PROTEIN FOLDING Thermal proximitycoaggregation for system ... · bility (fig. S4C) for proteins...

8
PROTEIN FOLDING Thermal proximity coaggregation for system-wide profiling of protein complex dynamics in cells Chris Soon Heng Tan, 1,2 * Ka Diam Go, 3 Xavier Bisteau, 1 Lingyun Dai, 3 Chern Han Yong, 4,5 Nayana Prabhu, 3 Mert Burak Ozturk, 1,6 Yan Ting Lim, 3 Lekshmy Sreekumar, 3 Johan Lengqvist, 7 Vinay Tergaonkar, 1,6,8 Philipp Kaldis, 1,6 Radoslaw M. Sobota, 1,2 Pär Nordlund 3,1,7 * Proteins differentially interact with each other across cellular states and conditions, but an efficient proteome-wide strategy to monitor them is lacking. We report the application of thermal proximity coaggregation (TPCA) for high-throughput intracellular monitoring of protein complex dynamics. Significant TPCA signatures observed among well-validated protein-protein interactions correlate positively with interaction stoichiometry and are statistically observable in more than 350 annotated human protein complexes. Using TPCA, we identified many complexes without detectable differential protein expression, including chromatin-associated complexes, modulated in S phase of the cell cycle. Comparison of six cell lines by TPCA revealed cell-specific interactions even in fundamental cellular processes. TPCA constitutes an approach for system-wide studies of protein complexes in nonengineered cells and tissues and might be used to identify protein complexes that are modulated in diseases. A living cell arises from a myriad of biomol- ecule interactions occurring in time and space among proteins, nucleic acids, metab- olites, and lipids. Central to this intricate biological network are protein complexes that mediate the biochemical processes and the structural organization of the cell. They assemble and dissociate dynamically according to cellular needs and are implicated in many different dis- eases (1, 2). Large-scale studies using specific cell lines (39), complemented by focused efforts, have contributed to a large protein-protein interaction network depicting the plausible cellular wiring and functional organization of the human pro- teome. However, the conservation of the assembled protein network and identified protein complexes across cell types, physiological states, and diseased conditions is unclear. Methods that permit effi- cient, system-wide, and hypothesis-free identifi- cation of differentiated protein complexes in nonengineered and diseased cells will expedite biological studies. Here, we explored the cellular thermal shift assay (CETSA) for the study of protein complex dynamics ( 10). By using protein mass spectrometry (MS) with multiplexed quantification for CETSA (MS-CETSA or thermal proteome profiling), melt- ing curves are generated for thousands of proteins (1113). In this work, we analyzed MS-CETSA data generated from samples without exogenous ligands. We validated thermal proximity coag- gregation (TPCA) as an approach for system- wide intracellular monitoring of protein complex dynamics. TPCA is based on the hypothesis that interact- ing proteins coaggregate upon heat denaturation, leading to similar solubility across different tem- peratures (Fig. 1A). We first investigated TPCA with the well-characterized Cdk2cyclin E1 com- plex, deriving the melting curves using immuno- blots for both proteins overexpressed in human embryonic kidney (HEK) 293T cells (fig. S1, A and B). Individually expressed cyclin E1V5 and Cdk2-HA (hemagglutinin) display distinct melt- ing curves, but they interact and are stabilized with similar melting curves when coexpressed (Fig. 1B and fig. S1A), consistent with the TPCA hypothesis. To evaluate the generality of TPCA, we ob- tained the melting curves for 7693 human pro- teins (table S1) collated from eight MS-CETSA experiments of the same K562 lysate (Fig. 1C). We assembled 111,776 protein-protein interac- tions annotated in BioGRID (14), InAct (15), and MINT (16) databases occurring among the 7693 proteins (table S2). Using Euclidean distance as an inverse measure of curve similarity, we ob- served that interacting protein pairs generally have higher curve similarity than all protein pairs (P < 2.2 × 10 -16 , one-tailed Mann-Whitney test, Fig. 1D). This similarity is more pronounced for interactions reported by multiple publica- tions (Fig. 1D and fig. S2). We also observed high curve similarity for interactions from two recent large-scale studies (5, 6) using yeast two- hybrid (Y2H) and affinity purification (AP)MS/MS, and for subunit pairs of complexes in the CORUM database (Fig. 1E) (17). Thus, it is unlikely that the observed TPCA signatures arose from ascertainment bias and the experimental methods used. We computed the average curve similarity among all subunit pairs of each protein complex and assessed TPCA signatures at the protein complex level. For the 558 nonredundant human complexes having at least three subunits with melting curves, 160 exhibited nonrandom TPCA signatures among subunits collectively (P < 0.05, table S3, Fig. 1F, and fig. S3). Next, we obtained MS-CETSA data from the K562 cell lysate that was depleted of lowmolecular weight (LMW) ligands by desalting (table S4). We observed de- creased average curve similarity for most protein complexes (~88%, fig. S4A and table S5), suggest- ing that LMW ligand depletion caused increased complex dissociation. However, we observed mul- tiple protein complexes with negligible changes in TPCA but with melting curves of all subunits shifted similarly, such as the PA700 proteasome subcomplex that could be due to adenosine 5´-triphosphate (ATP) depletion (Fig. 2A and fig. S5). Thus, whole-protein complexes can remain intact yet thermally destabilized by LMW ligand depletion. In comparison, MS-CETSA data from a new batch of K562 lysate shared good repro- ducibility with the previous lysate (Pearsons R = 0.88, fig. S3B). We observed a higher reproduci- bility (fig. S4C) for proteins with at least three quantified peptides in each data set, and thus combined existing and new data sets for subse- quent analysis (table S6). Six MS-CETSA experiments were performed on intact K562 cells (table S7). We observed melt- ing curves of all protein pairs to be statistically more similar in the lysate data (Fig. 2B), whereas those for interacting protein pairs are statisti- cally more similar in intact cell data (Fig. 2B). We also observed that protein complexes have better average curve similarity among subunits in intact cell data than in lysate data (figs. S6, A to D, and S7). Overall, intact cell data reveal more protein complexes with nonrandom TPCA sig- natures than the lysate data (Fig. 2C; fig. S6, E and F; and table S8). TPCA-significant protein complexes in stress response, cell cycle, and DNA processing path- ways are statistically enriched in intact cell data over lysate data (fig. S6G). Many DNA-chromatinassociated complexes and membrane-associated complexesas exemplified by the Nup 107-160 nu- clear pore subcomplex and the origin recognition complex, respectively (Fig. 2D)exhibit signif- icant TPCA signatures only in intact cells, pre- sumably due to disruption of DNA-chromatin and native membranes in cell lysate. Our analysis suggests that the replication factor complex C (RFC) remains assembled in both intact cell and RESEARCH Tan et al., Science 359, 11701177 (2018) 9 March 2018 1 of 7 1 Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore. 2 Institute of Medical Biology (IMB), A*STAR (Agency for Science, Technology and Research), Singapore. 3 School of Biological Sciences, Nanyang Technological University, Singapore. 4 Program in Cancer and Stem Cell Biology, DukeNational University of Singapore (NUS) Medical School, Singapore. 5 Centre for Computational Biology, DukeNUS Medical School, Singapore. 6 Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore. 7 Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska Institutet, Stockholm, Sweden. 8 Centre for Cancer Biology (University of South Australia and SA Pathology), Adelaide, Australia. *Corresponding author. Email: [email protected] (C.S.H.T.); [email protected] (P.N.) on March 19, 2020 http://science.sciencemag.org/ Downloaded from

Transcript of PROTEIN FOLDING Thermal proximitycoaggregation for system ... · bility (fig. S4C) for proteins...

