP-His8 tag and a tobacco etch virus (TEV) cleavage site, and was

expressed in HEK293S GnTI- (N-acetylglucosaminyl-transferase I-negative) cells

(ATCC, cat. no. CRL-3022)29. Cells were collected by centrifugation (6000×g, 10 min,

4 °C) and disrupted by sonication in buffer (50 mM Tris, pH 8.0, 150 mM NaCl)

supplemented with 5.2 µg/ml aprotinin, 2 µg/ml leupeptin, and 1.4 µg/ml pepstatinA

(all from Calbiochem). Cell debris was removed by centrifugation (10,000×g, 10 min,

4 °C). The membrane fraction was collected by ultracentrifugation (138,000×g, 1 h,

4 °C). The membrane fraction was solubilized for 1 h at 4 °C in buffer (50 mM Tris, pH

8.0, 150 mM NaCl, 5 mM dithiothreitol (DTT), 1% digitonin (Calbiochem)). Insoluble

materials were removed by ultracentrifugation (138,000×g, 1 h, 4 °C). The

detergent-soluble fraction was incubated with CNBr-Activated Sepharose 4 Fast Flow

beads (GE Healthcare) coupled with an anti-GFP nanobody (GFP enhancer)30, and

incubated for 2 h at 4 ºC. The beads were washed with SEC buffer (50 mM Tris, pH 8.0,

150 mM NaCl, 5 mM DTT, 0.1% digitonin), and further incubated overnight with TEV

protease to remove the GFP-His8 tag. After TEV protease digestion, the flow-through

was collected, concentrated, and purified by size-exclusion chromatography on a

Superose 6 Increase 10/300 GL column (GE Healthcare), equilibrated with SEC buffer.

The peak fractions of the protein were collected and concentrated to 15 mg/ml, using a

centrifugal filter unit (Merck Millipore, 100 kDa molecular weight cutoff). A 3 µl

portion of the concentrated HsLRRC8A protein (15 mg/ml) was applied to a

— 19 — bioRxiv preprint doi: https://doi.org/10.1101/331207; this version posted May 25, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

glow-discharged Quantifoil R1.2/1.3 Cu/Rh 300 mesh grid (Quantifoil), blotted using

Vitrobot Mark IV (FEI) under 4ºC and 100% humidity conditions, and then frozen in

liquid ethane.

EM image acquisition and data processing

The grid images were obtained with a Tecnai Arctica transmission electron microscope

(FEI) operated at 200 kV, and recorded by a K2 Summit direct electron detector (Gatan)

operated in the super-resolution mode with a binned pixel size of 1.490 Å. The dataset

was acquired with the SerialEM software31. Each image was dose-fractionated to 40

frames at a dose rate of 6-8 e-/pixel/sec, to accumulate a total dose of ~50 e-/Å2. In total,

5,305 super-resolution movies were collected. The movie frames were aligned in 5 x 5

patches, dose weighted and binned by two in MotionCor2 (ref. 32), and down-sampled

by two. Defocus parameters were estimated by CTFFIND 4.1 (ref. 33). First,

template-based auto-picking was performed with 2D class averages of a few hundred

manually picked particles as templates34. A total of 1,421,314 particles were extracted

in 3.25 Å/pix. These particles were divided into four batches and subjected to three

rounds of 2D classification in RELION 2.1 (refs. 34,35). The initial model was

generated in RELION. Subsequently, 664,971 good particles were further classified in

3D without symmetry. As indicated in Extended Data Fig. 3b, some classes exhibited

very weak density for one or two of the six subunits. Therefore, 164,749 particles in

classes with six intact subunits were re-extracted in the original pixel size of 1.49 Å/pix

and refined in C3 symmetry. The overall gold-standard resolution was 4.25 Å, with the

local resolution in the core TM region extending to 3.8 Å and that in the peripheral

region extending to 6.5 Å (Extended Data Fig. 3c-d).

— 20 — bioRxiv preprint doi: https://doi.org/10.1101/331207; this version posted May 25, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

Model building

The extracellular, transmembrane, and intracellular regions of HsLRRC8A were built

de novo into the electron density map in COOT36, using the Innexin-6 (PDB ID: 5H1Q)

structure as a guide. The LRR region was initially generated by creating a homology

model based on the LRR protein from L. interrogans (PDB ID: 4U08), using the Phyre2

server 24. The initial model was then refined, using PHENIX37 with secondary structure

restraints. For cross-validation, the final model was refined against one of the half maps

generated in RELION, and FSC curves were calculated between the refined model and

the half map 1, and between the refined model and the half map 2 (Extended Data Fig.

3e).

Liposome reconstitution

The purified HsLRRC8A protein was incorporated into artificial liposomes, using a

modified sucrose procedure38. Briefly, a 200 µl aliquot of a 25 mg/ml solution of

azolectin in chloroform (Avanti Polar Lipids) was dried in a glass test tube under a

stream of N2 while rotating the tube to form a homogeneous lipid film. After the lipid

was dried, 5 µl of pure water was added to the bottom (pre-hydration), followed by 1 ml

of 0.4 M sucrose. The solution was incubated for 3 h at 50 °C until the lipid was

resuspended. After cooling the solution to room temperature, the purified HsLRRC8A

protein was added to achieve the desired protein-to-lipid ratio (protein: lipid = 1:1000,

w/w). The glass tube containing the protein-lipid solution was shaken gently on an

orbital mixer for 3 h at 4 °C. After this procedure, the sample was ready for patch

clamping.

— 21 — bioRxiv preprint doi: https://doi.org/10.1101/331207; this version posted May 25, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

Patch clamping recordings

An aliquot of liposomes (2-4 µl) was taken from the liposome cloud and introduced to

the recording bath. All recordings were performed with the excised liposome patched.

For recording under asymmetric conditions, the pipette buffer (70 mM KCl, 1.5 mM

CaCl2, 1 mM MgCl2, 10 mM dextrose, and 10 mM HEPES, pH 7.4) and the bath buffer

(500 mM KCl, 1.5 mM CaCl2, 1 mM MgCl2, 10 mM dextrose, and 10 mM HEPES, pH

7.4) were used. For recording under symmetric conditions, the bath and pipette buffers

were identical and consisted of a solution of 70 mM KCl, 1.5 mM CaCl2, 1 mM MgCl2,

10 mM dextrose, and 10 mM HEPES, pH 7.4. The pH of buffers was adjusted by KOH.

For patch pipette preparation, the borosilicate glass pipettes (BF150-86-10, Sutter

Instrument, Novato) were pulled using a Flaming/Brown micropipette puller (P-97,

Sutter Instrument, Novato) and polished to a diameter corresponding to a pipette

resistance in the range of 4.0-6.0 MΩ. The recordings were performed when the pipette

was tightly sealed with the liposome membrane and the patch resistance increased

toward ~2 GΩ. The data were acquired at 50 kHz with a 0.5-kHz filter and a 50 Hz

notch filter, using an EPC-10 amplifier (HEKA, Lambrecht, Germany). All experiments

were performed at room temperature (21-24 ºC). All electrophysiological data were

analyzed by using the pCLAMP10 software (Axon Instruments, Foster City, CA).

Cell line formation for measurements of VRAC activity HEK293A cells (Invitrogen) and LRRC8A-knockout HEK293A cells were cultured in

DMEM-high glucose (Sigma-Aldrich, Cat. #D5671), supplemented with 10% fetal

bovine serum (FBS; BioWest, Cat. #S1560-500) and 100 units/ml penicillin G (Meiji

— 22 — bioRxiv preprint doi: https://doi.org/10.1101/331207; this version posted May 25, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

Seika, Cat. #6111400D2039). All cells were cultured in a 5% CO2 atmosphere at 37ºC

and verified to be negative for mycoplasma. LRRC8A-knockout HEK293A cells were

established using the CRISPR-Cas9 system39. Briefly, the targeting sequence for human

LRRC8A, 5′-GCACAACATCAAGTTCGACGTGG-3′, was designed using the

CRISPR design tool (http://crispr.mit.edu/), and oligo DNAs for its sense and anti-sense

strands were purchased from Eurofins Genomics. The sgRNA expression construct was

generated by inserting the annealed guide oligo duplex with BbsI into

pSpCas9(BB)-2A-Puro (PX459) V2.0, a gift from Dr. Feng Zhang (Addgene plasmid

#62988). The targeting sequence of the generated constructs was verified by sequencing.

Subsequently, HEK293A cells were transfected with the LRRC8A sgRNA expression

plasmid, using Lipofectamine 2000 (Invitrogen, Cat. #11668-019). After 29 h, the cells

were selected by 1 µg/ml puromycin for 44 h, followed by limiting dilution to establish

clonal cell lines. The generated LRRC8A-knockout cell lines were validated by checking

for the depletion of the LRRC8A proteins and the absence of novel truncated LRRC8A

proteins by western blotting.

Expression plasmids for this study were constructed by standard molecular biology

techniques, and all constructs were verified by sequencing. The human LRRC8A cDNA

(CDS of NM_019594.3 with the silent mutation c.1509C>T) was cloned from a cDNA

pool derived from HEK293A cells and subcloned into pcDNA3. LRRC8A mutants

(D102A, R103A, H104A, H105A, and Y106A) were constructed from LRRC8A

wild-type (WT) and subcloned into pcDNA3 (Invitrogen). A halide-sensitive YFP

(H148Q/I152L)26 mutant was generated from YFP (WT) and subcloned into pcDNA3.

