Research Article Identification of Posttranslational ... - BioMedSearch

0 downloads 0 Views 2MB Size Report
Jun 25, 2014 - electron transfer dissociation (ETD) MS/MS spectra gener- ated from the 20 highest intensity precursor ions. The voltage between the ion spray ...
Hindawi Publishing Corporation PPAR Research Volume 2014, Article ID 468925, 8 pages http://dx.doi.org/10.1155/2014/468925

Research Article Identification of Posttranslational Modifications in Peroxisome Proliferator-Activated Receptor 𝛾 Using Mass Spectrometry Shogo Katsura,1 Tomoko Okumura,1 Ryo Ito,1,2 Akira Sugawara,2 and Atsushi Yokoyama1,2 1 2

Institute of Molecular and Cellular Biosciences, University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-0032, Japan Department of Molecular Endocrinology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan

Correspondence should be addressed to Atsushi Yokoyama; [email protected] Received 31 March 2014; Accepted 19 May 2014; Published 25 June 2014 Academic Editor: Elisabetta Mueller Copyright © 2014 Shogo Katsura et al. This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. Posttranslational modification (PTM) of proteins is critical for various cellular processes. However, there are few studies examining PTMs in specific proteins using unbiased approaches. Here we report the attempt to identify the PTMs in peroxisome proliferatoractivated receptor 𝛾 (PPAR𝛾) proteins using our previously established PTM analysis system. In this study, we identified several PTMs in exogenously expressed PPAR𝛾2 proteins from 293T cells as well as endogenous PPAR𝛾1 proteins from a Caco-2 colon cancer cell line. The identified PTMs include phosphorylation of serine 112 and serine 81 in PPAR𝛾2 and PPAR𝛾1, respectively, both of which are well-known mitogen-activated protein kinase- (MAP kinase-) mediated PTMs in PPAR𝛾 proteins, thus confirming our experimental approach. Furthermore, previously unknown PTMs were also identified, demonstrating that this method can be applied to find previously unidentified PTMs in PPAR𝛾 proteins and other proteins including nuclear receptors.

1. Introduction Peroxisome proliferator-activated receptor 𝛾 (PPAR𝛾 and NR1C3) is a ligand-dependent nuclear receptor, which was initially established as the dominant regulator of adipocyte differentiation [1, 2]. PPAR𝛾 is activated by natural ligands, such as polyunsaturated fatty acids and eicosanoids, plays a dominant role in adipose cell differentiation, modulates metabolism and inflammation in immune cells, and has strong antigrowth properties. Although PPAR𝛾 levels are the highest in adipose cells, substantial levels are also found in certain other cell types, such as the colonic epithelium and epithelial cells of the breast and prostate. The PPAR𝛾 gene encodes two main splicing isoforms: PPAR𝛾1 and PPAR𝛾2. PPAR𝛾1 is expressed in many tissues and organs including stomach and small and large intestine, whereas PPAR𝛾2 is expressed specifically in adipocytes [1, 2]. Each PPAR𝛾 protein binds as a heterodimer with retinoid X receptor (RXR) to its recognition site called PPRE, with the sequence TGACCTxTGACCT. On binding to DNA, PPAR𝛾

recruits transcriptional coactivators such as steroid receptor coactivator 1 (SRC1) and positively regulates the expression of target genes [3, 4]. Though the underlying molecular mechanisms are unclear, PPAR𝛾-mediated transrepression for inflammatory genes has also been described [5]. PPAR𝛾 also undergoes several PTMs in response to exogenous signals such as growth factors and adipokines, resulting in modulation of its functions [6]. For example, mitogen-activated protein kinases (MAP kinases) phosphorylate the N-terminal AF-1 domain of PPAR𝛾 (serine 84 or 112 in human PPAR𝛾1 or PPAR𝛾2, resp.), and this PTM inhibits ligand-dependent PPAR𝛾 transcriptional activity [7–11]. Moreover, PPAR𝛾 also undergoes SUMOylation at the C-terminal ligand-binding domain (LBD) in a liganddependent manner. SUMOylated PPAR𝛾 binds to NCoR corepressor complex and thereby transrepresses inflammatory genes such as iNOS genes in macrophages [12]. In the postgenomic era, it has become obvious that diversity of biological phenomena cannot be explained only by the number of different genes. Protein posttranslational

2 modifications (PTMs) are one of the most efficient biological signals for expanding the genetic code and play key roles in diverse cellular processes such as protein transportation, DNA repair, and gene transcription [13–15]. We have previously developed a comprehensive PTM analysis system for proteins, combining biochemical and proteomic approaches [16]. In this system, we utilized nonrestrictive sequence alignment for PTM analysis and this method made it possible to identify PTMs without prior specification, enabling unbiased analysis of PTMs [17, 18]. Recent findings have revealed that the functions of epigenetic regulators, including PPAR𝛾, are under control of PTMs in response to cellular signaling and nutrients [6, 19]. Here we applied the unrestricted comprehensive analysis to PTMs in PPAR𝛾 and identified a subset of PTMs from overexpressed PPAR𝛾2 from 293T cells and endogenous PPAR𝛾1 protein from Caco-2 colon cancer cells.

