defining human erad networks through an integrative...

32
RESOURCE Defining human ERAD networks through an integrative mapping strategy John C. Christianson 1,2,5 , James A. Olzmann 1,5 , Thomas A. Shaler 3 , Mathew E. Sowa 4 , Eric J. Bennett 4,6 , Caleb M. Richter 1 , Ryan E. Tyler 1 , Ethan J. Greenblatt 1 , J. Wade Harper 4 and Ron R. Kopito 1,7 Proteins that fail to correctly fold or assemble into oligomeric complexes in the endoplasmic reticulum (ER) are degraded by a ubiquitin- and proteasome-dependent process known as ER-associated degradation (ERAD). Although many individual components of the ERAD system have been identified, how these proteins are organized into a functional network that coordinates recognition, ubiquitylation and dislocation of substrates across the ER membrane is not well understood. We have investigated the functional organization of the mammalian ERAD system using a systems-level strategy that integrates proteomics, functional genomics and the transcriptional response to ER stress. This analysis supports an adaptive organization for the mammalian ERAD machinery and reveals a number of metazoan-specific genes not previously linked to ERAD. Approximately one-third of the eukaryotic proteome consists of secreted and integral membrane proteins that are synthesized and inserted into the ER, where they must correctly fold and assemble to reach functional maturity 1 . ER quality control refers to the processes simultaneously monitoring deployment of correctly folded proteins and assembled complexes to distal compartments, while diverting folding-incompetent, mutant or unassembled polypeptides for proteasomal degradation through the process of ERAD (reviewed in refs 24). An ever-growing list of sporadic and genetic human disorders have been associated with ER quality control, illustrating the pivotal role these processes play in governance of protein trafficking 5 . Many of the individual components thought to underlie ERAD have been identified through genetic and biochemical analyses in Saccha- romyces cerevisiae and mammals 4,6 and point towards a mechanism mediated by a network of topologically and compartmentally restricted, partially redundant protein complexes 2,4,7–9 . ERAD is a vectorial process whereby coordination of ERAD components across three subcellular compartments (ER lumen, lipid bilayer and cytoplasm) must occur to effectively distinguish, target and deliver misfolded substrates for degradation. Exclusion of the ubiquitinproteasome system (UPS) from the ER lumen necessitates that substrates traverse the ER membrane to be degraded, but the molecular identity and mechanism of the required dislocation apparatus remains controversial 7,10,11 . Ubiquitin E3 ligases play central functional and organizational roles in ERAD (ref. 9). In yeast, the E3s Hrd1 and Doa10, which contain cytoplasmically oriented RING domains that recruit distinct ubiquitin- 1 Department of Biology & Bio-X Program, Stanford University, Lorry Lokey Building, 337 Campus Drive, Stanford, California 94305, USA. 2 Ludwig Institute for Cancer Research, University of Oxford, ORCRB, Headington, Oxford OX3 7DQ, UK. 3 SRI International, Menlo Park, California 94025, USA. 4 Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, USA. 5 These authors contributed equally to this work. 6 Present address: Division of Biological Sciences, UC San Diego, La Jolla, California 92093, USA. 7 Correspondence should be addressed to R.R.K. (e-mail: [email protected]) Received 18 April 2011; accepted 21 October 2011; published online 27 November 2011; DOI: 10.1038/ncb2383 conjugating enzymes and form functional complexes by scaffolding shared ERAD-related factors 12–16 , seem to be sufficient to degrade all ERAD substrates 16,17 . ERAD substrates with luminal or membrane folding lesions utilize Hrd1 (refs 16,18), whereas those with cytoplasmic lesions rely on Doa10 (refs 16,17,19). In contrast to yeast, at least ten different E3s have been implicated in mammalian ERAD (ref. 20), possi- bly reflecting an evolutionary adaptation to the broader substrate range imposed by the more complex metazoan proteome. Three mammalian E3s, gp78, Hrd1 and TEB4, share similar domain and topological organization, but scant sequence homology, with their yeast ortho- logues Hrd1 (orthologue of gp78 and Hrd1) and Doa10 (orthologue of TEB4). Uncovering how the organization of E3-containing membrane complexes allows them to access substrates in the ER lumen/membrane and recruit the cytoplasmic dislocation/extraction apparatus is crucial to establishing a comprehensive understanding of ERAD. Here, we have employed a systematic, multilayered approach that integrates high-content proteomics, functional genomics and gene expression to elucidate the interconnectivity and organization of ERAD in mammals (Supplementary Fig. S1). These studies have allowed us to generate an integrated physical and functional map of the ERAD system in the mammalian ER. RESULTS Mapping the mammalian ERAD interaction network We employed a high-content proteomics strategy to map the mammalian ERAD interaction network, starting with 15 S-tagged NATURE CELL BIOLOGY ADVANCE ONLINE PUBLICATION 1 © 2011 Macmillan Publishers Limited. All rights reserved.

Upload: doandang

Post on 17-Apr-2018

219 views

Category:

Documents


2 download

TRANSCRIPT

RESOURCE

Defining human ERAD networks through an integrativemapping strategyJohn C. Christianson1,2,5, James A. Olzmann1,5, Thomas A. Shaler3, Mathew E. Sowa4, Eric J. Bennett4,6,Caleb M. Richter1, Ryan E. Tyler1, Ethan J. Greenblatt1, J. Wade Harper4 and Ron R. Kopito1,7

Proteins that fail to correctly fold or assemble into oligomeric complexes in the endoplasmic reticulum (ER) are degraded by aubiquitin- and proteasome-dependent process known as ER-associated degradation (ERAD). Although many individualcomponents of the ERAD system have been identified, how these proteins are organized into a functional network that coordinatesrecognition, ubiquitylation and dislocation of substrates across the ER membrane is not well understood. We have investigated thefunctional organization of the mammalian ERAD system using a systems-level strategy that integrates proteomics, functionalgenomics and the transcriptional response to ER stress. This analysis supports an adaptive organization for the mammalian ERADmachinery and reveals a number of metazoan-specific genes not previously linked to ERAD.

Approximately one-third of the eukaryotic proteome consists ofsecreted and integral membrane proteins that are synthesized andinserted into the ER, where they must correctly fold and assembleto reach functional maturity1. ER quality control refers to theprocesses simultaneously monitoring deployment of correctly foldedproteins and assembled complexes to distal compartments, whilediverting folding-incompetent, mutant or unassembled polypeptidesfor proteasomal degradation through the process of ERAD (reviewed inrefs 2–4). An ever-growing list of sporadic and genetic human disordershave been associated with ER quality control, illustrating the pivotalrole these processes play in governance of protein trafficking5.Many of the individual components thought to underlie ERAD have

been identified through genetic and biochemical analyses in Saccha-romyces cerevisiae and mammals4,6 and point towards a mechanismmediated by a network of topologically and compartmentally restricted,partially redundant protein complexes2,4,7–9. ERAD is a vectorial processwhereby coordination of ERAD components across three subcellularcompartments (ER lumen, lipid bilayer and cytoplasm) must occurto effectively distinguish, target and deliver misfolded substrates fordegradation. Exclusion of the ubiquitin–proteasome system (UPS)from the ER lumen necessitates that substrates traverse the ERmembrane to be degraded, but the molecular identity and mechanismof the required dislocation apparatus remains controversial7,10,11.Ubiquitin E3 ligases play central functional and organizational roles

in ERAD (ref. 9). In yeast, the E3s Hrd1 and Doa10, which containcytoplasmically oriented RING domains that recruit distinct ubiquitin-

1Department of Biology & Bio-X Program, Stanford University, Lorry Lokey Building, 337 Campus Drive, Stanford, California 94305, USA. 2Ludwig Institute for CancerResearch, University of Oxford, ORCRB, Headington, Oxford OX3 7DQ, UK. 3SRI International, Menlo Park, California 94025, USA. 4Department of Cell Biology,Harvard Medical School, Boston, Massachusetts 02115, USA. 5These authors contributed equally to this work. 6Present address: Division of Biological Sciences, UCSan Diego, La Jolla, California 92093, USA.7Correspondence should be addressed to R.R.K. (e-mail: [email protected])

Received 18 April 2011; accepted 21 October 2011; published online 27 November 2011; DOI: 10.1038/ncb2383

conjugating enzymes and form functional complexes by scaffoldingshared ERAD-related factors12–16, seem to be sufficient to degrade allERAD substrates16,17. ERAD substrates with luminal or membranefolding lesions utilizeHrd1 (refs 16,18), whereas those with cytoplasmiclesions rely on Doa10 (refs 16,17,19). In contrast to yeast, at least tendifferent E3s have been implicated inmammalian ERAD (ref. 20), possi-bly reflecting an evolutionary adaptation to the broader substrate rangeimposed by the more complex metazoan proteome. Three mammalianE3s, gp78, Hrd1 and TEB4, share similar domain and topologicalorganization, but scant sequence homology, with their yeast ortho-logues Hrd1 (orthologue of gp78 and Hrd1) and Doa10 (orthologue ofTEB4). Uncovering how the organization of E3-containing membranecomplexes allows them to access substrates in the ER lumen/membraneand recruit the cytoplasmic dislocation/extraction apparatus is crucialto establishing a comprehensive understanding of ERAD.Here, we have employed a systematic, multilayered approach that

integrates high-content proteomics, functional genomics and geneexpression to elucidate the interconnectivity and organization of ERADin mammals (Supplementary Fig. S1). These studies have allowed usto generate an integrated physical and functional map of the ERADsystem in the mammalian ER.

RESULTSMapping the mammalian ERAD interaction networkWe employed a high-content proteomics strategy to map themammalian ERAD interaction network, starting with 15 S-tagged

NATURE CELL BIOLOGY ADVANCE ONLINE PUBLICATION 1

© 2011 Macmillan Publishers Limited. All rights reserved.

RESOURCE

baits consisting of proteins previously identified as ERAD pathwaycomponents in biochemical studies or by orthology to componentsidentified in yeast (Supplementary Table S1, Primary). After confirmingER localization in HeLa cells (Supplementary Fig. S2) and stableexpression of each full-length S-tagged bait in HEK293 cells (datanot shown), protein complexes captured by S-protein affinitypurification from detergent-solubilized lysates were analysed byliquid chromatography–tandem mass spectrometry (LC–MS/MS).Interactions were initially assessed for all baits by independentlyanalysing pulldowns from cells lysed in digitonin (SupplementaryTable S2) or themore stringent detergent Triton X-100 (SupplementaryTable S3). Total spectral counts for each captured protein weresubsequently evaluated with the Comparative Proteomics AnalysisSoftware Suite21,22 (CompPASS; SupplementaryMethods). CompPASSemploys a database of interacting proteins (including data frombaits in this study and 102 unrelated proteins described previously21)and comparative metrics to determine the likelihood of validityof interactors. The CompPASS parameter WDN-score22, whichintegrates the abundance, uniqueness and reproducibility of aninteracting protein, was used to identify high-confidence candidateinteracting proteins (HCIPs) for the ERAD network. Previous studiesdemonstrated that >68% of identified HCIPs were validated insubsequent biochemical analyses21, a rate of validation that is wellabove other high-throughput approaches to study protein–proteininteractions23. In our study, interacting proteins surpassing astringent threshold score of WDN > 1.0 were designated as HCIPs(Supplementary Tables S2a and S3a). Interacting proteins scoringbelow this cutoff may still represent bona fide interactions (full listin Supplementary Tables S2b and S3b).In addition to revealing interconnections among primary baits,

this analysis uncovered 10 HCIPs that had no previous relationshipwith ERAD. Seven (FAM8A1, UBAC2, KIAA0090, TTC35, C15orf24,TMEM111 and COX4NB) are functionally uncharacterized openreading frames and two (E-Syt1 andMMGT1, also known as TMEM32)are implicated in cellular processes unrelated to quality control. TheHCIP TXD16 (also known as ERp90) was recently suggested tobe involved in ERAD (ref. 24). On the basis of their identificationas HCIPs with multiple ERAD components in both digitonin andTriton X-100, high spectral counts and predicted ER localization(criteria described in Methods), these 10 HCIPs were introducedinto the proteomic workflow to iteratively expand and validate thenetwork (Supplementary Fig. S1, Secondary). Three proteins previouslyimplicated in mammalian ERAD (TEB4, RNF5 and HERP) couldnot be sufficiently expressed and were omitted. Ultimately, ourERAD network analysis included 25 baits, of which 9 seem to beunique to metazoans. No correlation was observed between baitabundance and the total number of interactions and HCIPs identified(Supplementary Fig. S3a). From 3,325 individual proteins identified byMASCOT in digitonin and 2,971 in Triton X-100 (SupplementaryTables S2b and S3b), CompPASS identified 320 and 202 HCIPs,respectively, for the 25 baits (Supplementary Tables S2a and S3a).These HCIPs correspond to 143 and 97 non-redundant proteinswith 71 HCIPs of interest, previously uncharacterized for a role inERAD (see Methods for selection criteria, Supplementary Table S4and Fig. S3b). Over 50% of HCIPs are ER/membrane localized, andgene ontology analysis indicates diverse functionality with significant

over-representation in folding, ubiquitin and catabolic processes(Supplementary Fig. S3c,d).

Overview of the mammalian ERAD interaction networkUnbiased hierarchical clustering of all HCIPs identified for eachbait in both detergents was used to assemble interaction data intoa coherent network (Fig. 1). Four of the digitonin clusters definesubnetworks organized around the E3s Hrd1 (clusters 1D and 6D)and gp78 (clusters 3D and 8D), indicating a central role in organizationof the mammalian ERAD system. Both Hrd1 and gp78 clusteredwith established integral membrane, luminal and cytoplasmic ERADcomponents (clusters 1D and 8D) as well as, separately, with most26S proteasome subunits (clusters 3D and 6D). Cluster 2D defines amacromolecular complex of previously uncharacterized proteins thatwe have designated the mammalian ER membrane complex (mEMC,discussed below) to reflect its orthology to a complex associatedwith theunfolded protein response (UPR) in yeast25. Cluster 4D confirms thepreviously reported interactions of theAAA+ATPaseVCP (also knownas p97) with an integral membrane binding partner VIMP (refs 26,27)and cytosolic NGly1 (ref. 28), while revealing interactions of VIMP andUBXD2 with the VCP accessory protein UBE4A, a Ufd2 orthologueimplicated in ubiquitin chain extension29. In addition to confirmingthe ERFAD–SEL1L interaction30, cluster 5D validated the recentlyreported interaction of ERFAD with TXD16 (ref. 24), reinforcing theconnection between ERAD and oxidative protein folding/unfolding.Cluster 7D contains several HCIPs in complex with Derlin-1 andDerlin-2, including the Ca2+-sensing protein extended-synaptotagmin1 (E-Syt1; ref. 31), the Ras superfamily member ARL6IP and YIF1B,both implicated in protein trafficking32,33.Of the eight prominent clusters identified in digitonin-solubilized

cells, only cluster 2 remained intact with Triton X-100 lysis. Fourclusters (1, 3, 5 and 8) were fragmented into discrete subclusters,and three (4, 6 and 7) were fully disrupted. On the basis of theseclusters (Fig. 1), we merged individual interactomes (SupplementaryFig. S4) to construct a topologically rendered, detailed interactionmap of the mammalian ERAD network in digitonin and Triton X-100(Fig. 2). The interaction network for ERAD (INfERAD) was arrangedaround clusters identified for Hrd1–SEL1L, gp78 and the mEMCsubnetworks, with those components located centrally reflecting sharedinteractions between clusters.As with any systems-based analysis of interaction networks, our

analysis was not exhaustive, and therefore we sought to integratedata from public protein–protein interaction resources (STRING).However, as ERAD components are poorly represented in this database(Supplementary Table S5) and other online resources (BIOGRID andMINT), interactions identified in this study were instead mappedtogether with pairwise interactions reported previously (SupplementaryTable S5 and Fig. S5). These combined data sets contain over250 interactions, reflecting the organizational complexity of themammalian ERAD system.

The Hrd1–SEL1L subnetworkOur proteomic analysis confirmed the E3 Hrd1 and its establishedcofactor SEL1L as a prominent nexus for ERAD. Nearly all previouslyreported interactions of the Hrd1–SEL1L complex (SupplementaryTable S5) were validated by our data set, which also uncovered

2 NATURE CELL BIOLOGY ADVANCE ONLINE PUBLICATION

© 2011 Macmillan Publishers Limited. All rights reserved.