— 23 — bioRxiv preprint doi: https://doi.org/10.1101/331207; this version posted May 25, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

Western blotting and antibodies

Cells were lysed in lysis buffer (20 mM Tris-HCl, pH 7.5, 150 mM NaCl, 10 mM

EDTA, 1% sodium deoxycholate, and 1% Triton X-100), supplemented with 1 mM

phenylmethylsulfonyl fluoride (PMSF) and 5 µg/ml leupeptin. The cell extracts were

clarified by centrifugation, and aliquots of the supernatants were mixed with 2x SDS

sample buffer (80 mM Tris-HCl, pH 8.8, 80 µg/ml bromophenol blue, 28.8% glycerol,

4% SDS, and 10 mM dithiothreitol). After boiling at 98ºC for 3 min, the samples were

resolved by SDS-PAGE and electroblotted onto an Immobilon-P membrane (Millipore,

Cat. #IPVH00010). The membrane was blocked with 2.5% skim milk in TBS-T (50

mM Tris-HCl, pH 8.0, 137 mM NaCl, and 0.05% Tween 20) and probed with the

appropriate antibodies. Antibody-antigen complexes were detected on X-ray films

(FUJIFILM, Cat. #47410-07523, Cat. #47410-26615 or Cat. #47410-07595) using the

ECL system (GE Healthcare). Antibodies against LRRC8A (Cat. #A304-175A, 1:5,000),

YFP-tag (YFP; Clone #1E4, Cat. #M048-3, 1:10,000), and actin (Cat. #A3853, 1:5,000)

were purchased from Bethyl Laboratories, MBL, and Sigma-Aldrich, respectively.

HRP-conjugated secondary antibodies against rabbit IgG (Cat. #7074, 1:2,000) and

mouse IgG (Cat. #7076, 1:10,000–1:20,000) were purchased from Cell Signaling

Technology.

Cell-based measurements of VRAC activity The VRAC activity in LRRC8A-rescued LRRC8A-knockout HEK293A cells was

evaluated by the efficiency of iodide influx in the halide-sensitive YFP quenching

assay9,10,26. LRRC8A-knockout HEK293A cells were transfected with YFP

(H148A/I152L) and/or LRRC8A mutant expression plasmids, using Lipofectamine

— 24 — bioRxiv preprint doi: https://doi.org/10.1101/331207; this version posted May 25, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

2000. After 4 h, the cells were reseeded in 96-well black imaging plates (Falcon, Cat.

#353219) and cultured for 65 h. Prior to measurements, the culture medium was

–1 exchanged with 50 µl/well of isoosmotic buffer (332 mOsm (kg H2O) , pH 7.4; 145

mM NaCl, 5 mM KCl, 2 mM CaCl2, 1 mM MgCl2, 10 mM glucose, 10 mM HEPES).

The initial YFP fluorescence (Ex: 500 ± 6 nm, Em: 545 ± 6 nm) was measured 5 times

at 10-sec intervals using a Varioskan Flash luminometer (Thermo Fisher Scientific).

After the addition of 125 µl/well of iodide-containing hypoosmotic buffer (190 mOsm,

pH 7.4; 70 mM NaI, 5 mM NaCl, 5 mM KCl, 2 mM CaCl2, 1 mM MgCl2, 10 mM

glucose, 10 mM HEPES) or isoosmotic buffer (332 mOsm, pH 7.4; 70 mM NaI, 5 mM

NaCl, 5 mM KCl, 2 mM CaCl2, 1 mM MgCl2, 10 mM glucose, 10 mM HEPES, 140

mM mannitol), subsequent measurements were performed 45 times at 10-sec intervals.

For the data analysis, each fluorescent value was subjected to background subtraction

by the fluorescent values of non-YFP (H148Q/I152L)-transfected cells, followed by

normalization with the fluorescent value at the 1st time point after the addition of the

iodide-containing buffer. To evaluate the efficiency of iodide influx, the normalized

fluorescence values obtained after adding the iodide-containing buffer from sextuplicate

wells were fitted to a one-phase exponential decay curve y = A exp(-t/t) + B by

nonlinear least squares on R, using RStudio. Using the estimated parameters, the speed

of YFP quenching at time t was calculated as -dy/dt = A/t exp(-t/t), and the maximum

speed of YFP quenching was defined as the speed of YFP quenching at the 1st time

point after the addition of the iodide-containing buffer: A/t. Finally, the mean of the

maximum speed of YFP quenching was computed from 4 independent experiments, for

the comparisons between the samples.

— 25 — bioRxiv preprint doi: https://doi.org/10.1101/331207; this version posted May 25, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

Immunocytochemistry LRRC8A-knockout HEK293A cells were transfected with the LRRC8A mutant

expression plasmids, using Lipofectamine 2000. After 5 h, the cells were reseeded on a

1% gelatin-coated cover slip (Matsunami Glass, Cat. #C015001) and cultured for 65 h.

The cells were fixed by 4% formaldehyde in PBS (137 mM NaCl, 2.7 mM KCl, 8.1

mM Na2HPO4•12H2O, 1.47 mM KH2PO4) for 15 min at room temperature,

permeabilized with 1% Triton X-100 in PBS for 15 min at room temperature, blocked

with 5% skim milk in TBS-T for 30 min at room temperature, and incubated with the

primary antibody for LRRC8A overnight at 4ºC. The samples were incubated with an

Alexa Fluor 594-conjugated secondary antibody against rabbit IgG (Molecular Probes,

Cat. #R37117) for 2 h at room temperature, followed by nuclear staining with Hoechst

33258 (Dojindo, Cat. #343-07961) for 30 min at room temperature. The cover slip was

mounted on a microscope slide (Matsunami Glass, Cat. #S2444) with Fluoromount

(Diagnostic BioSystems, Cat. #K024). The images were collected with a confocal

microscope, TCS-SP5 (Leica), with an HCX PL APO 63x/1.40–0.60 oil objective lens.

Statistical analysis The data are summarized as the mean ± s.e.m. No statistical method was utilized to

predetermine sample size. Homoscedasticity was assumed unless P < 0.01 in Levene’s

test based on the absolute deviations from the median. Statistical tests and sample size n

are indicated in the figure legends. Statistical tests were performed using R with

RStudio (http://www.rstudio.com/), and P < 0.05 was considered statistically significant.

No samples were excluded from statistical tests. The investigators were not blinded to

allocation during experiments and outcome assessment. The experiments were not

— 26 — bioRxiv preprint doi: https://doi.org/10.1101/331207; this version posted May 25, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

randomized.

Data availability

The raw image of HsLRRC8A after motion correction has been deposited in the

Electron Microscopy Public Image Archive (EMPIAR), under the accession number

XXXX. The cryo-EM density map of HsLRRC8 has been deposited in the Electron

Microscopy Data Bank (EMDB), under the accession number YYYY. The molecular

coordinates of HsLRRC8A have been deposited in the RCSB Protein Data Bank (PDB),

under the accession number ZZZZ.

— 27 — bioRxiv preprint doi: https://doi.org/10.1101/331207; this version posted May 25, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

Acknowledgements

We thank the members of the Nureki lab, the Kikkawa lab, and the RIKEN Center for

Life Science Technologies, especially Dr. Mizuki Takemoto and Dr. Reiya Taniguchi

(Nureki lab) for assistance with the manuscript preparation and Dr. Hideki Shigematsu

(RIKEN) for microscope maintenance. We also thank Dr. Yuhei Araiso (Kyoto Sangyo

University) for instructions regarding the digitonin preparation; Mr. Kouki Touhara

(The Rockefeller University) for advice about the anti-GFP nanobody preparation; and

Dr. Kenji Iwasaki, Dr. Naoyuki Miyazaki, and Dr. Akihiro Kawamoto (Osaka

University) for optimization of the cryo-EM experiment. This work was supported by a

grant from the National Key R&D Program of China (2016YFA0502800); by the

RIKEN Pioneering Project “Dynamic Structural Biology”; by the Platform for Drug

Discovery, Informatics and Structural Life Science from Japan Agency for Medical

Research and Development (AMED) to O.N.; by AMED (Grant No. JP17gm5010001)

to H.I.; by a MEXT Grant-in-Aid for Specially Promoted Research (Grant No.

16H06294) to O.N.; by the National Natural Science Foundation of China (Grant No.

31571083) to Z.Y.; by the Program for Professor of Special Appointment (Eastern

Scholar of Shanghai, TP2014008) to Z.Y.; by the Shanghai Rising-Star Program

(4QA1400800) to Z.Y.; by the Young 1000 Talent Program of China to Z.Y.; and by

JSPS KAKENHI (Grant Nos. 17J06101 to G.K.; 17K15086 to K.W.; 25221302 to H.I.;

17H03640 to R.I.; 24227004 to O.N.). This research was also supported by the Platform

Project for Supporting Drug Discovery and Life Science Research (Basis for Supporting

Innovative Drug Discovery and Life Science Research (BINDS)) from AMED.

— 28 — bioRxiv preprint doi: https://doi.org/10.1101/331207; this version posted May 25, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

Author contributions

G.K. and O.N. conceived and designed the project. G.K. screened and purified the

protein, and prepared cryo-EM samples. N.D. and R.N. assisted with the preparation of

cryo-EM samples. G.K., T.Nakane, T.Y., and M.S. performed cryo-EM data collection

and processing. T.Nishizawa, T.K., N.M., A.T., H.Y., and M.K. assisted with the

optimization of the cryo-EM data collection. G.K. and R.I. performed the model

building, with assistance from T.Nishizawa. Y.J., M.H., and Z.Y. performed the patch

clamping recordings. M.I., K.W., and H.I. performed the cell-based quenching assay.

G.K., R.I., and O.N. wrote the manuscript. O.N. supervised all of the research.

