You are on page 1of 13

ARTHRITIS & RHEUMATOLOGY

Vol. 67, No. 7, July 2015, pp 1891–1903


DOI 10.1002/art.39125
C 2015, American College of Rheumatology
V

Epitope Specificity Determines Pathogenicity and Detectability


of Anti–Platelet-Derived Growth Factor Receptor a
Autoantibodies in Systemic Sclerosis

Gianluca Moroncini,1 Antonella Grieco,1 Giulia Nacci,2 Chiara Paolini,1 Cecilia Tonnini,1
Katarzyna N. Pozniak,1 Massimiliano Cuccioloni,3 Matteo Mozzicafreddo,3
Silvia Svegliati,1 Mauro Angeletti,3 Andrius Kazlauskas,4 Enrico V. Avvedimento,5
Ada Funaro,2 and Armando Gabrielli1

Objective. To identify the epitopes recognized by for expression of IgG binding to PDGFRa and induction
autoantibodies targeting platelet-derived growth factor of reactive oxygen species in fibroblasts. The variable
receptor a (PDGFRa) in systemic sclerosis (SSc) and regions of anti-PDGFRa IgG were cloned into an IgG
develop novel assays for detection of serum anti- expression vector to generate distinct recombinant human
PDGFRa autoantibodies. monoclonal autoantibodies (mAb), which were character-
Methods. Epstein-Barr virus–immortalized B cells ized by binding and functional assays. The epitopes of
from 1 patient with SSc (designated PAM) were screened anti-PDGFRa recombinant human mAb were defined by
molecular docking, surface plasmon resonance binding
assays, screening of a conformational peptide library
Supported by the Gruppo Italiano Lotta alla Sclerodermia (Gils
Young Investigator Award to Dr. Moroncini), the Ministero Italiano per
spanning the PDGFRa extracellular domains, and expres-
l’Universita e la Ricerca Scientifica (grants to Drs. Avvedimento, Funaro, sion analyses of alanine-scanned PDGFRa mutants. Direct
and Gabrielli), the Italian Association for Cancer Research (grant or competitive enzyme-linked immunosorbent assays were
IG111364 to Dr. Avvedimento), the Fondazione Cariverona and
Fondazione Italiana Ricerca Artrite (grants to Dr. Gabrielli), and the
established to detect all serum anti-PDGFRa autoanti-
Fondazione di Medicina Molecolare e Terapia Cellulare (research bodies or, selectively, the agonistic ones.
funding to the Universita Politecnica delle Marche, Ancona, Italy). Results. Three types of anti-PDGFRa recombi-
Dr. Nacci is recipient of a fellowship from the Fondazione Umberto
Veronesi. nant human mAb, with the same VH but distinct VL
1
Gianluca Moroncini, MD, PhD, Antonella Grieco, PhD, chains, were generated. Nonagonistic VHPAM–Vk13B8
Chiara Paolini, PhD, Cecilia Tonnini, PhD, Katarzyna N. Pozniak, recognized 1 linear epitope, whereas agonistic VHPAM–
PhD, Silvia Svegliati, PhD, Armando Gabrielli, MD: Universit a Poli-
tecnica delle Marche and Ospedali Riuniti Ancona, Ancona, Italy; Vl16F4 and VHPAM–Vk16F4 recognized 2 distinct con-
2
Giulia Nacci, PhD, Ada Funaro, PhD: Universita di Torino, Turin, formational epitopes. Serum anti-PDGFRa antibodies
Italy; 3Massimiliano Cuccioloni, PhD, Matteo Mozzicafreddo, PhD, were detected in 66 of 70 patients with SSc, 63 of
Mauro Angeletti, PhD: Universita di Camerino, Camerino, Italy;
4
Andrius Kazlauskas, PhD: Schepens Eye Research Institute and Har- 130 healthy controls, 11 of 26 patients with primary
vard Medical School, Boston, Massachusetts; 5Enrico V. Avvedimento, Raynaud’s phenomenon (RP), and 13 of 29 patients
MD, PhD: Universita Federico II, Naples, Italy. with systemic lupus erythematosus (SLE). Serum
Drs. Grieco, Nacci, and Paolini contributed equally to this
work. VHPAM–Vk16F4-like antibodies were found in 24 of 34
Drs. Moroncini, Svegliati, Avvedimento, Funaro, and Gabrielli patients with SSc, but not in healthy controls, patients
hold a patent application (US Patent Application no. 13/812,764; Epi-
topes of the human PDGF Receptor able to bind human autoantibod-
with primary RP, or patients with SLE. Peptides com-
ies, antibodies and uses thereof). Dr. Gabrielli has received consulting posing the VHPAM–Vk16F4 epitope inhibited PDGFRa
fees, speaking fees, and/or honoraria from Actelion Pharmaceuticals signaling triggered by serum IgG from SSc patients.
(less than $10,000).
Address correspondence to Gianluca Moroncini, MD, PhD,
Conclusion. Agonistic anti-PDGFRa autoantibod-
or to Armando Gabrielli, MD, Universita Politecnica delle Marche, ies are enriched in SSc sera and recognize specific confor-
Dipartimento di Scienze Cliniche e Molecolari, Via Tronto 10, 60126 mational epitopes that can be used to discriminate agonistic
Ancona, Italy. E-mail: g.moroncini@univpm.it or a.gabrielli@univpm.it.
Submitted for publication July 17, 2014; accepted in revised from nonagonistic antibodies and block PDGFRa sig-
form March 17, 2015. naling in patients with SSc.
1891
1892 MORONCINI ET AL

