structural biology cryo-em structure of a mitochondrial ... · cryo-em structure of a mitochondrial...

7
STRUCTURAL BIOLOGY Cryo-EM structure of a mitochondrial calcium uniporter Jiho Yoo 1 *, Mengyu Wu 2 *, Ying Yin 1 , Mark A. Herzik Jr. 2 , Gabriel C. Lander 2 , Seok-Yong Lee 1 Calcium transport plays an important role in regulating mitochondrial physiology and pathophysiology. The mitochondrial calcium uniporter (MCU) is a calcium-selective ion channel that is the primary mediator for calcium uptake into the mitochondrial matrix. Here, we present the cryoelectron microscopy structure of the full-length MCU from Neurospora crassa to an overall resolution of ~3.7 angstroms. Our structure reveals a tetrameric architecture, with the soluble and transmembrane domains adopting different symmetric arrangements within the channel. The conserved W-D-F-F-E-P-V-T-Y sequence motif of MCU pore forms a selectivity filter comprising two acidic rings separated by one helical turn along the central axis of the channel pore.The structure combined with mutagenesis gives insight into the basis of calcium recognition. M itochondrial Ca 2+ transport is critical for shaping the dynamics of intracellular calcium signaling, regulating energy me- tabolism, generating reactive oxygen species, and modulating cell death (1, 2). Ca 2+ uptake across the mitochondrial inner mem- brane was shown to occur via a uniporter(3), and electrophysiological studies of the mitoplast inwardly-rectifying Ca 2+ current (I MICA ) showed that this uniporteris an ion channel that ex- hibits a remarkable selectivity for Ca 2+ (4). Re- cent genomics studies have identified the key components of the uniporter holocomplex (uni- plex) (59). In vertebrates, it comprises the mitochondrial calcium uniporter (MCU), an auxiliary transmembrane protein essential MCU regulator (EMRE) (8), and the auxiliary EF handcontaining proteins mitochondrial calcium uptake 1 (MICU1) and MICU2 (6, 10). EMRE plays a dual role in maintaining MCU in an open state and recruiting MICU1 and MICU2 (11), which regulate the activity of MCU in a Ca 2+ -dependent manner (6, 1215). Whereas MCU is widely dis- tributed across all major branches of eukaryotes (16), EMRE is metazoan-specific (8). The MCU ortholog from Dictyostelium discoideum, an or- ganism lacking EMRE, alone is capable of re- constituting channel activity in yeast (17). In fungi, MCU is typically present without the MICU1/2 homologs (16). Recent studies have further shown that fungal MCU homologs are also able to re- constitute channel activity in vitro and in vivo on their own (18, 19). Taken together, these data establish that MCU is the Ca 2+ -conducting pore- forming unit of the uniplex. A single protomer of MCU is predicted to possess two transmembrane helices (TM1 and TM2), two coiled-coils (CC1 and CC2), and an N-terminal domain (NTD) located on the matrix side. All MCU homologs contain a highly con- served sequence motif W-D- F- F-E-P-V-T-Y ( F denotes hydrophobic amino acids) located between TM1 and TM2. This motif is oriented facing the intermembrane space and is thought to form the selectivity filter in the oligomeric channel (5, 7, 9). The NTD is composed of ~100 residues and extends into the mitochondrial matrix. Crystallographic studies of the human MCU NTD revealed a distinct structural fold similar to a b-grasp, and subsequent functional studies revealed its modulatory role in MCU function (20, 21). A structure of the NTD-deleted MCU homolog from Caenorhabditis elegans (cMCU-DNTD) was recently determined by using nuclear magnetic resonance (NMR) and negative- stain electron microscopy, revealing a pentameric architecture (22), although the absence of the NTD limited further structural insights into the full-length channel assembly. We therefore con- ducted structural studies of a full-length MCU homolog using cryoelectron microscopy (cryo- EM). After screening a number of MCU homologs based on phylogenetic analyses (16), we found that a homolog from Neurospora crassa (MCU NC ) was suitable for structural studies. To prevent proteolysis, we introduced a Tyr 232 Ala mutation into a flexible loop within the NTD (fig. S1). This Tyr 232 Ala mutant eluted at a similar volume as that of wild-type MCU NC during size-exclusion chromatography and exhibited a similar overall architecture, as determined with negative-stain EM (fig. S2). MCU NC was prepared under two different Ca 2+ conditions for EM studies, referred to as low and high Ca 2+ (supplementary materials, materials and methods). In both conditions, MCU NC was reconstituted in amphipol and sub- jected to single-particle three-dimensional (3D) cryo-EM analysis. MCU NC unambiguously showed by means of EM a tetrameric arrangement under both Ca 2+ conditions (fig. S3). The attainable resolution of these reconstructions (4.7 to 7 Å) (fig. S3 and supplementary materials, materials and methods) appeared to be limited by flexibility of the NTD. Therefore, we attempted to cross-link the NTD of MCU NC using the water-soluble cross- linker bis-sulfosuccinimidyl suberate (BS3), in the presence of high Ca 2+ , and reconstituted the BS3cross-linked MCU NC into nanodiscs for EM analy- ses. The 3D reconstruction of cross-linked MCU NC in nanodiscs was determined to an overall re- solution of ~3.7 Å (fig. S4). Comparison of the 3D reconstructions of cross-linked MCU NC and native MCU NC showed that cross-linking did not appreciably affect the structure of MCU NC (fig. S4). Thus, data of cross-linked and native MCU NC were combined to yield an improved reconstruction at an overall resolution of ~3.7 Å (fig. S4 and supplementary materials, materials and methods) that allowed for de novo model building (table S1 and supplementary materials, materials and methods). SDSpolyacrylamide gel electrophoresis (SDS- PAGE) analysis of BS3cross-linked MCU NC in- dicated a complete conversion of the monomeric MCU band to a single band with an apparent molecular weight most compatible with tetrameric MCU NC (fig. S2). Both cross-linked and native MCU NC in detergent micelles elute at similar volumes during size-exclusion chromatography, suggesting that cross-linking did not alter the oligomeric state of MCU NC . Last, 2D classifica- tion and asymmetric 3D classification of both the native and cross-linked MCU NC samples un- ambiguously showed a tetrameric organization of MCU NC , with no evidence for a pentamer, strongly supporting the tetrameric architecture of MCU NC in our final structures (figs. S3 and S4). The overall shape of the MCU NC homo- tetramer is a prolate spheroid with dimensions of ~40 by 48 by 130 Å (Fig. 1). The transmembrane domain (TMD) is formed by TM1 and TM2 helices, and the matrix region comprises the coiled-coil domain (CCD) and the NTD. TM1 and CC1 form a long and continuous helix at the periphery of the channel, while the TM2 helices line the central symmetry axis (Fig. 1B). The TM helices are arranged so that TM1 from one protomer pri- marily interacts with TM2 from the adjacent protomer (Fig. 1E). TM2 is followed by ~15 amino acids near the matrix-membrane interface but could not be accurately modeled (fig. S6). Im- mediately C-terminal to this region, a short helix, which we termed the junctional helix (JH), is positioned nearly perpendicular to TM1 and TM2 and forms a junction between TM2 and CC2. In the CCD, CC1 and CC2 form a dimeric coiled-coil, resulting in four dimeric coiled-coils within the tetramer. Because of the uncertainty of the con- nection between TM2 and JH, we cannot rule out the possibility that CC2 interacts with CC1 from the neighboring protomer (fig. S6). Unlike human MCU (hMCU), CC2 of MCU NC is followed by three putative helical regions at its C terminus, which is not resolved in the 3D reconstruction (fig. S1). The NTD comprises six b-strands (b1 to b6) and two a-helices (a1 and a2) and is con- nected to CC1 of the CCD. a2, located between the CCD and the rest of the NTD, positions the RESEARCH Yoo et al., Science 361, 506511 (2018) 3 August 2018 1 of 6 1 Department of Biochemistry, Duke University School of Medicine, Durham, NC 27710, USA. 2 Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA. *These authors contributed equally to this work. Corresponding author. Email: [email protected] (S.-Y.L.); [email protected] (G.C.L.) on February 1, 2021 http://science.sciencemag.org/ Downloaded from