RESOURCE

ERLIN2gp78

UBAC2UBXD8

3D

C15orf24KIAA0090COX4NBTMEM111MMGT1TTC35

2D1D

C15orf24KIAA0090COX4NBTMEM111MMGT1TTC35

2T

7D Derlin-2Derlin-1E-Syt1

8D

ERLIN2UBAC2UBXD8

5D ERFAD

TXD16EN

O1

TXD16

TAGLN

2

RPL35

PPIBM

ARCKS

HM

RM

PU

HM

RM

PA2B1

FLNA

ATP5F1

USH

2A

NIPSN

AP1

LIN7C

GAN

AB

GALK1

DPY30

CASK

ACTG

2

ERFAD

MCM

2

CAN

XEDEM1ERFADTXD16

5T

Hrd1

6D

2D

3D

1D

1 10+

WDN -score

2T

Baits

7D

8D

5D

1T b

5T

8T

6D

Baits

*

AUP1AU

P1

UBE2G

2

PRKD

C

SURF4

CKAP5

HCIPs HCIPs

FAM8A1

Hrd1Hrd1

FAM8A1

4D

VCPVIM

P

Derlin-2

SELK

UBXD

6

KLHDC2

UBXD

2

UBE4A

4D

OS-9

SEL1L

XTP3-B

gp78

8TUBAC2

UBXD8UBXD

8

Derlin-2

gp78ERLIN2UBAC2UBXD8

ERLIN

1

ERLIN

2

*Baits are arranged in the same hierarchical cluster as for digitonin treatment

gp78

EDEM1ERFADTXD16

AUP1OS-9SEL1LXTP3-BFAM8A1Hrd1

Derlin-2Derlin-1FAM62AC15orf24KIAA0090COX4NBTMEM111MMGT1TTC35NGly1VIMPUBXD2

ERLIN2gp78

UBAC2UBXD8

Digitonin solubilizationTriton X-100 solubilization

EDEM1ERFADTXD16

AUP1OS-9SEL1LXTP3-BFAM8A1Hrd1

Derlin-2Derlin-1FAM62AC15orf24KIAA0090COX4NBTMEM111MMGT1TTC35NGly1VIMPUBXD2

ERLIN2gp78

UBAC2UBXD8

PSMD7

FAM8A1

UBE2J1

XTP3-B

SLC27A3

CPVL

Hrd1

SEL1L

UBE2G

2

GRP94

PSMD1

HM

GCR

OS-9

MYH

9

LONP2

FDFT1

DIP2B

AUP1

CAPR

IN2

1T a

1T c

3T b

3T a

1T c

1T aAUP1

OS-9SEL1LXTP3-BFAM8A1Hrd1

KIAA0090

COX4N

B

TTC35

TMEM

111

C19orf63

MM

GT1

C15orf24

TMEM

93

PSMD4

PSMD6

PSMD3

PSMD14

PSMC2

PSMB5

PSMA4

OSB

PL8

MYH

10

BRI3B

P

gp78

PSMB7

PSMA1

PSMA7

PSMA2

PSMB6

PSMB1

PSMB4

PSMB2

TMUB1

PSMA6

PSMB3

PSMA5

PSMA3

ERLIN

1

ERLIN

2

YIF1B

ARL6IP5

SYNJ2B

P

E-Syt2

E-Syt1

RTN

4

PGRM

C2

EPHX1

HM

OX1

Derlin-1

POR

NGly1VIMPUBXD2

1T b

LONP2

XTP3-B

CPVL

SEL1L

SERPIN

H1

OS-9

GRP94

RPN

1

GGH

3T a

KIAA0090

COX4N

B

TTC35

TMEM

111

C19orf63

MM

GT1

C15orf24

TMEM

93

C14orf122

TMEM

85

3T b

PSMB7

PSMA1

PSMA7

PSMA2

PSMB6

PSMB1

PSMB4

PSMB2

PSMA6

PSMB3

PSMA5

PSMA3

ERLIN

1

ERLIN

2

PSMA4

UBE2G

2

VCP

gp78

PSME4

PSMA1

PSMA7

PSMA2

PSMB6

PSMB1

PSMB4

PSMB2

PSMA6

PSMB3

PSMA5

PSMA3

ACTG

2

NIPSN

AP1

SLC25A10

FLNA

RAB

9A

KRT18

EMD

TXD16

ACTA1

EDEM

1

E-Syt1

CAN

X

LIN7C

CASK

ERFAD

MTC

H1

MCM

2

UBAC

2

TUBA1A

TMEM

43

TMED

9

RAB

7A

PEX19

GCN1L1

HNRNPM

SEC22B

VCPVIM

P

Derlin-2

Figure 1 Hierarchical cluster analysis of CompPASS-identifiedhigh-confidence candidate interaction proteins (HCIPs). Hierarchicalclustering of HCIPs for interactions present in digitonin (left) and

Triton X-100 (right). Prominent HCIP clusters identified in digitonin(1–8D) and Triton X-100 (1–3, 5 and 8T) were manually selected andare highlighted below. The colour of the square indicates the WDN-score.

several unreported interactors including FAM8A1; LONP2, a putativeLon-protease (with OS-9); CPVL, a putative carboxypeptidase (withXTP3-B); the stress-inducible haem oxygenase HMOX1 (also knownas HO1); and two components of the sterol biosynthetic pathway,HMG-CoA reductase (HMGCR) and squalene synthetase (FDFT1).The rate-limiting enzyme in cholesterol synthesis, HMGCR, is subjectto strict feedback regulation whereby sterol end products induce itsdegradation by ERAD (refs 12,34,35). Although evidence supports arole for gp78 in the degradation of HMGCR in mammalian cells36,the Hrd1 pathway degrades HMGCR in both yeast (Hmg2; ref. 12)and Drosophila37. Identification of HMGCR as a Hrd1 HCIP lendsstrength to the possibility that Hrd1 also plays a role in HMGCRdegradation in mammals38.The integrity of theHrd1–SEL1L subnetworkwas strongly influenced

by solubilization conditions. All Hrd1 HCIPs except FAM8A1 (Fig. 1a,cluster 1Ta) were lost in Triton X-100, whereas the complexescontaining SEL1L, OS-9 and other luminal components werepreserved (Fig. 1a, cluster 1Tb), consistent with all upstream (luminal)interactions being mediated through SEL1L, which is linked to Hrd1by a Triton X-100-labile association39.

The gp78 subnetworkCluster analysis exposed a reciprocally co-precipitating complexconsisting of the E3 gp78, an uncharacterized UBA domain-containingpolytopic protein UBAC2, the membrane-embedded, VCP-bindingprotein UBXD8, and Derlin-1 and Derlin-2. The high degree ofinterconnectivity indicates that gp78 and its cognate E2 (UBE2G2)and UBAC2 comprise a transmembrane pathway for ERAD that sharesessential cytoplasmic (for example, VCP and 26S proteasomes) andintegral membrane components (UBXD8 and Derlin-2) with theHrd1–SEL1L cluster. The recently described protein TMUB1 (ref. 40)was found in the gp78 cluster, as was BRI3BP, hitherto unlinked to ER.Signal peptide peptidase (SPP, also known as HM13) and a numberof poorly characterized integral membrane proteins (TMEM201,TMEM43, LRRC59 and CLPTM1) were also linked through UBAC2.In contrast to the Hrd1–SEL1L cluster, most HCIPs associated withthe gp78 subnetwork were stable in both detergents (SupplementaryTable S2, S3 and S6). Disruption of the Hrd1–SEL1L cluster in TritonX-100 caused the shared cytoplasmic interactors VCP and UBE2G2to cluster with gp78, probably reflecting their direct binding to thecarboxy-terminal cytoplasmic domain of gp78 (refs 41,42). These data

NATURE CELL BIOLOGY ADVANCE ONLINE PUBLICATION 3

© 2011 Macmillan Publishers Limited. All rights reserved.

RESOURCE

CPVL

XTP3-B

ERFAD

TXD16

TTC35

FAM8A1

UBXD8

UBE2G2

UBE2J1

EDEM1

ERLIN2

UBXD2E-Syt2E-Syt1 SPP

C14orf122

CANX

C19orf63

Hrd1

Derlin-2 Derlin-1

Lumen

LONP2

ER localized Other

MMGT1

C15orf24

Locale SC SGD ID

- -

- -

- -

- -

Cytoplasm

Unidirectional (only in digitonin)ReciprocalIn digitonin and Triton X-100Only in Triton X-100

InteractorBait

PSMA 1–7PSMB 1–7PSMC 2PSMD 1–3,6,7,14PSME4

Protein name

Length

(aa)

GRP94

SEL1L

AUP1 ERLIN1

VCP

NGly1

OS-9

COX4NB

TMEM93

TMEM85

KIAA0090

TMEM111

UBAC2

VIMP

hHR23B UBE4A

SERPINH1* RPN1* GGH* CIB2*HMOX OSBPL8DUSP3LIN7* KLHDC2HMGCR LRRC59

SURF4*YIF1B TMUB1 CAPRIN2c19orf6 DPY30NIPSNAP1* UBXD6CLPTM1 TBC1D15*TMEM201

TBC1D17*RTN4*BRI3BP DIP2BSYNJ2BPCASK*TMEM43 SELKFDFT1 TMED9 ARL6IP5

KIAA0090

MMGT1

TMEM85

TTC35

TMEM93

C15orf24

C14orf122

COX4NB

C19orf63

TMEM111

993

131

183

261

297

242

208

210

262

110

IMP

IMP

IMP

IMP

IMP

IMP

IMP

Cyto.

Cyto.

Cyto.

Emc1

Emc5

Emc4

Emc3

Emc2

Emc6

YCL045C

YIL027C

YGL231C

YKL207W

YJR088C

YLL014W

Figure 2 The INfERAD. Interaction network for ERAD isolated in digitoninand Triton X-100 represented by baits (squares) and their HCIPs (circles).Unidirectional (dashed, single arrow) and reciprocal (solid black, doublearrows) interactions are shown. Each bait protein is rendered in a uniquecolour and line colour reflects the bait protein used to identify theinteraction with the HCIP. Dotted lines marked with a circle indicateinteractions detected in both digitonin and Triton X-100, and longdashed lines represent those found only in Triton X-100. The inset table

lists the determined constituents of the mammalian ER membranecomplex (mEMC), their size (in amino acids, aa), cellular localization(IMP, integral membrane protein; Cyto., cytosolic), and correspondingyeast orthologues (SC) and ID in the Saccharomyces Genome Database(SGD). For clarity, a selection of additional digitonin HCIPs not includedin the map is shown on the bottom, with a circle’s colour correspondingto the bait for which the HCIP was observed and asterisks denoting anHCIP also detected in Triton X-100.

establish gp78 as the core of a detergent-stable E3 subnetwork thatshares several components with theHrd1 complex, and identify UBAC2as a central element in the gp78 complex.

ERAD E3s are associated with the 26S proteasomeStrikingly, nearly all subunits of the 26S proteasome were capturedwith Hrd1 and gp78 in digitonin lysates. Although not all of theseinteractions reached our stringent criteria to qualify as HCIPs (19/32for gp78; 15/31 for Hrd1; Supplementary Table S6), the fact thatgp78 captured all subunits of the 20S core particle and most subunitsof the 19S regulatory particle indicates a significant connectionbetween the ERAD E3s and 26S proteasomes (SupplementaryTable S6 and Fig. S6). Persistence of these interactions in TritonX-100, together with the observation that proteasome subunitswere not identified as HCIPs of other ERAD interaction networkcomponents, indicates an intimate, perhaps direct interaction ofgp78 (and possibly Hrd1) with the 26S proteasome. Proteasomestability requires ATP and without it, dissociation into 20S coreand 19S regulatory particles can occur43,44. The excess of 20Score particle subunits over 19S regulatory subunits captured withgp78 and Hrd1 (Supplementary Fig. S6) raises the possibility thatE3–proteasome connections may be linked independently of the 19Sregulatory particle, perhaps through direct interactions with 20S orthrough alternative adaptors.

The gp78 HCIP PSME4 (also known as PA200) was identified in ascreen for 26S proteasome activators and originally reported as a nu-clear protein with a possible role in DNA repair45. PA200 can assemblewith 20S and 19S subunits to form hybrid 26S proteasomes46 and acrystal structure of the apparent yeast orthologue Blm10 indicates thatit interacts directly with proteasomeα-subunits47. The functional signif-icance of the PA200 interaction is unclear, but its presence in gp78 (butnot Hrd1) complexes indicates that there may be heterogeneous 26Sproteasome populations associatedwith the ERmembrane andERAD.

The mEMC subnetworkThe detergent-stable mEMC (Fig. 1, cluster 2) was initially identifiedthrough KIAA0090, an uncharacterized, putative type I integralmembrane glycoprotein detected as a Derlin-1/2 HCIP (Fig. 1 andSupplementary Table S2). With KIAA0090 as bait, we identifiedfive additional HCIPs (TTC35, MMGT1, TMEM85, C15orf24 andCOX4NB), which reciprocally co-precipitated each other, and fouradditional proteins (TMEM111, C19orf63, C14orf122 and TMEM93;Supplementary Fig. S4). The mEMC comprises ten unique subunits,whereas its yeast counterpart seems to contain six (Fig. 2). Althoughthe function of the mEMC is unknown, three subunits (KIAA0090,TMEM111 and TTC35) were identified as HCIPs of UBAC2 andDerlin-2, indicating a close link between this complex and ERAD componentsimplicated in ubiquitin recognition and protein dislocation.

4 NATURE CELL BIOLOGY ADVANCE ONLINE PUBLICATION

© 2011 Macmillan Publishers Limited. All rights reserved.

RESOURCE

SEL1L

XTP3-B

OS-9

FAM8A1

AUP1 UBE2G2

UBE2J1

Hrd1

c

d

a

b

50

75

75

100

+ + + ++ + + ++

++

+– –– –

XTP3-B–Smyc–UBE2J1

FF Luc shRNASEL1L shRNA

SEL1L

myc–UBE2J1

Input AP: S-prot

50 XTP3-B–S

100M

r (K)

Mr (K)

Mr (K)

Mr (K)

Mr (K)

Mr (K)

Mr (K)

Mr (K)

Mr (K)

Mr (K)

Mr (K)

Mr (K)

Mr (K) M

r (K)

75

7550

+ + + ++

++

+– –– –

XTP3-B–SFF Luc shRNASEL1L shRNA

XTP3-B–S

SEL1LHrd1

k

d

b e

50

7575

100

+ + + ++

+

++– –

– –XTP3-B–S

FF Luc shRNASEL1L shRNA

SEL1L

FAM8A1

50XTP3-B–S

75

75

35

+ + + + + +

+ +

++

++– –

– – ––

– – ––––

S–FAM8A1FF Luc shRNASEL1L shRNA

Hrd1 shRNA

S–FAM8A1

Hrd1

SEL1L 75

50

75

+ + + +

+ + + +

++

++–

Hrd1–Smyc–UBE2J1

FF Luc shRNASEL1L shRNA

Hrd1–S

myc–UBE2J1

SEL1L 75

50

75

+ + + ++ + + ++

++

+– –– –

S–SEL1Lmyc–UBE2J1

FF Luc shRNAHrd1 shRNA

S–SEL1L

myc–UBE2J1

Hrd1

75

75

75

35

35

75

50

25

50

75

50

35 35

50

20

35

+ + + ++ + + ++

++

+- -- -

AUP1–Smyc–UBE2G2FF Luc shRNA

Hrd1 shRNA

AUP1–S

myc–UBE2G2

Hrd1

75

75

35

+ + + ++

++

+– –– –

AUP1–SFF Luc shRNA

Hrd1 shRNA

AUP1-S

SEL1L

Hrd1

j

10075

10075

+ + + ++

++

+– –– –

S–OS9.2FF Luc shRNASEL1L shRNA

S–OS9.2

SEL1LHrd1

SEL1L

XTP3-B

OS-9

FAM8A1

AUP1 UBE2G2

UBE2J1

Hrd1

SEL1L

XTP3-B

OS-9

FAM8A1

AUP1 UBE2G2

UBE2J1

Hrd1

fg

hi

j

SEL1L

XTP3-B

OS-9

FAM8A1

AUP1 UBE2G2

UBE2J1

Hrd1

+ + + ++

++

+– –– –

UBXD8–SFF Luc shRNAUBAC2 shRNA

+ + + ++

++

+– –– –

+ + + ++

++

+– –– –

q

gp78

UBAC2

Derlin-2

UBXD8–S

gp78

UBXD8

UBAC2

VCP

UBE2G2

Derlin-2

n ol

l

p

mgp78

UBXD8

UBAC2

VCP

UBE2G2

Derlin-2

gp78

UBAC2–SUBAC2

UBXD8

UBAC2–SFF Luc shRNA

gp78 shRNA

UBXD8

UBAC2–SUBAC2

gp78

UBAC2–SFF Luc shRNAUBXD8 shRNA

+ + + ++

++

+– –– –

UBXD8–SFF Luc shRNA

gp78 shRNA

gp78

UBXD8–S

UBAC2

Input AP: S-prot

Input AP: S-prot

Input AP: S-prot

Input AP: S-prot

Input AP: S-prot

Input AP: S-prot

Input AP: S-prot

Input AP: S-protInput AP: S-prot

Input AP: S-prot

Input AP: S-protInput AP: S-prot

50

2575

p

+ + + ++

++

+- -- -

UBXD8–SFF Luc shRNA

Derlin-2 shRNA

Derlin-2

UBXD8–S

gp78

Input AP: S-prot

g h

nm

a

c

i

o

f

l

Figure 3 shRNA-mediated refinement of ERAD E3 ligase subnetworks.(a–q) S-tagged ERAD baits were transiently co-expressed with theindicated shRNAs in HEK293 cells. All complexes were affinitypurified (AP) in 1% digitonin and analysed by immunoblotting.S-prot, S-protein; FF, firefly. (a) XTP3-B–S expression, probe for Hrd1and SEL1L simultaneously. (b) Co-expression of myc–UBE2J1 andXTP3-B–S, probe for myc and SEL1L; (c) XTP3-B–S expression, probefor FAM8A1 and SEL1L. (d) S–OS-9 expression, probe for Hrd1 andSEL1L simultaneously. (e) Incorporation of refinements (a–d) to theHrd1 complex. (f) S–FAM8A1 expression, probe for Hrd1 and SEL1L.(g) Co-expression of myc–UBE2J1 and Hrd1–S, probe for myc and

SEL1L. (h) Co-expression of myc–UBE2J1 and S–SEL1L, probe formyc and Hrd1. (i) AUP1–S expression, probe for Hrd1 and SEL1L.(j) Co-expression of myc–UBE2G2 and AUP1–S, probe for myc andHrd1. (k) Refined interaction map for Hrd1 complex. (l) UBAC2–Sexpression with UBXD8 knockdown, probe for gp78. (m) UBAC2–Sexpression with gp78 knockdown, probe for UBXD8. (n) UBXD8–Sexpression with gp78 knockdown, probe for UBAC2. (o) UBXD8–Sexpression with UBAC2 knockdown, probe for Derlin-2, UBAC2 andgp78. (p) UBXD8–S expression with Derlin-2 knockdown, probe forgp78. (q) Refined interaction map for the gp78 complex. Uncroppedimages of blots are shown in Supplementary Fig. S12.

NATURE CELL BIOLOGY ADVANCE ONLINE PUBLICATION 5© 2011 Macmillan Publishers Limited. All rights reserved.

RESOURCE

Deconvolving the ERAD interaction network with RNAinterferenceTo begin to decipher the organization within the mammalian ERADinteraction network, we systematically analysed the Hrd1–SEL1L andgp78 subnetworks. Co-expression of S-tagged proteins with shorthairpin RNAs (shRNAs) targeting central subnetwork nodes wasused to ascertain the requirement of each component to maintainindividual interactions (Fig. 3). Following SEL1L knockdown, XTP3-Binteractions with Hrd1 (Fig. 3a), UBE2J1 (Fig. 3b) and FAM8A1(Fig. 3c) were abolished, and OS-9 lost its connection to Hrd1 (Fig. 3d).LC–MS/MS analyses confirmed that XTP3-B andOS-9 affinity-purifiedcomplexes from cells lacking SEL1L lost their interactions with alldownstream membrane and cytosolic components (for example Hrd1,data not shown). These data verify the essential role that SEL1Lplays in scaffolding luminal, substrate-recognition elements to theHrd1 transmembrane complex30,39,48,49, and reveal the independentinteractions of XTP3-B andOS-9with theHrd1–SEL1Lnode (Fig. 3e).Hrd1–SEL1L subnetwork connections to integral membrane and

cytosolic ERAD components differed in that SEL1L knockdown didnot affect the Hrd1–FAM8A1 (Fig. 3f) or Hrd1–UBE2J1 interactions(Fig. 3g). Similarly, loss of Hrd1 failed to sever the connections betweenSEL1L–UBE2J1 (Fig. 3h), SEL1L–AUP1 (Fig. 3i) or AUP1–UBE2G2(Fig. 3j). Thus, both Hrd1 and SEL1L bind to UBE2J1, eitherdirectly or through a factor not identified in our proteomic analysis.Hrd1 knockdown abolished the SEL1L–FAM8A1 interaction (Fig. 3f),indicating that SEL1L and FAM8A1 independently bind to Hrd1. Thisconclusion is reinforced by the maintenance of the Hrd1–FAM8A1interaction in Triton X-100 where SEL1L is lost (Fig. 1). These datarefine themolecular topology of theHrd1–SEL1L complex, and identifyFAM8A1 as an obligate, SEL1L-independent partner ofHrd1 (Fig. 3k).A second prominent, highly interconnected subnetwork is composed

of gp78, Derlin-2, UBAC2 and UBXD8 (Fig. 2). Knockdown ofUBXD8 did not disrupt the gp78–UBAC2 interaction (Fig. 3l), nordid knockdown of gp78 affect UBXD8–UBAC2 (Fig. 3m,n). Althoughgp78 binding was lost, maintenance of the UBXD8–UBAC2 interactionin Triton X-100 indicates that their organization occurs independentlyof gp78 (Fig. 1). However, the UBXD8–gp78 interaction was abrogatedby knockdown of UBAC2 (Fig. 3o) but not Derlin-2 (Fig. 3p). Thesedata allow refinement of the gp78 subnetwork topology (Fig. 3q)and identify a role for UBAC2 in the recruitment of UBXD8 to thegp78 complex.