— 29 — bioRxiv preprint doi: https://doi.org/10.1101/331207; this version posted May 25, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

Figure Legends

Figure 1. Overall structure and subunit interactions of the HsLRRC8A

hexamer.

(a) Local resolution of the HsLRRC8A structure, estimated by RELION. (b)

Representative density maps of the transmembrane and LRR regions. The cartoon

models of the TM1, EL1 and the following TM2, and LRR regions were fitted to the

density maps (colored gray). (c) Overall structure of the HsLRRC8A hexamer, viewed

parallel to the membrane (left). Overall structure of the HsLRRC8A hexamer with

molecular envelope, viewed from the extracellular side (right, upper) and the

intracellular side (right, lower). (d) The subunit structures of HsLRRC8A, viewed

parallel to the membrane. Each region is colored as follows: TM1, purple; EL1, blue;

TM2, light blue; IL1, cyan; TM3, green; EL2, light green; TM4, yellow; IL2, orange;

LRRNT, red; LRR1-15, pink; and LRRCT, brown. The N- and C- termini are indicated

by ‘N’ and ‘C’, respectively. (e) Schematic representation of the HsLRRC8A membrane

topology, colored as in (d). The dotted lines indicate the disordered linkers. The

molecular graphics were illustrated with CueMol (http://www.cuemol.org/).

Figure 2. Subunit Interface.

(a) The tight and loose subunit interfaces (left and right panels, respectively) between

the three neighboring subunits (α, β, and ζ) of HsLRRC8A. The structures of the

subunits are shown as the solvent-accessible surface models. (b) Two adjacent subunits

(α and β subunits) of HsLRRC8A are superimposed on each other, using only the TM

region. The RMSD value for the 553 Cα atoms from the subunits is 3.79 Å. (c)

Close-up view of the superimposed intracellular IL2 regions from two adjacent subunits.

— 30 — bioRxiv preprint doi: https://doi.org/10.1101/331207; this version posted May 25, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

The residues involved in helix formation are depicted in stick representations. The

RMSD value for the 66 Cα atoms from the subunits of the IL2 regions is 0.88 Å.

Figure 3. pore.

(a) Cross-sections of the surface representation for the N-terminal half of the

HsLRRC8A subunits, showing the ion channel pore. The four subunits are colored

according to Fig. 1d, and are shown in cartoon representations. (b) The pore radius for

the HsLRRC8A structure along the pore center axis. The pore size was calculated with

the program HOLE40. (c-e) Close-up views of the channel pore forming regions. In the

left views, only two diagonal subunits viewed parallel to the membrane are shown, for

clarity. The side chains of the pore-lining amino residues are depicted in stick

representations. The distances between the residues involved in the pore constrictions

are shown. In the right views, all six subunits viewed from the extracellular side are

shown.

Figure 4. Pore constriction site.

(a) Close-up view of the neighboring EL1 helices forming the pore constriction site.

The side chains are depicted by stick models. (b) Sequence alignment around the pore

constriction site, depicted according to Extended Data Fig. 2. (c) VRAC activity in the

LRRC8A-rescued LRRC8A-knockout HEK293A cells. Each line indicates the mean ±

s.e.m. of the normalized YFP fluorescence (n = 4). Hypotonicity after the addition of

iodide: 231 mOsm. WT (+) and WT (++++) represent the different expression levels of

the LRRC8A wild-type protein detected by western blotting, according to Extended

Data Fig. 6c. (d) Maximum speed of hypotonicity-induced YFP quenching. Data are

— 31 — bioRxiv preprint doi: https://doi.org/10.1101/331207; this version posted May 25, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

presented as the mean ± s.e.m. with each individual light grey point (n = 4). ** P < 0.01,

*** P < 0.001 by Dunnett’s test.

Figure 5. Compact and relaxed structures of HsLRRC8A.

(a,b) Overall structure of the HsLRRC8A hexamer in the relaxed form (a), and in the

high-resolution compact form (b), viewed parallel to the membrane (left), and from the

extracellular side (right). The structures are depicted in cylinder representations. (c)

Schematic drawing depicting the structural transition between the compact and relaxed

forms of HsLRRC8A.

— 32 — bioRxiv preprint doi: https://doi.org/10.1101/331207; this version posted May 25, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

Extended Data Figure Legends

Extended Data Figure 1. Functional properties of the HsLRRC8A protein.

(a) FSEC profiles on a Superose 6 Increase 10/300 GL column (GE Healthcare) for the

EGFP-fused HsLRRC8A expressed in HEK293S GnTI- cells. The arrows indicate the

estimated elution positions of the void volume, the EGFP-fused HsLRRC8A, and the

free EGFP. (b) Single-channel current recordings of HsLRRC8A after reconstitution in

liposomes under asymmetric salt conditions (500 mM KCl in bath, 70 mM KCl in

pipette), and the normalized all-points amplitude histogram analysis of HsLRRC8A in

the open and closed states at 100 mV. The gray dashed lines indicate the closed (C) and

open (O) states. For the plot, the bin width is set at 0.1 pA/bin and the total count of

events is normalized to 1.0. The distribution data were fit by the sum of two Gaussians,

and the peaks correspond to the closed (C) and open (O) states. (c) Single-channel

current recordings of HsLRRC8A after reconstitution in liposomes under symmetric salt

conditions (70 mM KCl in bath, 70 mM KCl in pipette), and the normalized all-points

amplitude histogram analysis of HsLRRC8A at 100 mV.

Extended Data Figure 2. Sequence alignment of LRRC8 isoforms

The amino acid sequences of LRRC8 isoforms were aligned using Clustal Omega41, and

are shown using ESPript3 (ref. 42). The secondary structure elements and the colors of

the domains from HsLRRC8A are labeled above the alignments. For the sequence

alignment, the human and mouse LRRC8 isoforms were used: human (HsLRRC8A,

NCBI Reference sequence number: NP_062540.2; HsLRRC8B, NP_056165.1;

HsLRRC8C, NP_115646.2; HsLRRC8D, NP_060573.2; HsLRRC8E, AAH70089.1)

and mouse (MmLRRC8A, NP_808393.1; MmLRRC8B, NP_001028722.1;

— 33 — bioRxiv preprint doi: https://doi.org/10.1101/331207; this version posted May 25, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

MmLRRC8C, NP_598658.1; MmLRRC8D, NP_848816.3; MmLRRC8E,

NP_082451.2).

Extended Data Figure 3. Cyro-EM analysis of HsLRRC8A

(a) A representative cryo-EM micrograph of HsLRRC8A. Red circles indicate

individual particles. The white scale bar represents 50 nm. (b) Flow chart of cryo-EM

data processing of the HsLRRC8A structure, including particle picking, classification,

and 3D refinement. (c) Angular distribution plot of particles included in the final 3D

reconstruction of HsLRRC8A, with C3 symmetry imposed. (d) Fourier Shell

Correlation (FSC) curve of the final 3D reconstruction model calculated using

“relion_postprocess” with masked marked 4.25 Å resolution, corresponding to the FSC

= 0.143 gold standard cut-off criterion. (e) Cross-validation FSC curves for the refined

model versus half maps (Half maps 1 and 2), and versus summed map.

Extended Data Figure 4. Structural comparison of two adjacent subunits.

(a) Two adjacent subunits (α and β subunits) of HsLRRC8A are superimposed on each

other. The RMSD value for the 553 Cα atoms from the subunits is 3.79 Å. For clarity,

the overall subunits superimposed using only the TM region are presented. (b) The

extracellular regions from two adjacent subunits are superimposed on each other. The

RMSD value for the 68 Cα atoms from the two extracellular regions is 0.39 Å. (c) The

transmembrane regions from two adjacent subunits are superimposed on each other.

The RMSD value for the 110 Cα atoms from the two extracellular regions is 0.40 Å. (d)

The intracellular regions from two adjacent subunits are superimposed on each other.

The RMSD value for the 119 Cα atoms from the two extracellular regions is 0.98 Å. (e)

— 34 — bioRxiv preprint doi: https://doi.org/10.1101/331207; this version posted May 25, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

The LRR regions from two adjacent subunits are superimposed on each other. The

RMSD value for the 395 Cα atoms from the two extracellular regions is 0.47 Å.

Extended Data Figure 5. Subunit interfaces between two adjacent

subunits.

(a,b) Close-up views between two adjacent subunits at the extracellular region, showing

the tight (a) and loose (b) interactions. (c,d) Close-up views between two adjacent

subunits at the transmembrane region, showing the tight (c) and loose (d) interactions.

(e,f) Close-up views between two adjacent subunits at the intracellular region, showing

the tight (e) and loose (f) interactions. (g,h) Close-up views between two adjacent

subunits at the LRR region, showing the tight (g) and loose (h) interactions. A surface

model with the side chains of the residues lining the subunit interfaces depicted by stick

models is presented for each panel. Three adjacent subunits (α, β, and ζ subunits) of

HsLRRC8A are colored according to Fig. 1d.

Extended Data Figure 6. Validation of LRRC8A mutants and YFP

quenching assay.