Page 1: PROTEIN FOLDING Thermal proximitycoaggregation for system ... · bility (fig. S4C) for proteins with at least three quantified peptides in each data set, and thus combined existing

PROTEIN FOLDING

Thermal proximity coaggregation forsystem-wide profiling of proteincomplex dynamics in cellsChris Soon Heng Tan,1,2* Ka Diam Go,3 Xavier Bisteau,1 Lingyun Dai,3

Chern Han Yong,4,5 Nayana Prabhu,3 Mert Burak Ozturk,1,6 Yan Ting Lim,3

Lekshmy Sreekumar,3 Johan Lengqvist,7 Vinay Tergaonkar,1,6,8 Philipp Kaldis,1,6

Radoslaw M. Sobota,1,2 Pär Nordlund3,1,7*

Proteins differentially interact with each other across cellular states and conditions, but anefficient proteome-wide strategy to monitor them is lacking. We report the application ofthermal proximity coaggregation (TPCA) for high-throughput intracellular monitoring ofprotein complex dynamics. Significant TPCA signatures observed among well-validatedprotein-protein interactions correlate positively with interaction stoichiometry and arestatistically observable in more than 350 annotated human protein complexes. UsingTPCA, we identified many complexes without detectable differential protein expression,including chromatin-associated complexes, modulated in S phase of the cell cycle.Comparison of six cell lines by TPCA revealed cell-specific interactions even infundamental cellular processes. TPCA constitutes an approach for system-wide studiesof protein complexes in nonengineered cells and tissues and might be used to identifyprotein complexes that are modulated in diseases.

Aliving cell arises from amyriad of biomol-ecule interactions occurring in time andspace amongproteins, nucleic acids,metab-olites, and lipids. Central to this intricatebiological network are protein complexes

that mediate the biochemical processes and thestructural organization of the cell. They assembleand dissociate dynamically according to cellularneeds and are implicated in many different dis-eases (1, 2).Large-scale studies using specific cell lines

(3–9), complemented by focused efforts, havecontributed to a large protein-protein interactionnetwork depicting the plausible cellular wiringand functional organization of the human pro-teome.However, the conservation of the assembledprotein network and identified protein complexesacross cell types, physiological states, and diseasedconditions is unclear. Methods that permit effi-cient, system-wide, and hypothesis-free identifi-cation of differentiated protein complexes innonengineered and diseased cells will expeditebiological studies.

Here, we explored the cellular thermal shiftassay (CETSA) for the study of protein complexdynamics (10). By using proteinmass spectrometry(MS) with multiplexed quantification for CETSA(MS-CETSA or thermal proteome profiling), melt-ing curves are generated for thousands of proteins(11–13). In this work, we analyzed MS-CETSA datagenerated from samples without exogenousligands. We validated thermal proximity coag-gregation (TPCA) as an approach for system-wide intracellularmonitoring of protein complexdynamics.TPCA is based on the hypothesis that interact-

ing proteins coaggregate upon heat denaturation,leading to similar solubility across different tem-peratures (Fig. 1A). We first investigated TPCAwith the well-characterized Cdk2–cyclin E1 com-plex, deriving themelting curves using immuno-blots for both proteins overexpressed in humanembryonic kidney (HEK) 293T cells (fig. S1, Aand B). Individually expressed cyclin E1–V5 andCdk2-HA (hemagglutinin) display distinct melt-ing curves, but they interact and are stabilizedwith similar melting curves when coexpressed(Fig. 1B and fig. S1A), consistent with the TPCAhypothesis.To evaluate the generality of TPCA, we ob-

tained the melting curves for 7693 human pro-teins (table S1) collated from eight MS-CETSAexperiments of the same K562 lysate (Fig. 1C).We assembled 111,776 protein-protein interac-tions annotated in BioGRID (14), InAct (15), andMINT (16) databases occurring among the 7693proteins (table S2). Using Euclidean distance asan inverse measure of curve similarity, we ob-served that interacting protein pairs generallyhave higher curve similarity than all proteinpairs (P < 2.2 × 10−16, one-tailedMann-Whitney

test, Fig. 1D). This similarity ismore pronouncedfor interactions reported by multiple publica-tions (Fig. 1D and fig. S2). We also observedhigh curve similarity for interactions from tworecent large-scale studies (5, 6) using yeast two-hybrid (Y2H) and affinity purification (AP)–MS/MS, and for subunit pairs of complexes inthe CORUM database (Fig. 1E) (17). Thus, it isunlikely that the observed TPCA signatures arosefrom ascertainment bias and the experimentalmethods used.We computed the average curve similarity

among all subunit pairs of each protein complexand assessed TPCA signatures at the proteincomplex level. For the 558 nonredundant humancomplexes having at least three subunits withmelting curves, 160 exhibited nonrandom TPCAsignatures among subunits collectively (P < 0.05,table S3, Fig. 1F, and fig. S3). Next, we obtainedMS-CETSA data from the K562 cell lysate thatwas depleted of low–molecular weight (LMW)ligands by desalting (table S4). We observed de-creased average curve similarity for most proteincomplexes (~88%, fig. S4A and table S5), suggest-ing that LMW ligand depletion caused increasedcomplex dissociation. However, we observedmul-tiple protein complexes with negligible changesin TPCA but with melting curves of all subunitsshifted similarly, such as the PA700 proteasomesubcomplex that could be due to adenosine5´-triphosphate (ATP) depletion (Fig. 2A and fig.S5). Thus, whole-protein complexes can remainintact yet thermally destabilized by LMW liganddepletion. In comparison, MS-CETSA data froma new batch of K562 lysate shared good repro-ducibility with the previous lysate (Pearson’s R =0.88, fig. S3B). We observed a higher reproduci-bility (fig. S4C) for proteins with at least threequantified peptides in each data set, and thuscombined existing and new data sets for subse-quent analysis (table S6).Six MS-CETSA experiments were performed

on intact K562 cells (table S7). We observed melt-ing curves of all protein pairs to be statisticallymore similar in the lysate data (Fig. 2B), whereasthose for interacting protein pairs are statisti-cally more similar in intact cell data (Fig. 2B).We also observed that protein complexes havebetter average curve similarity among subunitsin intact cell data than in lysate data (figs. S6, Ato D, and S7). Overall, intact cell data reveal moreprotein complexes with nonrandom TPCA sig-natures than the lysate data (Fig. 2C; fig. S6, Eand F; and table S8).TPCA-significant protein complexes in stress

response, cell cycle, and DNA processing path-ways are statistically enriched in intact cell dataover lysate data (fig. S6G).ManyDNA-chromatin–associated complexes and membrane-associatedcomplexes—as exemplified by the Nup 107-160 nu-clear pore subcomplex and the origin recognitioncomplex, respectively (Fig. 2D)—exhibit signif-icant TPCA signatures only in intact cells, pre-sumably due to disruption of DNA-chromatinandnativemembranes in cell lysate. Our analysissuggests that the replication factor complex C(RFC) remains assembled in both intact cell and