Upload: others

Post on 29-Sep-2020

0 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: STRUCTURAL BIOLOGY Cryo-EM structure of a mitochondrial ... · Cryo-EM structure of a mitochondrial calcium uniporter Jiho Yoo 1*, Mengyu Wu 2*, Ying Yin , Mark A. Herzik Jr. , Gabriel

STRUCTURAL BIOLOGY

Cryo-EM structure of a mitochondrialcalcium uniporterJiho Yoo1*, Mengyu Wu2*, Ying Yin1, Mark A. Herzik Jr.2,Gabriel C. Lander2†, Seok-Yong Lee1†

Calcium transport plays an important role in regulating mitochondrial physiology andpathophysiology.The mitochondrial calcium uniporter (MCU) is a calcium-selective ionchannel that is the primary mediator for calcium uptake into the mitochondrial matrix. Here,we present the cryo–electron microscopy structure of the full-length MCU from Neurosporacrassa to an overall resolution of ~3.7 angstroms. Our structure reveals a tetramericarchitecture, with the soluble and transmembrane domains adopting different symmetricarrangements within the channel.The conserved W-D-F-F-E-P-V-T-Ysequence motif of MCUpore forms a selectivity filter comprising two acidic rings separated by one helical turn alongthe central axis of the channel pore.The structure combined with mutagenesis gives insightinto the basis of calcium recognition.

Mitochondrial Ca2+ transport is critical forshaping the dynamics of intracellularcalcium signaling, regulating energyme-tabolism, generating reactive oxygenspecies, and modulating cell death (1, 2).

Ca2+ uptake across themitochondrial innermem-brane was shown to occur via a “uniporter” (3),and electrophysiological studies of the mitoplastinwardly-rectifying Ca2+ current (IMICA) showedthat this “uniporter” is an ion channel that ex-hibits a remarkable selectivity for Ca2+ (4). Re-cent genomics studies have identified the keycomponents of the uniporter holocomplex (uni-plex) (5–9). In vertebrates, it comprises themitochondrial calcium uniporter (MCU), anauxiliary transmembrane protein essential MCUregulator (EMRE) (8), and the auxiliary EFhand–containing proteins mitochondrial calciumuptake 1 (MICU1) andMICU2 (6, 10). EMRE playsa dual role in maintaining MCU in an open stateand recruiting MICU1 and MICU2 (11), whichregulate the activity ofMCU in a Ca2+-dependentmanner (6, 12–15). Whereas MCU is widely dis-tributed across all major branches of eukaryotes(16), EMRE is metazoan-specific (8). The MCUortholog from Dictyostelium discoideum, an or-ganism lacking EMRE, alone is capable of re-constituting channel activity in yeast (17). In fungi,MCU is typically present without the MICU1/2homologs (16). Recent studies have further shownthat fungal MCU homologs are also able to re-constitute channel activity in vitro and in vivoon their own (18, 19). Taken together, these dataestablish that MCU is the Ca2+-conducting pore-forming unit of the uniplex.A single protomer of MCU is predicted to