(a) Subcellular localization of LRRC8A mutants rescued in LRRC8A-knockout

HEK293A cells. The white dashed square indicates a region around the cell membrane;

the magnified image of the LRRC8A-derived fluorescent channel is located in the

corner. The white scale bar indicates 25 µm. (b) FSEC profiles on a Superose 6 Increase

10/300 GL column (GE Healthcare) for the EGFP-fused HsLRRC8A mutants expressed

in HEK293S GnTI- cells. The arrows indicate the estimated elution positions of the void

volume, the EGFP-fused HsLRRC8A, and the free EGFP. The analysis of the mutants

— 35 — bioRxiv preprint doi: https://doi.org/10.1101/331207; this version posted May 25, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

showed symmetric and monodisperse peaks, similar to those of the wild-type channel,

confirming the proper structural integrities of these mutant channels. (c) Amounts of

HsLRRC8A expression in rescued LRRC8A-knockout HEK293A cells. † indicates

non-specific bands. (d) Comparison between individual time courses and the fitting

curve of the normalized YFP fluorescence. Grey lines and light blue lines indicate

individual time courses of normalized YFP fluorescence (n = 4). Blue lines indicate the

mean of one-phase exponential decay-fitting curves. I–-iso: 332 mOsm, I–-hypo: 231

mOsm.

Extended Data Figure 7. Structural similarity between HsLRRC8A, innexin,

and connexin.

(a-c) The N-terminal halves of the HsLRRC8A subunit (a), the CeInnexin-6 (PDB ID:

5H1Q) subunit (b), and the HsConnexin-26 (PDB ID: 2ZW3) subunit (c) are presented.

The subunits of CeInnexin-6 and HsConnexin-26 are colored according to the

HsLRRC8A coloring. The N- and C- termini are indicated by ‘N’ and ‘C’, respectively.

In each structure, the Cys residues forming disulfide bonds at the extracellular region

are depicted by stick models. In each panel, a close-up view of the disulfide bonds is

shown in the inset. (d-f) The solvent-excluded molecular surfaces of HsLRRC8A (d),

CeInnexin-6 (e), and HsConnexin-26 (f). The surfaces are colored according to the

subunits. For HsLRRC8A, the extracellular, TM, and intracellular regions are shown.

(g-i) The channel pore and N-terminal region of HsLRRC8A (d), CeInnexin-6 (e), and

HsConnexin-26 (f). The protein main chains are shown as cylinder models. (j) The

secondary structure prediction of the N-terminal regions of HsLRRC8A, CeInnexin-6,

and HsConnexin-26. The prediction was performed by the program PSIPRED43. The

— 36 — bioRxiv preprint doi: https://doi.org/10.1101/331207; this version posted May 25, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

actual locations of the corresponding helices are indicated by the red and pink boxes for

NTH and TM1, respectively.

Extended Data Figure 8. Model of the heteromeric LRRC8 protein.

(a) The conservation score mapped on the molecular surface of HsLRRC8A. The score

was calculated by the program CONSURF27, based on the multiple-sequence alignment

of the A-E isomers in Extended Data Fig. 2. The molecular surface is colored according

to the normalized conservation score, from 0.752 (pink) to -0.983 (purple). (b) The

model structure of the (LRRC8A)5·LRRC8D heterohexamer, viewed parallel to the

membrane (left), and from the extracellular side (right). The homology model of human

LRRC8D was generated by the program MODELLER44, using the alignment in

Extended Data Fig. 2, and then the α subunit of the present structure was replaced by

this model. In the inset, a close-up view of the channel pore forming regions from the

extracellular side is shown, and the side chains of pore-lining amino residues are

depicted in stick representations. The five LRRC8A subunits are colored light blue,

while the LRRC8D isoform is colored light brown.

Extended Data Table 1. Data collection, refinement and validation

statistics.

— 37 — bioRxiv preprint doi: https://doi.org/10.1101/331207; this version posted May 25, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

References

1. Hoffmann, E. K., Lambert, I. H. & Pedersen, S. F. Physiology of cell volume

regulation in vertebrates. Physiol. Rev. 89, 193–277 (2009).

2. Pedersen, S. F., Kapus, A. & Hoffmann, E. K. Osmosensory Mechanisms in

Cellular and Systemic Volume Regulation. J. Am. Soc. Nephrol. 22, 1587–1597

(2011).

3. Hoffmann, E. K. et al. Role of volume-regulated and calcium-activated anion

channels in cell volume homeostasis , cancer and drug resistance. Channels 6950,

380–396 (2015).

4. Okada, Y. et al. Receptor-mediated control of regulatory volume decrease (RVD)

and apoptotic volume decrease (AVD). J. Physiol. 532, 3–16 (2001).

5. Jentsch, T. J. VRACs and other ion channels and transporters in the regulation of

cell volume and beyond. Nat. Rev. Mol. Cell Biol. 17, 293–307 (2016).

6. Stauber, T. The volume-regulated anion channel is formed by LRRC8

heteromers-molecular identification and roles in membrane transport and

physiology. Biol. Chem. 396, 975–990 (2015).

7. Pedersen, S. F., Klausen, T. K. & Nilius, B. The identification of a

volume-regulated anion channel: An amazing Odyssey. Acta Physiol. 213, 868–

881 (2015).

8. Sawada, A. et al. A congenital mutation of the novel gene LRRC8 causes

agammaglobulinemia in humans. J. Clin. Invest. 112, 1707–1713 (2003).

9. Qiu, Z. et al. SWELL1, a plasma membrane protein, is an essential component of

volume-regulated anion channel. Cell 157, 447–458 (2014).

10. Voss, F. K. et al. Identification of LRRC8 Heteromers as an Essential

— 38 — bioRxiv preprint doi: https://doi.org/10.1101/331207; this version posted May 25, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

Component of the Volume-Regulated Anion Channel VRAC. Science 344, 634–

638 (2014).

11. Kubota, K. et al. LRRC8 involved in B cell development belongs to a novel

family of leucine-rich repeat proteins. FEBS Lett. 564, 147–152 (2004).

12. Smits, G. & Kajava, A. V. LRRC8 extracellular domain is composed of 17

leucine-rich repeats. Mol. Immunol. 41, 561–562 (2004).

13. Abascal, F. & Zardoya, R. LRRC8 proteins share a common ancestor with

, and may form hexameric channels involved in cell-cell

communication. BioEssays 34, 551–560 (2012).

14. Oshima, A., Tani, K. & Fujiyoshi, Y. Atomic structure of the innexin-6 gap

junction channel determined by cryo-EM. Nat. Commun. 7, 13681 (2016).

15. Maeda, S. et al. Structure of the connexin 26 gap junction channel at 3.5 A

resolution. Nature 458, 597–602 (2009).

16. Bennett, B. C. et al. An electrostatic mechanism for Ca2+ -mediated regulation of

gap junction channels. Nat Commun 7, 8770 (2016).

17. Syeda, R. et al. LRRC8 Proteins Form Volume-Regulated Anion Channels that

Sense Ionic Strength. Cell 164, 499–511 (2016).

18. Gradogna, A., Gavazzo, P., Boccaccio, A. & Pusch, M. Subunit-dependent

oxidative stress sensitivity of LRRC8 volume-regulated anion channels. J.

Physiol. 595, 6719–6733 (2017).

19. Planells-Cases, R. et al. Subunit composition of VRAC channels determines

substrate specificity and cellular resistance to Pt-based anti-cancer drugs. EMBO

J. 34, 2993–3008 (2015).

20. Schober, A. L., Wilson, C. S. & Mongin, A. A. Molecular composition and

— 39 — bioRxiv preprint doi: https://doi.org/10.1101/331207; this version posted May 25, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

heterogeneity of the LRRC8-containing swelling-activated osmolyte channels in

primary rat astrocytes. J. Physiol. 595, 6939–6951 (2017).

21. Lutter, D., Ullrich, F., Lueck, J. C., Kempa, S. & Jentsch, T. J. Selective transport

of neurotransmitters and-modulators by distinct volume-regulated LRRC8 anion

channels. J. Cell Sci. 130, 1122–1133 (2017).

22. Kawate, T. & Gouaux, E. Fluorescence-Detection Size-Exclusion

Chromatography for Precrystallization Screening of Integral Membrane Proteins.

Structure 14, 673–681 (2006).

23. Hattori, M., Hibbs, R. E. & Gouaux, E. A fluorescence-detection size-exclusion

chromatography-based thermostability assay for membrane protein

precrystallization screening. Structure 20, 1293–1299 (2012).

24. Kelley, L. A., Mezulis, S., Yates, C. M., Wass, M. N. & Sternberg, M. J. E. The

Phyre2 web portal for protein modeling, prediction and analysis. Nat. Protoc. 10,

845–858 (2015).

25. Ullrich, F., Reincke, S. M., Voss, F. K., Stauber, T. & Jentsch, T. J. Inactivation

and anion selectivity of volume-regulated anion channels (VRACs) depend on

c-terminal residues of the first extracellular loop. J. Biol. Chem. 291, 17040–

17048 (2016).

26. Galietta, L. J. V, Haggie, P. M. & Verkman, A. S. Green fluorescent

protein-based halide indicators with improved chloride and iodide affinities.

FEBS Lett. 499, 220–224 (2001).

27. Ashkenazy, H., Erez, E., Martz, E., Pupko, T. & Ben-tal, N. ConSurf 2010 :

calculating evolutionary conservation in sequence and structure of proteins and

nucleic acids. Nucleic Acids Res. 38, W529–W533 (2010).

— 40 — bioRxiv preprint doi: https://doi.org/10.1101/331207; this version posted May 25, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

28. Brohawn, S. G., Campbell, E. B. & MacKinnon, R. Physical mechanism for

gating and mechanosensitivity of the human TRAAK K+ channel. Nature 516,

126–130 (2014).

29. Goehring, A. et al. Screening and large-scale expression of membrane proteins in

mammalian cells for structural studies. Nat. Protoc. 9, 2574–2585 (2014).

30. Kirchhofer, A. et al. Modulation of protein properties in living cells using

nanobodies. Nat. Struct. Mol. Biol. 17, 133–138 (2010).