Functional genomic analysis of ERAD componentsTo assess their functional roles in substrate degradation, we monitoredthe effect of RNA interference (RNAi)-mediated knockdown ofindividual ERAD components on steady-state fluorescence levels offluorescent ERAD substrate reporters21,50–53. Cell lines stably expressingGFP fusions representing three major topological classes of ERADsubstrates: luminal-glycosylated (null Hong Kong variant of α1-anti-trypsin (A1ATNHK)), luminal-non-glycosylated (A1ATNHK-QQQ andmutant transthyretin TTRD18G) and integral membrane-glycosylated(CFTR1F508; Fig. 4a) substrates were employed. We also included theAMPA-type glutamate receptor subunit GluR1, as it is retained inthe ER and degraded in a UPS-dependent manner (SupplementaryFig. S7). Cell lines expressing the cytosolic proteasome substrateGFPu (ref. 54) and GFP served as controls for ERAD-independent

effects that might alter UPS function, reporter gene expression or GFPfluorescence intensity.All substrate reporter lines responded to proteasome inhibition

with time-dependent increases in mean GFP fluorescence (Fig. 4b).Expression of a dominant-negative VCP mutant (H317A; ref. 52)severely impaired degradation of only the ERAD substrates, but notGFPu (Fig. 4c), in agreement with the strong dependence of ERADpathways on VCP and 26S proteasomes. The mannosidase inhibitorkifunensine selectively inhibited the degradation of A1ATNHK (Fig. 4c),consistent with an established requirement for mannose trimming ofthis glycoprotein for ERAD (refs 55,56). GluR1 and CFTR1F508 arealso glycoproteins (Supplementary Fig. S7c), but were unaffected bykifunensine (Fig. 4c), indicating that mannose trimming is unlikelyto be the dominant signal committing them to degradation, or thatthere is redundant, glycan-independent targeting for these polytopicproteins. These data reflect an implicit requirement for multiple,substrate-specific recognition elements within the ERAD interactionnetwork to deliver substrates to shared degradationmachinery.To identify the individual factors required for substrate degradation,

we generated an shRNA library targeting genes implicated in ERAD(Fig. 4d and Supplementary Table S5) and monitored their impact onthe mean GFP fluorescence of reporter cell lines (Fig. 4e and Supple-mentary Table S7). Any shRNA that significantly stabilized an ERADreporter was selected for further validation by re-screening throughall other reporter lines and confirmation of knockdown (see Methodsand Supplementary Fig. S8). Each substrate seemed to rely on a uniqueset of individual ERAD components for degradation (SupplementaryTable S7), which is illustrated as a hierarchically clustered heat map forcomparison (Fig. 4f). Of the 59 components our library targeted, onlythe non-ATPase subunit of the 19S regulatory particle PSMD2 andVCPwere essential for all ERAD substrates (Fig. 4f). GFPu was stabilized byknockdown of PSMD2, but not VCP, mimicking the effects of MG132and VCPH317A (Fig. 4b,c) and validating the strategy of using shRNA-mediated gene silencing with ERAD reporters to interrogate the contri-bution of individual components to the overall degradation process. Hi-erarchical cluster analysis demonstrated that substrates were segregatedby topology (luminal versus integral membrane), but not by glycosy-lation (Fig. 4f). Moreover, a surprising degree of heterogeneity withineach substrate’s requirement profile was observed, especially for sub-strates utilizing the same central ERAD components (for example Hrd1,discussed below). These characteristic patterns indicate that the ERADsystem operates largely as an adaptive network, in which unique combi-nations of common components process individual substrates. Such anadaptive mechanism could be explained by the formation of substrate-specific subcomplexes or by a multisubunit complex that utilizesdiscrete sets of components to achieve substrate-specific degradation.

An adaptive mechanism for Hrd1-dependent degradationWe merged the heat map of shRNA-mediated impairment for eachsubstrate (Fig. 4f) with the comprehensive ERAD interaction network(Supplementary Fig. S5) to generate integrated substrate-specificsnapshots of the physical and functional networks responsible for degra-dation (Supplementary Figs S9–S11). Loss of either E3 in the networkimpacted degradation in a substrate-specific manner. Hrd1 knockdownstabilized topologically disparate substrates including A1ATNHK,A1ATNHK-QQQ, TTRD18G and GluR1, but had little effect on GFPu or

6 NATURE CELL BIOLOGY ADVANCE ONLINE PUBLICATION

© 2011 Macmillan Publishers Limited. All rights reserved.

RESOURCE

Derlin-2NGly1UBE2G2ERFADOS-9EmptyDerlin-3JAMPUBE4BAha1FF LucUBE4AERDJ5XTP3-BUBXD2AUP1TTC35gp78VIMPTXD16E-Syt1EDEM1KIAA0090C15orf24

VCPHrd1UBAC2FAM8A1HERPVCIP135SVIPLONP2UBXD8COX4NBUBE2J1SEL1LDerlin-1

PSMD2

TT

RD

18

G

A1

AT

NH

K

A1

AT

NH

K-Q

QQ

Glu

R1

GF

Pu

CF

TR

ΔF5

08

ER lumen

Cytosol

GF

P

GFP

GFP

GFP

TTRD18G –GFPA1ATNHK – GFP

GFP– CFTR ΔF508

GFP–GluR1

GFPGFPu

GFPGFP

GFP

A1AT NHK-QQQ–GFP

GF

Pu

Glu

R1

TT

RD

18

G

A1

AT

NH

K

A1

AT

NH

K-Q

QQ

0

1.0

2.0

3.0

4.0

Fo

ld c

han

ge in

mean

GF

P f

luo

rescen

ce in

ten

sity

CF

TR

ΔF5

08

0 h1 h2 h3 h

MG132

Total number of shRNA constructs 309

Total number of target genes 59

Reported ERAD components 45

Potential ERAD components 14

Average number of shRNAs per target ~5

CFTRΔΔF508

SEL1L

XTP3-B OS-9

FAM8A1

AUP1

UBE2G2

UBE2J1

Hrd1

GFPu

SEL1L

XTP3-B OS-9

FAM8A1

AUP1

UBE2G2

UBE2J1

Hrd1

TTRD18G

SEL1L

XTP3-B OS-9

FAM8A1

AUP1

UBE2G2

UBE2J1

Hrd1

A1ATNHK

SEL1L

XTP3-B OS-9

FAM8A1

AUP1

UBE2G2

UBE2J1

Hrd1

A1ATNHK-QQQ

SEL1L

XTP3-B OS-9

FAM8A1

AUP1

UBE2G2

UBE2J1

Hrd1

GluR1

SEL1L

XTP3-B OS-9

FAM8A1

AUP1

UBE2G2

UBE2J1

Hrd1

Wild-type VCP

VCPH317A

GF

Pu

Glu

R1

TT

RD

18

G

A1

AT

NH

K

A1

AT

NH

K-Q

QQ

Cyto.

Integral

membrane Luminal

024

6Kifu

nen

sin

e(h

)

CF

TR

ΔF5

08

Degradation score+3.0Impaired

–3.0Enhanced

Transfect shRNA

library into ERAD

reporter lines

Measure GFP

fluorescence intensity

by flow cytometry

Incubate cells

for 72 h

GFP fluorescence intensity

ControlERADimpairment

Confirm knockdown

of target

Re-screen

positive hits

59 targets

309 shRNA

Cells

Select positive

hits

shRNA

z sco

re

GFP fluorescence intensity

ControlERADimpairmentC

ells

100Mr (K)

Lysate

shRNA

Target

Tubulin

FF

Luc

sh

RN

A 1

sh

RN

A 2

50

a

d

e

f

g

b c

Cyto. Integral

membrane

Luminal

Figure 4 Functional genomic screen to identify essential substrate-specific ERAD components. (a) Localization and topology of GFPreporters (TTRD18G–GFP, A1ATNHK–GFP, A1ATNHK-QQQ–GFP, GFP–GluR1,GFP–CFTR1F508 and GFPu) and GFP. (b) Time course of relative mean GFPfluorescence intensity levels for each ERAD reporter cell line treated withMG132 (10 µM). Cyto., cytosolic. (c) Heat maps reflecting the normalizedfold change in mean GFP fluorescence intensity of ERAD reporter linestransfected with wild-type or dominant-negative VCP (wild-type or H317A,top panel) and time course of treatment with kifunensine (30 µM, bottompanel). Fold change in mean GFP fluorescence intensity was normalizedto the levels measured for each reporter at the 3 h time point of MG132

treatment, and thus a degradation score of 3 is equivalent to the impairmentinduced by 3 h MG132 treatment. (d) Target composition of the shRNAlibrary. (e) Overview of the functional genomic screen. (f) Hierarchicallyclustered heat map of the normalized fold change in mean GFP fluorescenceintensity of ERAD reporter lines in response to shRNA-mediated knockdownof ERAD components. The normalization and colour scale are the same as inc. FF, firefly. (g) Functional data from the heat map shown in f were mappedonto the refined Hrd1 physical interaction network (Fig. 3k) to providean integrated snapshot of substrate-specific functional requirements forHrd1 network components. Uncropped images of blots are shown inSupplementary Fig. S12.

CFTR1F508 (Fig. 4f,g). Instead, CFTR1F508 was stabilized following gp78knockdown, as previously reported57. Substrates utilizing Hrd1 did not

share a common dependence onHrd1–SEL1L subnetwork components.Whereas FAM8A1 and SEL1L were essential for degradation of

NATURE CELL BIOLOGY ADVANCE ONLINE PUBLICATION 7

© 2011 Macmillan Publishers Limited. All rights reserved.

RESOURCE

b

Fo

ld in

du

ctio

n (lo

g2)

a

0

–1

1

ERFAD

BiP

GRP94

ORP150CNX

OS-9CANXXTP3-BEDEM2EDEM3

EDEM1NGly1

UGT2ERMan1

UGT1GLT25D1

UBE4BUFD1p47VCP

Npl4UBE4A

VIMP

UBXD8UBXD2

AUP1hHR23B

UBAC2

hHR23ATMUB1

HERP

UBE2J1

UBE2G2

Hrd1

ParkinFbx6CHIPRNF5gp78TRC8

RFP2TEB4

USP13VCIP135ATX3YOD1

COX4NBTTC35KIAA0090

C15orf24TMEM85C14orf122TMEM111ccdc47

TMEM93

C19orf63

SPPDerlin-2Derlin-1FAM8A1

Derlin-3

Red

ox

Glyca

n bin

ding

and p

roce

ssing

VCP c

ofac

tors

VCP re

cruitin

g

fact

ors U

biquitin

bindin

g

E2 en

zym

es

E3 en

zym

es

Deu

biquity

latin

g

enzy

mes

EMC

Oth

er

ER c

haper

ones

ERLIN2E-Syt2CPVLAha1JAMPPSMD2E-Syt1

ERLIN1

LONP2

BAP31

MMGT1

SVIP

SIL1

TXD16

2

3

5

6

GRP94CPVL

XTP3-B

ERFAD

OS-9

TXD16

AUP1

NGly1

VCP

UBE2G2

hHR23B UBE4A

Unidirectional

Reciprocal

EDEM1 CANX

PSMD2

Bait Interactor

ERdj5

TEB4

Npl4

Ufd1

VCIP135

YOD1ATX3

BiP

CHIP

EDEM2

EDEM3

AHA1

Derlin-3

ERMan1

HACE1

Fbx6

ORP150RFP2

SIL1

TMUB1

TRC8

UBE4B

UGT1

UGT2

Parkin

LumenCytoplasmMembrane

Fold induction (log2)

LONP2

COX4NB

TTC35

KIAA0090

TMEM93

TMEM111

C14orf122

C19orf63

MMGT1

TMEM85

C15orf24

Lumen

Cytoplasm

SEL1L

gp78

UBXD8

UBAC2

UBE2J1

Derlin-2

ERLIN2

UBXD2

E-Syt2E-Syt1

VIMP

Derlin-1

SPP

Hrd1

HERP

RNF5JAMPBAP31

SVIP

ERLIN1

FAM8A1

–3 3

Figure 5 Coordinated ER stress response of ERAD genes. qRT–PCR resultsfor validated and suspected ERAD components following treatment ofHEK293 cells with tunicamycin (10 µgml−1, 6 h). Data are presented asfold induction (log2) normalized to β-actin. Tunicamycin-induced expression

changes in ERAD genes plotted as groups according to: (a) fold induction ofgene expression represented by functional category, and (b) fold inductionof gene expression from a mapped onto the ERAD interactome from Fig. 2.Additional genes of interest are presented alongside the induction map.

A1ATNHK and TTRD18G and dispensable for GluR1, the converse wastrue for AUP1 and UBE2G2 (Fig. 4f,g). Furthermore, despite sharinga common requirement for VCP, ERAD substrates were differentiallydependent on VCP-interacting proteins such as SVIP, UBE4A andVCIP135 (Fig. 4f), perhaps indicating additional heterogeneity amongthe VCP-containing complexes employed for dislocation.

Coordinate regulation of ERAD genes by the unfoldedprotein responseExpression of more than half of the ERAD genes in our network,including the Hrd1–SEL1L subnetwork (Fig. 5) and other knownUPR targets (for example, BiP and HERP), was induced bytunicamycin (Fig. 5). In contrast, gp78 and other ER-resident

8 NATURE CELL BIOLOGY ADVANCE ONLINE PUBLICATION

© 2011 Macmillan Publishers Limited. All rights reserved.

RESOURCE

Wild-type Hrd1

Wild-type Hrd1 + SEL1LWild-type FAM8A1

DFPDFP–FAM8A1Δ1–229

FAM8A1Δ230–413

Hrd1C294A

10%

40%

1Fractions 2 3 4 5 6 7 8 9 10 11 12

Hrd1–S

10

% A

P

FAM8A1

SEL1L

GFPu GluR1 TTR D18G

FAM8A1

VCP

SEL1L

NaCl SDS

S PS PS P

Na2CO3

S–FAM8A1

KDEL

Overlay

S–FAM8A1

KDEL

Overlay

Hrd1–S

KDEL

Overlay

KDEL

Overlay

Hrd1–S

SEL1L

XTP3-B OS-9

FAM8A1

FAM8A1

Hrd1

413 aa

Amino-acid position

Relia

bili

ty

CONSENSUS TM1: 255–275TM2: 306–326 TM3: 371–391

RDD

Degradation score

10

% in

pu

t

Co

ntr

ol

FA

M8

A1

IP

Hrd1

SEL1L

Tubulin

FAM8A1

100

100

Mr (K)

Mr (K)

Mr (K)

50

50

0

0.5

1.0

0 100 200 300 400

Cyto. Lumen TMD

50

100

100

a b c d

Cytosolic and luminal epitopesCytosolic epitopes

75

35

100

ImpairedEnhanced+3.0-3.0

e f g

CONSENSUS

Figure 6 Characterization of the Hrd1-binding partner FAM8A1.(a) Domain structure and interaction network of FAM8A1. aa, aminoacids. (b) Immunoprecipitation (IP) with anti-FAM8A1 from HEK293digitonin-soluble lysates was analysed by immunoblotting with theindicated antibodies. (c) Consensus TOPCONS prediction of FAM8A1membrane orientation (http://topcons.cbr.su.se). The reliability indexindicates the likelihood for consensus prediction at each position usinga sliding 21 amino-acid window. Cyto., cytosolic; TMD, transmembranedomain. (d) HEK293 membrane fractions incubated with 1M NaCl,0.1M Na2CO3 at pH12 or 1% SDS. Following 100,000g centrifugation,equal volumes of soluble (S) and pellet (P) fractions were analysed bywestern blotting with anti-FAM8A1. (e) HeLa cells expressing S–FAM8A1or Hrd1–S were permeabilized with digitonin or Triton X-100 to allow

antibody access to cytosolic epitopes or cytosolic and luminal epitopes,respectively, immunostained and analysed by fluorescence microscopy.Scale bar, 10 µm. (f) Hrd1–S-expressing HEK293 cell lysates separatedon a continuous 10–40% sucrose gradient. S-tagged Hrd1 proteincomplexes were affinity purified from each 1ml fraction (fractions 1–12)or from 150mg whole-cell lysate (10% AP), and analysed by westernblotting for Hrd1 (S-tag), SEL1L and FAM8A1. (g) Heat map representingthe normalized change in mean GFP fluorescence intensity (20,000 cells,n=3) of the indicated ERAD reporter cell lines following transfection withthe indicated Hrd1, SEL1L and FAM8A1 plasmids. DFP indicates deadfluorescent protein, a non-fluorescent GFP variant. Data are representedas a normalized heat map as in Fig. 4c. Uncropped images of blots areshown in Supplementary Fig. S12.

E3s responded only weakly (Fig. 5). All but one of the mEMCcomponents were transcriptionally upregulated by tunicamycin(Fig. 5); in yeast, only EMC3 is upregulated by the UPR (refs 25,58).The selective response to ER stress indicates a previously unrecognized,coordinate transcriptional regulation of this physically and functionallyintegrated network. The contributions of the Hrd1–SEL1L and mEMCsubnetworks to the cellular response to ER stress underscore theimportant role ERAD plays in this process.

ERAD components identified within the Hrd1–SEL1L andgp78 subnetworksFAM8A1 was identified as a previously uncharacterized componentof the Hrd1–SEL1L subnetwork (Fig. 6a). Immunoprecipitation ofendogenous FAM8A1 captured Hrd1 and SEL1L (Fig. 6b), confirmingthat FAM8A1 is a bona fide interactor of both components.Resistance to extraction from purified microsomes by high saltconcentration or pH conditions support predictions for FAM8A1 as anintegral membrane protein with three membrane-spanning domains(TOPCONS, Fig. 6c,d), and limited proteolysis of FAM8A1-containingmicrosomes (data not shown) and immunodetection of an amino-terminal epitope tag in semipermeabilized cells (Fig. 6e) establishedthe cytoplasmic localization of the N terminus. A complex isolatedwith S-tagged Hrd1 contained both FAM8A1 and SEL1L, confirmingFAM8A1 as a component of this E3 ligase complex (Fig. 6f).Disrupting the stoichiometry of the Hrd1 E3 complex by FAM8A1

knockdown (Fig. 3f) or wild-type Hrd1 overexpression (Fig. 6g)

impaired degradation of TTRD18G while enhancing that of GluR1.TTRD18G degradation was restored or enhanced when Hrd1 wasco-expressed with SEL1L (Fig. 6g). Similarly, FAM8A1 overexpression(or its RDD domain, amino acids 230–413) impaired TTRD18G

but not GluR1 degradation, whereas a cytoplasmic N-terminalfragment (FAM8A11230-413) affected neither (Fig. 6g). The dominant-negative effect of FAM8A111–229) on TTRD18G stability implies thatHrd1 interacts with FAM8A1 through its RDD domain and thatits cytoplasmic N-terminal region is required for Hrd1-mediateddegradation of luminal substrates. Collectively, our results establishFAM8A1 as a binding partner and potential regulator of Hrd1-dependent ERAD.UBAC2, identified as a UBXD8 interaction partner (Fig. 7a), is

predicted to be a rhomboid family pseudoprotease similar to theDerlin proteins59 that also contains a putative C-terminal UBA domain.A native interaction between the two was validated by endogenousco-precipitation (Fig. 7b). Functionally, UBAC2 knockdown stabilizedthe Hrd1 substrate TTRD18G–GFP (Fig. 7c), indicating potentialcoordination between the two ubiquitin ligase complexes. Both theUBAC2 C terminus (amino acids 304–344) and the N terminus ofUBXD8 (2–52) show a high degree of conservation with residuesessential for ubiquitin binding in UBA domains (Fig. 7d), andtheir predicted cytosolic localization positions them appropriatelyfor ubiquitin binding (Fig. 7e). Whereas a recombinant UBAC2C-terminal fragment (amino acids 293–344) was sufficient to capturepolyubiquitin chains from HEK293 cell lysates at a level comparable

NATURE CELL BIOLOGY ADVANCE ONLINE PUBLICATION 9

© 2011 Macmillan Publishers Limited. All rights reserved.