possess two transmembrane helices (TM1 and

TM2), two coiled-coils (CC1 and CC2), and anN-terminal domain (NTD) located on the matrixside. All MCU homologs contain a highly con-served sequence motif W-D-F-F-E-P-V-T-Y(F denotes hydrophobic amino acids) locatedbetween TM1 and TM2. This motif is orientedfacing the intermembrane space and is thoughtto form the selectivity filter in the oligomericchannel (5, 7, 9). The NTD is composed of ~100residues and extends into the mitochondrialmatrix. Crystallographic studies of the humanMCU NTD revealed a distinct structural foldsimilar to a b-grasp, and subsequent functionalstudies revealed its modulatory role in MCUfunction (20, 21). A structure of the NTD-deletedMCU homolog from Caenorhabditis elegans(cMCU-DNTD)was recently determined by usingnuclearmagnetic resonance (NMR) and negative-stain electron microscopy, revealing a pentamericarchitecture (22), although the absence of theNTD limited further structural insights into thefull-length channel assembly. We therefore con-ducted structural studies of a full-length MCUhomolog using cryo–electron microscopy (cryo-EM). After screening a number of MCU homologsbased on phylogenetic analyses (16), we foundthat a homolog fromNeurospora crassa (MCUNC)was suitable for structural studies. To preventproteolysis, we introduced a Tyr232Ala mutationinto a flexible loop within the NTD (fig. S1). ThisTyr232Ala mutant eluted at a similar volume asthat of wild-type MCUNC during size-exclusionchromatography and exhibited a similar overallarchitecture, as determined with negative-stainEM (fig. S2). MCUNC was prepared under twodifferent Ca2+ conditions for EM studies, referredto as low andhighCa2+ (supplementarymaterials,materials and methods). In both conditions,MCUNC was reconstituted in amphipol and sub-jected to single-particle three-dimensional (3D)cryo-EM analysis. MCUNC unambiguously showedby means of EM a tetrameric arrangement underboth Ca2+ conditions (fig. S3). The attainableresolution of these reconstructions (4.7 to 7 Å)

(fig. S3 and supplementary materials, materialsandmethods) appeared to be limited by flexibilityof the NTD. Therefore, we attempted to cross-linkthe NTD of MCUNC using the water-soluble cross-linker bis-sulfosuccinimidyl suberate (BS3), in thepresence of high Ca2+, and reconstituted the BS3–cross-linkedMCUNC into nanodiscs for EM analy-ses. The 3D reconstruction of cross-linkedMCUNC

in nanodiscs was determined to an overall re-solution of ~3.7 Å (fig. S4). Comparison of the3D reconstructions of cross-linked MCUNC andnative MCUNC showed that cross-linking didnot appreciably affect the structure of MCUNC

(fig. S4). Thus, data of cross-linked and nativeMCUNC were combined to yield an improvedreconstruction at an overall resolution of ~3.7 Å(fig. S4 and supplementary materials, materialsand methods) that allowed for de novo modelbuilding (table S1 and supplementary materials,materials and methods).SDS–polyacrylamide gel electrophoresis (SDS-

PAGE) analysis of BS3–cross-linked MCUNC in-dicated a complete conversion of the monomericMCU band to a single band with an apparentmolecular weightmost compatiblewith tetramericMCUNC (fig. S2). Both cross-linked and nativeMCUNC in detergent micelles elute at similarvolumes during size-exclusion chromatography,suggesting that cross-linking did not alter theoligomeric state of MCUNC. Last, 2D classifica-tion and asymmetric 3D classification of boththe native and cross-linkedMCUNC samples un-ambiguously showed a tetrameric organization ofMCUNC,with no evidence for a pentamer, stronglysupporting the tetrameric architecture of MCUNC

in our final structures (figs. S3 and S4).The overall shape of the MCUNC homo-

tetramer is a prolate spheroid with dimensionsof ~40 by 48 by 130Å (Fig. 1). The transmembranedomain (TMD) is formedby TM1 andTM2helices,and the matrix region comprises the coiled-coildomain (CCD) and the NTD. TM1 and CC1 forma long and continuous helix at the periphery ofthe channel,while the TM2helices line the centralsymmetry axis (Fig. 1B). The TM helices arearranged so that TM1 from one protomer pri-marily interacts with TM2 from the adjacentprotomer (Fig. 1E). TM2 is followed by ~15 aminoacids near the matrix-membrane interface butcould not be accurately modeled (fig. S6). Im-mediately C-terminal to this region, a short helix,which we termed the junctional helix (JH), ispositioned nearly perpendicular to TM1 and TM2and forms a junction between TM2 and CC2. Inthe CCD, CC1 and CC2 form a dimeric coiled-coil,resulting in four dimeric coiled-coils within thetetramer. Because of the uncertainty of the con-nection between TM2 and JH, we cannot ruleout the possibility that CC2 interacts with CC1from the neighboring protomer (fig. S6). UnlikehumanMCU (hMCU), CC2 ofMCUNC is followedby three putative helical regions at its C terminus,which is not resolved in the 3D reconstruction(fig. S1). The NTD comprises six b-strands (b1 tob6) and two a-helices (a1 and a2) and is con-nected to CC1 of the CCD. a2, located betweenthe CCD and the rest of the NTD, positions the