RESEARCH

Tan et al., Science 359, 1170–1177 (2018) 9 March 2018 1 of 7

1Institute of Molecular and Cell Biology (IMCB), A*STAR(Agency for Science, Technology and Research), Singapore.2Institute of Medical Biology (IMB), A*STAR (Agency forScience, Technology and Research), Singapore. 3School ofBiological Sciences, Nanyang Technological University,Singapore. 4Program in Cancer and Stem Cell Biology,Duke–National University of Singapore (NUS) Medical School,Singapore. 5Centre for Computational Biology, Duke–NUSMedical School, Singapore. 6Department of Biochemistry,Yong Loo Lin School of Medicine, National University ofSingapore, Singapore. 7Department of Oncology-Pathology,Cancer Center Karolinska, Karolinska Institutet, Stockholm,Sweden. 8Centre for Cancer Biology (University of SouthAustralia and SA Pathology), Adelaide, Australia.*Corresponding author. Email: [email protected](C.S.H.T.); [email protected] (P.N.)

on March 19, 2020

http://science.sciencem

ag.org/D

ownloaded from

Page 2: PROTEIN FOLDING Thermal proximitycoaggregation for system ... · bility (fig. S4C) for proteins with at least three quantified peptides in each data set, and thus combined existing

lysate (Fig. 2E, top) but that the whole complexis thermally destabilized in lysate (Fig. 2E, topleft), presumably due to the absence of DNA-chromatin.

We observed many instances in which sub-complexes exhibit distinct TPCA signaturesbetween intact cell and lysate data sets. For ex-ample, the 40S and 60S ribosomal subcom-

plexes each have subunits with similar curvesthat shifted closer to the other subcomplex inintact cell data (Fig. 2F), presumably becausemore fully assembled ribosomes are translating

Tan et al., Science 359, 1170–1177 (2018) 9 March 2018 2 of 7

100%

100%

50% 50%50% 50%

100%

100%

soluble soluble

37°CTemperature

Fra

ctio

n o

f S

olu

ble

Pro

tein

1

insolubleaggregate

0

insolubleaggregate

100%

50%50%

100%

soluble

insolubleco-aggregate

37°CTemperature

Fra

ctio

n o

f S

olu

ble

Pro

tein

1

0

insolubleco-aggregate

CORUM

AP−MS/MS

Y2H

Distribution of Euclidean Distance Among Protein Pairs

40 45 50 55

0.0

0.2

0.4

0.6

0.8

1.0

60

Fra

ctio

n o

f S

olu

ble

Pro

tein

Cyclin E1-V5 CDK2-HA

Cyclin E1-V5 (+ CDK2-HA) CDK2-HA (+ Cyclin E1-V5)

Temperature (°C)

higher pairwise curve similarity

All Pairs

P−value < 0.0001

0.00

0.25

0.50

0.75

1.00

1.25

40 45 50 55 60 65

ESCRT−II complex

P−value < 0.0001

0.00

0.25

0.50

0.75

1.00

1.25

40 45 50 55 60 65

Temperature (°C) Temperature (°C) Temperature (°C) Temperature (°C)

Sol

uble

Fra

ctio

n

Anaphase−promoting complex

P−value < 0.0001

0.00

0.25

0.50

0.75

1.00

1.25

40 45 50 55 60 65

Conserved oligomeric Golgi (COG) complex

P−value < 0.0001

0.00

0.25

0.50

0.75

1.00

1.25

40 45 50 55 60 65

Multisynthetase complex

P−value < 0.0001

0.00

0.25

0.50

0.75

1.00

1.25

40 45 50 55 60 65

Sol

uble

Fra

ctio

n

20S proteasome

P−value < 0.0001

0.00

0.25

0.50

0.75

1.00

1.25

40 45 50 55 60 65

BAF complex

P−value < 0.0001

0.00

0.25

0.50

0.75

1.00

1.25

40 45 50 55 60 65

Arp2/3 protein complex

P−value < 0.0001

0.00

0.25

0.50

0.75

1.00

1.25

40 45 50 55 60 65

BLOC−1 (biogenesis of lysosome− related organelles complex 1)

0.0 0.2 0.4 0.6 0.8 1.0 1.2 1.4 1.6 1.8

0.0 0.2 0.4 0.6 0.8 1.0 1.2 1.4 1.6 1.8

inte

nsi

tyin

tensi

ty

Quantitative Proteomics

(TMT labelling)

Fra

cti

on

of

So

lub

le P

rote

in

1

0

Protein Breporter ions

Protein Areporter ions

m/z

m/z

All Pairs

0.0 0.2 0.4 0.6 0.8 1.0 1.2 1.4 1.6 1.8

Inte

ract

ing

P

rote

in P

airs

Distribution of Euclidean Distance Among Protein Pairs

higher pairwise curve similarity

0.0 0.2 0.4 0.6 0.8 1.0 1.2 1.4 1.6 1.8

CellLysate

IntactCells cell lysis

Fig. 1. Interacting protein pairs exhibit strong TPCA signature. (A)Principle of TPCA for monitoring protein-protein interactions: Coaggregationand precipitation among interacting proteins result in similar protein solubilityacross different denaturing temperatures. (B) Cointeracting cyclin E1 andCdk2 exhibit similar melting curves: HEK 293Tcells were transfected withplasmids encodingV5-tagged cyclin E1 and/orHA-taggedCdk2, followedby theintact cell CETSA experiment. Data are from immunoblots of three biologicalreplicates (mean ± SD) and are fitted with a three-parameter log-logisticfunction. Representative immunoblots are shown in fig. S1B. (C) Schematic

overview of MS-CETSA experiment. m/z, mass/charge ratio; TMT, tandemmass tags. (D) Distribution of melting curve similarities between knowninteracting protein pairs according to number of reporting publications: PMID,PubMed identifier. Red dots indicate the median of all protein pairs of eachrespective protein subset. (E) Distribution of melting curve similarities ofknown interacting protein pairs reported in Rolland et al. (6), Huttlin et al. (5),and CORUM. (F) Protein melting curves of selected protein complexes: redlines, complex subunits. For the leftmost plot, the solubility curves of ~4000other proteins are plotted in gray.