2. Materials and Methods 2.1. Plasmids, Antibodies, and Reagents. The expression plasmid for full-length PPAR𝛾2 cDNA was cloned from a cDNA library of human adipocyte [20] and inserted into a pcDNA3 vector (Life Technologies, Carlsbad, CA) with a FLAG tag sequence. Anti-PPAR𝛾 antibody (#sc-7273) was purchased from Santa Cruz Biotechnology (Santa Cruz, CA). Troglitazone (Sigma, St. Louis, MO) was dissolved in DMSO. 2.2. Cell Culture and Transfection. T and Caco-2 cells were cultured in Dulbecco’s modified Eagle medium (DMEM) plus 10% fetal bovine serum (FBS) and antibiotics. THP-1 cells were cultured in RPMI plus 10% FBS and antibiotics. For transfection of FLAG-PPAR𝛾2 into 293T cells in the 10 cm dishes, we used Lipofectamine 2000 (Life Technologies) according to the manufacturer’s instructions and incubated the cells for 24 h. 2.3. Purification of the PPAR𝛾 Proteins and Immunoprecipitation. Purification of PPAR𝛾 protein was performed as previously described [16]. Briefly, we cross-linked 2 𝜇g of antibody with 30 𝜇L of Protein G Dynabeads (Invitrogen) in freshly dissolved 20 mM dimethyl pimelimidate (DMP) in a 0.2 M triethanolamine buffer (pH 8.2). The cross-linking reaction was performed for 1 h at room temperature. The reaction was stopped by replacing the buffer with 50 mM Tris-HCl (pH 7.5). Cell lysates of 293T transfected with FLAG-PPAR𝛾2 and Caco-2 cell (from a 10 cm dish and 48 dishes, resp.) were prepared with 1% NP-40 buffer (10 mM Tris [pH 7.8], 1 mM EDTA, 150 mM NaCl, and 1% NP-40). Lysates were subjected to immunoprecipitation with Protein G Dynabeads coupled to each antibody for 3 h. The immunoprecipitates were washed by 1% NP-40 buffer and eluted with 0.1 M glycine-HCl buffer (pH 2). The eluates were boiled with Laemmli sample buffer and then subjected to sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) and then Colloidal Blue (Life Technologies), silver staining (Silver

PPAR Research Quest, Life Technologies), or Western blotting with the indicated antibodies. 2.4. Mass Spectrometric Analysis. Mass spectrometric (MS) analysis of PPAR𝛾 protein and PTM analysis were performed as previously described [16]. Briefly, PPAR𝛾 protein was excised from the gel, reduced with 10 mM dithiothreitol solution in 0.1 M ammonium bicarbonate for 60 min at 56∘ C, alkylated with a 55 mM solution of iodoacetamide in 0.1 M ammonium bicarbonate in darkness for 45 min at room temperature, and in-gel digested with 25 ng/𝜇L trypsin gold (Promega, Madison, WI) in 50 mM ammonium bicarbonate for 16 h at 37∘ C. Digested peptides were extracted, replaced with 0.1% formic acid in 2% acetonitrile (ACN), and subjected to analysis by electrospray ionization- (ESI-) MS/MS using an LTQ velos Orbitrap with ETD (Thermo Fisher Scientific). The nano-LC used was a DiNa system (KYA TECH Corporation, Tokyo, Japan) equipped with a C-18 ESI capillary column (100 𝜇m × 150 mm, NIKKYO Technos, Tokyo, Japan). The gradient consisted of (A) 0.1% formic acid in 2% ACN and (B) 0.1% formic acid in 80% ACN: 0–100% B from 0 to 110 min, 100% B from 111 to 115 min, and 0% B from 116 to 120 min. The flow rate was 300 nL/min from 0 to 120 min. MS spectra were recorded from a range of m/z 350–1500 at 100,000 resolution, followed by data-dependent collision-induced dissociation (CID) MS/MS spectra and electron transfer dissociation (ETD) MS/MS spectra generated from the 20 highest intensity precursor ions. The voltage between the ion spray tip and the transfer tube was set to 1800 V. Peptides with +2 or greater charge were chosen for MS/MS experiments. 2.5. Computational Analysis for Protein Identification and PTM Analysis. Protein identification and PTM analysis of PPAR𝛾 protein were performed as previously described [16]. Briefly, for protein identification, spectra were processed using Proteome Discoverer ver. 1.3 (Thermo Fisher Scientific) against SEQUEST and subjected to a cutoff of 5% false discovery rate (FDR). The NCBI human protein database was used with a 10 ppm mass accuracy cutoff for parental MS and FT MS/MS and a 0.8 Da cutoff for ion trap MS/MS spectra. Carbamidomethylation (cysteine) was set as a fixed modification and oxidation (methionine) was set as a variable modification. For PTM identification, spectra were processed using the MODIRO ver. 1.1 (Protagen, Bochum, Germany) software against FASTA format of human PPAR𝛾1 and human PPAR𝛾2 amino acid sequences. Search parameters were set as follows: two maximum missing cleavage sites, a peptide mass tolerance of 15 ppm for peptide tolerance, 1.5 Da for fragment mass tolerance (for ion trap MS/MS), 15 ppm for fragment mass tolerance (for FT MS/MS), and modification 1 of carbamidomethyl (cysteine). In all the identified PTMs, PTMs which might have occurred during sample preparations such as methylation of glutamic acid [21] were excluded from the list.