RESEARCH

Yoo et al., Science 361, 506–511 (2018) 3 August 2018 1 of 6

1Department of Biochemistry, Duke University School ofMedicine, Durham, NC 27710, USA. 2Department ofIntegrative Structural and Computational Biology, TheScripps Research Institute, La Jolla, CA 92037, USA.*These authors contributed equally to this work.†Corresponding author. Email: [email protected] (S.-Y.L.);[email protected] (G.C.L.)

on February 1, 2021

http://science.sciencem

ag.org/D

ownloaded from

Page 2: STRUCTURAL BIOLOGY Cryo-EM structure of a mitochondrial ... · Cryo-EM structure of a mitochondrial calcium uniporter Jiho Yoo 1*, Mengyu Wu 2*, Ying Yin , Mark A. Herzik Jr. , Gabriel

NTD relative to the rest of the channel (Fig. 1, Cto E). Despite the low sequence identity (~17%for the structured regions), the NTD is struc-turally very similar to the hMCU NTD fragment[Protein Data Bank (PDB) ID: 5KUJ, Ca rootmean square deviation 1.8 Å] (figs. S1 and S7)(20, 21).All MCU homologs contain the highly con-

served sequencemotif W-D-F-F-E-P-V-T-Y, whichhas been proposed to form the selectivity filter(5, 7, 22). In our MCUNC structure, this sequencemotif is located in the N-terminal region of TM2(Fig. 2), with the carboxylate side chains of con-served acidic residues Asp355 and Glu358 fromeach protomer directed toward the central sym-metry axis, forming two acidic rings along thechannel pore. The first acidic ring formed byAsp355, which we term site 1 (S1), is located atthe mouth of the pore and exposed to the inter-membrane space. The distances between diago-nally positioned Asp355 are ~8.8 Å, indicatingthat a hydrated Ca2+ is likely to bind to this ring(Fig. 2C and fig. S1). The second acidic ringformed by Glu358 is termed site 2 (S2). There isan extensive network of interactions surround-ing Glu358 involving residues in the W-D-F-F-E-P-V-T-Y motif that position the carboxylate groupof Glu358 toward the central symmetry axis.Specifically, Pro359 appears to make interpro-tomer CH-p interactions with Trp354 from theadjacent subunit, the amide nitrogen in the in-dole ring of Trp354 hydrogen bonds to the car-boxylate group of Glu358 from an adjacentprotomer, and Tyr362 is involved in both p-pinteractions with Trp354 as well as hydrogenbonding interactions with Thr361 from the ad-jacent protomer (Fig. 2, C and D, and fig. S4).Although the EM density for the MCU motif issufficiently high to place side-chain atoms, theexact chemical nature of these interactionsshould be interpretedwith caution. The distancebetween the carboxylate groups of Glu358 fromdiagonally opposing protomers is ~4.8 Å, sug-gesting that only dehydrated Ca2+ can be co-ordinated at S2. There is a strong EM density atthe center of S2 (>17s) (fig. S8A) that we ten-tatively assign to Ca2+ because (i) the densitypeak is present in both an asymmetric recon-struction of MCUNC as well as the correspond-ing half maps (fig. S8, B and C), and (ii) calciumwas present in high concentration during bothprotein purification and EM grid preparation(supplementarymaterials,materials andmethods).The coordination of dehydrated Ca2+ by acidicresidues has been observed in both TRPV6and Orai1 and was proposed to be key to theCa2+ selectivity of these channels (23, 24). Wetherefore suggest that together, this S2 acidicring plays an important role in the selectiveCa2+ transport by MCU. Consistent with ourstructural observations, mutation of Glu358 hasbeen shown to abolish hMCU activity (22). Wealso observed two polar amino acids, Thr361

and Tyr362, located one helical turn below S2that line the central axis of the pore, which wetentatively term “site 3” (S3). The diagonal dis-tances between the side chains are large (13 to

14 Å), so S3 may play a role in hydrating Ca2+

ions exiting S2. We observed an EM density atthe center of S3, the identity of which is unclear(fig. S8, A to C). The selectivity filter organizationof MCUNC is in contrast with other classicaltetrameric cation channels, where the selectiv-ity filter is formed by loops connecting a TMhelix and a pore helix (25, 26).In order to investigate the role of the above-

described interactions of the W-D-F-F-E-P-V-T-Y

motif in Ca2+-permeation byMCU, we used site-directed mutagenesis and Ca2+-uptake assays.Although functional assays are not currentlyavailable for MCUNC, we exploited the high con-servation of the selectivity filter within the MCUfamily and used an established mitochondrialCa2+-uptake assay in human embryonic kidney(HEK) 293 cells lacking hMCU to test the activityof transfected hMCUmutants designed based onourMCUNC structure (11). Specifically, we tested