RESEARCH | REPORTon M

arch 19, 2020

http://science.sciencemag.org/

Dow

nloaded from

Page 3: PROTEIN FOLDING Thermal proximitycoaggregation for system ... · bility (fig. S4C) for proteins with at least three quantified peptides in each data set, and thus combined existing

mRNA in cells than in lysate. Another example istheNDC80 kinetochore complexwith two distinctsubunit groups based on melting curves that aremore similar to each other in intact cell data thanin lysate data (Fig. 2E, bottom). The two distinct

subunit groups correspond to the two stableheterodimers that are associated with each otherthrough a short tetramerization protein region(Fig. 2G) (18, 19). TPCA analysis suggests that thetwo subcomplexes had dissociated in lysate.

These observations suggest that TPCA can revealfundamental differences in protein complex or-ganization and their functional states.The stoichiometry of interaction and abundance

between subunits in a complex can potentially

Tan et al., Science 359, 1170–1177 (2018) 9 March 2018 3 of 7

0.0 0.2 0.4 0.6 0.8 1.0 1.2 1.4 1.6 1.8

0.0 0.2 0.4 0.6 0.8 1.0 1.2 1.4 1.6 1.8

LYSATE CELL

0.01 0.02 0.03 0.04 0.05

50

100

150

200

0<10-4

P-value Threshold

Nu

mb

er o

f P

rote

in C

om

ple

xm

eeti

ng

P-v

alu

e T

hre

sho

ld

LYSATE

CELL

All Pairs

Pro

tein

-Pro

tein

Inte

ract

ion

s

Nup 107-160 Subcomplex

LYSATE CELL

Origin Recognition Complex LYSATE CELL

40S Ribosomal Subunit / 60S Ribosomal Subunit

LYSATE CELL

40 45 50 55 60 65

Temperature (°C)40 45 50 55 60 65

0.00

0.25

0.50

0.75

1.00

1.25

So

lubl

e F

ract

ion

NDC80NUF2SPC24SPC25

0.00

0.25

0.50

0.75

1.00

1.25

So

lubl

e F

ract

ion

NDC80 Kinetochore

NDC80NUF2SPC24SPC25

LYSATE CELL

Ndc80

Nuf2

Spc24

Spc25

tetramerizationdomain

40 45 50 55 60 65

PA700 complex

0.00

0.25

0.50

0.75

1.00

1.25

So

lubl

e F

ract

ion

40 45 50 55 60 65

Temperature (°C)

Median Melting CurvesOriginal LysateDesalted Lysate

Original LysateDesalted Lysate

Distribution of Euclidean Distance Among Protein Pairs

0.00

0.25

0.50

0.75

1.00

1.25

So

lubl

e F

ract

ion

0.00

0.25

0.50

0.75

1.00

1.25

40 45 50 55 60 6540 45 50 55 60 65

Temperature (°C)

40 45 50 55 60 65 40 45 50 55 60 65

Temperature (°C)

BRD4-RFC Complex

0.00

0.25

0.50

0.75

1.00

1.25

BRD4RFC1RFC2RFC3RFC4RFC5

BRD4RFC1RFC2RFC3RFC4RFC5

LYSATE CELL

Fig. 2. TPCA signature is stronger in data from intact cells. (A) Effectof desalting cell lysate on subunit melting curves of PA700 proteasomesubcomplex. (B) Distribution of melting curve similarities among knowninteracting protein pairs computed from cell lysate and intact cell data,respectively. Red dots indicate the median of all protein pairs of each

respective protein subset. (C) Number of CORUM protein complexes withnonrandom TPCA behavior at different statistical thresholds. (D) Exampleof DNA-chromatin– and membrane-associated complexes. (E and F) Examplesof subcomplexes that differ in cell and in lysate. (G) Schematics ofstructure for NDC80 kinetochore.

RESEARCH | REPORTon M

arch 19, 2020

http://science.sciencemag.org/

Dow

nloaded from

Page 4: PROTEIN FOLDING Thermal proximitycoaggregation for system ... · bility (fig. S4C) for proteins with at least three quantified peptides in each data set, and thus combined existing

influence its TPCA signature. We analyzed suchdata for the interacting protein pairs reportedwith highest confidence in Hein et al. (9) andobserved positive correlation of the TPCA signa-ture with interaction stoichiometry (Spearman’sR = –0.22, P < 0.001, Fig. 3A and fig. S8A) andabundance stoichiometry (Spearman’s R = –0.21,P < 0.001, Fig. 3B and fig. S8B). The protein pairswith both interaction stoichiometry and abun-dance stoichiometry greater than 80% exhibitmuch stronger TPCA signatures than those meet-ing the two criteria separately (Fig. 3C). In com-parison, proteinspaired solely by similar abundancedo not exhibit strong TPCA signatures [fig. S9, Ato C, and table S9 (20)]. Accordingly, multiplyinginteraction stoichiometry with abundance stoi-chiometry results in values that correlate betterwith TPCA signature (Spearman’s R = –0.27, P <0.001, Fig. 3D and fig. S8C). Thus, interactionand abundance stoichiometry between interact-ing proteins are intrinsically captured by TPCA,albeit semiquantitatively.Under the core-attachment model, subsets of

proteins form the stable core of protein com-plexes that are differentially or temporally boundby other proteins (21, 22). We investigated thismodel with TPCA, comparingmelting curves ofprotein pairs from the complex core to curvesfor pairs between the core and the attachmentsubunits (Fig. 3E).We identified the core-core pairsin each complex as those found in two or moreother CORUM complexes and core-attachmentpairs as those found in that complex only. Formost of the complexes, the TPCA signature ofcore-core pairs is either similar to ormuch strongerthan that of the core-attachment pairs (Fig. 3F).This is in accordance with the core-attachmentmodel in which core-attachment interactionsgenerally occur at lower stoichiometry. Thus,TPCA signature potentially encapsulates the tem-poral and core-attachment organization of manycomplexes.Next, we explored TPCA for system-wide mon-

itoring of protein complex dynamics between cellstates. We obtained intact cell MS-CETSA datafromK562 cells arrested in the S phase of the cellcycle using methotrexate (fig. S10) and from di-methyl sulfoxide (DMSO)–treated unsynchron-ized cells (tables S10 and S11). We assessedstatistical significance for enhanced TPCA (i.e.,higher average curve similarity) observed for anyCORUM complex (see materials and methods).We identified 18 protein complexes, some con-taining overlapping subunits, that have statis-tically enhanced TPCA signature across bothbiological replicates in S phase–synchronizedcells (tables S12 to S14).All but three identified modulated protein

complexes had previously been implicated in theS phase. They include the CAF-1 complex (Fig.3G), which forms and localizes to the replicationfork during S phase (23); the TREX-THO com-plex (Fig. 3G, bottom right) required for repli-cation fork progression (24, 25); and the TRAPcomplex required for S phase progression (26).We also identified many chromatin-nucleosomeremodeling and associated complexes such as