RESOURCE

gp78

UBXD8

UBAC2

E-Syt1 SPPERLIN1 ERLIN2

UBAC2–S

KDEL

Overlay

UBAC2–S

KDEL

Overlay

gp78–S

KDEL KDEL

OverlayOverlay

gp78–S Beadalone

UBAC2293–344

UBXD81–77

hPlic2575–624

Anti-ubiquitin

MG132 – + – + – + – + – + – + – + – +

Beadalone

UBAC2293–344

UBXD81–77

hPlic2575–624

344 aa

UBA

Cytosolic and luminal epitopesCytosolic epitopes

a b

d

c

e f

UBAC2

UBAC2

Dsk2

hPlic-2

hHR23A-1

hHR23A-2

hHR23B-2

hHR23B-1

UBXD8

UBAC2_HUMAN/304–344

DSK2_YEAST/327–371

UBQL2_HUMAN/581–621

RD23A_HUMAN/161–201

RD23A_HUMAN/318–358

RD23B_HUMAN/364–404

RD23B_HUMAN/190–228

UBXD8_HUMAN/11–53

CONSENSUS

Input Pulldown

MMGT1

COX4NB

TMEM85

TTC35

C15orf24KIAA0090

TMEM93

TMEM111

C14orf122

C19orf63

10

% in

pu

t

Co

ntr

ol

UB

XD

8

IP

UBXD8

UBAC2

Tubulin

Fo

ld c

han

ge in

mean

GF

P f

luo

rescen

ce in

ten

sity

shRNA:

50

Mr (K)

50

25

1.0

1.5

0.5

0

2.0

FF

Luc

Hrd

1

UB

AC

2-1

UB

AC

2-2

...EVSEEQVARLMEM.GFSR.GDALEALRASNNDLNVATNFLLQH

PPEERYEHQLRQLNDM.GFFDFDRNVAALRRSGGSVQGALDSLLNG

....RFQQQLEQLNAM.GFLNREANLQALIATGGDINAAIERLLGS

...SEYETMLTEIMSM.GYER.ERVVAALRASYNNPHRAVEYLLTG

...PQEKEAIERLKAL.GFPE.SLVIQAYFACEKNENLAANFLLSQ

...PQEKEAIERLKAL.GFPE.GLVIQAYFACEKNENLAANFLLQQ

...QSYENMVTEIMSM.GYER.EQVIAALRASFNNPDRAVEYLLMG

...QEQTEKLLQFQDLTGIESMDQCRHTLEQHNWNIEAAVQDRLNE

100

75

50

50

250Mr (K)

150

α1 α2 α3

Anti-tubulin

Figure 7 Characterization of UBAC2, a ubiquitin-binding ERADcomponent. (a) Predicted domain structure and interaction network ofUBAC2. aa, amino acids. (b) Immunoprecipitation (IP) with anti-UBXD8from HEK293 digitonin-soluble lysates was analysed by western blottingwith the indicated antibodies. (c) Analysis of multiple UBAC2 -targetingshRNAs on the Hrd1 substrate TTRD18G–GFP by flow cytometry. FF, firefly.(d) Sequence alignment of the predicted UBA domains from UBAC2(304–344) and UBXD8 (8–53) with characterized human and yeast

UBA domains. (e) HeLa cells expressing C-terminally S-tagged UBAC2 orgp78 were permeabilized, immunostained and analysed by fluorescencemicroscopy as in Fig. 6e. Scale bar, 10 µm. (f) Recombinantly expressedUBA domains of hPlic2, UBXD8 and UBAC2 were coupled to Affi-Geland incubated with HEK293 cell lysates (±10 µM MG132, 6 h). Sampleswere separated by SDS–PAGE, and ubiquitin binding was determined byimmunoblotting with anti-ubiquitin. Uncropped images of blots are shownin Supplementary Fig. S12.

to the well-characterized hPlic2 UBA (ref. 60), under these conditionsthe UBXD8 UBA domain was not (Fig. 7f). Thus, it is UBAC2 ratherthan UBXD8 that adds polyubiquitin-binding capabilities to thegp78 subnetwork.

DISCUSSIONThe application of high-content proteomics to identify interconnec-tivity within defined functional networks has been used with successto map high-resolution interaction landscapes for several complexmammalian protein networks21,22,61. In this study, we have integratedthe mammalian ERAD interaction landscape with gene expressiondata and substrate-specific functional ‘fingerprints’ of the mappedcomponents to generate a multidimensional view of this dynamic andcomplex network. Our data indicate that the mammalian ERAD systemmay accommodate the diverse array of potential substrates by using

combinatorial interactions of the two central E3s, Hrd1 and gp78, witha palette of accessory factors (Fig. 8).Although many individual components of the mammalian ERAD

system have been previously identified, so far there has been nosystematic effort to place them into an integrated interaction landscape.Our study confirmed many of the interactions previously reported inmammals62 and those inferred from yeast16,63 (shown in Fig. 8; blacklines), validating our approach and allowing us to arrange componentsinto a topologically and functionally coherent model (Fig. 8). Thisanalysis also identified 71 HCIPs that are either uncharacterized orhave not previously been linked to ERAD (Fig. 8, only selected nodesand interactions shown), illustrating the ability of focused proteomicstrategies to uncover new components. Our analysis integratesinteraction and functional data from the present study into a frameworkconsisting of six functional modules that execute the principal ERAD

10 NATURE CELL BIOLOGY ADVANCE ONLINE PUBLICATION

© 2011 Macmillan Publishers Limited. All rights reserved.

RESOURCE

HRD1 complex

Deubiquitylating

Recruitment factors

Substrate extraction

RINGE3 ligase

E2 enzyme

FAM8A1

Shuttling factor?UBA/UBL

hHR23B

UBE2J1

UBE2G2

HERP

ERLIN2

ERLIN1

VCIP135

Usp13 Deglycosylating

NGly1Glycanase /

PUB

ATX3

YOD1

UBXUBA/UBX

CPVLERdj5

Reductase

ReductaseThioredoxin

TXD16

Chaperone Chaperone Chaperone

Lectin Lectin

Protease

Substrate recognition and processing

Ubiquitin

extension

Adaptor

E2 enzyme

CUE

UBL

Hrd1 regulator?

VCPAAA+

ATPase

U-boxE4enzyme

OTU/UBXL

Josephine /

UIM

UBP/UBA

OTU/UBXL

Lumen

Cytosol

Membrane

gp78 complex

UBA

SPFH

SPFH

SPFH

SPFH

E2 enzyme

UBE2G2

BiP GRP94

Ubiquitin ligase

Ubiquitin ligase

UPR induced

GFPu

GluR1TTRD18G

A1ATNHK

A1ATNHK-QQQ

CFTRΔF508

Gene induction

Functional requirement

Legend

1.1–1.5× >1.5×Impact scale

Inter-module

Intra-module

Interactions

XTP3-B OS-9

Trafficking? DerlinsDislocationDerlin-3

protease

ProcessingESYT1 C2

C2ESYT2

gp78

CANX

Lectin Lectin

Hrd1

ERFAD

EDEM1

SEL1L

UBAC2

SPP

AUP1

CUE /

RING

E3 ligase

UBE4A

Derlin-2Derlin-1mEMC complex

WD40

repeat

MPN

MPN

TPRrepeat

C19orf63 TMEM85

TMEM93 Rab5IP

MMgT

COX4NB

TTC35

C15orf24

C14orf122

MMGT1

TMEM111

KIAA0090

ERLIN2

ERLIN1

UBXD8 VIMP UBXD

Proteasome

19S regulatory particle

20S core particle

Activator

PSME4

PSMA1,2,3,4,5,6,7

PSMB1,2,3,4,5,6,7,8

PSMC1*,2,3*,4*,5*,6*

PSMD1*,2*,3,4*,6*,7*,8*,11*,12*,13*,14

Proteasome

PSMA1,2,3,4*,5,6,7*

PSMB1,2,3,4,5*,6,7,8

PSMC1*,2*,3*,4*,5*,6*

PSMD1,2*,3*,4*,6*,7,

8*,11*,12*,13*,14*

19S regulatory particle

20S core particle

∗ BiP∗

EDEM1

complex

OS-9 complexXTP3-B complexERFAD complex

Figure 8 Functional integration of mammalian ERAD networks. Theschematic model of the ERAD protein interaction network is topologicallyorganized with respect to the ER membrane and arranged as anarray of six colour-coded functional modules. Individual componentsfrom this study (baits or HCIPs) are indicated as nodes with reportedcomponents (black) and previously unknown components (red). Similarly,reported interactions confirmed in this study (black lines) and previouslyunknown interactions (red lines) are shown. Symbols for protein–protein

interactions, UPR induction and functional requirements are indicatedin the legend. Inter-module interactions represented terminate eitherat the specific node within a module that establishes the link with themodule periphery or at the module itself (where there are interactionswith multiple components and that module is a single complex; forexample, the mEMC or proteasome). Asterisks indicate components thatwere identified by proteomics, but exhibited a subthreshold CompPASSscore (WDN-score<1.0).

activities: substrate recognition, dislocation, extraction, ubiquitylationand degradation (proteasome), as well as the EMC whose function isunknown at present. Submodules are grouped together on the basisof predicted structural and topological features, and on an unbiasedanalysis of network interconnectivity of the proteins represented ineach group. The ERAD system can thus be viewed as a distributednetwork, organized around central ubiquitin ligase modules for Hrd1and gp78 that cooperate with components of themembrane-embeddeddislocation and the cytoplasmically-oriented substrate extractionmodules. These interconnections are likely to ensure secure couplingbetween substrate dislocation/extraction and ubiquitin conjugation.The Hrd1 and gp78 complexes contain submodule-specific factors andshare interactions with ERLIN1/2 and UBE2G2. The Hrd1 submodulehas four main connections to other modules. Three are mediated

through SEL1L, which connectsHrd1 to the upstream luminal substraterecognition machinery, as well as to the downstream dislocationmodule through Derlin-2 and the substrate extraction module throughUBXD8. Direct interactions between the last two proteins and VCPprovide an extended pathway from the luminal substrate-bindinglectins OS-9 and XTP3-B to cytoplasmic VCP. The third connection isa direct link between this E3 and the 26S proteasome.The gp78 submodule seems to connect to the ERADnetwork through

UBAC2. This protein interacts with UBXD8 and Derlin-1/2, and hasa functional polyubiquitin-binding domain, indicating that it mayfunction as a membrane nexus integrating ubiquitin conjugation,dislocation and extraction. A VCP-binding site within the cytoplasmicdomain of gp78 (ref. 42) means that this complex can associate withVCP in at least two ways. VCP interacts directly with multiple compo-

NATURE CELL BIOLOGY ADVANCE ONLINE PUBLICATION 11

© 2011 Macmillan Publishers Limited. All rights reserved.

RESOURCE

nents of the ERADmachinery including Derlin-1 and Derlin-2 (refs 27,59,64), VIMP (refs 26,27), gp78 (ref. 42), UBXD2 (ref. 65) and UBXD8(refs 66,67). With at least six different recruitment sites for VCP withinthe ERAD network, it is not surprising that disruption or silencing ofthis cytoplasmic AAA+ATPase has amore universal effect on the degra-dation of diverse ERAD substrates when compared with the loss of anindividual factor (Fig. 4f).Multiple recruitment avenues at the ERmem-branemay reflect an acquired adaptability of VCP to accommodate andengage the diverse substrates it encounters. Moreover, VCP accessoryfactors (for example, UBE4A, VCIP135 and SVIP), functionally essen-tial for specific substrates (Fig. 4f), could confer an added level of speci-ficity or may reflect a requirement for different VCP configurations atdifferent steps of the dislocation andmembrane extraction processes.‘Input’ of luminal substrates into the Hrd1 submodule occurs

through the well-established interaction with SEL1L. A capacity ofthe Hrd1 submodule to engage substrates independently of SEL1L isalso supported by several observations: SEL1L is dispensable for GluR1degradation (Fig. 4f); Hrd1 overexpression enhanced GluR1 degrada-tion while stabilizing TTRD18G (Fig. 6g); and co-expression of SEL1LwithHrd1 resulted in enhanced degradation of both substrates (Fig. 6g).One hypothesis is that Hrd1 recognizes substrates directly throughits membrane-spanning region, as suggested from studies in yeast68.We speculate that, given its close interaction with Hrd1, FAM8A1may regulate the partitioning of Hrd1 between SEL1L-dependentand -independent modes of substrate recognition. This model forFAM8A1 regulation of Hrd1 partitioning is supported by the opposingeffects that FAM8A1 depletion has on SEL1L-dependent (A1ATNHK,A1ATNHK-QQQ, TTRD18G) and SEL1L-independent (GluR1) substrates.How substrates are directed to the gp78 submodule is less clear,

as no high-confidence interactions between components of this E3submodule and components of the substrate recognition module weredetected in our study or have been reported. Given the large number ofcommon components within the dislocation and substrate extractionmodules that interact with both E3 submodules, it is possible that thesetwo principal ERAD E3s cooperate to degrade substrates, consistentwith some ERAD substrates being partially stabilized by knockdown ofeither Hrd1 or gp78. Indeed, several examples of cooperative functionby pairs of mammalian ERAD-associated E3s have been reported,including RMA1–CHIP andRMA1–gp78 in the ubiquitylation of CFTR(refs 57,69) and also Hrd1–gp78 (ref. 70).Our data support an organizational model for ERAD where a

dynamic network of interacting functional modules facilitate therecognition, recruitment, dislocation, extraction, ubiquitylation anddegradation of the diverse classes of secretory pathway proteins. Thiswork should provide a resource for future analysis of this cellularquality-control system. �

METHODSMethods and any associated references are available in the onlineversion of the paper at http://www.nature.com/naturecellbiology

Note: Supplementary Information is available on the Nature Cell Biology website

ACKNOWLEDGEMENTSThis work was supported by grants from the NIH to R.R.K. and J.W.H. J.C.C. wassupported by funding from the Ludwig Institute for Cancer Research. J.A.O. andR.E.T were supported by NRSA fellowships from NIH. E.J.B. was supported by afellowship from the Damon Runyon Cancer Research Foundation (DRG 1974-08).

We thank the members of the Kopito laboratory for helpful discussion, and M.Pearce, J. Hwang and C. Beveridge for critical reading of the manuscript.

AUTHOR CONTRIBUTIONSThe manuscript was written collectively by J.C.C., J.A.O. and R.R.K. Experimentsand data analysis were carried out by J.A.O. and J.C.C. with assistance from C.M.R.R.E.T. and E.J.G. LC–MS/MS analysis was carried out by T.A.S. CompPASS analysiswas carried out by M.E.S. and E.J.B with support from J.W.H.

COMPETING FINANCIAL INTERESTSThe authors declare no competing financial interests.

Published online at http://www.nature.com/naturecellbiologyReprints and permissions information is available online at http://www.nature.com/reprints

1. Ghaemmaghami, S. et al. Global analysis of protein expression in yeast. Nature 425,737–741 (2003).

2. Hebert, D. N., Bernasconi, R. & Molinari, M. ERAD substrates: which way out?Semin. Cell Dev. Biol. 21, 526–532 (2010).

3. Buchberger, A., Bukau, B. & Sommer, T. Protein quality control in the cytosol andthe endoplasmic reticulum: brothers in arms. Mol. Cell 40, 238–252 (2010).

4. Vembar, S. S. & Brodsky, J. L. One step at a time: endoplasmic reticulum-associateddegradation. Nat. Rev. Mol. Cell Biol. 9, 944–957 (2008).

5. Aridor, M. Visiting the ER: the endoplasmic reticulum as a target for therapeutics intraffic related diseases. Adv. Drug Deliv. Rev. 59, 759–781 (2007).

6. Xie, W. & Ng, D. T. ERAD substrate recognition in budding yeast. Semin. Cell Dev.Biol. 21, 533–539 (2010).

7. Bagola, K., Mehnert, M., Jarosch, E. & Sommer, T. Protein dislocation from the ER.Biochim. Biophys. Acta 1808, 925–936 (2011).

8. Hoseki, J., Ushioda, R. & Nagata, K. Mechanism and components of endoplasmicreticulum-associated degradation. J. Biochem. 147, 19–25 (2010).

9. Kostova, Z., Tsai, Y. C. & Weissman, A. M. Ubiquitin ligases, critical mediatorsof endoplasmic reticulum-associated degradation. Semin. Cell Dev. Biol. 18,770–779 (2007).

10. Carvalho, P., Stanley, A. M. & Rapoport, T. A. Retrotranslocation of a misfoldedluminal ER protein by the ubiquitin-ligase Hrd1p. Cell 143, 579–591 (2010).

11. Ploegh, H. L. A lipid-based model for the creation of an escape hatch from theendoplasmic reticulum. Nature 448, 435–438 (2007).

12. Hampton, R. Y., Gardner, R. G. & Rine, J. Role of 26S proteasome and HRDgenes in the degradation of 3-hydroxy-3-methylglutaryl-CoA reductase, an integralendoplasmic reticulum membrane protein. Mol. Biol. Cell 7, 2029–2044 (1996).

13. Bordallo, J., Plemper, R. K., Finger, A. & Wolf, D. H. Der3p/Hrd1p is required forendoplasmic reticulum-associated degradation of misfolded lumenal and integralmembrane proteins. Mol. Biol. Cell 9, 209–222 (1998).

14. Bays, N. W., Gardner, R. G., Seelig, L. P., Joazeiro, C. A. & Hampton, R. Y.Hrd1p/Der3p is a membrane-anchored ubiquitin ligase required for ER-associateddegradation. Nat. Cell Biol. 3, 24–29 (2001).

15. Swanson, R., Locher, M. & Hochstrasser, M. A conserved ubiquitin ligase of thenuclear envelope/endoplasmic reticulum that functions in both ER-associated andMatα2 repressor degradation. Genes Dev. 15, 2660–2674 (2001).