PPAR Research

3

BSA 0.1 𝜇g

THP-1

293T (FLAG-PPAR𝛾2)

THP-1

Caco-2

Caco-2

𝛼-PPAR𝛾

IgG THP-1 293T (FLAG-PPAR𝛾2)

Caco-2

293T (FLAG-PPAR𝛾2)

Input

∙ 293T (FLAG-PPAR𝛾2) cell lysates

150

∙ Caco-2 cell lysates (kDa)

100 Protein G Dynabeads cross-linked anti-PPAR𝛾 antibody

75 PPAR𝛾2 PPAR𝛾1 50 37

Acid elution

Silver staining

SDS-PAGE (a)

(b)

THP-1

Caco-2

THP-1

Caco-2

293T (FLAG-PPAR𝛾2)

𝛼-PPAR𝛾

IgG 293T (FLAG-PPAR𝛾2)

THP-1

Caco-2

293T (FLAG-PPAR𝛾2)

Input

PPAR𝛾2 PPAR𝛾1 IB: PPAR𝛾 (c)

Figure 1: Purification of PPAR𝛾 proteins from 293T and Caco-2 cell lysates. (a) Schematic diagram of the purification of the exogenous and endogenous PPAR𝛾 proteins. 293T and Caco-2 cell lysates were incubated with anti-PPAR𝛾 cross-linked Protein G Dynabeads as described in Section 2. Rabbit IgG cross-linked Protein G Dynabeads were used as a negative control. Bound proteins were eluted with 0.1 M glycineHCl buffer (pH 2). (b) Isolated exogenous and endogenous PPAR𝛾 proteins. Eluted proteins were subjected to SDS-PAGE, followed by silver staining. The molecular masses of PPAR𝛾1 and PPAR𝛾2 are shown in the right side of the gel. (c) Enrichment of PPAR𝛾 proteins were also confirmed by Western blotting using anti-PPAR𝛾 specific antibodies.

3. Results 3.1. Purification of Exogenous PPAR𝛾2 from 293T and Endogenous PPAR𝛾1 from Caco-2 Cells. For comprehensive analysis of PTMs in PPAR𝛾 protein, we performed the affinity purification of PPAR𝛾 proteins from 293T cells and Caco-2 colon cancer cells, which were expressing exogenous FLAG-tagged PPAR𝛾2 and endogenous PPAR𝛾1 protein, respectively, using the scheme shown in Figure 1(a). 1% NP-40 soluble fraction of cell lysates prepared from 293T and Caco-2 cells without PPAR𝛾 ligand was prepared and incubated with Protein G Dynabeads cross-linked with anti-PPAR𝛾 antibodies. Purified protein-antibody complexes were washed with lysis buffer and then eluted from the antibodies by adding acidic glycine-HCl buffer; eluates were then subjected to SDS-PAGE and visualized by silver staining. As shown in Figure 1(b), we successfully detected exogenous FLAG-PPAR𝛾2 in

the anti-PPAR𝛾 affinity purified eluates at the expected molecular size. We also detected endogenous PPAR𝛾1 proteins in the anti-PPAR𝛾 affinity purified eluates from Caco-2. THP-1 human monocyte cells were used as indicators of PPAR𝛾1 molecular size [22]. Enrichment of FLAG-PPAR𝛾2 and endogenous PPAR𝛾1 proteins was also confirmed by Western blotting using anti-PPAR𝛾 specific antibodies (Figure 1(c)). 3.2. Identification of PPAR𝛾 Proteins Using SEQUEST Algorithm. The purified FLAG-PPAR𝛾2 from 293T cells and PPAR𝛾1 protein from Caco-2 cells were cut from the colloidal blue-stained gel, in-gel digested with trypsin, and subjected to analysis by LC-MS/MS using a combination of collisioninduced dissociation (CID) and electron transfer dissociation (ETD) activation. Precursor MS spectra were detected by Orbitrap mass analyzers (a Fourier transform (FT) mass analyzer that can measure peptide m/z with high accuracy)