Yoo et al., Science 361, 506–511 (2018) 3 August 2018 2 of 6

90°

B

A

90°TM2

TM1

TM2

TM1 Matrix

IMS

Transmembranedomain

Coiled coil domain

N-terminaldomain

130

Å40 Å

48 Å

C D

TM2

CC1 CC2

NTD

C

α1

α2

β1 β2 β3

β4β5β6

N

CCD

TMD TM1

TM1 TM2

TMD

CC1 CC2

CCD

α2

α1

NTD

C

N

JH

JH

E

TM1

TM2

TM1

TM2

Intermembrane Space

Mitochondrial Matrix

90° 90°

α2

CC1

CC2

NTD

D E

Fig. 1. Overall architecture of MCUNC. (A) Single-particle cryo-EM reconstruction and (B) modelof MCUNC viewed from the intermembrane space (left), within the inner mitochondrial membrane(middle), and from the mitochondrial matrix (right). (C) Illustration outlining the protein domains andsecondary structures. (D) Detailed view of an MCU protomer as colored in (C). Dashed lines indicateparts that were not modeled in the structure. (E) Close-up views of interactions between TM2 andTM1 from neighboring protomers (top), between CCD and a2 from NTD (bottom).

RESEARCH | REPORTon F

ebruary 1, 2021

http://science.sciencemag.org/

Dow

nloaded from

Page 3: STRUCTURAL BIOLOGY Cryo-EM structure of a mitochondrial ... · Cryo-EM structure of a mitochondrial calcium uniporter Jiho Yoo 1*, Mengyu Wu 2*, Ying Yin , Mark A. Herzik Jr. , Gabriel

Yoo et al., Science 361, 506–511 (2018) 3 August 2018 3 of 6

B

C

TM2TM1

E358

Y362

D355

T361

S2

S1

TM2TM1

TM1TM2

Y362

T361TM2

E358W354

TM1

S1

S2E358

D355

L356

V357

T361Y362

A

TM2

TM1

D355

S1 S2

P359

P359

TM2TM1

TM2TM1

E

0.0

0.2

0.4

0.6

0.8

1.0

Rel

ativ

e C

a2+2+ u

ptak

e

hMC

U

W260A

W260F

T267A

Y268A

Y268F

HE

K293T

KO

hMC

U-R

u360

hMCU

W260AW260FT267AY268AY268F

HEK293T

KOhMCU-Ru360

CaCl2

0.0 0.5 1.0 1.5 2.0 2.5 3.00.0

0.2

0.4

0.6

0.8

1.0

1.2

Time (min)

Ext

ram

itoch

ondr

ial [

Ca2+

] (N

orm

aliz

ed A

.U.)

hMC

U

W26

0A

W26

0F

T26

7A

Y26

8A

Y26

8F

Anti-FLAG

β-actin

β-actin

Anti-hMCU

HE

K29

3T

KO

F

D

D355

TM2 (B) TM2 (A)

W354E358

Y362

T361

P359

S3

90°

45°

S3

0 1 2 3 4 5 60

5

10

15

20

25

Po

siti

on

alo

ng

th

e p

ore

)

Position radius (Å)

D355

E358

Y362

Fig. 2. Ion conduction pore and selectivity filter. (A) The ion conductionpathway of MCUNC and the pore radius along the central axis [generated withHOLE software (30)] indicate three constrictions at Asp355, Glu358, and Tyr362.Front and rear protomers were removed for clarity. (B) Putative Ca2+

coordination sites, among which a strong cryo-EM density peak was observedand tentatively modeled as Ca2+ at site 2. (C) Top views of the putativeCa2+ coordination sites constituted of Asp355 at S1 (left); Trp354, Glu358, andPro359 at S2 (middle); and Thr361 and Tyr362 at S3 (right). (D) Detailed viewfrom the membrane plane showing the extensive network of interactionsengaged by residues in the “W-D-F-F-E-P-V-T-Y” motif. The CH-p (Pro359

and Trp354) and p-p (Trp354 and Tyr362) interactions are highlighted by bluedashed lines. (E) Mitochondrial calcium uptake of human MCU (hMCU)

mutants from MCNNC structure-based mutagenesis at the ion conductionpore. Representative traces of calcium uptake in digitonin-permeabilizedcells after 10 mM CaCl2 was added. Mutation of Trp260 to Ala or Phe suppresseshMCU channel function, whereas mutation of Thr267 or Tyr268 to Ala solelyreduces the activity. Mutation of Tyr268 to Phe shows calcium uptake to thecomparable extent as wild-type hMCU (hMCU) expressed in MCU knockout(KO) cells. (F) Bar graph showing the calcium uptake of hMCUmutants relativeto the wild-type hMCU between 0.5 and 3 min time points (mean ± SEM,n ≥ 4 independent measurements).To detect the expression of hMCUmutants,cell lysates were analyzed by means of immunoblotting with antibody toFLAG.Wild-type HEK293Tcells and MCU knockout cells were confirmed bymeans of antibody to hMCU. b-actin was used as the loading control.