16. Carvalho, P., Goder, V. & Rapoport, T. A. Distinct ubiquitin-ligase complexes defineconvergent pathways for the degradation of ER proteins. Cell 126, 361–373 (2006).

17. Ravid, T., Kreft, S. G. & Hochstrasser, M. Membrane and soluble substrates ofthe Doa10 ubiquitin ligase are degraded by distinct pathways. EMBO J. 25,533–543 (2006).

18. Vashist, S. & Ng, D. T. Misfolded proteins are sorted by a sequential checkpointmechanism of ER quality control. J. Cell Biol. 165, 41–52 (2004).

19. Deng, M. & Hochstrasser, M. Spatially regulated ubiquitin ligation by an ER/nuclearmembrane ligase. Nature 443, 827–831 (2006).

20. Mehnert, M., Sommer, T. & Jarosch, E. ERAD ubiquitin ligases: multifunctionaltools for protein quality control and waste disposal in the endoplasmic reticulum.Bioessays 32, 905–913 (2010).

21. Sowa, M. E., Bennett, E. J., Gygi, S. P. & Harper, J. W. Defining the humandeubiquitinating enzyme interaction landscape. Cell 138, 389–403 (2009).

22. Behrends, C., Sowa, M. E., Gygi, S. P. & Harper, J. W. Network organization of thehuman autophagy system. Nature 466, 68–76 (2010).

23. Braun, P. et al. An experimentally derived confidence score for binary protein–proteininteractions. Nat. Methods 6, 91–97 (2009).

24. Riemer, J., Hansen, H. G., Appenzeller-Herzog, C., Johansson, L. & Ellgaard, L.Identification of the PDI-family member ERp90 as an interaction partner of ERFAD.PLoS One 6, e17037 (2011).

25. Jonikas, M. C. et al. Comprehensive characterization of genes required for proteinfolding in the endoplasmic reticulum. Science 323, 1693–1697 (2009).

26. Ye, Y., Shibata, Y., Yun, C., Ron, D. & Rapoport, T. A. A membrane protein complexmediates retro-translocation from the ER lumen into the cytosol. Nature 429,841–847 (2004).

27. Lilley, B. N. & Ploegh, H. L. Multiprotein complexes that link dislocation,ubiquitination, and extraction of misfolded proteins from the endoplasmic reticulummembrane. Proc. Natl Acad. Sci. USA 102, 14296–14301 (2005).

12 NATURE CELL BIOLOGY ADVANCE ONLINE PUBLICATION

© 2011 Macmillan Publishers Limited. All rights reserved.

RESOURCE

28. Li, G., Zhao, G., Zhou, X., Schindelin, H. & Lennarz, W. J. The AAA ATPase p97 linkspeptide N-glycanase to the endoplasmic reticulum-associated E3 ligase autocrinemotility factor receptor. Proc. Natl Acad. Sci. USA 103, 8348–8353 (2006).

29. Koegl, M. et al. A novel ubiquitination factor, E4, is involved in multiubiquitin chainassembly. Cell 96, 635–644 (1999).

30. Riemer, J. et al. A luminal flavoprotein in endoplasmic reticulum-associateddegradation. Proc. Natl Acad. Sci. USA 106, 14831–14836 (2009).

31. Min, S. W., Chang, W. P. & Sudhof, T. C. E-Syts, a family of membranousCa2+-sensor proteins with multiple C2 domains. Proc. Natl Acad. Sci. USA 104,3823–3828 (2007).

32. Pettersson, M., Bessonova, M., Gu, H. F., Groop, L. C. & Jonsson, J. I.Characterization, chromosomal localization, and expression during hematopoieticdifferentiation of the gene encoding Arl6ip, ADP-ribosylation-like factor-6 interactingprotein (ARL6). Genomics 68, 351–354 (2000).

33. Carrel, D. et al. Targeting of the 5-HT1A serotonin receptor to neuronal dendrites ismediated by Yif1B. J. Neurosci. 28, 8063–8073 (2008).

34. Goldstein, J. L. & Brown, M. S. Regulation of the mevalonate pathway. Nature 343,425–430 (1990).

35. Ravid, T., Doolman, R., Avner, R., Harats, D. & Roitelman, J. The ubiquitin-proteasome pathway mediates the regulated degradation of mammalian 3-hydroxy-3-methylglutaryl-coenzyme A reductase. J. Biol. Chem. 275, 35840–35847 (2000).

36. Song, B-L., Sever, N. & DeBose-Boyd, R. A. Gp78, a membrane-anchored ubiquitinligase, associates with Insig-1 and couples sterol-regulated ubiquitination todegradation of HMG CoA reductase. Mol. Cell 19, 829–840 (2005).

37. Nguyen, A. D., Lee, S. H. & DeBose-Boyd, R. A. Insig-mediated, sterol-accelerateddegradation of the membrane domain of hamster 3-hydroxy-3-methylglutaryl-coenzyme A reductase in insect cells. J. Biol. Chem. 284, 26778–26788 (2009).

38. Kikkert, M. et al. Human HRD1 is an E3 ubiquitin ligase involved in degradation ofproteins from the endoplasmic reticulum. J. Biol. Chem. 279, 3525–3534 (2004).

39. Christianson, J. C., Shaler, T. A., Tyler, R. E. & Kopito, R. R. OS-9 and GRP94deliver mutant α1-antitrypsin to the Hrd1–SEL1L ubiquitin ligase complex for ERAD.Nat. Cell Biol. 10, 272–282 (2008).

40. Jo, Y., Sguigna, P. V. & DeBose-Boyd, R. A. Membrane-associated ubiqui-tin ligase complex containing gp78 mediates sterol-accelerated degradationof 3-hydroxy-3-methylglutaryl-coenzyme A reductase. J. Biol. Chem. 286,15022–15031 (2011).

41. Chen, B. et al. The activity of a human endoplasmic reticulum-associateddegradation E3, gp78, requires its Cue domain, RING finger, and an E2-bindingsite. Proc. Natl Acad. Sci. USA 103, 341–346 (2006).

42. Ballar, P., Shen, Y., Yang, H. & Fang, S. The role of a novel p97/valosin-containingprotein-interacting motif of gp78 in endoplasmic reticulum-associated degradation.J. Biol. Chem. 281, 35359–35368 (2006).

43. Glickman, M. H., Rubin, D. M., Fried, V. A. & Finley, D. The regulatory particle ofthe Saccharomyces cerevisiae proteasome. Mol. Cell Biol. 18, 3149–3162 (1998).

44. Verma, R. et al. Proteasomal proteomics: identification of nucleotide-sensitiveproteasome-interacting proteins by mass spectrometric analysis of affinity-purifiedproteasomes. Mol. Biol. Cell 11, 3425–3439 (2000).

45. Ustrell, V., Hoffman, L., Pratt, G. & Rechsteiner, M. PA200, a nuclear proteasomeactivator involved in DNA repair. EMBO J. 21, 3516–3525 (2002).

46. Blickwedehl, J. et al. Role for proteasome activator PA200 and postglutamylproteasome activity in genomic stability. Proc. Natl Acad. Sci. USA 105,16165–16170 (2008).

47. Sadre-Bazzaz, K., Whitby, F. G., Robinson, H., Formosa, T. & Hill, C. P. Structureof a Blm10 complex reveals common mechanisms for proteasome binding and gateopening. Mol. Cell 37, 728–735 (2010).

48. Hosokawa, N. et al. Human XTP3-B forms an endoplasmic reticulum quality controlscaffold with the HRD1-SEL1L ubiquitin ligase complex and BiP. J. Biol. Chem. 283,20914–20924 (2008).

49. Cormier, J. H., Tamura, T., Sunryd, J. C. & Hebert, D. N. EDEM1 recognition anddelivery of misfolded proteins to the SEL1L-containing ERAD complex. Mol. Cell34, 627–633 (2009).

50. Hampton, R. Y., Koning, A., Wright, R. & Rine, J. In vivo examination of membraneprotein localization and degradation with green fluorescent protein. Proc. Natl Acad.Sci. USA 93, 828–833 (1996).

51. Fiebiger, E. et al. Dissection of the dislocation pathway for type I membraneproteins with a new small molecule inhibitor, eeyarestatin. Mol. Biol. Cell 15,1635–1646 (2004).

52. Delabarre, B., Christianson, J., Kopito, R. & Brunger, A. Central pore residuesmediate the p97/VCP activity required for ERAD. Mol. Cell 22, 451–462 (2006).

53. Stagg, H. R. et al. The TRC8 E3 ligase ubiquitinates MHC class I molecules beforedislocation from the ER. J. Cell Biol. 186, 685–692 (2009).

54. Bence, N. F., Sampat, R. M. & Kopito, R. R. Impairment of the ubiquitin-proteasomesystem by protein aggregation. Science 292, 1552–1555 (2001).

55. Hosokawa, N. et al. Enhancement of endoplasmic reticulum (ER) degradation ofmisfolded Null Hong Kong α1-antitrypsin by human ERmannosidase I. J. Biol. Chem.278, 26287–26294 (2003).

56. Liu, Y., Choudhury, P., Cabral, C. M. & Sifers, R. N. Intracellular disposal ofincompletely folded human α1-antitrypsin involves release from calnexin andpost-translational trimming of asparagine-linked oligosaccharides. J. Biol. Chem.272, 7946–7951 (1997).

57. Morito, D. et al. Gp78 cooperates with RMA1 in endoplasmic reticulum-associateddegradation of CFTR 1 F508. Mol. Biol. Cell 19, 1328–1336 (2008).

58. Travers, K. J. et al. Functional and genomic analyses reveal an essential coordinationbetween the unfolded protein response and ER-associated degradation. Cell 101,249–258 (2000).

59. Greenblatt, E. J., Olzmann, J. A. & Kopito, R. R. Derlin-1 is a rhomboidpseudoprotease required for the dislocation of mutant α-1 antitrypsin from theendoplasmic reticulum. Nat. Struct. Mol. Biol. 18, 1147–1152 (2011).

60. Bennett, E. J. et al. Global changes to the ubiquitin system in Huntington’s disease.Nature 448, 704–708 (2007).

61. Bennett, E. J., Rush, J., Gygi, S. P. & Harper, J. W. Dynamics of cullin-RINGubiquitin ligase network revealed by systematic quantitative proteomics. Cell 143,951–965 (2010).

62. Mueller, B., Klemm, E. J., Spooner, E., Claessen, J. H. & Ploegh, H. L. SEL1Lnucleates a protein complex required for dislocation of misfolded glycoproteins. Proc.Natl Acad. Sci. USA 105, 12325–12330 (2008).

63. Goder, V., Carvalho, P. & Rapoport, T. A. The ER-associated degradation componentDer1p and its homolog Dfm1p are contained in complexes with distinct cofactors ofthe ATPase Cdc48p. FEBS Lett. 582, 1575–1580 (2008).

64. Oda, Y. et al. Derlin-2 and Derlin-3 are regulated by the mammalian unfoldedprotein response and are required for ER-associated degradation. J. Cell Biol. 172,383–393 (2006).

65. Liang, J. et al. Characterization of erasin (UBXD2): a new ER protein that promotesER-associated protein degradation. J. Cell Sci. 119, 4011–4024 (2006).

66. Lee, J. N., Zhang, X., Feramisco, J. D., Gong, Y. & Ye, J. Unsaturated fatty acidsinhibit proteasomal degradation of Insig-1 at a postubiquitination step. J. Biol. Chem.283, 33772–33783 (2008).

67. Alexandru, G. et al. UBXD7 binds multiple ubiquitin ligases and implicates p97 inHIF1α turnover. Cell 134, 804–816 (2008).

68. Sato, B. K., Schulz, D., Do, P. H. & Hampton, R. Y. Misfolded membrane proteins arespecifically recognized by the transmembrane domain of the Hrd1p ubiquitin ligase.Mol. Cell 34, 212–222 (2009).

69. Younger, J. M. et al. Sequential quality-control checkpoints triage misfolded cysticfibrosis transmembrane conductance regulator. Cell 126, 571–582 (2006).

70. Bernardi, K. M. et al. The E3 ubiquitin ligases Hrd1 and gp78 bind to and promotecholera toxin retro-translocation. Mol. Biol. Cell 21, 140–151 (2010).

NATURE CELL BIOLOGY ADVANCE ONLINE PUBLICATION 13

© 2011 Macmillan Publishers Limited. All rights reserved.

METHODS DOI: 10.1038/ncb2383

METHODSPlasmids and constructs. ERAD component complementary DNAs (Supple-mentary Table S10) were cloned into the pcDNA3.1 vector in frame with anS-tag (KETAAAKFERQHMDS) either at the N or C terminus. For some, theendogenous signal sequence was replaced by bovine preprolactin followed by theS-peptide. ERAD reporters were constructed by fusing eGFP in frame with theC termini of transthyretin (TTRD18G), an α1-anti-trypsin NHK variant (A1ATNHK)and an NHK variant lacking consensus glycosylation sites (A1ATNHK-QQQ), giftfrom N. Hosokawa, Kyoto University, Japan). An N-terminal eGFP fusion of ratGluR1 (GFP–GluR1–flop) was subcloned into pcDNA3.1(-) (gift from R. Malinow,University of California, San Diego, USA). GFP–CFTR1F508; ref. 71) and GFPu

(ref. 54) have been described previously. HA–GluR1 (rat) was a gift fromW. Green,University of Chicago, USA.

Bait selection. Primary baits were selected on the basis of previous reports oron orthology to known yeast ERAD components. Secondary baits were selectedusing multiple criteria, including: identification as an HCIP with several primarybaits; identification as an HCIP in both digitonin and Triton X-100; presence inLC–MS/MS analyses with high total spectral counts; a predicted ER localization; anda domain structure or previous reports suggesting a potential function of relevanceto ER quality control.

Mass spectrometry. Cells were collected in PBS and solubilized in lysis buffer(50mM Tris–HCl (pH 7.4), 150mM NaCl, 5mM EDTA) containing Completeprotease inhibitor cocktail (Roche), and either 1% digitonin or 1% Triton X-100.Lysates were spun twice, first at 1,000g and the supernatant was respun at 20,000g . Aquantity of 1–1.5mg of total protein from cleared lysates was affinity purified with S-protein agarose (Novagen). Bead-bound complexes were washed three times in lysisbuffer containing 0.1% digitonin or Triton X-100 then twice in 50mM ammoniumbicarbonate, at pH 8. Bound proteins were eluted by overnight treatment withRapigest (Waters) and subjected to trypsin digestion (Promega) before injection intothe mass spectrometer. Samples were analysed in duplicate/triplicate on a systemconsisting of a CTC-PAL autosampler (Leap Technologies), a capillary gradientHPLC pump (Agilent Model 1100) and a linear-ion-trapmass spectrometer (ModelLTQ, ThermoFisher Scientific). The acquired MS/MS spectra were searched usingthe MASCOT protein database search program (Matrix Science) against a fulldatabase of human protein sequences to which the set of S-tagged protein sequencesand S-protein sequence were added.

Data deposition. The spectral files reported in this article have beendeposited in the Proteome Commons Tranche repository (http://tranche.proteomecommons.org) and can be accessed using the following hash:ZPwKzx2i + VY1M2U0GB450u9kH7MFcrfOjXFam9/xha4QUN/6O5 + KLj/NnuEYMBTJPSwId/rJFvjsYbPbQrg8mDys7F8AAAAAAABMXA==.

CompPASS analysis. A MASCOT-generated list of all interacting proteins andtheir corresponding total spectral counts from duplicate MS analyses for each baitaffinity complex was merged with a database containing 102 unique baits analysedpreviously21. CompPASS output metrics were then adjusted using both a weightingfactor and normalization such that a WDN score greater than 1 signifies a HCIP.Briefly, the CompPASS-calculatedWD score assesses interacting protein abundance(peptide number), uniqueness (number of baits that interact with the protein) andreplication (number of experiments the interaction is observed in) to determineHCIPs (ref. 21). To analyse data sets containing high numbers of shared interactors(for example, autophagy network and ERAD network), a normalized WD score(WDN) was developed and has been described in extensive detail previously22. Thisscore includes a normalization factor based on the standard deviation of the scannumber for the interactor across the bait proteins, which was found to correlate withbona fide interacting proteins22. All MASCOT and CompPASS data can be accessedthrough INfERAD, an interactive web-based portal at http://falcon.hms.harvard.edu/ipmsmsdbs/comppass.html. The hierarchically clustered heatmap representingHCIP data was generated using MultiExperimental Viewer v4.7. It should be notedthat the high WDN score for the bait protein could be due to self-identificationor a self-interaction (Fig. 1 and Supplementary Tables S2 and S3) and these twopossibilities could not be distinguished in the present study. HCIP clusters wereselected manually by encompassing the largest number of HCIPs proximal to eachbait with a minimum of two HCIPs.

ERAD shRNA library. Target gene selection was based on a reported/suspectedrole in ERAD or on identification as an HCIP in proteomic analyses. shRNA targetsequences were selected from the literature, the RNAi codex online repository, orgenerated using siRNA selection programs as indicated (Supplementary Table S7).shRNAs were cloned into the pSUPERSTAR expression vector52. The librarycontains 309 shRNA constructs, including 3 negative control constructs, 222constructs targeting 45 reported ERAD components, and 87 constructs targeting 14potential ERAD components. Each gene is targeted by an average of∼5 shRNAs.

Cell culture, transfection and stable cell lines. HEK293 and HeLa cells weremaintained in DMEM (Mediatech) +10% animal serum complex (Gemini Bio-Products) at 37 ◦C and 5% CO2. Cells were transfected by the calcium-phosphateco-precipitation technique or with FuGENE6 (Roche). Stable HEK293 clones/poolsexpressing S-tagged ERAD components were selected by G418 resistance andlimiting dilution. Clonal HEK293 cell lines expressing GFP-fusion ERAD reportersare described below.

Functional genomic analysis of the ERAD network. Clonal HEK293 celllines expressing GFP-tagged substrates (GFPu, GFP–GluR1, TTRD18G–GFP,A1ATNHK–GFP, A1ATNHK-QQQ–GFP and GFP–CFTR1F508) were obtained by G418selection followed by limiting dilution and/or sorting by FACS as describedpreviously52. Cell line selection was based on multiple criteria including: expressionof full-length protein; a single GFP peak as measured by flow cytometry; andaccumulation with MG132 (10 µM for 12 h). For primary screening, reportercells were seeded in 96-well plates (15,000 cells per well) and reverse-transfectedusing FuGENE6 with individually arrayed pSUPERSTAR-shRNA plasmids(175 ng) and pcDNA3–mCherry (ChFP) (25 ng). After 72 h, cells were analysedby high-throughput flow cytometry (LSR-II, Becton Dickinson). Mean GFPfluorescence intensity was determined for ∼2,000 ChFP-positive cells andnormalized to the mean GFP fluorescence intensity of pSUPERSTAR emptyvector (n = 3). Potential positive hits (Z scores > 1.5) were re-screenedthrough all reporters, and the mean GFP fluorescence intensity for 20,000cells was measured. shRNAs scoring positive in three independent experimentsand also depleted for the target transcript were considered bona fide hits.The heat map (Fig. 4f) used mean GFP fluorescence values for each shRNAthat were normalized to values resulting from a 3 h MG132 treatment, soas to facilitate comparison of each shRNAs effect between reporter cell lines.