4 (ΔMS < 5 ppm), and MS/MS spectra were detected using the ion trap analyzer (ΔMS < 0.5 Da). MS spectral data were first analyzed using the SEQUEST algorithm in Proteome Discoverer ver. 1.3 and the PPAR𝛾 protein was identified with high accurate molecular weight (sequence coverage 67.92%, FDR < 5% for PPAR𝛾2 from 293T cells, coverage 49.79%, FDR < 5% for PPAR𝛾1 from Caco-2 cells), suggesting successful protein purification and mass measurement for the PPAR𝛾 proteins (Figures 2(a), 2(b) and 2(c)). 3.3. Nonrestrictive PTM Analysis for Purified PPAR𝛾 Proteins Using the MODIRO Algorithm. Next, the spectral data were searched using the MODIRO algorithm, which enables unrestricted identification of all possible PTMs in targeted proteins. As a result, various PTMs were identified in peptides derived from exogenous PPAR𝛾2 protein from 293T cells (Figures 3(a) and 3(b)), and identified PTMs with significance score >90 were presented in Figures 3(b) and 3(c). At the top of the list, phosphorylation of serine 112 in PPAR𝛾2 was identified with significance score = 100. This phosphorylation site is one of the well-characterized PTM sites in PPAR𝛾2 protein, catalyzed by MAPKs and inhibiting the liganddependent transcription function of PPAR𝛾 [7–11]. This result confirms that our experimental approach detects the PTMs of PPAR𝛾 protein correctly. Furthermore, this PTMs list also included some PTMs such as methylation of 487 threonine residue and ubiquitination of lysine 160, which were novel PTMs for PPAR𝛾 proteins. To further confirm these identified PTMs, we analyzed the peptides by LC-MS/MS again. In this measurement, to obtain more rigorous MS/MS data and thereby more reliable PTM search results, we utilized the FT analyzer for both parental MS and MS/MS spectra. As shown in Figure 3(d), two of the three PTMs including S112 phosphorylation and T487 methylation were identified again with SEQUEST algorithm, thus confirming the initial result. Next, PPAR𝛾1 proteins from Caco-2 cells were analyzed in the same way. As shown in Figures 4(a), 4(b) and 4(c), phosphorylation of serine 84 was identified with significance score = 100. This serine residue corresponds to serine 112 of PPAR𝛾2. We further tested PPAR𝛾1 proteins from 10 𝜇M Troglitazone-treated Caco-2 cells (24 h). However, no other PTMs besides S84 phosphorylation were identified from this analysis (data not shown).

4. Discussion Here, we described the first report of comprehensive PTM analysis of PPAR𝛾 protein using mass spectrometry and succeeded in identifying some previously known and unknown PTMs including serine phosphorylation and threonine methylation. Among the identified PTMs, phosphorylation of S112 and S84 in PPAR𝛾2 and PPAR𝛾1, respectively, was the one of the well-known PTMs in PPAR𝛾 proteins. It is reported that phosphorylation at S112 in PPAR𝛾2 was catalyzed by a mitogen-activated protein (MAP) kinase and was involved in

PPAR Research repression of transcriptional activity in adipocyte [7]. However, whether this modification exists in the PPAR𝛾1 protein in colon cancer cells was elusive. Thus, in this study we could confirm the existence of S84 phosphorylation in PPAR𝛾1 in colon cancer cells. Although the role of S84 phosphorylation in colon cancer is unclear, the phosphorylation mimic mutant of PPAR𝛾1 (S84D) slightly derepressed PPAR𝛾-mediated transrepression of 𝛽-catenin using the TOPflash reporter system [23], and further characterization with regard to the effect in transcriptional function is needed (Shogo Katsura, unpublished result). In the present study, we could also identify the K160 ubiquitination and T487 methylation of PPAR𝛾 proteins, although there are few reports about these modifications. Protein ubiquitination can be targeted to a number of biological pathways such as proteasomal degradation and signal transduction depending on the numbers and linkage types of the conjugated ubiquitins [24]. Because the number and linkage types of the conjugated ubiquitins were not clear from present experiment, further detailed analyses are required to characterize the function of K160 ubiquitination in PPAR𝛾2 protein. As for threonine methylation, it is still unclear whether this modification occurs in the sample preparation prior to MS analysis or is posttranslational [14]. We avoided methanol in each step of our experimental procedure so that proteins were not nonenzymatically methylated, although still we should be careful about this modification. However, this is a novel modification of PPAR𝛾 protein and thus our comprehensive analysis of PTMs can be applied to find previously unidentified PTMs in PPAR𝛾 proteins and other proteins including nuclear receptors. Along with the identified phosphorylation, ubiquitination, and methylation, PPAR𝛾 protein is known to also undergo several PTMs, such as phosphorylation at other regions [25, 26], SUMOylations [12], O-GlcNAcylations [27], and ubiquitination [28, 29], although these PTMs were not identified in this study. Generally, as mass spectrometric analysis can detect relatively highly concentrated or efficiently ionized peptides [15, 17], it is possible to speculate that there are many more PTMs than what have been identified in PPAR𝛾 proteins. Because purified PPAR𝛾 proteins might be mixtures of variously modified states, we presume that, with further purification of the eluted proteins such as by isoelectric fractionation of purified PPAR𝛾 proteins or enrichment of specific modifications with PTM-recognizing antibodies, we could extend the variations of identified peptides and thus detect greater numbers of PTMs in PPAR𝛾 proteins. Those PTMs might be new candidates for drug targets to control PPAR𝛾 activities.