Systemic sclerosis (SSc; scleroderma) is a clini- flow cytometry, using, as target cells, mouse embryonic fibro-
cally heterogeneous disease of the connective tissue blasts derived from PDGFR-knockout mice (F2/2 cells) or
mouse embryonic fibroblasts transfected with full-length
characterized by vascular, immune/inflammatory, and
human PDGFRa (Fa cells) (12), as described previously (3).
fibrotic manifestations, in which oxidative stress is a Generation of recombinant human mAb. Total RNA
prominent feature (1,2). Despite extensive investiga- was extracted from each anti-PDGFRa–positive B cell line
tions, the key pathogenic links between these disease (;300,000 cells) with an RNeasy Micro kit and reverse-
hallmarks remain obscure, which hampers the develop- transcribed with an Omniscript RT kit (Qiagen). Complemen-
tary DNA (cDNA) was amplified with polymerase chain reaction
ment of tools for early diagnosis, adequate disease mon-
(PCR) primers specific for rearranged human IgG variable-
itoring, and effective therapies. We have previously region and constant-region heavy- and light-chain genes (13)
reported the presence of agonistic anti–platelet-derived (details available upon request from the corresponding author).
growth factor receptor (anti-PDGFR) autoantibodies Amplified heavy- and light-chain variable regions (VH and VL,
that stimulate reactive oxygen species (ROS) in the respectively) were sequenced using a TOPO TA Cloning kit
(Invitrogen). The unique VH-chain sequence and the 4 differ-
serum IgG of 46 patients with SSc (3). However, other
ent VL-chain sequences amplified in the 3 PAM B cell lines
studies (4,5) failed to detect these stimulatory antibod- showing PDGFRa binding were alternatively paired to replace
ies in the serum, and found nonstimulatory anti- the VH and VL chains of antibody b12 (14), thereby generating
PDGFR antibodies both in SSc patients and in healthy 4 distinct human IgG1 constructs. VH and VL sequences were
controls (5) or in patients with systemic lupus erythema- fused with the respective leader peptide sequences using a 3-
step overlap extension PCR (15), and then independently
tosus (SLE) (6). Therefore, the presence of stimulatory
inserted into the Xba I–Sac I and Hind III–Eco RI restriction
anti-PDGFR autoantibodies in SSc remains controver- sites of the pDR12 vector containing the parental human IgG1
sial (7). constant-region heavy- and light-chain genes, including the Fc
To address this issue, we generated combinatorial sequence.
monoclonal autoantibodies (mAb) using immunoglobu- Upon sequencing, the constructs were stably trans-
fected into Chinese hamster ovary (CHO) cells with Lipofect-
lin sequences, identified in memory B cells obtained
amine 2000 (Invitrogen). Expression of recombinant IgG in
from the peripheral blood of 1 patient with SSc, that the culture supernatants was confirmed by human IgG–specific
were specific for PDGFRa, the isoform that was enzyme-linked immunosorbent assay (ELISA). Cell cultures
reported to better discriminate SSc IgG from healthy were expanded in bioreactors (CELLine CL-1000; Integra Bio-
control IgG (3). With the use of these reagents, we sciences), and secreted recombinant human mAb were purified
from the supernatants by Protein A–affinity and size-exclusion
obtained a novel map of the functional epitopes of
chromatography. The purity of the recombinant human mAb
PDGFRa selectively driving oxidative stress and collagen was checked by sodium dodecyl sulfate–polyacrylamide gel elec-
gene expression increase. Moreover, we assessed the clin- trophoresis (SDS-PAGE) Coomassie brilliant blue protein
ical relevance of these findings by developing novel bind- staining.
ing assays for anti-PDGFRa autoantibodies. Immunoprecipitation and immunoblotting. Each re-
combinant human mAb (10 mg/ml) was incubated for 4 hours
at 48C with 100 mg of total protein extracts from human fibro-
MATERIALS AND METHODS blasts. The mixture was incubated overnight at 48C with 20 ml
of agarose–Protein A/G (Santa Cruz Biotechnology). The
Ethics statements. Use of human material was ap- immunocomplexes were then subjected to SDS-PAGE and
proved by the Institutional Ethics Committee of Universita immunoblotting, as described previously (3).
Politecnica delle Marche (Ancona, Italy). Informed consent Functional assays. Primary human fibroblasts from
was obtained from all subjects participating in this study. adult healthy skin (Life Technologies) were cultured in Dulbecco’s
All assays were performed in a blinded manner on coded modified Eagle’s medium (DMEM) with 10% fetal calf serum
samples. (FCS) for 2 passages. When subconfluence was reached, the
Screening of immortalized memory B cells. CD22- cells were maintained in 0.2% FCS for 24 hours, before the
positive memory B cells were purified by magnetic selection addition of human PDGF-BB (15 ng/ml), transforming growth
(Miltenyi Biotech) from peripheral blood mononuclear cells factor b (2 ng/ml) (each from R&D Systems), or recombinant
(PBMCs) obtained from 1 patient with diffuse SSc designated human mAb (10 mg/ml). Protein extracts (30 mg) were sepa-
PAM (47-year-old woman with esophagus and lung involve- rated on 4–12% SDS-PAGE under reducing conditions and
ment, a Rodnan skin thickness score of 17, positivity for anti- transferred to PVDF membranes. After blocking, the membranes
nuclear antibodies and anti–Scl-70 antibodies, and negativity were probed with anti-pAkt (sc-439), anti–pERK-1/2 (sc-7783),
for anticentromere antibodies) (8). These cells (designated the and anti–Ha-Ras (sc-520) antibodies, and then stripped and
PAM B cells) were immortalized using Epstein-Barr virus, as reprobed with antibodies to total Akt, ERK-1/2, and b-actin
described previously (9–11). Three days postinfection, the cells (Santa Cruz Biotechnology), followed by horseradish peroxi-
were manually counted and seeded (5 cells/well) in 96-well dase (HRP)–conjugated antibodies.
plates in the presence of irradiated (30 Gy) allogenic PBMCs. To quantify collagen gene transcription, total RNA
After 2 weeks, B cell supernatants were screened for the pres- was extracted from the primary human fibroblasts using the
ence of anti-PDGFRa antibodies by immunofluorescence and Aurum total RNA Mini kit (Bio-Rad) at baseline and after 1
EPITOPE SPECIFICITY OF ANTI-PDGFRa AUTOANTIBODIES IN SSc 1893