Immunopurification, affinity purification and immunoblotting. Cells werecollected and affinity purified (S-protein) as described above. For immunoprecipi-tations, anti-FAM8A1 or anti-UBXD8 antibodies were conjugated to beads using thePierce direct immunoprecipitation kit (Thermo Scientific) and incubated with celllysates. All samples were washed three times in lysis buffer, resuspended in Laemmlibuffer +10mM dithiothreitol, separated by SDS–PAGE and transferred to PVDFmembrane for western blotting.

Antibodies. Experiments used the following antibodies: anti-S-tag (1:5,000),anti-myc (9E10; 1:2,000), anti-Hrd1 (1:50; gift from R. Wojcikiewicz, SUNYUpstate Medical University, USA), anti-SEL1L, anti-UBXD8, anti-AUP1 (gifts fromH. Ploegh,Whitehead Institute, USA), anti-VCP (1:1,000; Novus), anti-HA (12CA5;1:1,000), anti-KDEL (1:500; Stressgen), anti-calnexin (1:500; Assay Designs), anti-GFP (1:1,000; Roche), anti-tubulin (gift from T. Stearns, Stanford University, USA)and anti-ubiquitin (FK2; 1:2,000; BioMol). The polyclonal rabbit anti-FAM8A1(1:20,000) antibody was generated against a synthetic human FAM8A1 peptide(residues 65–78, CDKLEPPRELRKRGE) by conjugation to KLH and immunizationof two rabbits (Proteintech Group).

Measurement of shRNA efficacy by qRT–PCR and western blotting. Fortarget gene messenger RNA depletion, cells seeded in 12-well plates were transfectedwith the pSUPERSTAR-shRNA construct. After 24 h, cells were divided betweentwo plates. Following an additional 48 h of growth, total RNA from plate onewas isolated using RNeasy columns (Qiagen). HEK293 cells from the duplicateplate were collected, stained with anti-CD4-allophycocyanin and analysed using aFACSCalibur (Becton Dickenson) to determine transfection efficiency. Transcriptlevels were subsequently measured by quantitative real-time PCR with reversetranscription (qRT–PCR) with an IQ5 Real Time PCR Detection System (Bio-Rad),using the iScript One-Step RT-PCR kit with SYBR green (Bio-Rad), in triplicatereactions according to the manufacturer’s instructions. Primer sequences are listedin Supplementary Table S9.

For analysis of shRNA-mediated effects on target protein levels, HEK293 cellswere transfected with the expression plasmid encoding S-tagged target protein alongwith the corresponding control or shRNA-targeting plasmid at a ratio of 1:3. Levelsof the S-tagged target protein were subsequently analysed by immunoblotting ofeither SDS-solubilized cell lysates or of S-protein agarose affinity-purified proteins.

Measurement of UPR induction of the ERAD network. HEK293 cells wereincubated in the absence or presence of 10 µgml−1 tunicamycin and total RNAwas isolated using RNeasy columns (Qiagen). Transcript levels were analysed byqRT–PCRwith an IQ5 Real Time PCRDetection System (Bio-Rad) using the iScriptOne-Step RT-PCR kit with SYBR green (Bio-Rad), in triplicate reactions, accordingto the manufacturer’s instructions. Individual reaction efficiencies were determinedusing the LinRegPCR software and fold change in gene expression was calculated

NATURE CELL BIOLOGY

© 2011 Macmillan Publishers Limited. All rights reserved.

DOI: 10.1038/ncb2383 METHODS

using the Pfaffl method with β-actin as the reference gene. Primer sequences arelisted in Supplementary Table S9.

Immunofluorescence microscopic analysis of protein localization andtopology. HeLa cells grown on poly-l-lysine-coated glass coverslips were fixedwith4% paraformaldehyde, permeabilized with 0.1% Triton X-100 and blocked by 1%bovine serum albumin. Primary antibody incubation (2 h at room temperature) wasfollowed by incubation with AlexaFluor secondary antibodies (Invitrogen, 1 h atroom temperature). Nuclei were stained with 10 µgml−1 bisbenzamide for 10minbefore mounting. For analysis of protein topology, HeLa cells grown and fixed asabove were permeabilized with either 20 µM digitonin for 1.5min or 0.1% TritonX-100 for 3min at room temperature to allow permeabilization of the plasmamembrane or permeabilization of both the plasma and ERmembranes, respectively.Cells were washed three times with PBS and immunostained as described above.Stained cells were visualized on a Zeiss Axiovert 200M (×40 air objective) and theresulting images were acquired digitally (Roper Scientific).

Sucrose gradient fractionation. HEK293 cells were lysed as described above. Aquantity of 1.5mg of total protein from cleared lysates was adjusted to 5% sucroseand loaded onto 10–40% continuous sucrose gradients (containing 0.1% digitonin)prepared using aGradientMaster (BioComp). Samples were centrifuged in an SW41rotor at 39,000 r.p.m. for 14.25 h at 4 ◦C. Fractions of 1ml in volume were collected,sucrose concentrations were adjusted to approximately 20% with lysis buffer, andS-tagged proteins were affinity purified with S-protein agarose.

Metabolic labelling and pulse-chase assay. Pulse-chase assays of GFP–GluR1reporter cells were carried out as described previously39.

Proteomic interactiondata set analysis. LC–MS/MS analysis of replicate affinity-purified protein complexes from 25 bait proteins identified a total of 10,481 proteinsin digitonin and 8,262 in Triton X-100. Approximately two-thirds were redundantentries, with the number of proteins having unique GenInfo Identifier numbersreduced to 3,325 (digitonin, 31.7%) and 2,917 (Triton X-100, 35.9%) for theentire analysis. A total of 320 (digitonin) and 202 (Triton X-100) entries weredesignated as HCIPs by CompPASS (WDN-score > 1) and included redundantentries representing shared interactors (Supplementary Tables S2 and S3). Abreakdown of digitonin and Triton X-100 HCIPs is presented in SupplementaryTable S4, including specific analyses for primary and secondary baits. The totalnumber of CompPASS-identified HCIPs represents all bait-interacting proteins.Bait proteins often had WDN-score > 10, but as we could not distinguish baitfrom an endogenous, homo-oligomerized counterpart, HCIPs representing theiridentical bait proteins were not considered (bait self-identification), neither weretrypsin (introduced during the experimental method (trypsin)) and proteins pulled

down non-specifically by S-protein agarose from untransfected HEK293 cells (beadcontrol). The result yielded 267 (digitonin) and 153 (Triton X-100) interactions(cellular HCIPs/ERAD interactions) with 143 (digitonin) and 97 (Triton X-100)corresponding to unique HCIPs. Proteins whose gene ontology assignments werecytoskeletal, mitochondrial, peroxisomal or nuclear (for example, MYH9, KRT8,PEX19, TOMM20 and MCM2) were deemed unlikely to be ERAD-related andremoved from subsequent analysis (likely false positives) and indicated as suchin individual interactomes (Supplementary Fig. S4). To determine the numberof new HCIPs identified, all primary and secondary baits were subtracted (otherprimary/secondary baits as prey), as well as proteins implicated in ERAD (forexample, VCP, GRP94 and proteasome subunits) but not used as bait (reportedERAD factors). Ultimately, we identified 59 (digitonin) and 30 (Triton X-100)uncharacterized proteins of interest with a wide range of topologies and functionaldomains, and a potential role within the ERAD network (Supplementary Fig. S3d,not including the 10 secondary baits).

Comparison and integration of proteomic data set with reportedinteractions. A list of reported pairwise interactions for ERAD components frompublished data was manually curated and used to generate a more comprehensivepicture of the mammalian ERAD interaction network (Supplementary Table S5).This table includes: an assigned ‘interaction number’; bait and prey definingthe interaction and whether our study used the bait (red); the method(s) ofidentification (for example, affinity capture-MS, two-hybrid, and so on) and therelevant reference(s) for the reported interaction; whether the reciprocal interactionwas shown and its corresponding ‘interaction number’; detection of the reportedinteraction in our digitonin or Triton X-100 interaction networks; and the presenceof this interaction in the STRING online protein interaction database. STRINGlists only 33/131 reported interactions (∼25%), supporting the use of a manuallycurated list rather an online resource. Of the 81 reported interactions using the25 primary/secondary baits, 31 were confirmed by our proteomic analysis with anadditional 3 being identified in a reciprocal interaction not previously reported.Seven more reported interactions were observed but were subthreshold (WDN< 1).We identified 21 reciprocal and 36 unidirectional interactions between baits andERAD components, and interactions with 71 uncharacterized proteins of interestthat scored above the threshold for HCIP classification (Supplementary Table S5and Fig. S3d). Combining reported pairwise interactions (Supplementary TableS5) with INfERAD (Fig. 2) and the RNAi-mediated ‘epistasis-like’ experiments(Fig. 3) yielded an up-to-date, comprehensive view of the ERAD interaction network(Supplementary Fig. S5).

71. Ward, C. L., Omura, S. & Kopito, R. R. Degradation of CFTR by the ubiquitin-proteasome pathway. Cell 83, 121–127 (1995).

NATURE CELL BIOLOGY

© 2011 Macmillan Publishers Limited. All rights reserved.

S U P P L E M E N TA RY I N F O R M AT I O N

WWW.NATURE.COM/NATURECELLBIOLOGY 1

DOI: 10.1038/ncb2383

Validated ERADcomponent

S-tagging and affinity capture

shotgunLC-MS/MS

Functional assay withERAD reporters

Identification ofInteractors

CompPASSanalysis

S-tagS

59 targets309 shRNA Tunicamycin

1° baits(15)

Stable expression in HEK293 cells

duplicate or triplicate runs

GFP fluorescence Cycle

PROTEOMICS FUNCTIONALGENOMICS

ER Stress

Quantitative RT-PCR

GENEEXPRESSION

Biochemical interactions

Functionalinteractions

Coordinate geneinduction

ERAD functionalnetworks

2° baits(10)

PS

MD

2V

CP

UB

E4A

ER

DJ5

XT

P3-

BU

BX

D2

AU

P1

Der

lin2

NG

LY1

UB

E2G

2E

RFA

DO

S9

Em

pty

Der

lin3

JAM

PU

BE

4AA

HA

1F.

Luc

.G

P78

VIM

PT

XD

16E

SY

T1

ED

EM

1K

IAA

0090

HR

D1

UB

AC

2FA

M8A

1H

ER

PV

CP

IP1

SV

IPLO

N2P

UB

XD

8C

OX

4NB

UB

E2J

1S

EL1

LT

TC

35T

ME

M33

Der

lin1

c15o

rf24

RNAi

Novel componentidentification

Kopito Supplemental Figure S1

Figure S1 Strategy to elucidate functional complexes of the mammalian ERAD pathway. Flow chart illustrating the strategy used to identify functional ERAD complexes by integrating high-content proteomics, functional genomics and gene expression data sets (see Methods).

© 2011 Macmillan Publishers Limited. All rights reserved.

S U P P L E M E N TA RY I N F O R M AT I O N

2 WWW.NATURE.COM/NATURECELLBIOLOGY

S U P P L E M E N TA RY I N F O R M AT I O N

A

B UBAC2-S KDEL Overlay

OverlayKDELS-FAM8A1

Hrd1-S KDEL Overlay

gp78-S KDEL Overlay

UBXD8-S KDEL Overlay

UBXD2-S KDEL Overlay

EDEM1-S

KDELS-ERFAD Overlay

KDELAUP1-S Overlay

KDEL Overlay

XTP3-B-S KDEL Overlay

S-OS9.2 KDEL Overlay

Derlin2-S

ERLIN2-S KDEL Overlay

KDEL OverlayVIMP-S

KDEL OverlayNGly1-S

OverlayKDELS-SEL1L

KDEL Overlay

Derlin1-S KDEL Overlay

TXD16-S KDEL Overlay

KDEL OverlayS-E-Syt1

TMEM111-S KDEL Overlay

KIAA0090-S KDEL Overlay

S-TTC35 KDEL Overlay

COX4NB-S KDEL Overlay

C15orf24-S KDEL Overlay

Kopito Supplemental Figure S2

Figure S2 Subcellular localisation of S-tagged ERAD components. HeLa cells, transiently expressing individual S-tagged ERAD components, were immunostained simultaneously with anti-S-tag (green) and anti-KDEL (red)

antibodies, with nuclei identified by bisbenzamide (blue). (a) Primary baits employed for initial proteomic analyses. (b) Secondary baits identified as HCIPs from initial proteomic screening (MMGT1-S not shown)

© 2011 Macmillan Publishers Limited. All rights reserved.

S U P P L E M E N TA RY I N F O R M AT I O N

WWW.NATURE.COM/NATURECELLBIOLOGY 3

Total InteractorsHCIPs

40

35

30

25

20

15

10

5

0

800

700

600

500

400

300

200

100

0

Tota

l Int

erac

tors

HC

IPs

Digitonin

Total InteractorsHCIPs

TritonX-10030

25

20

15

10

5

0

HC

IPs

600

500

400

300

200

100

0

Tota

l Int

erac

tors

gp78AUP1

bead

CTRL

c15o

rf24

XTP3-B

COX4NB

Derlin-

1

Derlin-

2

EDEM1Erlin

2

E-Syt1

FAM8A

1

ERFAD

KIAA00

90NGly1

OS-9

SEL1LVIM

PHrd1

TMEM111

TMEM32TTC35

TXD16

UBAC2

UBXD2

UBXD8gp

78AUP1

bead

CTRL

c15o

rf24

XTP3-B

COX4NB

Derlin-

1

Derlin-

2

EDEM1Erlin

2E-S

yt1

FAM8A

1

ERFAD

KIAA00

90NGly1

OS-9

SEL1LVIM

PHrd1

TMEM111

TMEM32TTC35

TXD16

UBAC2

UBXD2

UBXD8

B

A

C

!"

#"

$"

%"

&"

'!"

'#"

'$"

'%"

'&"

()*)+*,-,"

.-*/010,-,"

.-*,21+3045"

5(+(.*6-5"

70806*)901+"

7-:0;014(4*1"

<=>"7(9(/0"

<=>"?0)6-5(4*1"

@*67-1/"

90+(.*6-5"

9-+*,-,"

ABCD?"

)3*,)3*;26(4*1"

);*+0*62,-,"

?=>"E;*50,,-1/"

,-/1(6"+;(1,7F54*1"

+;(1,5;-)4*1"

+;(1,6(4*1"

F.-GF-41"

80,-560"+;(1,)*;+"

<-/-+*1-1"

B;-+*1HI'!!"

Fold

cha

nge

in re

pres

enta

tionD

!"#$%&'()*)#+,#-*.*/+%*%#0123)# !"#"$#%&%'(&#)*+*%&%'(&#%,+$-*./'/!$!(#0&/'1*2*0#"3*+$'1&4*5*+.!.#+'678'1!3!)*'678'9*"0&/!.#+':#01&+)'3*$!(#0&/'3&$#%&%';<=>9'"-#%"-#5,0!.#+'"5#$*#0,%&%'978'?5#/*%%&+)'%&)+!0'$5!+%1@/.#+'$5!+%/5&".#+'$5!+%0!.#+'@(&A@&.+'2*%&/0*'$5!+%"#5$'

TX-100

41 18(28) 12

Uncharacterized HCIPs of interest identified by CompPASS including the 10 HCIPs

that became secondary bait (grey)

ARF3DNAJA1FAM186BGGHHSP90AB2PRAB9ARPN1SERPINH1SLC25A10SURF4TBC1D17TMEM85

C14orf122C19orf63CAND1CASKCPVLEMDENO1LIN7CLONP2

NIPSNAP1NPEEPSPFKLPFKPRTN4SAE1TBC1D15TMEM93UBQLN2

Both

FAM8A1UBAC2FAM62ATXN1DC16KIAA0090

COX4NBTTC35C15orf24TMEM111MMGT1

Kopito Supplemental Figure S3

Digitonin

ARF5ARL6IP1ARL6IP5BRI3BPC19orf6CAPRIN2CLPTM1CLPTM1L

DIP2BDPY30DUSP3EPHX1FAM62BFDFT1GALK1GANAB

HMGCRHMOX1KLHDC2KIAA0746LRRC59MARCK5OSBPL8PGRMC2

RAB7ARPN2SEC22BSELKSLC27A3STT3ASYNJ2BPTAGLN2

TMED9TMEM201TMEM43TMUB1UBXD6USH2AVAPAVAPBYIF1B

!"#$%&'()*)#+,#-.*/+%01233#4567)# !"#"$#%&%'(&#)*+*%&%'(&#%,+$-*./'/!$!(#0&/'1*2*0#"3*+$'1&4*5*+.!.#+'678'1!3!)*'678'9*"0&/!.#+':#01&+)'3*$!(#0&/'3&$#%&%';<=>9'"-#%"-#5,0!.#+'"5#$*#0,%&%'978'"5#/*%%&+)'%&)+!0'$5!+%1?/.#+'$5!+%/5&".#+'$5!+%0!.#+'?(&@?&.+'2*%&/0*'$5!+%"#5$'

Figure S3 Analysis of HCIP abundance and gene ontology (GO). (a) The total number of interactors (red) and identified HCIPs (blue) are plotted for individual baits solubilised in DIG (top) or TX-100 (bottom). (b) Venn diagram showing the uncharacterised HCIPs obtained in DIG (red), TX-100 (blue), or in both (purple). 28 HCIPs were identified in both detergents, with

the 10 HCIPs boxed in grey selected and designated as secondary baits. (c) Individual pie graphs reflecting the relative percentage of HCIPs belonging to each designated GO annotation in DIG (top) or TX-100 (bottom). (d) Enrichment of HCIPs in GO defined categories. Determined as the ratio of the representative percentage of HCIPs vs. entire proteome.

© 2011 Macmillan Publishers Limited. All rights reserved.