BAF (Fig. 3G, top right), LARC, and BRG1-SIN3A(27), consistent with the expected remodeling ofchromatin during the S phase. Histone mRNAswereup-regulatedprior to Sphasebutwere rapidlydegraded with halted DNA replication (28). Weidentified three modulated protein complexes—themRNA decay complex, the exosome, and theintegrator complex—that are involved in his-tone mRNA degradation. Deletion of RRP6, a sub-unit of exosome, was reported to increase histonemRNA HTB1 during S phase (29), thus high-lighting the role of this complex during Sphase. Integrator complex was implicated inthe processing of replication-dependent his-tone mRNAs (30).Twoof the three identified complexesnot known

to be implicated in S phase are the MDC1-MRN-ATM-FANCD2 and the DNA ligase III–XRCC–PNK–polymerase III complexes. However, theyare involved in the DNA damage response inaccordance with the known DNA-damaging ef-fect of methotrexate (31). The third complex notimplicated in S phase is the tumor necrosis factor–a (TNF-a)–nuclear factor kB (NF-kB) signalingcomplex (Fig. 3G).We validated thatmethotrexateincreases the assembly of the associated IkBa-p65-p50 protein complex by suppressing activationof the NF-kB signaling pathway, in both the pres-ence and absence of TNF, through inhibitingphosphorylation of the IkB kinase a/b (IKKa/b)complex, IkBa, and p65 in K562 cells (Fig. 3, Hand I).We subsequently combined data frombothreplicates—the better data coverage and precision(table S15) allowed us to uncover more complexesthat are differentially modulated between S phaseand unsynchronized cells (table S16 and fig. S11).The enhanced TPCA signature of identified

complexes largely arose from curve convergenceofmost subunits (Fig. 4A). Nevertheless, this couldarise from synchronized changes in protein expres-sion. Thus, we quantified relative protein abundancebetween the DMSO- and methotrexate-treatedcells usingMS. We observed high reproducibilityin relative abundance (Pearson’sR = 0.87, fig. S12and table S17) across biological replicates andidentified only five subunits (P < 0.05, or nine atP < 0.1) with differential protein expression (Fig.4A and table S18) that are mostly parts of themediator and integrator megacomplexes. Thus,most subunits exhibit enhanced TPCA signaturewith each other without differential protein ex-pression (Fig. 4A).Next, we analyzed TPCA signature acrossmulti-

ple cell lines. We generated MS-CETSA data fromHEK 293T, A375, HCT116,MCF7, andHL60 intactcells. Combining data from two biological repli-cates, each with two technical MS runs (averagePearson’sR = 0.92 between biological replicates,fig. S13), we obtained melting curves for ~7600proteins on average for each cell line (fig. S14Aand tables S19 to S23). On average, 33.8% of thequalified CORUMcomplexes exhibit a nonrandomTPCA signature (P < 0.05) in each cell line (figs.S14B and S15 to S20), with ~70%overlap betweencell lines. We also obtained data frommouse liverand observed ~37.7% of qualified protein com-plexes (three ormore subunitswithmelting curves)

with nonrandom TPCA signatures (tables S24 toS26 and figs. S21 and S22). Thus, TPCA is ob-servable across multiple cell lines and from tissuesamples. Many protein complexes exhibit strongTPCA behavior across the six cell lines (fig. S23),but we also found many complexes with highquality but distinct curves across cell lines (figs.S24 and S25). This suggests plausible variationin protein complex stoichiometry and composi-tion among cell lines, even for fundamental andabundant protein complexes, that could arisefrom changes in interaction stoichiometry and/or protein abundance.Subsequently, we generatedweightednetworks

of reported interactions among proteins iden-tified in all the six cell lines with TPCA-derivedz-scores (table S26). A basal network averagingthe z-scores, after removing the highest andthe lowest z-scores for each interaction, is alsoconstructed. Comparing the basal network withcell-specific TPCA-weighted networks facilitatesidentifying potentially differentiated interac-tions, pathways, and functional modules (tableS27). Focusing on HCT116, we found that theRAS-RAF-MEK-ERKpathwaycontainsmany inter-actionswith highly differentiated TPCA signatures(Fig. 4B). Retrospectively,we observedmarked sim-ilarity in melting curves of BRAF and RAF1 pro-teins in HCT116 cells compared to other cell lines(Fig. 4C), consistent with expected dimerizationof BRAF and RAF1 driven by active KRASG13D inHCT116. TPCA analysis also suggests that BRAF-CRAF interaction with MEK is up-regulated inHCT116 cells but is down-regulated in A375 cells,which express BRAFV600E (Fig. 4B). This is in linewith the recent finding that the interaction is up-regulated in cells expressing mutant KRAS andwild-type BRAF but suppressed in cells withmutant BRAF (32). Thus, TPCA could potentiallycapture cell-specific interactions and pathways.Lastly, we validated that known and poten-

tially previously unknown protein complexes canbe identified from the existing human interac-tome map using graph or network clusteringalgorithms (22, 33–36) with TPCA-based scoringof interactions (Fig. 4D, figs. S26 and S27, andsupplementary text). The best performance wasobtained with the COACH algorithm, which in-corporates the core-attachment model of proteincomplexes (22). Using this algorithm and TPCA-based scoring of interactions gives a performancecomparable to that of published interaction re-liability scores that incorporate publication countand functional similarity (37). We also observedthat TPCA profiling in its current format carriespredictive power for protein-protein interactions,with the area under the curve ranging from 0.62to 0.79 depending on the interaction data sets(Fig. 4, E and F), which collectively suggests thatTPCA profiles could also serve to discover newinteractions and protein complexes in combina-tion with other approaches.TPCA enables the intracellular study of the

dynamics of multiple protein complexes simulta-neously in intact nonengineered cells and tissues.About one-third of the qualified CORUM com-plexes in each cell line exhibit nonrandom TPCA

Tan et al., Science 359, 1170–1177 (2018) 9 March 2018 4 of 7

RESEARCH | REPORTon M

arch 19, 2020

http://science.sciencemag.org/

Dow

nloaded from

Page 5: PROTEIN FOLDING Thermal proximitycoaggregation for system ... · bility (fig. S4C) for proteins with at least three quantified peptides in each data set, and thus combined existing

Tan et al., Science 359, 1170–1177 (2018) 9 March 2018 5 of 7

Complex A Complex B

Core Complex

Non-uniqueCore-Core

Unique Core-Attachment

IKBα

p65

C T M T+M

p50

Hsp90

Input

C T M T+M C T M T+M C T M T+M

IP: p65 IP: p50 IP: IgG

IB: IKBα

IB: p50

IB: p65

IB: IKBα

IB: p65

CHAF1ACHAF1BRBBP4

0.00

0.25

0.50

0.75

1.00

1.25

So

lubl

e F

ract

ion

40 45 50 55 60 65 40 45 50 55 60 65Temperature (°C)

CHAF1ACHAF1BRBBP4

Chromatin Assembly Complex (CAF-1)