RESEARCH | REPORTon F

ebruary 1, 2021

http://science.sciencemag.org/

Dow

nloaded from

Page 4: STRUCTURAL BIOLOGY Cryo-EM structure of a mitochondrial ... · Cryo-EM structure of a mitochondrial calcium uniporter Jiho Yoo 1*, Mengyu Wu 2*, Ying Yin , Mark A. Herzik Jr. , Gabriel

the importance of Trp260, Thr267, and Tyr268

(Trp354, Thr361, and Tyr362 in MCUNC) for Ca2+

uptake by hMCU (Fig. 2, E and F). Mutation ofTrp260 (Trp354 in MCUNC) to either phenylalanineor alanine abolished hMCU-mediated Ca2+ up-take, andmutation of Thr267 (Thr361 inMCUNC)or Tyr268 (Tyr362 in MCUNC) to alanine reducedthe hMCU-mediated Ca2+ uptake substantially,indicating their importance for MCU function.Ca2+ uptake was restored in the Tyr268 (Tyr362 inMCUNC) to phenylalaninemutant, indicating thataromaticity is important for this position,which isin agreement with the above-described interac-tions. None of the tested mutations appreciablyaffected the expression level of hMCU comparedwith that of wild type (Fig. 2F).Viewing along the symmetry axis of the chan-

nel, a symmetry mismatch occurs in which theTMD adopts a fourfold symmetry, whereas theCCD and the NTD show a twofold symmetricorganization (Fig. 3). This observed symmetrymismatch is apparent in the 3D reconstructionsof both native MCUNC and BS3–cross-linkedMCUNC (fig. S3). Local and global alignmentsof protomers A and B in the MCUNC structureindicate that the domain rearrangements be-

tween the TMD and the CCD-NTD account forthe symmetry break (Fig. 3 and fig. S9), withthe departure from fourfold symmetry originat-ing from two distinct interaction networks en-gaged by neighboring NTDs (Fig. 3). With respectto the NTDs and CCDs, the channel adopts adimer-of-dimers assembly; within the NTD dimercomprising protomers A and B (the A/B dimer),there is a large dimer interface (~620 Å2) definedby both NTDs and CCDs, whereas there is acomparatively minimal dimer interface (~380 Å2)mediated by the NTD dimer comprising pro-tomers B and C (the B/C dimer).The dimer-of-dimers assembly of soluble do-

mains and the tetrameric assembly of the TMDin MCUNC is analogous to that of ionotropicglutamate receptors (iGluRs) (27). In iGluRs, theligand-binding domain (LBD) and amino terminaldomain (ATD) assume twofold symmetric ar-rangements, whereas the TMD adopts a four-fold symmetric arrangement. The LBD transitionsinto various conformations, including a pseudo-fourfold arrangement during the gating cycle ofiGluR (28, 29), indicating that the NTD dimer-of-dimers assemblymight play a comparable rolein MCU gating. This notion is further supported

by recent studies that suggest phosphorylation ofthe NTD or divalent cation binding to the NTDmodulates MCU function (fig. S7) (20, 21).Many structural and architectural features ob-

served in our MCUNC structure contrast withthose of cMCU-DNTD (Fig. 4). First, cMCU-DNTDandMCUNC adopt distinct pentameric and tetra-meric stoichiometries, respectively, which couldresult from construct design (truncation of theNTD versus full-length protein), choice of deter-gents (zwitterionic Fos-choline-14 versus dodecyl-maltoside), and/or protein preparation (extractionfrom inclusion body versus the membrane). TheCa2+ channel function has not been shown ineither cMCU (22) norMCUNC. Therefore, the invivo oligomeric status of MCU has not beenestablished, and we cannot exclude the possibilitythat fungal and cMCUs may adopt differentoligomeric arrangements. These discrepanciesawait further validation. Second, the arrangementof TM1 and TM2 in MCUNC establishes an ex-tensive inter-protomer interface, whereas the TMhelices in cMCU-DNTD only form intra-protomerinteractions. Third, although the selectivity filtersequence is located on TM2 in MCUNC, the cor-responding residues in cMCU-DNTDarepositioned

Yoo et al., Science 361, 506–511 (2018) 3 August 2018 4 of 6

TMD

CCD

NTD TMD-CCD

CC1 (A)

CC2 (A)CC1 (B)

CC2 (B)

D407

TMD

TM1 (A)

TM2 (A)TM1 (B)

TM2 (B)

W354

CCD-NTD

NTD (A)

NTD (B)

S255

A

B C D

NTD (C) NTD (B)

CC1

CC2

CC2

CC1

90°

TM1

TM2

NTD

TM1

TM2

CC1CC2

JH

TM1 TM2

CC1CC2

NTD

CC1CC2

NTD

JH

NTD (B) NTD (A)

CC2

CC1

CC1

CC2α2 α2

H160

Q163

Y167

P164S166

S254

Q298Y294

Y295 E431R263

D264

T116

W252

Fig. 3. Hinge rotation enables the mismatch in channel symmetry.(A) Top-view comparison of channel symmetry sliced throughthe three layers depicted in the tetrameric MCUNC channel.The TMD exhibits fourfold symmetry, whereas both the CCD andNTD display twofold symmetry. The lines are drawn between Ca atoms of

Trp354, Asp407, and Ser255. (B and C) Alignment of protomer A(red) and protomer B (blue) at (B) NTD and (C) TMD, showingthe rigid body rotations of TMD and NTD around the hinge point JH.(D) Distinct interfacial networks between each NTD and itsneighboring partner.