31. Mastronarde, D. N. Automated electron microscope tomography using robust

prediction of specimen movements. J. Struct. Biol. 152, 36–51 (2005).

32. Zheng, S. Q. et al. MotionCor2: anisotropic correction of beam-induced motion

for improved cryo-electron microscopy. Nat. Methods 14, 331–332 (2017).

33. Rohou, A. & Grigorieff, N. CTFFIND4: Fast and accurate defocus estimation

from electron micrographs. J. Struct. Biol. 192, 216–221 (2015).

34. Scheres, S. H. W. Semi-automated selection of cryo-EM particles in

RELION-1.3. J. Struct. Biol. 189, 114–122 (2015).

35. Kimanius, D., Forsberg, B. O., Scheres, S. H. W. & Lindahl, E. Accelerated

cryo-EM structure determination with parallelisation using GPUS in RELION-2.

Elife 5, 1–21 (2016).

36. Emsley, P., Lohkamp, B., Scott, W. G. & Cowtan, K. Features and development

of Coot. Acta Crystallogr. D. Biol. Crystallogr. 66, 486–501 (2010).

37. Adams, P. D. et al. PHENIX: a comprehensive Python-based system for

macromolecular structure solution. Acta Crystallogr. D. Biol. Crystallogr. 66,

213–21 (2010).

38. Battle, A. R., Petrov, E., Pal, P. & Martinac, B. Rapid and improved

— 41 — bioRxiv preprint doi: https://doi.org/10.1101/331207; this version posted May 25, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

reconstitution of bacterial mechanosensitive ion channel proteins MscS and

MscL into liposomes using a modified sucrose method. FEBS Lett. 583, 407–412

(2009).

39. Ran, F. A. et al. Genome engineering using the CRISPR-Cas9 system. Nat.

Protoc. 8, 2281–2308 (2013).

40. Smart, O. S., Neduvelil, J. G., Wang, X., Wallace, B. A. & Sansomt, M. S. P.

HOLE : A program for the analysis of the pore dimensions of ion channel

structural models. J. Mol. Graph. 14, 354–360 (1996).

41. Sievers, F. et al. Fast, scalable generation of high-quality protein multiple

sequence alignments using Clustal Omega. Mol. Syst. Biol. 7, (2011).

42. Robert, X. & Gouet, P. Deciphering key features in protein structures with the

new ENDscript server. Nucleic Acids Res. 42, 320–324 (2014).

43. Buchan, D. W. A. et al. Protein annotation and modelling servers at University

College London. Nucleic Acids Res. 38, W563–W568 (2010).

44. Fiser, A. & Sali, A. Modeller: Generation and Refinement of Homology-Based

Protein Structure Models. Methods Enzymol. 374, 461–491 (2003).

— 42 — bioRxiv preprint doi: https://doi.org/10.1101/331207; this version posted May 25, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. A B D102 T48 R103

3.8 Å

4.2 Å

4.7 Å I135 W23 5.2 Å TM1 EL1-TM2 LRRNT,LRR1-15,LRRCT 5.7 Å (W23-T48) (D102-I135) (L413-D806)

C

Extracellular region (35 Å) Out

TM region (40 Å) 90°

Intracellular In region (35 Å)

α subunit LRR region (60 Å) β subunit 90° γ subunit δ subunit ε subunit 130 Å ζ subunit

D Extracellular E Extracellular region region EL1H EL1 EL1β EL2β2 EL2 EL2β1

Out Out TM region TM region TM3 TM1 TM2 TM1 TM1 TM3 TM3 TM4 TM4 TM2

In N In N Intracellular Intracellular N region region

IL2H2 IL2H3 IL1 IL2H3 IL1H1 IL2H1 IL2 180° IL1H2 IL1H3

IL2H4 LRRNT

LRR region LRR1 LRR2 LRR3 LRR4 LRR5 LRR6 LRR7 LRR8 LRR9 LRR10

C C LRRNT C

LRRCT LRR1-15 LRRCT LRR15 LRR14 LRR13 LRR12 LRR11

Kasuya et al. Figure 1 bioRxiv preprint doi: https://doi.org/10.1101/331207; this version posted May 25, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

A Tight interface Loose interface

60°

α subunit β subunit ζ subunit

B C

N IL2H2

IL2H3 IL2H1

60° IL2H4 ~11º

LRRNT α subunit β subunit C

~22º~22°

Kasuya et al. Figure 2 bioRxiv preprint doi: https://doi.org/10.1101/331207; this version posted May 25, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

A C Extracellular region Extracellular EL1H region

EL1H D102 R103 14.8 Å

TM region N107 D110 7.6 Å 180° R103 Y114 TM1 Intracellular IL1H1 region D TM region IL1H3 TM1 T48 T44 13.7 Å V40 M37 I33 180° T48 B Y30 V26 140

Extracellular 50.8 Å region 120 D102 R103 Constriction 100 T48 Intracellular region T44 TM region E 80 I33 IL1H1 S150 60 R18 Intracellular

region E154 D165 40 S158 Distance along pore (Å) K235 D165 K235

20 LRR region D414 K235

0 IL1H3 22.5 Å 180° 0 4 8 12 16 Pore radius (Å)

Kasuya et al. Figure 3 bioRxiv preprint doi: https://doi.org/10.1101/331207; this version posted May 25, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

A B H104 (α) EL1H R103 (α) 100 110

H104 (β) HsLRRC8A K YD L D RH QY NYV DA VCY E N R103 (β) D102 (α) MmLRRC8A K YD L D RH QY NYV DA VCY E N HsLRRC8B Q N DL HR QQY SYI DA VCY E K MmLRRC8B Q N DL HR QQY SYI DA VCY E K HsLRRC8C KT DL D L QQY SFI NQ MCY E R Q105 (α) MmLRRC8C KT DL D L QQY SFI NQ MCY E R L101 (α) Y106 (α) HsLRRC8D KT NL D F QQY V FI NQ MCY HL MmLRRC8D KT NL D F QQY V FI NQ MCY HL Q105 (β) HsLRRC8E KN NL D L QQY SFI NQ LCY E T D100 (α) MmLRRC8E KN NL D L QQY SFI NQ LCY E T

K98 (α) Y99 (α)

C Iodide-containing buffer D 0.006 ***

1.0 0.005 *** **

- 0.004 *** Y106A 0.8 Q105A 0.003 WT (+) D102A H104A 0.002 0.6 WT (++++)

R103A 0.001 Normalized YFP fluorescence (a.u.) Maximum speed of YFP quenching [a.u.] 0 100 200 300 400 0.000 - WT WT D102A R103A H104A Q105A Y106A Time after adding iodide-containing buffer (Sec) (+) (++++)

Kasuya et al. Figure 4 bioRxiv preprint doi: https://doi.org/10.1101/331207; this version posted May 25, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

A Relaxed form Extracellular region (35 Å) Out

TM region (40 Å)

Intracellular In region 90° (33 Å)

LRR region (52 Å)

150 Å B Extracellular Compact form region (35 Å) Out

TM region (40 Å)

Intracellular In region 90° (35 Å)

LRR region (60 Å)

130 Å

C Compact form Relaxed form Out

TM

In IL1 and IL2

LRR

Kasuya et al. Figure 5 bioRxiv preprint doi: https://doi.org/10.1101/331207; this version posted May 25, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

A B 1.0

0.8 Count (N) HsLRRC8A-EGFP O 0.6 C C 5 pA 0.4 0.5 s 0.2 O

-2 -1 0 1 2 3 4 5 EGFP Amplitude (pA) C 1.0 Void

0.8 Count (N)

0.6 10 pA Normalized fluorescence (a.u.) 0 1 s 0.4 0 5 10 15 20 25 0.2 Volume (ml)

-0.4 -0.3 -0.2 -0.1 0.0 0.1 0.2 0.3 0.4 Amplitude (pA)

Kasuya et al. Figure S1 bioRxiv preprint doi: https://doi.org/10.1101/331207; this version posted May 25, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

TM1 EL1β α1 β1 . 1 10 20 30 40 50 60 70 HsLRRC8A M I P V TE L R YFA D TQ P A YRI LKPWWDV F T DY ISIV ML MI A VF GG TL Q VTQDKMI .CLP C K WVTKD S CNDSFRGWA A .... . MmLRRC8A M I P V TE L R YFA D TQ P A YRI LKPWWDV F T DY ISIV ML MI A VF GG TL Q VTQDKMI .CLP C K WVTKD S CNDSFRGWA A .... . HsLRRC8B M I T L TE L K CLA D AQ SS YHI LKPWWDV F WY Y ITLI ML LV A V LAGA LQ LTQ S RVL CCLP C K VEFD N HCAVPWDILK A .... . MmLRRC8B M I T L TE L K CLA D AQ SS YHI LKPWWDV F WY Y ITLI ML LV A V LAGA LQ LTQ S RVL CCLP C K VEFD NQ CAVPWDLLKG.... . HsLRRC8C M I P V TE F R QFS E QQ P A FRV LKPWWDV F T DY LSVA ML MI G VF GC TL Q V M QDKII .CLP K R VQPA QN HSSLSNVSQ A .... . MmLRRC8C M I P V TE F R QFS E QQ P A FRV LKPWWDV F T DY LSVA ML MI G VF GC TL Q V M QDKII .CLP K R VQPA QN HSSVPNVSQ A .... . HsLRRC8D M FT L AE VASLN D IQ P T YRI LKPWWDV F M DY L A VV ML MV A IF AG TM Q LT K D Q VV .CLP VLPSPV NS KAHTPPGNAEVTTN I MmLRRC8D M FT L AE VASLN D IQ P T YRI LKPWWDV F M DY L A VV ML MV A IF AG TM Q LT K D Q VV .CLP VLPSPA NS KAHTPPGNADITTE V HsLRRC8E M I P V AE F K QFT E QQ P A FKV LKPWWDV LA EY LTVA ML MI G VF GC TL Q VTQDKII .CLP N H ELQE N L...... SE A .... . MmLRRC8E M I P V AE F K QFT E QQ P A FKV LKPWWDV LA EY LTVA ML MI G VF GC TL Q VTQDKII .CLP S H ESRE N I...... SG A .... .