ACTL6AARID1ASMARCA2SMARCC1SMARCC2SMARCD1SMARCE1

40 45 50 55 60 65 40 45 50 55 60 65Temperature (°C)

BAF Complex

ACTL6AARID1ASMARCA2SMARCC1SMARCC2SMARCD1SMARCE1

CHUKIKBKGKPNA3NFKB1NFKB2NFKBIBNFKBIERELRELA

0.00

0.25

0.50

0.75

1.00

1.25

So

lubl

e F

ract

ion

40 45 50 55 60 65 40 45 50 55 60 65

Temperature (°C)

CHUKIKBKGKPNA3NFKB1NFKB2NFKBIBNFKBIERELRELA

TNFα/NFκB signaling complex (CHUK, KPNA3, NFKB2, NFKBIB, REL, IKBKG, NFKB1, NFKBIE, RELB, NFKBIA, RELA, TNIP2)

40 45 50 55 60 65 40 45 50 55 60 65Temperature (°C)

TREX/THO Complex

Vehicle Methotrexate Vehicle Methotrexate

Vehicle Methotrexate

THOC1THOC2THOC3THOC5THOC6THOC7

THOC1THOC2THOC3THOC5THOC6THOC7

Vehicle Methotrexate

0.00

0.25

0.50

0.75

1.00

1.25

So

lubl

e F

ract

ion

0.00

0.25

0.50

0.75

1.00

1.25

So

lubl

e F

ract

ion

Core-CoreCORUM Pairs

Average Eucl. Distance between Protein Pairs in Each Complex

0.0 0.4 0.8 1.2

0.0

0.4

0.8

1.2

Un

iqu

e C

ore

-Att

ach

men

tC

OR

UM

Pai

rs

n=239

0.2

0.4

0.6

0.8

1.0

Eu

clid

ean

Dis

tan

ce b

etw

een

Inte

ract

ing

Pro

tein

Pai

rs

0-0.

2

0.2-

0.4

0.4-

0.6

0.6-

0.8

0.8-

1.0

All Pro

ten

Pairs

Interaction Stoichiometry

0.2

0.4

0.6

0.8

1.0

0-0.

2

0.2-

0.4

0.4-

0.6

0.6-

0.8

0.8-

1.0

All Pro

ten

Pairs

Abundance Stoichiometry

0.2

0.4

0.6

Abund

. Sto

i.> 0

.8

Int. S

toi <

0.8

Abund

. & In

t.

Stoi. >

0.8

Eu

clid

ean

Dis

tan

ce b

etw

een

Inte

ract

ing

Pro

tein

Pai

rs

5056 protein pairs5056 protein pairs

0.2

0.4

0.6

0.8

1.0

0-0.

2

0.2-

0.4

0.4-

0.6

0.6-

0.8

0.8-

1.0

All Pro

ten

Pairs

Abund. Stoi. × Int. Stoi.

C T M T+Mp-IKKα/β (S180/181)p-IKBα (S32)

IKBα

p-p65(S536)

Hsp90

Abund

. Sto

i.< 0

.8

Int. S

toi >

0.8

Fig. 3. TPCA signature interaction stoichiometry and abundancestoichiometry between interacting proteins. Correlation of TPCA signature(intact cell MS-CETSA data) with (A) protein interaction stoichiometry and(B) protein abundance stoichiometry of interacting protein pairs reported inHein et al. (9). (C) Protein pairs with high stoichiometry in both parametersexhibiting the highest TPCA signature. (D) Multiplication of interaction andabundance stoichiometry correlates better with TPCA signature. (E) Core-attachment model for functional organization of protein complexes. (F) Core-attachment protein pairs of a complex generally exhibit weaker TPCA signaturethan core-core protein pairs. Each dot represents a protein complex.

(G) Melting curves for subunits of the chromatin assembly complex 1 (CAF-1),the chromatin-remodeling BAFcomplex, the NF-kB complex, and the THOcomplex from methotrexate-treated and vehicle-treated K562 cells.(H) Assessment of the NF-kB signaling pathway uponmethotrexate treatmentin K562 cells.Western blotting analysis of canonical NF-kB signaling pathwaymembers in whole-cell lysate. (I) Immunoprecipitation of NF-kB p65 andp50 showing increased interaction with IkBa upon methotrexate treatment.C, control (DMSO); T,TNF-a; M, methotrexate; T+M,TNF-a+methotrexate.IP, immunoprecipitation; IB, immunoblotting; IgG, immunoglobulin G. Hsp90served as a loading control.

RESEARCH | REPORTon M

arch 19, 2020

http://science.sciencemag.org/

Dow

nloaded from

Page 6: PROTEIN FOLDING Thermal proximitycoaggregation for system ... · bility (fig. S4C) for proteins with at least three quantified peptides in each data set, and thus combined existing

Tan et al., Science 359, 1170–1177 (2018) 9 March 2018 6 of 7

40 45 50 55 60 65

Sol

uble

Fra

ctio

n

A375

0.00

0.25

0.50

0.75

1.00

1.25

40 45 50 55 60 65

HCT116

0.00

0.25

0.50

0.75

1.00

1.25

40 45 50 55 60 65

Sol

uble

Fra

ctio

n

HEK293T

40 45 50 55 60 65

HL60

40 45 50 55 60 65

MCF7

RAF1 BRAF

40 45 50 55 60 65

Temperature (°C)Temperature (°C) Temperature (°C)

Temperature (°C)Temperature (°C)Temperature (°C)