RESEARCH | REPORTon F

ebruary 1, 2021

http://science.sciencemag.org/

Dow

nloaded from

Page 5: STRUCTURAL BIOLOGY Cryo-EM structure of a mitochondrial ... · Cryo-EM structure of a mitochondrial calcium uniporter Jiho Yoo 1*, Mengyu Wu 2*, Ying Yin , Mark A. Herzik Jr. , Gabriel

within a loop, leading to different selectivity filterstructures (fig. S10). Last, the CCD inMCUNC con-sists of four dimeric coiled coils, formed by CC1and CC2, whereas the CC2 from each protomerin cMCU-DNTD forms a pentameric coiled coilin the CCD (Fig. 4).Our studies provide structural insights into

the design principle of the MCUNC selectivityfilter, which will serve as a platform to under-stand the mechanism of selective calcium per-meation by this channel family.

REFERENCE AND NOTES

1. C. Mammucari, G. Gherardi, R. Rizzuto, Front. Oncol. 7, 139(2017).

2. K. J. Kamer, V. K. Mootha, Nat. Rev. Mol. Cell Biol. 16, 545–553(2015).

3. T. E. Gunter, D. R. Pfeiffer, Am. J. Physiol. 258, C755–C786(1990).

4. Y. Kirichok, G. Krapivinsky, D. E. Clapham, Nature 427,360–364 (2004).

5. J. M. Baughman et al., Nature 476, 341–345 (2011).6. F. Perocchi et al., Nature 467, 291–296 (2010).7. D. De Stefani, A. Raffaello, E. Teardo, I. Szabò, R. Rizzuto,

Nature 476, 336–340 (2011).8. Y. Sancak et al., Science 342, 1379–1382 (2013).9. A. Raffaello et al., EMBO J. 32, 2362–2376 (2013).10. K. J. Kamer, V. K. Mootha, EMBO Rep. 15, 299–307 (2014).11. M. F. Tsai et al., eLife 5, e15545 (2016).12. N. E. Hoffman et al., Cell Reports 5, 1576–1588 (2013).13. K. J. Kamer, Z. Grabarek, V. K. Mootha, EMBO Rep. 18,

1397–1411 (2017).14. G. Csordás et al., Cell Metab. 17, 976–987 (2013).15. K. Mallilankaraman et al., Cell 151, 630–644 (2012).16. A. G. Bick, S. E. Calvo, V. K. Mootha, Science 336, 886 (2012).17. E. Kovács-Bogdán et al., Proc. Natl. Acad. Sci. U.S.A. 111,

8985–8990 (2014).18. J. Song, X. Liu, P. Zhai, J. Huang, L. Lu, Fungal Genet. Biol. 94,

15–22 (2016).19. G. Wu et al., Biochem. Biophys. Res. Commun. 496, 127–132

(2018).

20. Y. Lee et al., EMBO Rep. 16, 1318–1333 (2015).21. S. K. Lee et al., Cell Chem. Biol. 23, 1157–1169 (2016).22. K. Oxenoid et al., Nature 533, 269–273 (2016).23. X. Hou, L. Pedi, M. M. Diver, S. B. Long, Science 338,

1308–1313 (2012).24. K. Saotome, A. K. Singh, M. V. Yelshanskaya, A. I. Sobolevsky,

Nature 534, 506–511 (2016).25. D. A. Doyle et al., Science 280, 69–77 (1998).26. M. Hirschi et al., Nature 550, 411–414 (2017).27. A. I. Sobolevsky, M. P. Rosconi, E. Gouaux, Nature 462,

745–756 (2009).28. E. C. Twomey, M. V. Yelshanskaya, R. A. Grassucci, J. Frank,

A. I. Sobolevsky, Nature 549, 60–65 (2017).29. E. C. Twomey, A. I. Sobolevsky, Biochemistry 57, 267–276 (2018).30. O. S. Smart, J. G. Neduvelil, X. Wang, B. A. Wallace,

M. S. Sansom, J. Mol. Graph. 14, 354–360, 376 (1996).

ACKNOWLEDGMENTSCryo-EM data were collected at The Scripps Research Institute (TSRI)electron microscopy facility. We thank A. Kuk and Y. Suo at DukeUniversity, who helped with calcium flux experiments and preliminarynegative-staining EM analyses, respectively. We thank J.-C. Ducomat TSRI High Performance Computing facility for computational support

Yoo et al., Science 361, 506–511 (2018) 3 August 2018 5 of 6

90°

A 90° 90°

90°

(D)

TM1TM2

TM2

TM1

(A)

(C)

(B)

B

F

(A)

(E)

TM1

TM2

(D)

(C)

(B)

G

TM1 TM2

D240W239I241V242

E243P244

V245T246 Y247

H

MCUNC

cMCU-ΔNTD

TMD

TMD

CCD

CCD

D355

E358

Y362

T361

V360

TM1 TM2

C

W354L356

V357

P359

D (D)

CC2CC1

I(E)

CC1CC2

Matrix

IMS

Matrix

IMS

(A)CC1

CC2

(D)

(C)

(B)

E Matrix

(A)(E)

(B)

(C)

(D)

CC1CC2

J Matrix

Fig. 4. Comparison of cryo-EM structure MCUNC and NMR structurecMCU-DNTD. (A and B) Side and top views showing the tetramericconfiguration of MCUNC. (C) The selectivity filter sequence (yellow) of MCUNC

is located at the beginning of TM2. (D) Close-up view of the hydrophobicinteractions (silver spheres) between CC1 and CC2. Hydrophilic residues arecolored in teal. (E) Viewed from the intermembrane space, CC1 (orange), andCC2 (red) of MCUNC form dimeric coiled-coils within each protomer viaextensive hydrophobic interactions (highlighted by silver spheres).