hour of stimulation. The purity of the RNA templates, meas- Soluble peptides. Three peptides were synthesized and
ured as the ratio between sample absorbance at 260 and numbered from 1 to 3 following the amino acid sequence of
280 nm (Nanodrop; Thermo Scientific), was in the 1.8–2.1 human extracellular PDGFRa from the N- to the C-terminus.
range. RNA (1 mg) was reverse transcribed with an iScript Peptide 1 was ac-VIVEDDDSAIIPCRTTD-conh2, peptide 2
cDNA synthesis kit (Bio-Rad). The human type I collagen was ac-VVPASYDSRQGFNGTFTVGPYICE-conh2, and pep-
genes COL1A1 and COL1A2 were measured by real-time tide 3 was ac-CAARQATREVKEMKKVT-conh2. Scrambled
PCR with iQ SYBR Green Supermix and an iCycler (Bio- peptide 2 was ac-IASGCGTFTRVVFEQPNSPYYDGV-conh2.
Rad). The primers used were as follows: for human COL1A1, The purity of the peptides, determined by high-performance
forward 50 -AGGGCCAAGACGAAGACATC-30 and reverse liquid chromatography, was .95%.
50 -AGATCACGTCATCGCACAACA-30 ; for human COL1A2, In binding-competition experiments, VHPAM–Vk16F4
forward 50 -AGGTCAAACAGGAGCCCGTGGG-30 and reverse (10 mg/ml) (molar ratio of peptide to recombinant human
50 -GCACCTGGGAAGCCTGGAGGG-30 ; for human GAPDH, mAb 100:1) or a pool of IgG from the serum of 6 SSc patients
forward 50 -TGCACCACCAACTGCTTAGC-30 and reverse or 6 healthy controls (each 200 mg/ml) was incubated in solu-
50 -GGCATGGACTGTGGTCATGAG-30 ; for human 18S ribo- tion with single peptides for 1 hour at 378C. The mixtures were
somal RNA, forward 50 -TCCCCATGAACGAGGAATTC-30 then used to stimulate fibroblasts.
and reverse 50 -GTGTACAAAGGGCAGGGACTT-30 . PCR PDGFRa mutagenesis and expression of PDGFRa
conditions were 958C for 3 minutes, and then 958C for 15 sec- mutants in F2/2 mouse embryonic fibroblasts. Five different
onds and 588C for 60 seconds (45 cycles). sets of alanine mutations were introduced into the epitope of
The ROS bioassay was performed as described previ- VHPAM–Vk16F4. The template for mutagenesis (GenScript)
ously (3,16), using a fluorometric assay to determine the was the full-length human PDGFRa cDNA inserted into the
amount of intracellular ROS generated by adherent retroviral vector pLHDCX2 (22). The PDGFRa mutants were
fibroblasts. termed Ala-scans and numbered from the N- to the C-terminus
Homology modeling and molecular docking. Homol- according to the corresponding peptides: for Ala-scan 1,
109
ogy modeling to predict the unknown 3-dimensional structures EDDD112 to 109AAAA112; for Ala-scan 2, 149TFT151 to
149
of human PDGFRa and recombinant human mAb monovalent AAA151; and for Ala-scan 3, 262QATR265 to 262AAAA265. Two
fragments from their amino acid sequences was performed longer Ala-scans were generated, combining Ala-scans 1 and 2
using a Swiss-Model server (17). Swiss-Pdb Viewer software (Ala-scan 4) or Ala-scans 1, 2, and 3 (Ala-scan 5). The 5 Ala-scans
was used to create project files submitted to the server with were individually transfected into PA317 cells (ATCC) with Lipo-
default parameter settings (BLAST search P , 0.00001 and fectamine, to produce retroviral virions (23). Virus-containing
global degree of sequence identity .25% determined using the medium was collected, concentrated (at 25,000g for 90 minutes at
SIM alignment tool). PDGFRa query sequences were obtained 48C), and used to infect F2/2 mouse embryonic fibroblasts (12)
from the UniProt Knowledgebase (details available at http:// grown in DMEM with 8 mg/ml polybrene (hexadimethrine bro-
beta.uniprot.org/). Rigid protein–protein molecular docking mide; Sigma-Aldrich) for 24 hours. Successfully infected cells
between homology-modeled monomeric PDGF-BB (pdb identi- were selected in histidine-free DMEM containing 5 mM histidi-
fication no. 1PDG) (18), recombinant human mAb Fab frag- nol (Sigma-Aldrich) (24). Mutated PDGFRa expression was
ments, and monomeric PDGFRa was performed by uploading assessed in the cells by fluorescence-activated cell sorting analy-
the pdb files on a ClusPro 2.0 server (19) and using DOT software sis, using VHPAM–Vk16F4 and anti-PDGFRa mAb 1264 anti-
(version 1.0) as the docking program, with a clustering radius of bodies (R&D Systems).
5Å, electrostatic hits of 1,500, and 30 final resulting structures Immunoenzymatic assays (ELISAs). A recombinant
(details available upon request from the corresponding author). receptor fused to a poly-histidine tag (PDGFRa-His), either
Generation and screening of PDGFRa peptide library. produced in-house (25) or obtained commercially (amino acid
Eighty overlapping 15-oligomer synthetic peptides spanning Met-1–Glu-524, code no. 10556-H08H; Sino Biological), was
the first 3 N-terminal, extracellular Ig-like domains of immobilized (0.5 mg/ml in 100 ml HEPES buffered saline
PDGFRa were flanked by either glycines or cysteines. [HBS], pH 7.2) onto 96-well Nunc Maxisorp ELISA plates
Glycine-flanked peptides were left in linear conformation, overnight at 48C. After 2 hours of blocking at 378C with 200 ml
whereas cysteine-flanked peptides were imposed on a triva- of HBS containing 0.2% polyvinylpyrrolidone (Sigma-Aldrich)
lent (T3) chemical scaffold containing 3 PhCH2Br (1,3,5- and washing with HBS/0.05% Tween 20, serum (1:100 in HBS,
tribromomesitylene [T3]) groups, reacting 1:1 with the 3 100 ml/well) was added overnight at 48C. After extensive wash-
cysteines in the peptides, with the sequence Cys-15-oligomer- ing, HRP-conjugated anti-human IgG (1:10,000, 100 ml/well)
Cys-15-oligomer-Cys, displaying spatially defined conformations (Jackson ImmunoResearch) was added for 1 hour at 208C. The
to mimic a looped conformation. By virtue of this technology reaction was developed with 100 ml of tetramethylbenzidine
(Chemically Linked Peptides on Scaffolds [CLIPS]; Pepscan (BioFX) for 10 minutes at room temperature, and then stopped
Therapeutics) (20,21), the eighty 15-oligomer segments were with 50 ml of 0.5M H2SO4. The optical density (OD) was mea-
all combined with each other to obtain all of the possible com- sured at 450 nm.
binations of Cys-15-oligomer-Cys-15-oligomer-Cys bicyclic In the peptide-based ELISA, 20 mg/ml of each peptide
peptides (total number of 30-oligomer peptides identified by was immobilized onto 96-well plates and probed with 0.7 mg/ml
T3-CLIPS 5 6,400). The T3-CLIPS topology allows the pep- of the indicated recombinant human mAb. In the competitive
tides to have either a 2-looped folding or a loop plus partial ELISA, serum (1:100), VHPAM–Vk16F4 (0.7 mg/ml), or
sheet folding. The combinatory peptides were immobilized PDGF-BB (15 ng/ml) was preincubated with 25 mg/ml of
onto 455-well plates, and binding of recombinant human mAb the indicated peptide (molar ratio of peptide to PDGFRa 50:1)
and PDGF-BB was tested by Pepscan-based ELISA (Pepscan for 1 hour at 378C. Alternatively, serum (1:100) was incubated
Therapeutics). with immobilized PDGFRa-His overnight at 48C, before adding
1894 MORONCINI ET AL