EL1H α2

80 90 100 110 HsLRRC8A ...... PGPEPTYPN S TIL P T P D...... TGPT GIK YD L D RH QY NYV DA VCY E NR L MmLRRC8A ...... SSPEPTYPN S TVL P T P D...... TGPT GIK YD L D RH QY NYV DA VCY E NR L HsLRRC8B ...... SMNTSS.....NP G T P L...... PLPLR I Q N DL HR QQY SYI DA VCY E KQ L MmLRRC8B ...... SENASS.....NS G LLL...... PLPLR I Q N DL HR QQY SYI DA VCY E KQ L HsLRRC8C ...... VAS T TPL P P P K....PSPANPITVEMK GLKT DL D L QQY SFI NQ MCY E RA L MmLRRC8C ...... VIS T TPL P P P K....PSPTNPATVEMK GLKT DL D L QQY SFI NQ MCY E RA L HsLRRC8D PKMEAATNQDQDGRT.TNDISFGTSAVTPDIPLRATY P RTDFALPNQEAKKEKKDPT G R KT NL D F QQY V FI NQ MCY HLA L MmLRRC8D PRMETATHQDQNGQTTTNDVAFGTSAVTPDIPL Q ATH P HAESTLPNQEAKKEKRDPT G R KT NL D F QQY V FI NQ MCY HLA L HsLRRC8E ...... PC Q QLL P R G I....P.EQIGALQEVK GLKN NL D L QQY SFI NQ LCY E TA L MmLRRC8E ...... PC Q QLL P Q G I....S.EQMGGLRELS GLKN NL D L QQY SFI NQ LCY E TA L TM2 IL1H1 IL1H2

α3 α4 α5 α6

120 130 140 150 160 170 180 190 HsLRRC8A HW FA K YFPYL VLL HT LIFLACSN FW FKF P R TS S KL EHF VS IL LKCF DSPWTT RALSE TVV E E S DPKPAFSKMNGS.MDK K MmLRRC8A HW FA K YFPYL VLL HT LIFLACSN FW FKF P R TS S KL EHF VS IL LKCF DSPWTT RALSE TVV E E S DPKPAFSKMNGS.MDK K HsLRRC8B HW FA K FFPYL VLL HT LIFAACSN FW L HY P S TS S RL EHF V AIL HKCF DSPWTT RALSE TVA E Q S VRPLKLSKSKIL...L S MmLRRC8B HW FA K FFPYL VLL HT LIFAACSN FW L HY P S TS S RL EHF VS IL HKCF DSPWTT RALSE TVA E Q S VRPLKLSKSKTL...L S HsLRRC8C HW YA K YFPYL VLI HT LVFMLCSN FW FKF P G SS S KI EHF IS IL GKCF DSPWTT RALSE VSG E D S EEKDNRKNNMNRSNTI . MmLRRC8C HW YA K YFPYL VLI HT LVFMLCSN FW FKF P G SS S KI EHF IS IL GKCF DSPWTT RALSE VSG E D S EEKDNRKNNMNRSGTI . HsLRRC8D PW Y SK YFPYL ALI HT II L MV S SN FW FKY P K T CS KV EHF VS IL GKCF ESPWTT KALSE TAC E D S EENKQRITGAQTLPKH V MmLRRC8D PW Y SK YFPYL ALI HT II L MV S SN FW FKY P K T CS KV EHF VS IL GKCF ESPWTT KALSE TAC E D S EENKQRITGAQTLPKH V HsLRRC8E HW Y TK YFPYL VVI HT LIFMVCTS FW FKF P G TS S KI EHF IS IL GKCF DSPWTT RALSE VSG E N Q KGPAATERAAATIVAM A MmLRRC8E HW YA K YFPYL VVI HT LIFMVCTS FW FKF P G TS S KI EHF IS IL GKCF DSPWTT RALSE VSG E NHKGPASG.RAMVTTVTT T IL1H3 TM3

α7 α8

200 210 220 230 240 250 260 270 HsLRRC8A S S TVSEDVEA.....TVPMLQRTKSRI E QG IVD RSETG VLDKK EGEQAKA LFEKV KK FR THVE E GD IV Y R LY MR Q TII K V MmLRRC8A S S TVSEDVEA.....TVPMLQRTKSRI E QG IVD RSETG VLDKK EGEQAKA LFEKV KK FR THVE E GD IV Y R LY MR Q TII K V HsLRRC8B S S GCSADIDS.....GKQSLPYPQPGL E SAG IE SPTSS VLDKK EGEQAKA IFEKV KR FR MHVE QK D II Y R VY LK Q I IV K V MmLRRC8B T S GGSADIDA.....SKQSLPYPQPGL E SPG IE SPTSS VLDKK EGEQAKA IFEKV KR FR LHVE QR D II Y R VY LK Q I IV K V HsLRRC8C Q S GP...EGS...... LVNSQSLKSIP E KF VVD KSTAG ALDKK EGEQAKA LFEKV KK FR LHVE E GD IL Y A MY VR Q TVL K V MmLRRC8C Q S GP...EGN...... LVRSQSLKSIP E KF VVD KSAAG ALDKK EGEQAKA LFEKV KK FR LHVE E GD IL Y A MY VR Q TVL K V HsLRRC8D ..STSSDEGSPSASTPMINKTGFKFSA E KP VIE VPSMT ILDKK DGEQAKA LFEKV RK FR AHVE D SD LI Y K LY V VQ TVI K T MmLRRC8D ..STSSDEGSPSASTPMINKTGFKFSA E KP VIE VPSMT ILDKK DGEQAKA LFEKV RK FR AHVE D SD LI Y K LY V VQ TLI K T HsLRRC8E G T GP...GKA.....GEGEKEKVLAEP E KV V T E PPVVT LLDKK EGEQAKA LFEKV KK FR MHVE E GD IL Y T MY IR Q TVL K V MmLRRC8E GAGS...GKV.....GEGEKEKVLIEP E KV V S E PPVVT LLDKK EGEQAKA LFEKV KK FR VHVE E GD IL Y S MY IR Q TVL K V EL2β1 EL2β2 TM4

β2 β3 α9

280 290 300 310 320 330 340 350 HsLRRC8A IK F IL I I CY T VY YV HN I K F D V DC T VDI ES LT GY RT Y RC A H P LA T LF KI L A SF Y I SL V IF YG L I CM Y TL WW M L R RS L K KY S MmLRRC8A IK F AL I I CY T VY YV HN I K F D V DC T VDI ES LT GY RT Y RC A H P LA T LF KI L A SF Y I SL V IF YG L I CM Y TL WW M L R RS L K KY S HsLRRC8B I LF VL I I TY VPY FL TH IT LE I DC S VDV QAF TGY KR Y QC VY SL A E IF KV L A SF Y V IL V I LYG L TSS Y SL WW M L R SS L K QY S MmLRRC8B I LF VL I I TY VPY FL SY IT LE I DC S IDV QAF TGY KR Y QC VY SL A E IF KV L A SF Y V IL V M LYG L TSS Y SL WW M L R SS L K QY S HsLRRC8C IK F LI I I AY N SAL V SK VQF T V DC N VDI QD MT GY KN F SC N HTM A H LF SK L SFC Y L CF V S IYG L T CL Y TL YW L FYRS L R EY S MmLRRC8C IK F LI I I AY N SAL V SK VQF T V DC N VDI QD MT GY KN F SC N HTM A H LF SK L SFC Y L CF V S IYG L T CL Y TL YW L FYRS L R EY S HsLRRC8D AK F I FI L CY T AN FV NA ISF EHV C KPK V EH L IGY EV F EC T HNM A Y M LKK L L IS Y I SI I C VYG FI CL Y TL FW L F R IP L K EY S MmLRRC8D AK F I FI L CY T AN FV NA ISF EHV C KPK V EH LT GY EV F EC T HNM A Y M LKK L L IS Y I SI I C VYG FI CL Y TL FW L F R IP L K EY S HsLRRC8E C KF LA I L VY N LV YV EK ISF L V AC R VE TSE VT GY AS F CC N HT KA H LF SK L A FC Y I SF V C IYG L T CI Y TL YW L F H RP L K EY S MmLRRC8E C KF F AI L VY N LI YV EK ISF L V AC R VE TSE IT GY AS F CC N HT KA H LF SK L A FC Y I SF V C VYG I T CL Y TL YW L F H RP L K EY S IL2H1 IL2H2 IL2H3 IL2H4 LRRNT LRR1