5. Conclusions We have identified several modifications including phosphorylation of serines 112 and 81 in PPAR𝛾2 and PPAR𝛾1, respectively, and threonine methylation in PPAR𝛾2 using our previously established PTM analysis system. This method can be applied to find previously unidentified PTMs in PPAR𝛾 proteins and other proteins including nuclear receptors.

PPAR Research

5

Intensity (counts)

PPAR𝛾2 4200 4000 3800 3600 3400 3200 3000 2800 2600 2400 2200 2000 1800 1600 1400 1200 1000 800 600 400 200 0

S V

E

A

V

A

Y

Q E I

V

T

Q

E

E

I

T

E

E

981.5159

T Q

I

E V

Y

A

A b[+1] y[+1]

E

Y A K

853.4808 611.3323

640.9861

510.2648 381.3228

724.5114 1080.5537

486.2808 856.4898

369.3430 316.2491 218.1195 254.2223 324.3483

100

150

200

250

300

350

400

715.6851

511.3397 596.3229

458.3839

450

500

550

600

650

700

838.4752 734.7620 835.5722

750

800

957.5300

900

1280.6210 1320.5750

1068.5313 1086.5580

909.6925

850

1151.6089

1050.5863

1134.5054

1231.4612

1330.3995

950 1000 1050 1100 1150 1200 1250 1300 1350

(m/z)

Intensity (counts)

PPAR𝛾1 42000 40000 38000 36000 34000 32000 30000 28000 26000 24000 22000 20000 18000 16000 14000 12000 10000 8000 6000 4000 2000 0

A

Y

S V

E

A

V

Q T

V E

Q E I

T

E

I

E

E

T

I

981.6100 Q

E V

Y

A

A b[+1] y[+1]

E

Y A K

611.3849

387.3207

853.5764

641.0496

510.2936

724.5853

486.2864

150

200

250

300

350

1151.6803

838.6519 856.6251

369.2928 316.3759 298.3360 323.4264 209.2424 240.2159

100

1080.6870

939.5366

486.3477 596.4117 576.3777

458.3426

400

450

500

550

600

716.4139 829.6257 680.7112 743.4557

650

750

700

800

1068.6456

921.6993

850

900

950

1050.6719

1280.6987

1086.6199 1134.7570

1320.7329 1249.6603 1221.7404

1000 1050 1100 1150 1200 1250 1300

(m/z)

(a)

Coverage: 67.92% 1 11

21

31

41

51

61

71

1

MGETLGDSPI

DPESDSFTDT

LSANISQEMT

101

EYQSAIKVEP

ASPPYYSEKT

QLYNKPHEEP

SNSLMAIECR

GRMPQAEKEK LLAEISSDID

QLNPESADLR

ALAKHLYDSY

IKSFPLTKAK

ARAILTGKTT DKSPFVIYDM

IPGFVNLDLN

DQVTLLKYGV

HEIIYTMLAS

LMNKDGVLIS

201

VGMSHNAIRF

MVDTEMPFWP

QSKEVAIRIF

QGCQFRSVEA

VQEITEYAKS

401

KFNALELDDS

DLAIFIAVII

LSGDRPGLLN VKPIEDIQDN

501

YKDLY

301

TNFGISSVDL

SVMEDHSHSF

81

91

DIKPFTTVDF

SSISTPHYED

IPFTR TDPVV ADYKYDLKLQ

VCGDKASGFH YGVHACEGCK GFFRRTIRLK

LIYDRCDLNC

RIHKKSRNKC

LLQALELQLK LNHPESSQLF

QYCRFQKCLA

NSLMMGEDKI KFKHITPLQE

EGQGFMTREF LKSLRKPFGD FMEPKFEFAV

AKLLQKMTDL RQIVTEHVQL

LQVIKKTETD

MSLHPLLQEI

FDR < 0.01 (b)