Figure 1. Characterization of IgG in the PAM13B8, PAM16F4, and PAM17H8 B cell lines from the peripheral blood of a patient with systemic scle-
rosis. A, Left, Binding of supernatant IgG from the 3 B cell lines (white profile) to mouse embryonic fibroblasts transfected with full-length platelet-
derived growth factor receptor a (PDGFRa; Fa) or those from mice deficient in PDGFRa (F2/2) was assessed by flow cytometry. Shaded profile is
background fluorescence. Right, Production of reactive oxygen species (ROS) was assayed in Fa mouse fibroblasts treated with PDGF-BB or IgG
purified from supernatants of the 3 B cell lines. Results are the mean 6 SEM of 3 experiments, measured as relative fluorescence units (RFU) of
dihydrodichlorofluorescein diacetate fluorescence. B, Subgroup specificities of the polymerase chain reaction–amplified IgG cDNA fragments identified
in the 3 B cell lines are shown. VH or VL chain sequences with the same color encode for the same transcripts. The VH chain shared by the 3 cell
lines was designated VHPAM. The VL sequences were classified according to the k or l subgroups and the PAM cell line in which they were first
identified. C, PDGFRa was immunoprecipitated (IP) from human fibroblasts with the 4 recombinant human monoclonal antibodies (mAb), and
visualized by Western blotting (WB) using an anti-PDGFRa antibody. Anti-PDGFRa mAb 1264 was used as a positive control. D, Levels of ROS
(left), levels of pAkt, Akt, pERK-1/2, ERK-1/2, and Ha-Ras (middle), and induction of type I collagen genes (right) were assessed in human fibro-
blasts treated with recombinant human mAb, PDGF-BB, phosphate buffered saline (PBS), or transforming growth factor b (TGFb). In A and D,
results are the mean 6 SEM of 3 experiments performed in duplicate. * 5 P , 0.05 versus baseline.