S U P P L E M E N TA RY I N F O R M AT I O N

4 WWW.NATURE.COM/NATURECELLBIOLOGY

CLPTM1L

COX4NB

TTC35

TMEM111

C19orf63

C14orf122

TMEM93

TMEM32KIAA0090

UBQLN2

Hrd1

SLC27A3

OS-9Derlin-2

UBE2G2SEL1L

CPVLFAM8A1

RPL35

FLNA

PPIB

ENO1HNRNPU

HNRNPA2B1ATP5F1

ERFAD

PSMA7

PSMA2

PSMA3

PSMA5

PSMA6

PSMA1

PSMB1

PSMB2PSMB3

KLHDC2

SELK

UBXD6

Derlin-1Derlin-2

OS-9

UBE2J1

XTP3-B

GRP94

MYH9

SEL1LLONP2

hHR23B

VCP

Hrd1

OS-9

CAPRIN2SEL1L

SYNJ2BP

E-Syt2

ARL6IP5YIF1B

HNRNPM

C19orf6

DUSP3ERLIN1

SEL1L

XTP3-B

FAM8A1

AUP1

ERLIN2 UBXD2E-Syt1

VIMP

Derlin-1

Hrd1

EDEM1UBE2G2

DIP2B

SEL1L

CAPRIN2

YIF1B

E-Syt2

HMOX1

EPHX1

POR

E-Syt1

RTN4

SEC22BARL6IP5

VCPPGRMC2

UBXD8KIAA0090

Derlin-2

Derlin-1

UBXD2

TTC35

VIMP

POR

STT3Agp78

ARL6IP5

VCPSEL1L

TMEM111UBXD8

E-Syt1RPN2

XTP3-B

NGly1

OS-9

TXD16

C15orf24

VCPUBXD8

UBE4A

PSMB4

PSMB6PSMB7

PSMD7PSMD1

HMGCR

HMOX1AUP1

POROS-9

ERLIN2

UBE2J1

FDFT1

MYH9

SEL1L

ERLIN1

XTP3-BFAM8A1

MARCKS

UBE4A

PSMA7

PSMA2

PSMA3

PSMA5

PSMA6

PSMA1

PSMB1

PSMB2PSMB3

PSMB4PSMB6

PSMB7PSMD7PSMD1VCP

BRI3BPUBXD8

UBAC2OSBPL8

ERLIN2

UBE2G2

TMUB1

MYH9

MYH10

ERLIN1

PSMA4PSMD3

PSMD6 PSME4PSMB5 PSMD14

PSMC2

gp78

cytoplasmic/nuclear (GREY)peroxisomal (GREEN)mitochondrial (BLUE)

DIG and TX-100DIG onlyTX-100 only

ERAD or ER related (RED)

MTCH1

E-Syt1

CANX

PRKDC

CKAP5

SURF4

RTN4

MCM2

VDAC2KRT18

PRKDCRAB9A

C2orf30

ACTA1

NIPSNAP1

VDAC1

DPY30

EMD

GALK1

TXD16

CASK

RPL10ACTG2

CANXLIN7C

SEL1LUSH2A

ERFAD

GANAB

GGHRPN1

SERPINH1

TMEM85

KRT6A

FAM186B

TMEM85C14orf122

COX4NB

C15orf24

TTC35

TMEM111

C19orf63

TMEM93

TMEM32KIAA0090

UBQLN2CANX

KIAA0090

ENO1

COX4NB

C15orf24

TMEM111

TMEM32VDAC2

TMEM85

TTC35

CIB2

TMEM85

C15orf24

COX4NB

TMEM111

C19orf63

C14orf122

TMEM93

TMEM32KIAA0090

TTC35TMEM111

TMEM85

COX4NB

TTC35

C15orf24

C19orf63

C14orf122

TMEM32

KIAA0090UBQLN2

COX4NB

TTC35

TMEM111

C19orf63

C14orf122

TMEM93

KIAA0090UBQLN2

MMGT1C15orf24

HSP90AB2PTOMM40 SURF4

RAB35

TBC1D17CAND1

ACTA1TUBA8

TUBA4Q

VDAC1

HNRNPM

ANK2

TBC1D15

EMD

RAB7ARPL10

HM13TTC35

gp78ERLIN2 MLL2TMED9

TUBA1AGCN1L1

ERLIN1TMEM111

PEX19

UBXD8

TMEM43

CLPTM1

TMEM201

KIAA0090

LRRC59RPN2

UBAC2

SEC22BVAPA

COX2 COX4I1KRT18

EMD

ATP5B

SERPINH1

RAB9AARF3

ACTG1

ACTG2

TUBA1B1

UBXD8

KRT8

PFKP

TOMM40

VDAC3

OAT

ABCD3

ARL6IP1RAB7A

TOMM22VAPB

TOMM20VIMP

MCM2TMED9

SAE1

LARSPFKL

Derlin-1

OS-9ERLIN2

Derlin-2

ARF5

NPEPPS

GCN1L1

TUBA1AVCPCSE1L

SEL1LPEX11B

CAND1ERLIN1

TMEM43UBAC2

E-Syt1

ENO1

UQCRC2

IPO9

SEL1L3

PSMB5

TAGLN2

VDAC2

FLNA

Non-ER/Suspected Contaminants

HCIPs of interest

Kopito Supplemental Figure S4

Figure S4 Interaction maps for individual ERAD baits. Interactomes for each individual S-tagged ERAD component are shown, representing HCIPs detected in DIG only (dashed-green line), TX-100 only (dashed-purple line), and both DIG and TX-100 (solid-black line). ER-localised / ERAD-related HCIPs are indicated with red text and where appropriate, by their assigned colour from

Fig. 2. HCIPs of interest are depicted as dark brown circles with either red or black text, depending on their predicted localisation. HCIPs with alternative GO localisation (i.e. non-ER) and suspected non-specific contaminants are shown as: cytoplasmic/nuclear (grey circles, grey text), peroxisomal (beige circles, green text) and mitochondrial (light brown circles, light blue text).

© 2011 Macmillan Publishers Limited. All rights reserved.

S U P P L E M E N TA RY I N F O R M AT I O N

WWW.NATURE.COM/NATURECELLBIOLOGY 5

All Interactions-Pruned

LONP2GRP94CPVL

XTP3-BERFAD

OS-9

TXD16

MMGT1

COX4NB

TMEM85

TTC35

c15orf24KIAA0090

TMEM93

AUP1

NGly1 VCP

Ube2g2

hHR23B

UBE4A

EDEM1

TMEM111

c14orf122

CANX

PSMA1-7PSMB 1-7PSMC 2PSMD 1-3,6,7,14PSME4

BAIT INTERACTORERdj5

VCIP135

UBQLN1

BiP

hHR23A

PLAPCul2

REPORTED INTERACTOR

Npl4 Ufd1

SVIPATX3YOD1

Parkin

RFP2Usp13

LUMEN

CYTOPLASM

unidirectional (dashed)recipricol (solid)

MS analysisReported

Kopito Supplemental Figure S5

Both

Derlin-2Derlin-1

UbxD2

ERLIN2ERLIN1VIMP

SEL1L

gp78

UBAC2

c19orf63

Hrd1

UbxD8E-Syt2E-Syt1 SPP

RNF5

Derlin-3

Insig1

BAP31JAMP

FAM8A1

Ube2j1

Herp

Figure S5 Integration of previously reported interactions with the determined ERAD network. The previously reported interactions (Table S5) for ERAD baits and HCIPs were mapped onto the ERAD interactome from Fig. 1b with

the appropriate refinements from Fig. 2. Shown are reported interactions that were identified in our study (green), interactions only identified in our study (red), and additional reported interactions (blue).

© 2011 Macmillan Publishers Limited. All rights reserved.

S U P P L E M E N TA RY I N F O R M AT I O N

6 WWW.NATURE.COM/NATURECELLBIOLOGY

PS

MA

6P

SM

A2

PS

MA

4P

SM

A7

PS

MA

5P

SM

A1

PS

MA

3P

SM

B6

PS

MB

7P

SM

B3

PS

MB

2P

SM

B5

PS

MB

1P

SM

B4

PS

ME

4P

SM

D2

PS

MD

4P

SM

D14

PS

MD

8P

SM

D1

PS

MD

3P

SM

D12

PS

MD

11P

SM

D6

PS

MD

7P

SM

D13

PS

MC

2P

SM

C1

PS

MC

4P

SM

C6

PS

MC

3P

SM

C5

20S

19S

20S Core subunits

gp78 proteasome interactions (DIG) gp78 (DIG)

gp78 (TX)

Hrd1 (DIG)

Hrd1 (TX)

gp78 proteasome interactions (TX)

Hrd1 proteasome interactions (DIG)

Hrd1 proteasome interactions (TX)

Act

.

19S cap subunits

PS

MA

6P

SM

A2

PS

MA

4P

SM

A7

PS

MA

5P

SM

A1

PS

MA

3P

SM

B6

PS

MB

7P

SM

B3

PS

MB

2P

SM

B5

PS

MB

1P

SM

B4

PS

ME

4P

SM

D2

PS

MD

4P

SM

D14

PS

MD

8P

SM

D1

PS

MD

3P

SM

D12

PS

MD

11P

SM

D6

PS

MD

7P

SM

D13

PS

MC

2P

SM

C1

PS

MC

4P

SM

C6

PS

MC

3P

SM

C5

20S Core subunits Act

.

19S cap subunits

PS

MA

6P

SM

A2

PS

MA

4P

SM

A7

PS

MA

5P

SM

A1

PS

MA

3P

SM

B6

PS

MB

7P

SM

B3

PS

MB

2P

SM

B5

PS

MB

1P

SM

B4

PS

ME

4P

SM

D2

PS

MD

4P

SM

D14

PS

MD

8P

SM

D1

PS

MD

3P

SM

D12

PS

MD

11P

SM

D6

PS

MD

7P

SM

D13

PS

MC

2P

SM

C1

PS

MC

4P

SM

C6

PS

MC

3P

SM

C5

20S Core subunits Act

.

19S cap subunits

PS

MA

6P

SM

A2

PS

MA

4P

SM

A7

PS

MA

5P

SM

A1

PS

MA

3P

SM

B6

PS

MB

7P

SM

B3

PS

MB

2P

SM

B5

PS

MB

1P

SM

B4

PS

ME

4P

SM

D2

PS

MD

4P

SM

D14

PS

MD

8P

SM

D1

PS

MD

3P

SM

D12

PS

MD

11P

SM

D6

PS

MD

7P

SM

D13

PS

MC

2P

SM

C1

PS

MC

4P

SM

C6

PS

MC

3P

SM

C5

20S Core subunits Act

.

19S cap subunits

0.180.160.140.120.100.080.060.040.02

0

0.160.140.120.100.080.060.040.02

0

Spec

tral

abu

ndan

ce fa

ctor

Spec

tral

abu

ndan

ce fa

ctor

Spec

tral

abu

ndan

ce fa

ctor

Spec

tral

abu

ndan

ce fa

ctor

Ave

rage

spe

ctra

lab

unda

nce

fact

or

0.090.10

0.020.01

0.040.03

0.050.06

0.080.07

0

0.02

0.01

0.04

0.03

0.05

0.06

0

a

c

e

g

b

d

f

h

Kopito Supplemental Figure S6

0

0.02

0.04

0.06

0.08

0.10

0.12

20S

19S

Ave

rage

spe

ctra

lab

unda

nce

fact

or

0

0.02

0.04

0.06

0.08

0.10

0.01

0.03

0.05

0.07

0.09

0

0.02

0.04

0.06

0.08

0.10

0.01

0.03

0.05

0.07

0.09

20S

19S

Ave

rage

spe

ctra

lab

unda

nce

fact

or

20S

19S

Ave

rage

spe

ctra

lab

unda

nce

fact

or

0

0.02

0.03

0.01

Figure S6 Analysis of gp78 and Hrd1-associated proteasome compositions. (a, c, e, g) The calculated spectral abundance factor (Supplementary Table S6) for proteasome subunits co-precipitated with gp78 or Hrd1 in the indicated detergent is plotted. (b, d, f, h) The

average spectral abundance factor for 20S core subunits (PSMA and PSMB subunits) and 19S core subunits (PSMC and PSMD subunits) co-precipitated with gp78 or Hrd1 in the indicated detergent is plotted with the standard error.

© 2011 Macmillan Publishers Limited. All rights reserved.

S U P P L E M E N TA RY I N F O R M AT I O N

WWW.NATURE.COM/NATURECELLBIOLOGY 7

0.0

0.5

1.0

1.5

2.0

2.5

3.0

3.5

4.0

0 2 4 6 8 10 12

A B

C

D

E MG132

(Ub)n

IP: anti-GFP

- +

IB: α-Ub

GFP-GluR1hrs. 0 2 4 6 6

CTRL MG132

F

GluR1 calnexin overlay

MG132

Fold

cha

nge

in m

ean

F

post-drug addition (hrs.)

emetine

MG132

EndoHMG132

- + - +- - + +

IP: α-HAIB: α-HA

IB: α-GFP

CTRL

MG132+emetine

101 102 103 104100

GFP fluorescence

150

120

90

60

30

0

CTRL

coun

ts

ER lumen

cytosol

GFP GFP-GluR1

Kopito Supplemental Figure S7

Figure S7 Unassembled GluR1 is a bona fide ERAD substrate. (a) Illustration of GFP-GluR1 fusion protein topology. (b) Colocalisation of GFP-GluR1 (green) expressed in HEK293 cells with the ER marker calnexin (red). Nuclei are stained with DAPI (blue). (c) HA-GluR1 migration in the absence and presence of Endo H and MG132 (10 mM). EndoH sensitivity indicates that GluR1 is retained in the ER in its core glycosylated form and proteasome inhibition does not promote its maturation. (d) GFP-GluR1 pulse-chase assay. A t1/2 for GluR1 was

calculated to be ~2.5 hrs. and its degradation is mitigated by MG132. (e) GluR1 is ubiquitinated upon addition of MG132. GFP-GluR1 was immunoprecipitated with anti-GFP antibody, and Western blots were probed with anti-Ub (top) and anti-GFP antibodies (bottom). (f) Fluorescence histograms of the GFP-GluR1 stable cell line untreated (CTRL) and treated with MG132 (6 hrs, 10 mM) as measured by flow cytometry (left). Time course of GFP-GluR1 degradation after treatment with MG132 (10 mM) and/or emetine (10 mM) over 12 hrs (right).

© 2011 Macmillan Publishers Limited. All rights reserved.

S U P P L E M E N TA RY I N F O R M AT I O N

8 WWW.NATURE.COM/NATURECELLBIOLOGY

S-VIMP(22kDa)

shRNAF.F.

Luc

.

64 132

tubulin

C15orf24-S(23kDa)

shRNAF.F.

Luc

.

319

320

tubulin

UBXD2-S(71kDa)

shRNAF.F.

Luc

.

142

143

tubulin

S-SEL1L(100kDa)

shRNAF.F.

Luc

.

mNGly1(73kDa)

shRNAF.F.

Luc

.

24

tubulin

AUP1-S(45kDa)

shRNAF.F.

Luc

.

178

179

180

tubulin

S-OS9.2(~90kDa)

shRNAF.F.

Luc

.

5 67 88

tubulin

38 102

tubulin

TXD16-S(92kDa)

shRNAF.F.

Luc

.

217

tubulin

Derlin2-S(23kDa)

shRNAF.F.

Luc

.

148

tubulin

FAM8A1(50kDa)

shRNAF.F.

Luc

.

196

Hrd1-S

COX4NB-S(25kDa)

shRNAF.F.

Luc

.

327

329

tubulin

gp78-S(73kDa)

Ubn-gp78-S(>250kDa)

shRNAF.F.

Luc

.

37

tubulin

ERFAD-S(90kDa)

shRNAF.F.

Luc

.

161

tubulin

Hrd1-S(75kDa)

shRNA

Hrd1-S

HE

K

65 66 85 86 95Em

pty

3231

tubulin

S-TTC35(35kDa)

shRNAF.F.

Luc

.

313

316

tubulin

UBXD8-S(51kDa)

shRNA

UBXD8-S

HE

K

226

229

230

231

232

233

Em

pty

5148

tubulin(lysate)

S-UBE2G2(20kDa)

shRNAF.F.

Luc

.

36

tubulin

shRNA

S-JAMP(27kDa)

tubulin

HE

K

Em

pty

8374

myc-UBE2J1(39kDa)

shRNA

myc-UBE2J1

HE

K

Em

pty

170

171

173

tubulin

EDEM1-S(73kDa)

shRNAF.F.

Luc

.

72 73

tubulin

S-E-Syt1(120kDa)

shRNAF.F.

Luc

.

223

224

tubulin

UBAC2(35kDa)

shRNAF.F.

Luc

.

159

234

UBXD8-S(51kDa)

A

D H

L

P

T

W

U V

Q R

S

M

N O

I J K

E F G

B C

0

80 81P

SM

D2

SE

L1L

VIM

P

UB

XD

2

XTP

3-B

FAM

8A1

gp78

VC

P

KIA

A00

90

Hrd

1

Der

lin-3

HE

RP

SV

IP

Aha

1E

Rdj

5

LON

P2TMEM111 Derlin-1 Derlin-2

38 102

307

309

310

312

131

132

143 11 53 196 37 109

123

125

200 65 94 25 26 341 70 353 42 148

348 63 127

136

166

188

311

50

100

75

25

Nor

mal

ized

exp

ress

ion

leve

l

Kopito Supplemental Figure S8

Figure S8 Validation of shRNA knockdown by Western blotting and qRT-PCR. To determine the efficacy of shRNA constructs targeting the each gene, plasmids encoding the corresponding S-tagged or myc-tagged target protein were transiently co-expressed in HEK293 cells with a control shRNA construct or the targeting shRNA construct (indicated by the library reference number; Table S7a). SDS-solubilised cellular lysates (a-c) or S-protein agarose affinity-purified proteins from TX-100 lysates (d-v) were separated by SDS-PAGE and analysed by Western blotting with anti-S-peptide or anti-myc

antibodies and anti-tubulin antibodies as a loading control. (w) To determine the efficacy of shRNA constructs at depleting the target transcript, a control shRNA plasmid targeting firefly luciferase or shRNA plasmids targeting potential ERAD components (indicated by the library reference number; Table S7a) were transiently expressed in HEK293 cells. Transcript levels for the indicated components were determined by qRT-PCR, normalised to cells transfected with the control plasmid targeting firefly luciferase and to the percentage of transfection, and presented as the normalised expression level.

© 2011 Macmillan Publishers Limited. All rights reserved.

S U P P L E M E N TA RY I N F O R M AT I O N

WWW.NATURE.COM/NATURECELLBIOLOGY 9

TTR(D18G) Integrated degradation mapA

BGluR1 Integrated degradation map

SEL1L

GRP94CPVL

XTP3-BERFAD

OS-9

TXD16

gp78FAM8A1

UBXD8

UBAC2

AUP1

NGly1 VCP

UBE2G2

hHR23B

UBE4A

UBE2J1

EDEM1

ERLIN2

UBXD2E-Syt2E-Syt1 SPP

CANX

PSMA1-7PSMB 1-7PSMC 2PSMD 1-3,6,7,14PSME4

Hrd1

ERdj5

HERP

RNF5

VCIP135

UBQLN1

Derlin-3

Insig1

BiP

Derlin-1

hHR23A

PLAPCul2

Npl4 Ufd1

SVIPATX3

YOD1

BAP31JAMP

PARK2

VIMP

RFP2USP13

MMGT1

COX4NB

TMEM85

TTC35

C15orf24KIAA0090

TMEM93

TMEM111

C14orf122

C19orf63

LON2P

Derlin-2

ERLIN1

SEL1L

GRP94CPVL

XTP3-BERFAD

OS-9

TXD16

gp78FAM8A1

UBXD8

UBAC2

AUP1

NGly1 VCP

UBE2G2

hHR23B

UBE4A

UBE2J1

EDEM1

ERLIN2

UBXD2E-Syt2E-Syt1 SPP

CANX

PSMA1-7PSMB 1-7PSMC 2PSMD 1-3,6,7,14PSME4

Hrd1

BAIT INTERACTOR

ERdj5

HERP

RNF5

VCIP135

UBQLN1

Derlin-3

Insig1

BiP

Derlin-1

hHR23A

PLAPCul2

Npl4 Ufd1

SVIPATX3

YOD1

BAP31JAMP

PARK2

RFP2USP13

MMGT1

COX4NB

TMEM85

TTC35

C15orf24KIAA0090

TMEM93

TMEM111

C14orf122

C19orf63

LON2P

Derlin-2

ERLIN1

no effect

not determined

unidirectional (dashed)recipricol (solid)

MS analysisReportedBoth

VIMP

BAIT INTERACTOR no effect

not determined

unidirectional (dashed)recipricol (solid)

MS analysisReportedBoth

Kopito Supplemental Figure S9

Figure S9 Integrated substrate-specific degradation snapshots. The normalised fold change in mean GFP fluorescence of ERAD reporter lines in response to shRNA-mediated knockdown of ERAD components represented as a heat map (Fig. 3f) was directly mapped onto the

comprehensive ERAD interactome (Fig. S5) to construct an integrated snapshot of substrate-specific functional ERAD networks for two example substrates, TTR(D18G)-GFP (panel (a)) and GFP-GluR1 (panel (b)).