Coverage: 49.79% 1 11 21 51 71 91 31 41 61 81 1 MTMVDTEMPF WPTNFGISSV DLSVMEDHS H SFDIKPFTTV DFSSISTPHY EDIPFTRTDP VVADYKYDLK LQEYQSAIKV EPASPPYYSE KTQLYNKPHE 101

EPSNSLMAIE

CRVCGDKASG

201

IDQLNPESAD

LRALAKHLYD SYIK SFPLTK AKARAILTGK

301

KSIPGFVNLD

LNDQVTLLKY GVHEIIYTML

ASLMNKDGVL

ISEGQGFMTR

401

KPIEDIQDNL

LQALELQLKL NHPESSQLFA

KLLQKMTDLR

QIVTEHVQLL

FHYGVHACEG

CKGFFRRTIR LKLIYDRCDL TTDKSPFVIY

NCRIHKKSRN

RFGRMPQAEK

EKLLAEISSD

QEQSKEVAIR IFQGCQFRSV

EAVQ EITEYA

EFLKSLRKPF GDFMEPKFEF AVK FNALELD DSDLAIFIAV

IILSGGLLNV

DMNSLMMGED

QVIKKTETDM

KCQYCRFQKC LAVGMSHNAI KIKFKHITPL

SLHPLLQEIY K

FDR < 0.01 FDR < 0.05 (c)

Figure 2: Identification of PPAR𝛾 proteins using the SEQUEST algorithm. (a) Representative MS/MS spectra of PPAR𝛾2 proteins of the peptide [317-SVEAVQEITEYAK-329] and PPAR𝛾1 proteins of the peptide [289-SVEAVQEITEYAK-301] assigned by SEQUEST are shown. (b) Amino acid sequence coverage of identified exogenous PPAR𝛾2 proteins. The identified amino acid sequence is indicated. (c) Amino acid sequence coverage of identified endogenous PPAR𝛾1 proteins. The identified amino acid sequence is indicated.

6

PPAR Research

a b

L Q E YQ S A I KV E P A S P P Y Y S E K 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 21 20 19 18 17 16 15 14 13 12 11 10 9 8 7 6 5 4 3 2 1

0

200

400

600

800 1000 (m/z) (Da)

1200

1400

1600

y

1800

(a) Sequence

meas. Modification m/z(Da)

Position

Phosphorylation (STY) Methylation (C DEHKNQRST) Phosphorylation (STY) GlyGLy (K)

99–119

K.LQEYQSAIKVEPAS HPO3 PPYYSEK.T

K.TMe ETDMSLHPLLQEIYK.D

487–502

K.VEPAS HPO3 PPYYSEK.T

108–119

K.ASGFHYGVHACCAMe EGCKGlyGly GFFR.R

146–164

m/z theor. Error (Da) Charge (Da)

Sig.

1254.0911

1254.0907

0.0003

2

966.4921

966.4928

−0.0007

2

100

723.8124

723.8131

−0.0007

2

99.6

748.6661

748.6669

−0.0008

3

90.7

100

(b)

Methylation (T487)

Phosphorylation Ubiquitylation (S112) (K160) DBD 1

138

LBD

203

281

505

PPAR𝛾2

Intensity (counts)

(c) P

P

Y

50000 45000 40000 35000 30000 25000 20000 15000 10000 5000 0

K V E P A s

L Q E Y Q S A I

KV

S

P P Y Y S

AP

1205.6043 E

EV

PA

200

300

400

689.3065 755.3247

526.2537

500

600

700

800

IAS

y[+1]

1526.7864

1120.5297 933.4702 1020.5148

865.4089

b[+1]

1289.6722

1196.5986 429.0870

s

E K

883.4197

337.9508

K

900

1236.5692 1329.5365

1476.7433

1574.7175 1624.7579

1747.9325 1845.8789

1000 1100 1200 1300 1400 1500 1600 1700 1800

Intensity (counts)

(m/z) 130000 120000 110000 100000 90000 80000 70000 60000 50000 40000 30000 20000 10000 0

T

t

E T

D

I

D M S

L

H P

L

502.2951 245.1135 185.0917

100

200

346.1593

300

MSLH

EQ

456.2005

400

500

L

Q E I

618.3132

844.9412 L

A

736.4026

y[+1]

793.9154 852.4351

700

H

Y K

557.2935

600

b[+1] P

800

900

1003.5819 1140.6539

1791.5409

1000 1100 1200 1300 1400 1500 1600 1700

(m/z) (d)

Figure 3: Identified PTMs in PPAR𝛾2 proteins from 293T cells. (a) Representative MS/MS spectra of PPAR𝛾2 proteins of the peptide [99LQEYQSAIKVEPASHPO3 PPYYSEK-119] assigned by MODIRO are shown. (b) List of identified PTMs in exogenous PPAR𝛾2 proteins using ion trap MS/MS. Amino acid sequences, position of amino acids, identified PTMs theoretical mass, measured mass, error between measured and theoretical masses in Daltons, and significance score are listed. (c) Diagram for summary of identified PTMs in exogenous PPAR𝛾2 proteins. (d) Identified PTMs in exogenous PPAR𝛾2 proteins using FT MS/MS. Each responsible spectrum for S112 (top) and T487 (bottom) is shown.