biotinylated VHPAM–Vk16F4 (100 ng/ml) (ChromaLink Biotin RESULTS


Protein Labeling Kit Solulink; VWR International) for 2 hours
at 378C, followed by HRP-conjugated streptavidin (1:5,000)
(Bio-Rad) for 1 hour at room temperature. Cloning of agonistic and nonagonistic anti-
Statistical analysis. Data are presented as the mean 6 PDGFRa autoantibodies from the memory B cells of a
SEM or mean 6 SD, as indicated. Results were analyzed with patient with SSc. Immortalized memory B cells from 1
one-way analysis of variance for nonparametric data, with a patient with SSc (the PAM B cell lines) (9–11) were
Kruskal-Wallis test and Dunn’s test for multiple comparisons seeded in 96-well plates. Supernatants of ;2,000 wells
(analyses of peptide binding, direct and competitive ELISAs)
or 2-tailed unpaired t-test (with 95% confidence intervals were screened for the presence of IgG selectively binding
[95% CIs]) (functional analyses, cell-based assays). Spear- to Fa mouse embryonic fibroblasts, but not to fibroblasts
man’s rank correlation coefficients (for nonparametric data) derived from F2/2 mouse embryos (12), and inducing
were calculated to assess correlations between the percentage ROS in the fibroblasts. Three B cell cultures (namely,
of inhibition of VHPAM–Vk16F4 binding to PDGFRa-His PAM13B8, PAM16F4, and PAM17H8) contained
after incubation with sera, and the percentage of inhibition of
serum binding to PDGFRa after preincubation with peptide PDGFRa-binding IgG, of which the IgG from PAM16F4
2. Receiver operating characteristic (ROC) curves were gener- B cells induced ROS (Figure 1A).
ated to determine the area under the ROC curve with its 95% RNA was extracted from these B cells, reverse-
CI. The optimal cutoff values were chosen according to the transcribed, and amplified with primers designed to ana-
highest Youden index, representing sensitivity 1 (1 2 specific- lyze the human IgG gene repertoire (13). PCR analysis of
ity), which was calculated using MedCalc12 software. Graph-
Pad Prism 5 and MedCalc12 software were used for all rearranged IgG genes suggested the presence of a panel
statistical analyses. P values less than 0.05 were considered of VH and VL chains representative of most IgG sub-
statistically significant. groups (Figure 1B). However, subsequent sequencing
EPITOPE SPECIFICITY OF ANTI-PDGFRa AUTOANTIBODIES IN SSc 1895