© 2011 Macmillan Publishers Limited. All rights reserved.

S U P P L E M E N TA RY I N F O R M AT I O N

10 WWW.NATURE.COM/NATURECELLBIOLOGY

GFPu Integrated degradation mapA

BCFTR(∆F508) Integrated degradation map

SEL1L

GRP94CPVL

XTP3-BERFAD

OS-9

TXD16

gp78FAM8A1

UBXD8

UBAC2

AUP1

NGly1 VCP

UBE2G2

hHR23B

UBE4A

UBE2J1

EDEM1

ERLIN2

UBXD2E-Syt2E-Syt1 SPP

CANX

PSMA1-7PSMB 1-7PSMC 2PSMD 1-3,6,7,14PSME4

Hrd1

ERdj5

HERP

RNF5

VCIP135

UBQLN1

Derlin-3

Insig1

BiP

Derlin-1

hHR23A

PLAPCul2

Npl4 Ufd1

SVIPATX3

YOD1

BAP31JAMP

PARK2

VIMP

RFP2USP13

MMGT1

COX4NB

TMEM85

TTC35

C15orf24KIAA0090

TMEM93

TMEM111

C14orf122

C19orf63

LON2P

Derlin-2

ERLIN1

SEL1L

GRP94CPVL

XTP3-BERFAD

OS-9

TXD16

gp78FAM8A1

UBXD8

UBAC2

AUP1

NGly1 VCP

UBE2G2

hHR23B

UBE4A

UBE2J1

EDEM1

ERLIN2

UBXD2E-Syt2E-Syt1 SPP

CANX

PSMA1-7PSMB 1-7PSMC 2PSMD 1-3,6,7,14PSME4

Hrd1

BAIT INTERACTOR

ERdj5

HERP

RNF5

VCIP135

UBQLN1

Derlin-3

Insig1

BiP

Derlin-1

hHR23A

PLAPCul2

Npl4 Ufd1

SVIPATX3

YOD1

BAP31JAMP

PARK2

RFP2USP13

MMGT1

COX4NB

TMEM85

TTC35

C15orf24KIAA0090

TMEM93

TMEM111

C14orf122

C19orf63

LON2P

Derlin-2

ERLIN1

no effect

not determined

unidirectional (dashed)recipricol (solid)

MS analysisReportedBoth

VIMP

BAIT INTERACTOR no effect

not determined

unidirectional (dashed)recipricol (solid)

MS analysisReportedBoth

Kopito Supplemental Figure S10

Figure S10 Integrated substrate-specific degradation snapshots for GFPu and CFTR(∆F508). Integrated snapshots of substrate-specific functional ERAD networks as described in Fig. S9 are shown for GFPu (a) and CFTR(∆F508) (b).

© 2011 Macmillan Publishers Limited. All rights reserved.

S U P P L E M E N TA RY I N F O R M AT I O N

WWW.NATURE.COM/NATURECELLBIOLOGY 11

A1AT(NHK) Integrated degradation mapA

BA1AT(NHK-QQQ) Integrated degradation map

SEL1L

GRP94CPVL

XTP3-BERFAD

OS-9

TXD16

gp78FAM8A1

UBXD8

UBAC2

AUP1

NGly1 VCP

UBE2G2

hHR23B

UBE4A

UBE2J1

EDEM1

ERLIN2

UBXD2E-Syt2E-Syt1 SPP

CANX

PSMA1-7PSMB 1-7PSMC 2PSMD 1-3,6,7,14PSME4

Hrd1

ERdj5

HERP

RNF5

VCIP135

UBQLN1

Derlin-3

Insig1

BiP

Derlin-1

hHR23A

PLAPCul2

Npl4 Ufd1

SVIPATX3

YOD1

BAP31JAMP

PARK2

VIMP

RFP2USP13

MMGT1

COX4NB

TMEM85

TTC35

C15orf24KIAA0090

TMEM93

TMEM111

C14orf122

C19orf63

LON2P

Derlin-2

ERLIN1

SEL1L

GRP94CPVL

XTP3-BERFAD

OS-9

TXD16

gp78FAM8A1

UBXD8

UBAC2

AUP1

NGly1 VCP

UBE2G2

hHR23B

UBE4A

UBE2J1

EDEM1

ERLIN2

UBXD2E-Syt2E-Syt1 SPP

CANX

PSMA1-7PSMB 1-7PSMC 2PSMD 1-3,6,7,14PSME4

Hrd1

BAIT INTERACTOR

ERdj5

HERP

RNF5

VCIP135

UBQLN1

Derlin-3

Insig1

BiP

Derlin-1

hHR23A

PLAPCul2

Npl4 Ufd1

SVIPATX3

YOD1

BAP31JAMP

PARK2

RFP2USP13

MMGT1

COX4NB

TMEM85

TTC35

C15orf24KIAA0090

TMEM93

TMEM111

C14orf122

C19orf63

LON2P

Derlin-2

ERLIN1

no effect

not determined

unidirectional (dashed)recipricol (solid)

MS analysisReportedBoth

VIMP

BAIT INTERACTOR no effect

not determined

unidirectional (dashed)recipricol (solid)

MS analysisReportedBoth

Kopito Supplemental Figure S11

Figure S11 Integrated substrate-specific degradation snapshots for A1AT(NHK) and A1AT(NHK-QQQ). Integrated snapshots of substrate-specific functional ERAD networks as described in Fig. S9 are shown for A1AT(NHK) (a) and A1AT(NHK) (b).

© 2011 Macmillan Publishers Limited. All rights reserved.

S U P P L E M E N TA RY I N F O R M AT I O N

12 WWW.NATURE.COM/NATURECELLBIOLOGY

250150100

75

50

37

2520

250150100

75

50

37

25

20

250150100

75

50

37

25

20

250150100

75

50

37

25

20

250150100

75

50

37

25

20

250150100

75

50

37

25

20

250150100

75

50

37

25

20

250150100

75

50

37

25

20

250150100

75

5037

2520

250150100

75

50

37

25

250150100

75

5037

2520

75

50

37

25

20

250150100

75

250150100

75

50

37

IB: SEL1L and Hrd1

IB: myc IB: SEL1L and S-peptide

IB: FAM8A1

IB: SEL1L

IB: S-peptide

Figure 3A

Figure 3HFigure 3G

Figure 3I

Figure 3B Figure 3C

IB: SEL1L and Hrd1

IB: SEL1L

IB: S-peptide

IB: Hrd1

IB: S-peptide

Figure 3D

Figure 3F

IB: Hrd1IB: SEL1LIB: S-peptide

IB: Hrd1

IB: SEL1L

IB: S-peptide

IB: myc

IB: S-peptide

IB: myc

250150100

75

50

37

25

20

250150100

75

50

37

25

250150100

75

50

37

25

250150100

75

50

37

25

20

250150100

75

50

37

25

250150100

75

50

37

25

20

250150100

75

50

37

25

20

Kopito Supplemental Figure S12

Figure S12 Full scans of immunoblots.

© 2011 Macmillan Publishers Limited. All rights reserved.

S U P P L E M E N TA RY I N F O R M AT I O N

WWW.NATURE.COM/NATURECELLBIOLOGY 13

250150100

75

50

37

2520

250150100

75

50

37

25

250150100

75

50

37

25

250150100

75

50

37

25

250150100

75

50

37

25

20

250150100

75

50

37

25

20

250150100

75

50

37

25

20

250150100

75

50

37

25

20

250150100

75

50

37

25

20

250150100

75

50

37

2520

Figure 3L Figure 3M Figure 3N

Figure 3P Figure 3P

Figure 3J

Figure 3O Figure 3O

IB: S-peptide

IB: UBAC2

IB: gp78

IB: Derlin-2

IB: S-peptide

IB: myc

IB: Hrd1

IB: UBAC2

IB: gp78 and UBXD8

IB: UBAC2

IB: gp78

IB: UBXD8

IB: S-peptide

IB: S-peptide

IB: Derlin-2IB: gp78

IB: gp78

IB: UBAC2

250150100

75

50

37

2520

250150100

75

50

37

2520

250150100

75

50

37

2520

250150100

75

50

37

25

20

250150100

75

50

37

25

20

250150100

75

50

37

25

20

250150100

75

50

37

25

20

250150100

75

50

37

25

20

Kopito Supplemental Figure S12 (cont.)

Figure S12 Full scans of immunoblots continued

© 2011 Macmillan Publishers Limited. All rights reserved.

S U P P L E M E N TA RY I N F O R M AT I O N

14 WWW.NATURE.COM/NATURECELLBIOLOGY

250150100

75

50

37

25

250150100

75

50

20

250150100

75

50

37

25

250150100

75

50

37

2520

250150100

75

50

37

2520

250150100

75

50

37

2520

250150100

75

50

250150100

75

50

37

25

20

250150100

75

50

37

25

20

Figure 6B Figure 6B

Figure 6D Figure 6D

Figure 7BFigure 6F

IB: FAM8A1

IB: p97/VCP and FAM8A1

IB: S-peptide

IB: UBXD8

IB: UBAC2

IB: Tubulin

IB: FAM8A1

IB: SEL1LIB: SEL1L

IB: SEL1L

IB: Tubulin

IB: Hrd1

250150100

75

50

250150100

75

50

37

25

250150100

75

50

37

25

Kopito Supplemental Figure S12 (cont.)

250150100

75

50

37

25

20

250150

100

75

50

37

250150

100

75

50

37

250150100

75

5037

25

20

250150100

75

5037

25

20

Figure S8AFigure S7F

IB: ubiquitin

IB: ubiquitin

Figure S8B

IB: S-peptideIB: S-peptide

IB: tubulin

250150100

75

50

37

25

20IB: tubulin

Figure S8E

Figure S8J Figure S8K Figure S8L

Figure S8F Figure S8G

Figure S8H Figure S8I

IB: S-peptide

IB: S-peptideIB: S-peptide

IB: S-peptideIB: S-peptide

IB: tubulinIB: tubulin

IB: tubulin

IB: tubulin

IB: tubulin

IB: tubulin

IB: S-peptide IB: S-peptideIB: S-peptide

IB: tubulin

IB: tubulin

250150100

75

50

37

25

250150100

75

50

37

25

250150100

75

50

37

25

250150100

75

5037

25

75

50

37

25

250150100

75

50

37

25

250150100

75

50

37

25

250150

10075

50

37

25

250150100

75

50

37

25

250150100

75

50

37

25

250150100

75

50

37

25

250150100

75

50

37

25

250150100

75

50

37

25

250150100

75

50

37

25

250150100

75

50

37

25

250150100

75

5037

25

Kopito Supplemental Figure S12 (cont.)

250150100

75

5037

25

20

250150100

75

50

37

25

20

15

250150100

75

50

37

25

20

15

Figure S8C Figure S8D

IB: myc

IB: tubulin

IB: S-peptide

IB: tubulin

250150100

75

5037

25

20

Figure S12 Full scans of immunoblots continued

© 2011 Macmillan Publishers Limited. All rights reserved.

S U P P L E M E N TA RY I N F O R M AT I O N

WWW.NATURE.COM/NATURECELLBIOLOGY 15

Figure S8E

Figure S8J Figure S8K Figure S8L

Figure S8F Figure S8G

Figure S8H Figure S8I

IB: S-peptide

IB: S-peptideIB: S-peptide

IB: S-peptideIB: S-peptide

IB: tubulinIB: tubulin

IB: tubulin

IB: tubulin

IB: tubulin

IB: tubulin

IB: S-peptide IB: S-peptideIB: S-peptide

IB: tubulin

IB: tubulin

250150100

75

50

37

25

250150100

75

50

37

25

250150100

75

50

37

25

250150100

75

5037

25

75

50

37

25

250150100

75

50

37

25

250150100

75

50

37

25

250150

10075

50

37

25

250150100

75

50

37

25

250150100

75

50

37

25

250150100

75

50

37

25

250150100

75

50

37

25

250150100

75

50

37

25

250150100

75

50

37

25

250150100

75

50

37

25

250150100

75

5037

25

Kopito Supplemental Figure S12 (cont.)

250150100

75

5037

25

20

250150100

75

50

37

25

20

15

250150100

75

50

37

25

20

15

Figure S8C Figure S8D

IB: myc

IB: tubulin

IB: S-peptide

IB: tubulin

250150100

75

5037

25

20

Figure S12 Full scans of immunoblots continued

© 2011 Macmillan Publishers Limited. All rights reserved.

S U P P L E M E N TA RY I N F O R M AT I O N

16 WWW.NATURE.COM/NATURECELLBIOLOGY

Figure S8O

Figure S8T Figure S8U Figure S8V

Figure S8P Figure S8Q

Figure S8R Figure S8S

IB: S-peptide IB: S-peptideIB: S-peptide

IB: S-peptideIB: S-peptide

IB: tubulin

IB: tubulin

IB: tubulin

25015010075

50

37

20

25

25015010075

50

37

25

25015010075

50

37

2520

25015010075

50

37

25

20

IB: tubulin

IB: S-peptide

IB: S-peptide

IB: S-peptide

IB: tubulin

IB: tubulin

IB: tubulin

25015010075

50

37

25

25015010075

50

37

25

25015010075

50

37

25

250150100

75

50

37

25

25015010075

50

37

25

25015010075

50

37

25

250150100

75

50

37

25

IB: tubulin

25015010075

50

37

25

15010075

50

37

25

15010075

50

37

25

25015010075

50

37

2520

25015010075

50

37

25

20

Kopito Supplemental Figure S12 (cont.)

Figure S8M Figure S8N

IB: tubulin IB: S-peptide

IB: FAM8A1IB: S-peptide

25015010075

5037

20

25

25015010075

5037

20

25

250150

100

75

50

37

25

100

75

50

37

25

Figure S12 Full scans of immunoblots continued

© 2011 Macmillan Publishers Limited. All rights reserved.

S U P P L E M E N TA RY I N F O R M AT I O N

WWW.NATURE.COM/NATURECELLBIOLOGY 17

Supplementary Tables

Table S1 Listing of primary and secondary bait proteins. Primary (top) and secondary (bottom) S-tagged proteins employed as baits in the proteomic analyses are shown with relevant information including alternative names, known functions, protein domains/repeats, cellular localisation (ER-L: ER luminal, ER-M: ER integral membrane, C: cytoplasmic), and identified yeast orthologs (Standard and Systematic name) are also shown.

Table S2 CompPASS-identified HCIPs and complete MASCOT list of interactors in DIG. (a) Listing of proteins interacting in DIG that displayed a WDN-score above the threshold score of 1 and were therefore designated as HCIPs. Also shown are additional statistical measures of interactor confidence, including Z-score, WS-score, and ZD-score. Self-identification of the bait protein identified is shown in red. (b) Listing of all MASCOT-identified protein interactors and their respective number of peptide scans found in affinity purifications of bait proteins from DIG-solubilised cell lysates.

Table S3 CompPASS-identified HCIPs and complete MASCOT list of interactors in TX-100. (a) Listing of proteins interacting in TX-100 that displayed a WDN-score above the threshold score of 1 and were therefore designated as HCIPs. Also shown are additional statistical measures of interactor confidence, including Z-score, WS-score, and ZD-score. The bait protein identified in each purification is shown in red. (b) Listing of all MASCOT-identified protein interactors and their respective number of peptide scans found in affinity purifications of bait proteins from TX-100-solubilised cell lysates.

Table S4 Analysis of proteomic interaction data. (a) Analysis and breakdown of proteomic data obtained in DIG. The total number of HCIPs identified by CompPASS is shown as well as the number of scans contributed by Bait self-identification, Trypsin, and Bead controls that were removed to yield the total number of HCIPs/ERAD interactions. The total number HCIPs is further broken down into likely false positives, primary / secondary baits obtained as prey, reported ERAD factors, and uncharacterised proteins of interest. The number of HCIPs is also shown separately for primary and secondary baits, as well as the overlap between these two data sets. (b) Same as in panel (a), but for data obtained in TX-100.

Table S5 Previously reported ERAD component interactions. Listing of reported pairwise interactions. Each interaction between a bait and prey was assigned an ‘interaction number’. The method by which the interaction was identified and the relevant reference (blue text) are indicated. Proteins used as baits in this study are shown (red text), and gray shading indicates proteins that were not employed as baits or identified as HCIPs in this study. The identification of the reported interaction in our DIG or TX-100 analyses or in the STRING database is indicated as an ‘O’ (identified) or as an ‘X’ (not identified). A comparative analysis of the reported interactions literature and those identified in our study is shown in the inset table.

Table S6 E3-proteasome subunit interactions. Listing of all MASCOT-identified proteasome subunits, their number of peptide scans, and their spectral abundance factor (SAF) from affinity purifications of the E3s Hrd1 and gp78 in DIG or TX-100 as indicated. Proteasome subunits determined by CompPASS to be HCIPs (WDN-score > 1) are highlighted in blue.

Table S7 Raw substrate fluorescence data from the primary ERAD reporter screen. (a). Complete listing of all shRNA target sequences, library reference numbers, and their effects on the fluorescence levels of the ERAD reporter cell lines tested in the initial round of screening, including GFPu, GFP-GluR1, TTR(D18G)-GFP, A1AT(NHK)-GFP, and GFP-CFTR(∆F508). (b) shRNA constructs employed in the second round of screening in the following cell lines: GFPu, GFP-GluR1, TTR(D18G)-GFP, A1AT(NHK)-GFP, A1AT(NHK-QQQ)-GFP, GFP-CFTR(∆F508), and GFP cell lines. These data are represented as a heat map in Fig. 4f.

Table S8 ERAD component data and links. Listing of ERAD components and identified HCIPs of interest along with relevant information, including gene and protein names, known function, localisation, UNIPROT ID, and known yeast orthologs.

Table S9 qRT-PCR primers. Listing of qRT-PCR primer sequences used to assess the affect of UPR induction on transcript levels in Fig. 5 and to assess shRNA knockdown of target transcript in Supplementary Fig. S8.

Table S10 ERAD cDNA sources. Listing of cDNA sources for expression of the bait proteins employed in this study.

© 2011 Macmillan Publishers Limited. All rights reserved.