PPAR Research

7

a b

0

200

400

600

800

1000

L Q E Y Q S A I K V E P A S P P Y Y S E K 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 y 21 20 19 18 17 16 15 14 13 12 11 10 9 8 7 6 5 4 3 2 1

1200

1400

1600

1800

(m/z) (Da)

(a) Sequence K.LQEYQSAIKVEPAS

HPO3

Position

PPYYSEK.T 71–91

m/z meas. Modification (Da) Phosphorylation 836.3947 (STY)

m/z theor. Error (Da) (Da) 836.3962

−0.0015

Charge

Sig.

3

100

(b)

Phosphorylation (S84) DBD 1

108

LBD

173

251

477

PPAR𝛾1 (c)

Figure 4: Identified PTMs in PPAR𝛾1 proteins from Caco-2 cells. (a) Representative MS/MS spectra of PPAR𝛾1 proteins of the peptide harboring [80-VEPASHPO3 PPYYSEK-91] assigned by MODIRO are shown. (b) List of identified PTMs in endogenous PPAR𝛾1 proteins using ion trap MS/MS. (c) Diagram for summary of identified PTM in endogenous PPAR𝛾1 proteins.

Conflict of Interests The authors declare that there is no conflict of interests regarding the publication of this paper.

Acknowledgments The authors appreciate the technical assistance provided by Ms. Kei Izumikawa (University of Tokyo). This work was supported in part by priority areas from the Ministry of Education, Culture, Sports, Science and Technology (to Atsushi Yokoyama).

References [1] P. Tontonoz and B. M. Spiegelman, “Fat and beyond: the diverse biology of PPARgamma,” Annual Review of Biochemistry, vol. 77, pp. 289–312, 2008. [2] I. A. Voutsadakis, “Peroxisome proliferator-activated receptor 𝛾 (PPAR𝛾) and colorectal carcinogenesis,” Journal of Cancer Research and Clinical Oncology, vol. 133, no. 12, pp. 917–928, 2007. [3] J. Xu and Q. Li, “Review of the in vivo functions of the p160 steroid receptor coactivator family,” Molecular Endocrinology, vol. 17, no. 9, pp. 1681–1692, 2003. [4] M. G. Rosenfeld, V. V. Lunyak, and C. K. Glass, “Sensors and signals: a coactivator/corepressor/epigenetic code for integrating signal-dependent programs of transcriptional response,” Genes and Development, vol. 20, no. 11, pp. 1405–1428, 2006. [5] M. Ricote and C. K. Glass, “PPARs and molecular mechanisms of transrepression,” Biochimica et Biophysica Acta. Molecular and Cell Biology of Lipids, vol. 1771, no. 8, pp. 926–935, 2007.

[6] Z. E. Floyd and J. M. Stephens, “Controlling a master switch of adipocyte development and insulin sensitivity: covalent modifications of PPAR𝛾,” Biochimica et Biophysica Acta. Molecular Basis of Disease, vol. 1822, no. 7, pp. 1090–1095, 2012. [7] E. Hu, J. B. Kim, P. Sarraf, and B. M. Spiegelman, “Inhibition of adipogenesis through MAP kinase-mediated phosphorylation of PPAR𝛾,” Science, vol. 274, no. 5295, pp. 2100–2103, 1996. [8] B. Zhang, J. Berger, G. Zhou et al., “Insulin- and mitogenactivated protein kinase-mediated phosphorylation and activation of peroxisome proliferator-activated receptor 𝛾,” Journal of Biological Chemistry, vol. 271, no. 50, pp. 31771–31774, 1996. [9] H. S. Camp and S. R. Tafuri, “Regulation of peroxisome proliferator-activated receptor 𝛾 activity by mitogen-activated protein kinase,” Journal of Biological Chemistry, vol. 272, no. 16, pp. 10811–10816, 1997. [10] H. S. Camp, S. R. Tafuri, and T. Leff, “c-Jun N-terminal kinase phosphorylates peroxisome proliferator- activated receptor-𝛾1 and negatively regulates its transcriptional activity,” Endocrinology, vol. 140, no. 1, pp. 392–397, 1999. [11] M. Adams, M. J. Reginato, D. Shao, M. A. Lazar, and V. K. Chatterjee, “Transcriptional activation by peroxisome proliferatoractivated receptor 𝛾 is inhibited by phosphorylation at a consensus mitogen-activated protein kinase site,” Journal of Biological Chemistry, vol. 272, no. 8, pp. 5128–5132, 1997. [12] G. Pascual, A. L. Fong, S. Ogawa et al., “A SUMOylationdependent pathway mediates transrepression of inflammatory response genes by PPAR-𝛾,” Nature, vol. 437, no. 7059, pp. 759– 763, 2005. [13] R. J. Sims III and D. Reinberg, “Is there a code embedded in proteins that is based on post-translational modifications?” Nature Reviews Molecular Cell Biology, vol. 9, no. 10, pp. 815– 820, 2008.