α10 α11 α12 α13 α14 β4

360 370 380 390 400 410 420 430 HsLRRC8A F E S IR EES SY SDIPDVKNDFAF MLH LI DQYD PLYSKRF A VFLSEVSE NK L RQLN LN NEWT L DK L RQRLTK N A QD KL EL HL MmLRRC8A F E S IR EES SY SDIPDVKNDFAF MLH LI DQYD PLYSKRF A VFLSEVSE NK L RQLN LN NEWT L DK L RQRLTK N A QD KL EL HL HsLRRC8B F E A LR E K S NY SDIPDVKNDFAF ILH LA DQYD PLYSKRF S IFLSEVSE NK L KQIN LN NEWT V EK L KSKL V KN A QD KI EL HL MmLRRC8B F E A LR E K S NY SDIPDVKNDFAF ILH LA DQYD PLYSKRF S IFLSEVSE NK L KQIN LN NEWT V ER L KSKL V KN SQD KV EL HL HsLRRC8C F E Y VR Q ET GID DIPDVKNDFAF MLH MI DQYD PLYSKRF A VFLSEVSE NK L KQLN LN NEWT P DK L RQKLQ TN A HN RL EL PL MmLRRC8C F E Y VR Q ET GID DIPDVKNDFAF MLH MI DQYD PLYSKRF A VFLSEVSE NK L KQLN LN NEWT P DK L RQKLQ TN A HN RL EL PL HsLRRC8D F E K VR EES SF SDIPDVKNDFAF LLH MV DQYD QLYSKRF G VFLSEVSE NK L R E IS LN HEWT F EK L RQHISR N A QD K QEL HL MmLRRC8D F E K VR EES SF SDIPDVKNDFAF LLH MV DQYD QLYSKRF G VFLSEVSE NK L R E IS LN HEWT F EK L RQHVSR N A QD K QEL HL HsLRRC8E F RS VR EET GMG DIPDVKNDFAF MLH LI DQYD SLYSKRF A VFLSEVSE SR L KQLN LN HEWT P EK L RQKLQR N A AG RL EL AL MmLRRC8E F RS VR EET GM NDIPDVKNDFAF MLH LI DQYD SLYSKRF A VFLSEVSE SR L KQLN LN HEWT P EK L RQKLQR N M RG RL EL SL

Kasuya et al. Figure S2 bioRxiv preprint doi: https://doi.org/10.1101/331207; this version posted May 25, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

LRR2 LRR3 LRR4 η1 β5 η2 β6 α15 β7 TT TT 440 450 460 470 480 490 500 510 HsLRRC8A FML SG IPD TVF DL V ELEV L KLE LI P DV T IP PS I AQ L TG L KEL W L Y H TA AKI EAP A LA FL RE NL RALHI K F T DI K EI P LW I MmLRRC8A FML SG IPD TVF DL V ELEV L KLE LI P DV T IP PS I AQ L TG L KEL W L Y H TA AKI EAP A LA FL RE NL RALHI K F T DI K EI P LW I HsLRRC8B FML NG LPD NVF ELTEMEV L SLE LI P EV K LP SA V SQ L V NL KEL R V Y H SS L V V DHP A LA FL E E NL KILRL K F T EM GK IP RW V MmLRRC8B FML NG LPD NVF ELTEMEV L SLE LI P EV K LP AA V AQ L V NL REL H V Y H SS L V V DHP A LA FL E E NL RILRL K F T EM GK IP RW V HsLRRC8C IML SG LPD TVF EITEL QS L KLE II KN V M IP AT I AQ L D NL QEL S L HQCS VKI HSA A LS FL KE NL KVL S VK F D DM R EL P PW M MmLRRC8C IML SG LPD TVF EITEL QS L KLE II KN V M IP AT I AQ L D NL QEL C L HQCS VKI HSA A LS FL KE NL KVL S VK F D DM R EL P PW M HsLRRC8D FML SG VPD AVF DLTDLDV L KLE LI P EA K IP AK I SQ M T NL QEL H L C H CP AKV EQT A FS FL RD HL R C LHV K F T DV A EI P AW V MmLRRC8D FML SG VPD AVF DLTDLDV L KLE LI P EA K IP AK I SQ M T NL QEL H L C H CP AKV EQT A FS FL RD HL R C LHV K F T DV A EI P AW V HsLRRC8E CML PG LPD TVF ELSEVE SL RLE AI C DI TF P PG L SQ L VH L QEL S L L H SP ARL PFS L QV FL RD HL KVMRV K CE EL R EV P LW V MmLRRC8E CML PG LPD TVF ELSEVEA L RLE AI C DI SF P PG L SQ L V NL QEL S L L H SP ARL PFSSQI FL RD RL KVI C VK F E EL R EV P LW V LRR5 LRR6 LRR7 β8 α16 η3 β9 η4 β10 α17 TT TT TT TT 520 530 540 550 560 570 580 590 HsLRRC8A Y SL KT L EEL HL TG N L SA E NN R Y I V ID G LRE L K RL KV L R LK S NLSKL P Q V VT D V GV HL QKL S I NN EG TK L IV LN SLKK MA N MmLRRC8A Y SL KT L EEL HL TG N L SA E NN R Y I V ID G LRE L K RL KV L R LK S NLSKL P Q V VT D V GV HL QKL S I NN EG TK L IV LN SLKK MV N HsLRRC8B F HL KN L KEL YL SG C V LP E QLST M Q LE GFQ DL K NL R TL Y LK S SLSRI P Q V VT D L LPS L QKL S L DN EG SK L VV LN NLKK MV N MmLRRC8B F HL KN L KEL YL SG C V LP E QLSS L H LE GFQ DL K NL R TL Y LK S SLSRI P Q V VT D L LPS L QKL S L DN EG SK L VV LN NLKK MV N HsLRRC8C Y GL RN L EEL YL VG S L SH D IS R N V T LE S LRD L K SL KI L S IK S NVSKI P Q A V VD V SS HL QKM C I HN DG TK L VM LN NLKK M T N MmLRRC8C Y GL RN L EEL YL VG S L SH D IS K N V T LE S LRD L K SL KI L S IK S NVSKI P Q A V VD V SS HL QKM C V HN DG TK L VM LN NLKK M T N HsLRRC8D Y LL KN L REL YL IG N L NS E NN K M I G LE S LRE L R HL KI L H VK S NLTKV P S N IT D V AP HL TKL V I HN DG TK L LV LN SLKK MM N MmLRRC8D Y LL KN L REL YL IG N L NS E NN K M I G LE S LRE L R HL KI L H VK S NLTKV P S N IT D V AP HL TKL V I HN DG TK L LV LN SLKK MM N HsLRRC8E F GL R GL EEL HL EG LFPQ E LA R A A T LE S LRE L K QL KV L S LR S NA G KV P AS VT D V AG HL QRL S L HN DG A RL VA LN SLKK LA A MmLRRC8E F GL R GL EEL HL EG LFPP E MA R G A T LE S LRE L K QL KV L S LR S NA G KV P AS VT D V AG HL QRL S L HN DG A RL LA LN SLKK LA V LRR8 LRR9 LRR10 LRR11

β11 η5 β12 α18 β13 η6 β14 TT TT 600 610 620 630 640 650 660 670 HsLRRC8A L T ELEL I RC DLERIPH S IFSL H NL QE IDL KD N NLKTI EEI ISFQH LHR L T CL KLW YN HI AYI P IQ I GN L T NLE RL Y L NR N MmLRRC8A L T ELEL I RC DLERIPH S IFSL H NL QE IDL KD N NLKTI EEI ISFQH LHR L T CL KLW YN HI AYI P IQ I GN L T NLE RL Y L NR N HsLRRC8B L KS LEL I SC DLERIPH S IFSL N NL HE LDL RE N NLKTV EEI ISFQH L QN L S CL KLW HN NI AYI P AQ I GA L S NLE QL SLD HN MmLRRC8B L KS LEL L SC DLERIPH S IFSL N NL HE LDL KE N NLKTV EEI ISFQH L PS L S CL KLW HN NI AYI P AQ I GA L S NLE QL F LG HN HsLRRC8C L T ELEL V HC DLERIPH A VFSL L SL QE LDL KE N NLKSI EEI VSFQH LRK L T VL KLW HN SI T YI P EH I KK L T SLE RL SF SH N MmLRRC8C L T ELEL V HC DLERIPH A VFSL L SL QE LDL KE N NLKSI EEI VSFQH LRK L T VL KLW YN SI AYI P EH I KK L T SLE RL F F SH N HsLRRC8D V A ELEL QN C ELERIPH A IFSL S NL QE LDL K SN NIRTI EEI ISFQH LKR L T CL KLW HN KI V T IP PS I TH V K NLE SL Y F S NN MmLRRC8D V A ELEL QN C ELERIPH A IFSL S NL QE LDL K SN NIRTI EEI ISFQH LKR L T CL KLW HN KI V A IP PS I TH V K NLE SL Y F S NN HsLRRC8E L R ELEL V AC GLERIPH A VFSL GA L QE LDL KD N H LRSI EEI LSFQH C RK L VT L RLW HN QI AYV P EH V RK L R SLE QL Y L S YN MmLRRC8E L R ELEL V AC GLERIPH A IFSL GA L QE LDL KD N H LRSI EEI LSFQH C RK L VT L RLW HN QI AYV P EH V RK L R SLE QL Y L SH N LRR12 LRR13 LRR14