K562

BRAF MEK2

MEK1CRAF

ARAF

ERK1

ERK2

MK2p38α MEK1

ERK2

ARAF

CRAF ERK1

BRAF MEK2

MK2p38α

HCT116 A375

CRAF

MEK2

ERK1

ERK2

BRAF

MEK1

ARAF

MK2p38α

K562

CRAF

MEK2

ERK1

ERK2

BRAF

MEK1

ARAF

MK2p38α

HL60

ARAFBRAF MEK2

ERK2

MEK1CRAF ERK1 MK2p38α

MCF7

CRAF

MEK2

ERK1

ERK2

BRAF

MEK1

ARAF

MK2p38α

HEK293T

TPCA-derived Z-Score

MED17

BRD4

MED12CCNT1

CDK9

MED1

THOC1

ACAD8

MED4MED8

MED30

MED7 MED26

MED22

MED13L

MED15

MED9

THRAP3

MED13

MED10

MED29

CDK8

MED14

MED21

MED27HDAC1

SMARCD2

CCNC

SMARCC2

SMARCC1

ACTL6A

UPF2

DCP2

UPF1

XRN1

CHTF18

RFC4

RFC3

RFC5

RFC2

PCNA

NFKBIE

CCNH

GTF2F1

SIN3A

RBBP7

HDAC2

SMARCE1

SMARCA2

THOC3

THOC6

THOC7

CHAF1A

THOC2

PRMT5

RBBP4

CHAF1B

THOC5

ARID4B

UPF3B

EXOSC2

XRN2

EXOSC10EXOSC1

EXOSC4

PARNMED31

RELA

MED25

MED18MED20

MED16

CHUK

SMARCD1

SMARCD3

SAP30

ING1

ARID1A

MRE11A

RAD50

IKBKG

KPNA3

NFKB1

NBN

INTS12

INTS8

NFKB2INTS7

INTS6

RELINTS10

INTS2

CPSF3L

PLCG1

PIK3R1SOS1

NFKBIB

INTS3

EXOSC3

EXOSC8

DIS3

FANCD2

MDC1

EXOSC7

EXOSC6

EXOSC5

EXOSC9

ATMPOLR2A LYN

POLR2B

LCP2

PLCG2

GRB2

INTS9

INTS4

TREX/THOcomplex

TNFα/NFκB signaling complex

GRB2-associatedsignaling complexes

PCNA-CHL12-RFC2-5complex

MDC1-MRN-ATM-FANCD2complexIntegrator-RNAPII

complex

Mediator & associatedcomplexes

mRNA decaycomplex

Exosome

Chromatin remodeling & associated complexes

0.0 0.2 0.4 0.6 0.8 1.0

0.0

0.2

0.4

0.6

0.8

1.0

False Positive Rate

Pearson’s REuclidean

ROC curves

ROC curves

0.0 0.2 0.4 0.6 0.8 1.0

0.0

0.2

0.4

0.6

0.8

1.0

False Positive Rate

Tru

e p

osi

tive

rat

eT

rue

po

siti

ve r

ate

n=3n=2

n=1

Protein pairs occurring in at least n CORUM complexes

AUC0.670.69

AUC0.620.720.79

MCL COACH

Pre

cisi

on

Recall0 0.1 0.2

0

0.2

0.4

0.6

0.8

1.0

0

0.2

0.4

0.6

0.8

1.0

0 0.1 0.2 0.3 0.4

TPCAPublication CountPublication Count+ Function Similarity

0-1-2 1 2

Fig. 4. TPCA signature reveals cell-specific interactions and is predic-tive of protein complexes. (A) Network view of protein complexes withnonrandom increase in TPCA behavior in methotrexate-treated K562 cells.Subunits of protein complexes are visualized as nodes with the edgeconnecting every pair of subunits within a complex. A red node indicates thatthe protein has greater stability in methotrexate-treated cells than in vehicle-treated cells, whereas a blue node indicates the reverse. Color intensitycorrelates linearly with difference in stability. A red line or edge between twonodes indicates that melting curves of the two proteins are more similar inmethotrexate-treated cells, and a blue line or edge indicates the reverse. Lineor edge width correlates linearly with difference in melting curve similarity.Differentially expressed proteins are indicated in red (P < 0.10) and bold type(P < 0.05). (B) TPCA analysis reports differentiated interactions in RAF-MEK-ERK pathway in HCT116.TPCA-derived z-scores are used to facilitate

comparison of TPCA signatures across cell lines. A higher z-score impliesincreased interactions. (C) Melting curves of BRAFand RAF1 across the six celllines. (D) Prediction of CORUM protein complexes using graph or networkclustering algorithms on protein interaction network weighted with publicationcount, reliability score (publication count + function similarity) (33), andTPCA-based scoring. Precision and recall were evaluated against CORUMreference complexes (of size >3) with Jaccard similarity ≥ 0.5 considered asmatch. (E) Predictability of interacting protein pairs (table S2) reported by twoor more publications using TPCA. (F) Predictability of co-complex pairs usingTPCA. Melting curve data of intact K562 cells (table S7) are used. Euclideandistance between all protein pairs with melting curve is computed and rankedby ascending order (lower Euclidean distance first). All protein pairs areconsidered as negative data except those known to interact (table S2) orannotated as part of CORUM complexes.

RESEARCH | REPORTon M

arch 19, 2020

http://science.sciencemag.org/

Dow

nloaded from

Page 7: PROTEIN FOLDING Thermal proximitycoaggregation for system ... · bility (fig. S4C) for proteins with at least three quantified peptides in each data set, and thus combined existing

signatures, with 58% of the complexes exhibitingnonrandom TPCA signatures in at least one ofthe cell lines profiled. Membrane-embedded com-plexes are included in CORUM but are likely notamenable to the protocol adopted in this work(13). Nonsignificant TPCA signatures could arisefor protein complexes with very low interactionstoichiometry—increased assembly can be moni-tored by changes in TPCA signature. TPCA profil-ing suggests that many complexes can remainintact yet thermally destabilized in the absenceof interaction with DNA and LMW ligands suchas ATP (Fig. 2, A and E). We observedmanymito-chondrial proteins that seemmore temperature-resistant in intact cells, as observed previously(11). It is unclear whether TPCA is intrinsic andcaptured by the recent limited proteolysis andMS methodology (38).We observed many more complexes with sig-

nificant TPCA signature in cells than in lysates,suggesting that TPCA could potentially aid theintracellular studies of weak or transient protein-protein interactions that are not preserved inlysate, including protein complexes that dependon the integrity of chromatin-DNA, membrane,and associated structures for stability. TPCA couldbe used to validate complexes identified by otherproteome-wide methodologies and help in theirfunctional characterization across different cellstates and conditions. TPCA also permits study-ing chemical modulators of protein complexesand interactions directly in nonengineered cellsand tissues. Analogous to quantifying expressionof genes and proteins from a reference genomeand reference proteome, respectively, we envis-age TPCA-based profiling with reference inter-actomes as a system-wide discovery strategy formodulated cellular processes. The method there-fore can facilitate the discovery of protein com-plexes involved in diseases, some as potentialtherapeutic targets or, by TPCA profiles in patient

tissue samples, for prognosis of disease progres-sion or optimization of therapy.

REFERENCES AND NOTES

1. T. Ideker, R. Sharan, Genome Res. 18, 644–652 (2008).2. X. Wang et al., Nat. Biotechnol. 30, 159–164 (2012).3. A. Malovannaya et al., Cell 145, 787–799 (2011).4. P. C. Havugimana et al., Cell 150, 1068–1081 (2012).5. E. L. Huttlin et al., Cell 162, 425–440 (2015).6. T. Rolland et al., Cell 159, 1212–1226 (2014).7. R. M. Ewing et al., Mol. Syst. Biol. 3, 89 (2007).8. U. Stelzl et al., Cell 122, 957–968 (2005).9. M. Y. Hein et al., Cell 163, 712–723 (2015).10. D. Martinez Molina, P. Nordlund, Annu. Rev. Pharmacol. Toxicol.