revealed that most of these PCR products were similar,


although not identical, at the oligonucleotide level (details
available upon request from the corresponding author),
both within each of the individual B cell cultures and
among the 3 cell cultures, indicating that oligoclonal
restriction occurred. Indeed, PAM13B8, PAM16F4, and
PAM17H8 cells shared a single VH-chain amino acid
sequence, designated VHPAM, and a VL chain, designated
Vl13B8. Moreover, PAM13B8 and PAM17H8 shared a Vk
chain, designated Vk13B8, whereas the repertoire of
PAM16F4 included 2 additional VL chains, designated
Vk16F4 and Vl16F4 (Figure 1B).
The VHPAM cDNA and, alternatively, each of
the 4 VLPAM cDNAs were cloned into an expression
vector containing the human IgG1 constant region (15)
and transfected into CHO cells to produce 4 recombi-
nant human mAb, designated VHPAM–Vk13B8, VHPAM–
Vl13B8, VHPAM–Vk16F4, and VHPAM–Vl16F4. All
recombinant human mAb, except VHPAM–Vl13B8,
immunoprecipitated PDGFRa from human fibroblast
lysates (Figure 1C). VHPAM–Vk16F4 and VHPAM–Vl16F4,
but not VHPAM–Vk13B8 and VHPAM–Vl13B8,
induced ROS production and phosphorylation of ERK-
1/2 and Akt in vitro (Figure 1D). VHPAM–Vk16F4, but
not VHPAM–Vl16F4, also induced Ha-Ras and type I
collagen gene expression (Figure 1D). These data show
that PDGFRa binding and the biologic effects of these
recombinant human mAb can be dissociated, suggest-
ing that the PDGFRa epitopes mediating such func-
tions are distinct.
Epitope mapping of human PDGFRa. To iden-
tify the PDGFRa-specific epitopes of the recombinant
human mAb, we used in silico molecular docking, i.e., a
computer simulation of molecular interactions, as a
starting model to predict the binding regions between
homology-modeled monomeric human PDGFRa and
recombinant human mAb Fab fragments or monomeric
human PDGF-BB, the only PDGF isoform with resolved
crystal structure. PDGF-BB was predicted to bind a dis-
Figure 2. Epitope mapping of human PDGFRa. Molecular docking
continuous epitope encompassing 4 amino acid segments models show binding of monomeric PDGF-BB (A) or Fab fragments
lying between the second and third Ig-like extracellular of the recombinant human mAb VHPAM–Vk16F4 (B), VHPAM–
domains of PDGFRa (Figure 2A), which is consistent with Vk13B8 (C), and VHPAM–Vl16F4 (D) to the extracellular region of
the observations from previous studies (26,27). monomeric PDGFRa, encompassing the 5 Ig-like domains (labeled
I–V from the N- to the C-terminus). Below each model, the amino
The agonistic recombinant human mAb VHPAM–
acid sequence of the first 3 N-terminal extracellular domains of
Vk16F4 was predicted to bind to a discontinuous epitope PDGFRa is shown. Along this sequence, the amino acids composing
within the second and third PDGFRa extracellular the predicted PDGF-BB and recombinant human mAb binding sites
domains, largely overlapping the PDGF-BB binding site are highlighted in the indicated colors. The binding sites identified
(Figure 2B). Conversely, the predicted epitope of the in the PDGFRa peptide library are underlined. Insets in B–D show
binding curves (measured in arc/seconds [arcsec] over time) of the
nonagonistic recombinant human mAb VHPAM–Vk13B8
specific recombinant human mAb to PDGFRa-His immobilized on
encompassed a single linear amino acid sequence within the biosensor chip, alone or after saturation of PDGFRa-His with
the first PDGFRa extracellular domain (Figure 2C). Part PDGF-BB (#),VHPAM–Vk13B8 (##), or VHPAM–Vl16F4 (###).
of this sequence was shared by the larger, discontinuous See Figure 1 for other definitions.
1896 MORONCINI ET AL

Figure 3. Specific PDGFRa peptides interfere with the binding and functions of the agonistic VHPAM–Vk16F4 recombinant human mAb and
IgG from the serum of patients with systemic sclerosis (SSc). A, Binding of VHPAM–Vk16F4, VHPAM–Vl16F4, and VHPAM–Vk13B8 to immo-
bilized peptides 1, 2, and 3. B, Binding of VHPAM–Vk16F4 or PDGF-BB to immobilized PDGFRa-His, with or without preincubation with the
peptides (molar ratio of peptide to PDGFRa 50:1). *** 5 P , 0.001 versus without peptides. C, COL1A1 and COL1A2 gene induction in human
fibroblasts exposed to VHPAM–Vk16F4, with or without preincubation with the peptides (molar ratio of peptide to recombinant human mAb
100:1). * 5 P , 0.05; ** 5 P , 0.01 versus without peptides. D, Human COL1A1 gene expression (left) or ROS production (right) in fibroblasts
incubated with equal amounts of pooled IgG purified from the sera of 6 SSc patients or 6 healthy control (HC) subjects or SSc IgG with or with-
out preincubation with the peptides. * 5 P , 0.05 versus SSc IgG without peptides. Scrambled peptide 2 was used as a control. Results are the
mean 6 SEM of 3 independent experiments performed in duplicate. See Figure 1 for other definitions.