η7 β15 η8 β16 η9 β17 η10 TT TT TT 680 690 700 710 720 730 740 750 HsLRRC8A KI E K IP T Q LF Y C R KLR YLD LS HN N L TF LP A DIGL L QN LQ NLA IT AN R IE TLP PE L F QC RKL R AL H LG NN V LQS L PS RVG E MmLRRC8A KI E K IP T Q LF Y C R KLR YLD LS HN N L TF LP A DIGL L QN LQ NLA VT AN R IE ALP PE L F QC RKL R AL H LG NN V LQS L PS RVG E HsLRRC8B N IE N LP LQ LF L CTKLH YLD LS YN H L TF IP E EI QY L SN LQ YFA VT NN N IE MLP DG L F QC KKL QC L L LG KN S L M NL SP HVG E MmLRRC8B N IE S LP LQ LF L CTKLH YLD LS YN H L TF IP E EI QY L TN LQ YFA VT NN N IE MLP DG L F QC KKL QC L L LG RN S LT DL SPL VG E HsLRRC8C KI E V LP S H LF L CNKIR YLD LS YN D I RF IP P EIGV L QS LQ YFS IT CN K VE SLP DE L Y FC KKL K TL K IG KN S LS VL SP KIG N MmLRRC8C KV E V LP S H LF L CNKIR YLD LS YN D I RF IP P EIGV L QS LQ YFS IT CN K VE SLP DE L Y FC KKL K TL K IG KN S LS VL SP KIG N HsLRRC8D KL E S LP VA VF SL QKLR CLD VS YN N I SM IP I EIGL L QN LQ HLH IT GN K VD ILP KQ L F KC IKL R TL N LG QN C ITS L PE KVG Q MmLRRC8D KL E S LP T A VF SL QKLR CLD VS YN N I ST IP I EIGL L QN LQ HLH IT GN K VD ILP KQ L F KC VKL R TL N LG QN C I A SL PE KI S Q HsLRRC8E KL E T LP S Q L GL CS G LR LLD VS HN G L HS LP P EVGL L QN LQ HLA LS YN A LE ALP EE L F FC RKL R TL L LG DN Q LSQ L SP HVG A MmLRRC8E KL E T LP T Q L GQ C FG LR LLD LS HN G L RS LP P ELGL L QS LQ HLA LS YN A LE SLP DE L F FC HKL R TL L LG YN H LTQ L SPD V A A LRR15 LRRCT

β18 η11 η12 α19 α20

760 770 780 790 800 810 HsLRRC8A L T NL T Q IELR GN R LE CLP V EL G EC PL LK RSG L VVE E DL F N TLP P EV KE R L WRADKEQA.... MmLRRC8A L T NL T Q IELR GN R LE CLP V EL G EC PL LK RSG L VVE E DL F S TLP P EV KE R L WRADKEQA.... HsLRRC8B L S NL T H LEL IGN Y LE TLP P EL EG C QS LK RN CL IVE E NL LN TLP LP V T E R L QTCLDKC..... MmLRRC8B L S NL T H LEL TGN Y LE TLP V EL EG C QS LK RS CL IVE D SL LN SLP LP V A E R L QTCLDKC..... HsLRRC8C L LF L S Y LDVK GN HF E ILP P EL G DC RA LK R A GL VVE D AL F E TLP S DV RE Q M KTE...... MmLRRC8C L LF L S Y LDIK GN HF E VLP P EL G DC RA LK R A GL VVE D AL F E TLP S DV RE Q M KAD...... HsLRRC8D L S QL T Q LELK GN C LD RLP AQ L G QC RM LK KSG L VVE D HL F D TLP L EV KE A L NQDINIPFANGI MmLRRC8D L T QL T Q LELK GN C LD RLP AQ L G QC RM LK KSG L VVE D QL F D TLP L EV KE A L NQDVNVPFANGI HsLRRC8E L RA L S R LELK GN R LE ALP E EL G NC GG LK K A GL LVE D TL Y QG LP A EV RD K M EEE...... MmLRRC8E L Q AL S R LELK GN R LE TLP E EL G DC KG LK KSG L LVE D TL Y EG LP A EV RE K M EEE......

Kasuya et al. Figure S2 (continued) bioRxiv preprint doi: https://doi.org/10.1101/331207; this version posted May 25, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. A

50 nm

B 2D classification Manual screening Auto picking (3 rounds, 4 batches) 5,304 micrographs 5,284 micrographs 1,421,314 particles

3D classification (8 classes)

664,971 particles

Class #1 Class #2 Class #3 Class #4 Class #5 Class #6 Class #7 Class #8 (14.2%) (10.5%) (8.6%) (17.8%) (9.5%) (12.0%) (16.7%) (10.6%)

90° 90° 90° 90° 90° 90° 90° 90°

3D auto refinement 3D auto refinement

Overall (4.25 Å) C Overall (9.17 Å)

Kasuya et al. Figure S3 bioRxiv preprint doi: https://doi.org/10.1101/331207; this version posted May 25, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. D E

1.0 1.0 Model vs. Sum Model vs. Half map 1 Model vs. Half map 2 0.8 0.8

0.6 0.6

0.4 0.4

0.2 0.2 FSC = 0.143

Fourier Shell Correlation 0.0 Fourier Shell Correlation 0.0

0.00 0.05 0.10 0.15 0.20 0.25 0.30 0.35 0.00 0.05 0.10 0.15 0.20 0.25 0.30 0.35 Resolution (1/Å) Resolution (1/Å)

Kasuya et al. Figure S3 (continued) bioRxiv preprint doi: https://doi.org/10.1101/331207; this version posted May 25, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

A B C region Extracellular TM region

Extracellular TM region region

D E region Intracellular LRR region

α subunit β subunit Intracellular LRR region region

Kasuya et al. Figure S4 bioRxiv preprint doi: https://doi.org/10.1101/331207; this version posted May 25, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

A Tight interface B Loose interface TM2 (β) T316 TM2 (α) T316

Y124 I320 Y124 I320 Y127 F324 Y127 F324 TM region L131 L131

L134 V34 L134 V34

Y30 A138 Y30 A138 TM4 (α) TM4 (ζ) F27 F27 F142 F142 W23 W23 F146 F146 TM1 (α) P147 TM1 (ζ) C P147 D IL1H1 (β) IL2H2 (α) Y382 IL1H1 (α) S151 Y382 IL2H2 (ζ) E154 S151 D383 E154 H155 D383 H155 L385 L385 R389 Intracellular

R389 region K249 K249 T170 T170 E245 E245 S174 IL2H3 (α) S174 IL2H3 (ζ) E238 E238 IL1H2 (α) IL1H3 (ζ) IL1H3 (α) IL1H3 (β) D233 D233

E F

LRR (ζ) LRR region

LRR (α) LRR (α) LRRCT LRR (β)

G H

Y106 H104 L302 C57 EL2 (β) EL2 (α) C57

Y106 Extracellular H104 L302 region R309 R309 Y108 Y108

A111 A311 A111 A311 EL2 (α) P313 EL2 (ζ) P313 EL1H (β) E115 EL1H (α) E115

Tight interface Loose interface

Kasuya et al. Figure S5 bioRxiv preprint doi: https://doi.org/10.1101/331207; this version posted May 25, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

A WT D102A R103A H104A Q105A Y106A

Hoechst LRRC8A

DIC

B C YFP D102A (H148Q/I152L) R103A WT HsLRRC8A-EGFP LRRC8A: D102A R103A H104A Q105A Y106A H104A Q105A Y106A † 100 IB: LRRC8A 75

EGFP

IB: YFP 28 Void Normalized fluorescence

48 0 IB: Actin 0 5 10 15 20 25 (kDa) Volume (ml)

D WT (+) WT (++++) D102A 1.4 - I--iso I--hypo 1.2 Regression curve

1.0

0.8

0.6

0.4 R103A H104A Q105A Y106A 1.4

1.2

1.0 Normalized YFP fluorescence (a.u.)

0.8

0.6

0.4 0 100 200 300 400 0 100 200 300 400 0 100 200 300 400 0 100 200 300 400 Time after adding iodide-containing buffer (Sec)

Kasuya et al. Figure S6 bioRxiv preprint doi: https://doi.org/10.1101/331207; this version posted May 25, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

A HsLRRC8A B CeInnexin-6 C HsConnexin-26 c EL1 EL2 C310 C54 EL1 EL2 EL2 C295 EL1

N C113 TM3 TM3 TM3 TM1 TM1 TM1 NTH N TM4 TM4 TM2 TM2 TM4 NTH N C TM2

IL IL2 IL1 IL1 C IL2

C

D E F

HsLRRC8A CeInnexin-6 HsConnexin-26 G H I J HsLRRC8A EL1H EL1H Pred: TM1 EL1H AA: MIPVTELRYFADTQPAYRILKPWWDVFTDYISIVMLMIAV 10 20 30 40 N

TM1 TM1 TM1 CeInnexin-6 Pred: N NTH TM1 NTH AA: MASQVGAINSVNALISRVFVQPKGDLADRLNSRVTVVILA N 10 20 30 40 NTH HsConnexin-26 Pred: NTH TM1 AA: MDWGTLQTILGGVNKHSTSIGKIWLTVLFIFRIMILVVAA 10 20 30 40 HsLRRC8A CeInnexin-6 HsConnexin-26

Kasuya et al. Figure S7 bioRxiv preprint doi: https://doi.org/10.1101/331207; this version posted May 25, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

A

Out

In Conserved

180° Diversified

B

Out

In 90°

F143 (LRRC8D)

LRRC8A LRRC8D

Kasuya et al. Figure S8 bioRxiv preprint doi: https://doi.org/10.1101/331207; this version posted May 25, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

Table S1. Data collection and model statistics Data collection Microscope FEI Tecnai Arctica Voltage (kV) 200 Detector Gatan K2 Summit Pixel size (Å)* 1.490 Nominal magnification* 23,500x Electron dose (e-/Å2)* 50 Defocus range (mm) -0.5 ~ -3.5 Number of collected micrographs 5304 Number of selected micrographs 5284

Reconstitution Number of used particles 164,749 Symmetry imposed C3 Resolution 4.25

Refinement Number of protein residues 4,227 R.m.s. deviations Bond lengths (Å) 0.006 Bond angles (°) 1.124 Ramachandran (%) Favored 88.74 Allowed 11.26 Outlier 0.00 * Calibrated pizel size at the detector