(F and G) Side and top views showing cMCU-DNTD (PDB ID: 5ID3) forms apentamer. (H) The “D-F-F-E” motif (yellow) in cMCU is located at the loopconnecting TM1 and TM2. (I) Close-up view of the hydrophobic residues(silver spheres) located on CC1 and CC2. Hydrophilic residues are colored inteal. (J) Viewed from the intermembrane space, CC2 (red) forms apentameric helical bundle via hydrophobic interactions (silver spheres)pointing toward the central axis. Hydrophobic residues (silver spheres) onCC1 (orange) are exposed to the mitochondrial matrix.

RESEARCH | REPORTon F

ebruary 1, 2021

http://science.sciencemag.org/

Dow

nloaded from

Page 6: STRUCTURAL BIOLOGY Cryo-EM structure of a mitochondrial ... · Cryo-EM structure of a mitochondrial calcium uniporter Jiho Yoo 1*, Mengyu Wu 2*, Ying Yin , Mark A. Herzik Jr. , Gabriel

and W. Anderson for microscope support. We thank S. Y. Kim at DukeFunctional Genomics Shared Resource for generating the hMCUknockout cell line. Funding: This work was supported by the NationalInstitutes of Health (R35NS097241 to S.-Y.L.; DP2EB020402 andR21AR072910 to G.C.L.). G.C.L is supported as a Searle Scholar and aPew Scholar. Computational analyses of EM data were performed byusing shared instrumentation funded by NIH S10OD021634. Authorcontributions: J.Y. conducted all biochemistry of MCU and themitochondrial Ca2+ uptake assay under the guidance of S.-Y.L. M.A.H.

performed negative-staining EM experiments, and M.W. conductedall cryo-EM experiments under the guidance of G.C.L and M.A.H. J.Y.,Y.Y., M.A.H., and M.W. performed model building and refinement. Allauthors wrote the paper.Competing interests:The authors declareno competing interests. Data and materials availability: Thecoordinates are deposited in the Protein Data Bank with the PDB ID6DT0, and the electron density maps have been deposited in theElectron Microscopy Data Bankwith the ID EMD-8911. All other data areavailable in the manuscript or the supplementary materials.

SUPPLEMENTARY MATERIALS

www.sciencemag.org/content/361/6401/506/suppl/DC1Materials and MethodsFigs. S1 to S10Tables S1References (31–49)

16 April 2018; accepted 20 June 201810.1126/science.aar4056

Yoo et al., Science 361, 506–511 (2018) 3 August 2018 6 of 6

RESEARCH | REPORTon F

ebruary 1, 2021

http://science.sciencemag.org/

Dow

nloaded from

Page 7: STRUCTURAL BIOLOGY Cryo-EM structure of a mitochondrial ... · Cryo-EM structure of a mitochondrial calcium uniporter Jiho Yoo 1*, Mengyu Wu 2*, Ying Yin , Mark A. Herzik Jr. , Gabriel

Cryo-EM structure of a mitochondrial calcium uniporterJiho Yoo, Mengyu Wu, Ying Yin, Mark A. Herzik Jr., Gabriel C. Lander and Seok-Yong Lee

originally published online June 28, 2018DOI: 10.1126/science.aar4056 (6401), 506-511.361Science 

, this issue p. 506Sciencetogether with mutagenesis, suggests that two acidic rings inside the channel provide the selectivity for calcium.formed by four MCU protomers with differing symmetry between the soluble and membrane domains. The structure,

The channel isNeurospora crassa. report the high-resolution structure of MCU from et al.entry into mitochondria. Yoo for cellular physiology. A calcium-selective channel called the mitochondrial calcium uniporter (MCU) mediates calcium

Maintaining the correct balance of calcium concentrations between the cytosol and the mitochondria is essentialA channel for calcium

ARTICLE TOOLS http://science.sciencemag.org/content/361/6401/506

MATERIALSSUPPLEMENTARY http://science.sciencemag.org/content/suppl/2018/06/27/science.aar4056.DC1

CONTENTRELATED

http://stke.sciencemag.org/content/sigtrans/12/579/eaav1439.fullhttp://stke.sciencemag.org/content/sigtrans/11/553/eaaq1380.full

REFERENCES

http://science.sciencemag.org/content/361/6401/506#BIBLThis article cites 49 articles, 9 of which you can access for free

PERMISSIONS http://www.sciencemag.org/help/reprints-and-permissions

Terms of ServiceUse of this article is subject to the

is a registered trademark of AAAS.ScienceScience, 1200 New York Avenue NW, Washington, DC 20005. The title (print ISSN 0036-8075; online ISSN 1095-9203) is published by the American Association for the Advancement ofScience

Copyright © 2018, American Association for the Advancement of Science

on February 1, 2021

http://science.sciencem

ag.org/D

ownloaded from