epitope of the agonistic recombinant human mAb binding of VHPAM–Vk16F4 to PDGFRa (Figure 2B).
VHPAM–Vl16F4, which comprised additional amino acid These data confirm that VHPAM–Vk16F4 (inducing
stretches in the first and second PDGFRa domains both ROS and type I collagen genes) recognizes an epi-
(Figure 2D). tope that overlaps the PDGF binding site but is distinct
To experimentally validate these predictive data, from the epitopes of VHPAM–Vk13B8 (nonagonistic)
we screened with PDGF-BB and the recombinant and VHPAM–Vl16F4 (inducing ROS, but not type I
human mAb a conformational PDGFRa peptide library, collagen genes).
which was synthesized by Pepscan (28,29). This approach Conversely, binding of VHPAM–Vk13B8 and
highlighted a remarkable correspondence between the VHPAM–Vl16F4 to PDGFRa was not significantly
predicted (in silico) and the actual (in vitro) PDGFRa inhibited by PDGF-BB (Figures 2C and D). Moreover,
epitopes bound by the recombinant human mAb and by VHPAM–Vk13B8 and VHPAM–Vl16F4 showed partial
PDGF-BB (Figures 2A–C), except VHPAM–Vl16F4, reciprocal binding inhibition (Figures 2C and D), con-
which did not bind to any peptide, most likely sistent with the minimal overlap between their epitopes.
because of an incomplete representation of its con- We therefore conclude that there are 3 distinct
formational epitope in the library. Notably, the high- PDGFRa epitopes recognized by these antibodies. One
est binding peaks of PDGF-BB and VHPAM–Vk16F4 epitope, largely shared by PDGF-BB and by the ROS-
occurred with looped peptides (results available upon and collagen-inducing VHPAM–Vk16F4, is conforma-
request from the corresponding author), corrobo- tional and discontinuous. The second epitope, bound by
rating the conformational nature of their PDGFRa VHPAM–Vl16F4 and inducing ROS but not collagen, is
epitopes. also conformational and discontinuous. The third epi-
To further validate the PDGFRa epitope map- tope, bound by VHPAM–Vk13B8, is an apparently non-
ping, binding-competition experiments were performed functional single peptide in the first extracellular
on a surface plasmon resonance device, in which the domain of PDGFRa.
PDGFRa-His, immobilized and folded into a native-like Interference of specific PDGFRa peptides with
conformation (25), was saturated with PDGF-BB or a the binding and functions of VHPAM–Vk16F4 recombi-
single recombinant human mAb before each recombi- nant human mAb and serum IgG from SSc patients.
nant human mAb was added. The resulting sensorgrams The 3 peptides corresponding to the PDGFRa sequen-
(Figures 2B–D, insets) showed that binding of VHPAM– ces (Figure 2B), composing the discontinuous epitope
Vk16F4 to PDGFRa was completely prevented by of VHPAM–Vk16F4, bound VHPAM–Vk16F4, but
PDGF-BB, but only partially prevented by VHPAM– not VHPAM–Vl16F4 or VHPAM–Vk13B8 (Figure 3A).
Vk13B8 (Figure 2B), likely because of steric hindrance. Preincubation with any of these peptides significantly
Saturation with VHPAM–Vl16F4 only slightly inhibited inhibited VHPAM–Vk16F4 binding to PDGFRa
EPITOPE SPECIFICITY OF ANTI-PDGFRa AUTOANTIBODIES IN SSc 1897

Figure 4. Mutagenesis of the discontinuous epitope of the agonistic VHPAM–Vk16F4 recombinant human mAb. A, Visualization of the
PDGFRa conformational epitope of VHPAM–Vk16F4. Epitope surface amino acid residues composing the VHPAM–Vk16F4 binding site, as
predicted by molecular docking, are highlighted (rendered as light blue sticks). The corresponding translucent Van der Waals surface is superim-
posed to depict the VHPAM–Vk16F4 binding pocket. Secondary structures of PDGFRa are displayed using the ribbon representation. The dif-
ferent sites of alanine mutations (Ala-scans) are emphasized by the boxed areas. B, Fluorescence-activated cell sorting analysis of cell surface
PDGFRa expression by Fa mouse cells or those with alanine mutations (Ala-scans) of the VHPAM–Vk16F4 epitope, stained with VHPAM–
Vk16F4 or mAb 1264. F2/2 mouse cells were used as a negative control. C, Human COL1A1 gene induction measured by real-time quantitative
polymerase chain reaction in Fa, F2/2, and Ala-scan–expressing cells exposed to VHPAM–Vk16F4 for 1 hour. Values are the mean 6 SEM
relative fold increase in expression, calculated by the 22DDCt method, in 3 experiments performed in duplicate, normalized to the mean values of
endogenous cyclophilin A. * 5 P , 0.05 versus baseline. FITC 5 fluorescein isothiocyanate; PE 5 phycoerythrin (see Figure 1 for other
definitions).