56, 141–161 (2016).11. M. M. Savitski et al., Science 346, 1255784 (2014).12. K. V. Huber et al., Nat. Methods 12, 1055–1057 (2015).13. F. B. Reinhard et al., Nat. Methods 12, 1129–1131 (2015).14. A. Chatr-Aryamontri et al., Nucleic Acids Res. 43, D470–D478

(2015).15. H. Hermjakob et al., Nucleic Acids Res. 32, D452–D455 (2004).16. L. Licata et al., Nucleic Acids Res. 40, D857–D861 (2012).17. A. Ruepp et al., Nucleic Acids Res. 38 (suppl. 1), D497–D501

(2010).18. C. Ciferri et al., Cell 133, 427–439 (2008).19. H. W. Wang et al., J. Mol. Biol. 383, 894–903 (2008).20. J. R. Wiśniewski, M. Y. Hein, J. Cox, M. Mann, Mol. Cell.

Proteomics 13, 3497–3506 (2014).21. Z. Dezso, Z. N. Oltvai, A. L. Barabási, Genome Res. 13,

2450–2454 (2003).22. M. Wu, X. Li, C. K. Kwoh, S. K. Ng, BMC Bioinformatics 10, 169

(2009).23. M. Hoek, B. Stillman, Proc. Natl. Acad. Sci. U.S.A. 100,

12183–12188 (2003).24. B. Gómez-González, I. Felipe-Abrio, A. Aguilera, Mol. Cell. Biol.

29, 5203–5213 (2009).25. R. E. Wellinger, F. Prado, A. Aguilera, Mol. Cell. Biol. 26,

3327–3334 (2006).26. M. Ito, C. X. Yuan, H. J. Okano, R. B. Darnell, R. G. Roeder, Mol.

Cell 5, 683–693 (2000).27. H. Cho et al., Mol. Cell. Biol. 18, 5355–5363 (1998).28. W. F. Marzluff, E. J. Wagner, R. J. Duronio, Nat. Rev. Genet. 9,

843–854 (2008).29. R. Canavan, U. Bond, Nucleic Acids Res. 35, 6268–6279 (2007).30. J. R. Skaar et al., Cell Res. 25, 288–305 (2015).31. S. A. Martin et al., EMBO Mol. Med. 1, 323–337 (2009).32. J. R. Haling et al., Cancer Cell 26, 402–413 (2014).33. G. Liu, L. Wong, H. N. Chua, Bioinformatics 25, 1891–1897

(2009).

34. T. Nepusz, H. Yu, A. Paccanaro, Nat. Methods 9, 471–472 (2012).35. S. Pu, J. Vlasblom, A. Emili, J. Greenblatt, S. J. Wodak,

Proteomics 7, 944–960 (2007).36. M. Li, J. E. Chen, J. X. Wang, B. Hu, G. Chen, BMC

Bioinformatics 9, 398 (2008).37. H. N. Chua, W. K. Sung, L. Wong, Bioinformatics 22, 1623–1630

(2006).38. P. Leuenberger et al., Science 355, eaai7825 (2017).

ACKNOWLEDGMENTS

We thank H. Y. Chang for support and A. Larsson for suggestions.Funding: This research was funded directly by Young InvestigatorGrant (1610151038) awarded to C.S.H.T. by the BiomedicalResearch Council of the Agency for Science, Technology andResearch (A*STAR). Support for this research was provided by astartup grant from Nanyang Technological University and grantsfrom the Swedish Research Council, the Swedish Cancer Society,and the Knut and Alice Wallenberg foundation awarded to P.N., andby National Medical Research Council (NMRC) grantMOHIAFCAT2/004/2015 to P.N. and R.M.S. Research in the lab ofV.T. is supported by grant NRF2016NRF-CRP001-024 from theNational Research Foundation Singapore. P.K. and X.B. aresupported by the Biomedical Research Council, A*STAR, andNMRC-CBRG14nov086 grants. Authors contributions: P.N.initiated the study; C.S.H.T. conceptualized TPCA and designedand implemented associated algorithms; C.S.H.T, X.B., P.K., V.T.,R.M.S., and P.N. designed and supervised experiments; C.S.H.T.and C.H.Y. performed computational analysis; K.D.G., C.S.H.T.,X.B., M.B.O., L.D., N.P., Y.T.L., and L.S. performed experiments;R.M.S. and J.L. supervised MS analysis; C.S.H.T. wrote the originalmanuscript; and C.S.H.T. and P.N. reviewed and edited themanuscript. Competing interests: P.N. is the inventor of a patentcontrolled by Pelago Biosciences AB and Evitra Proteoma ABcovering the basic CETSA method. J.L. is a paid consultant ofPelago Biosciences AB for MS analysis. All other coauthors declareno competing interests. Data and materials availability: All dataare available in the supplementary materials.

SUPPLEMENTARY MATERIALS

www.sciencemag.org/content/359/6380/1170/suppl/DC1Materials and MethodsSupplementary TextFigs. S1 to S27Tables S1 to S27

22 February 2017; resubmitted 28 September 2017Accepted 27 January 2018Published online 8 February 201810.1126/science.aan0346

Tan et al., Science 359, 1170–1177 (2018) 9 March 2018 7 of 7

RESEARCH | REPORTon M

arch 19, 2020

http://science.sciencemag.org/

Dow

nloaded from

Page 8: PROTEIN FOLDING Thermal proximitycoaggregation for system ... · bility (fig. S4C) for proteins with at least three quantified peptides in each data set, and thus combined existing

cellsThermal proximity coaggregation for system-wide profiling of protein complex dynamics in

Ting Lim, Lekshmy Sreekumar, Johan Lengqvist, Vinay Tergaonkar, Philipp Kaldis, Radoslaw M. Sobota and Pär NordlundChris Soon Heng Tan, Ka Diam Go, Xavier Bisteau, Lingyun Dai, Chern Han Yong, Nayana Prabhu, Mert Burak Ozturk, Yan

originally published online February 8, 2018DOI: 10.1126/science.aan0346 (6380), 1170-1177.359Science 

, this issue p. 1170; see also p. 1105Scienceinteractions in six cell lines, highlighting the potential for identifying protein complexes that are modulated by disease.the curves. The method was validated by detection of many known protein complexes. It identified cell-specificto determine melting curves for thousands of proteins and assigns a TPCA signature on the basis of similarity between

assaythat proteins within a complex will coaggregate upon heat denaturation. It uses a previously described cellular shift ). The method is based on the ideaet al.the dynamics of native protein complexes inside cells (see the Perspective by Li

developed a method called thermal proximity coaggregation (TPCA) to monitoret al.depending on cellular needs. Tan Many of the processes in living cells are mediated by protein complexes that dynamically assemble and dissociate

Taking the heat together

ARTICLE TOOLS http://science.sciencemag.org/content/359/6380/1170

MATERIALSSUPPLEMENTARY http://science.sciencemag.org/content/suppl/2018/02/07/science.aan0346.DC1

CONTENTRELATED http://science.sciencemag.org/content/sci/359/6380/1105.full

REFERENCES

http://science.sciencemag.org/content/359/6380/1170#BIBLThis article cites 38 articles, 11 of which you can access for free

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

Terms of ServiceUse of this article is subject to the

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

Science. No claim to original U.S. Government WorksCopyright © 2018 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of

on March 19, 2020

http://science.sciencem

ag.org/D

ownloaded from