8 [14] C. T. Walsh, S. Garneau-Tsodikova, and G. J. Gatto Jr., “Protein posttranslational modifications: the chemistry of proteome diversifications,” Angewandte Chemie, vol. 44, no. 45, pp. 7342– 7372, 2005. [15] J. Seo, J. Jeong, M. K. Young, N. Hwang, E. Paek, and K.J. Lee, “Strategy for comprehensive identification of posttranslational modifications in cellular proteins, including low abundant modifications: application to glyceraldehyde-3phosphate dehydrogenase,” Journal of Proteome Research, vol. 7, no. 2, pp. 587–602, 2008. [16] A. Yokoyama, S. Katsura, R. Ito et al., “Multiple posttranslational modifications in hepatocyte nuclear factor 4𝛼,” Biochemical and Biophysical Research Communications, vol. 410, no. 4, pp. 749–753, 2011. [17] A. R. Farley and A. J. Link, “Identification and quantification of protein posttranslational modifications,” Methods in Enzymology, vol. 463, pp. 725–763, 2009. [18] E. Ahrn´e, M. M¨uller, and F. Lisacek, “Unrestricted identification of modified proteins using MS/MS,” Proteomics, vol. 10, no. 4, pp. 671–686, 2010. [19] E. Smith and A. Shilatifard, “The chromatin signaling pathway: diverse mechanisms of recruitment of histone-modifying enzymes and varied biological outcomes,” Molecular Cell, vol. 40, no. 5, pp. 689–701, 2010. [20] T. Yanase, T. Yashiro, K. Takitani et al., “Differential expression of PPAR 𝛾1 and 𝛾2 isoforms in human adipose tissue,” Biochemical and Biophysical Research Communications, vol. 233, no. 2, pp. 320–324, 1997. [21] R. Sprung, Y. Chen, K. Zhang et al., “Identification and validation of eukaryotic aspartate and glutamate methylation in proteins,” Journal of Proteome Research, vol. 7, no. 3, pp. 1001– 1006, 2008. [22] H. Shu, B. Wong, G. Zhou et al., “Activation of PPAR𝛼 or 𝛾 reduces secretion of matrix metalloproteinase 9 but not interleukin 8 from human monocytic THP-1 cells,” Biochemical and Biophysical Research Communications, vol. 267, no. 1, pp. 345–349, 2000. [23] T. Fujisawa, A. Nakajima, N. Fujisawa et al., “Peroxisome proliferator-activated receptor 𝛾 (PPAR𝛾) suppresses colonic epithelial cell turnover and colon carcinogenesis through inhibition of the 𝛽-catenin/T cell factor (TCF) pathway,” Journal of Pharmacological Sciences, vol. 106, no. 4, pp. 627–638, 2008. [24] D. Komander and M. Rape, “The ubiquitin code,” Annual Review of Biochemistry, vol. 81, pp. 203–229, 2012. [25] J. H. Choi, A. S. Banks, J. L. Estall et al., “Anti-diabetic drugs inhibit obesity-linked phosphorylation of PPAR𝛾 3 by Cdk5,” Nature, vol. 466, no. 7305, pp. 451–456, 2010. [26] J. H. Choi, A. S. Banks, T. M. Kamenecka et al., “Antidiabetic actions of a non-agonist PPAR𝛾 ligand blocking Cdk5mediated phosphorylation,” Nature, vol. 477, no. 7365, pp. 477– 481, 2011. [27] S. Ji, S. Y. Park, J. Roth, H. S. Kim, and J. W. Cho, “O-GlcNAc modification of PPAR𝛾 reduces its transcriptional activity,” Biochemical and Biophysical Research Communications, vol. 417, no. 4, pp. 1158–1163, 2012. [28] S. Hauser, G. Adelmant, P. Sarraf, H. M. Wright, E. Mueller, and B. M. Spiegelman, “Degradation of the peroxisome proliferatoractivated receptor 𝛾 is linked to ligand-dependent activation,” Journal of Biological Chemistry, vol. 275, no. 24, pp. 18527–18533, 2000.

PPAR Research [29] G. E. Kilroy, X. Zhang, and Z. E. Floyd, “PPAR-𝛾 AF-2 domain functions as a component of a ubiquitin-dependent degradation signal,” Obesity, vol. 17, no. 4, pp. 665–673, 2009.