(Figure 3B) and VHPAM–Vk16F4-mediated collagen Mutagenesis of the discontinuous epitope of the
gene induction (Figure 3C). Peptide 2 was the most agonistic recombinant human mAb VHPAM–Vk16F4.
effective inhibitor under these experimental conditions, A limited alanine-scanning mutagenesis of the essential
as it also significantly reduced PDGF-BB binding motifs composing the VHPAM–Vk16F4 epitope (Figures
(Figure 3B) and collagen gene induction and ROS 2B and 4A) was performed. Binding to VHPAM–Vk16F4
production triggered by SSc IgG (Figure 3D). Peptide 1, was retained by Ala-scans 1, 2, and 3 (Figure 4B); how-
but not peptide 3, inhibited SSc IgG–induced ROS ever, COL1A1 gene stimulation by VHPAM–Vk16F4
production (Figure 3D). The scrambled peptide 2 was observed with Ala-scans 1 and 3 only, whereas it was
did not bind to VHPAM–Vk16F4, PDGF-BB, or SSc completely impaired by Ala-scan 2 (Figure 4C). This
IgG, nor did it inhibit their agonistic effects (Figures result is consistent with the finding that peptide 2 is the
3A–D). most effective inhibitor of VHPAM–Vk16F4-induced
1898 MORONCINI ET AL

Figure 5. Detection of serum anti–platelet-derived growth factor receptor a (anti-PDGFRa) autoantibodies. A, Left, Direct enzyme-linked immu-
nosorbent assay (ELISA) of anti-PDGFRa autoantibodies in serum from patients with systemic sclerosis (SSc), healthy control (HC) subjects,
and patients with primary Raynaud’s phenomenon (PRP). The optimal cutoff value for PDGFRa binding between positive and negative sera is
indicated by the broken line. Right, The optimal cutoff for binding was determined by receiver operating characteristic (ROC) curve analyses of
the area under the ROC curve (AUC) for SSc patients versus healthy controls (sensitivity 94.4%, specificity 51.5%, AUC 0.801 [95% confidence
interval 0.793–0.854]). B, Left, Inhibition of binding of SSc serum to PDGFRa by peptide 2. The optimal cutoff value for discriminating sera
whose binding to PDGFRa is significantly inhibited by peptide 2 is indicated by the broken line. Right, The optimal cutoff for inhibition was
determined by ROC curve analyses (sensitivity 70.6%, specificity 88.2%, AUC 0.764 [95% confidence interval 0.645–0.858]). C, Left, Binding
inhibition of VHPAM–Vk16F4 to PDGFRa by sera. The optimal cutoff value for discriminating sera inhibiting VHPAM–Vk16F4 binding to
PDGFRa from those weakly or not providing inhibition is indicated by the broken line. Right, The optimal cutoff for inhibition was determined by
ROC curve analyses (sensitivity 70.6%, specificity 94.1%, AUC 0.760 [95% confidence interval 0.641–0.856]). D, Direct ELISA or competitive
ELISAs (with peptide 2 or scrambled peptide 2 control) of SLE serum. Each dot represents the mean optical density of each sample from triplicate
experiments. Symbols indicate individual samples; horizontal line indicates the median. ** 5 P , 0.001; *** 5 P , 0.0001, versus SSc serum.

collagen stimulation (Figure 3C) and indicates that the patients with a definite diagnosis of SSc (8) (details on
TFT amino acid motif is essential for collagen induction. the patients’ clinical data are available upon request
Expression of Ala-scan 4, spanning motifs 1 and from the corresponding author), from 130 sex- and age-
2, or Ala-scan 5, spanning motifs 1, 2, and 3 (Figure matched healthy control subjects, and from 26 consecu-
4A), impaired both VHPAM–Vk16F4 binding and col- tive patients with a definite diagnosis of primary Ray-
lagen gene stimulation (Figures 4B and C). These naud’s phenomenon (RP) (30). Based on the optimal
results independently validate the findings from OD cutoff value for autoantibody positivity, as deter-
PDGFRa epitope mapping, and highlight the minimal mined by ROC curve analysis of SSc and healthy control
extracellular PDGFRa motif required for collagen serum, 94.3% of SSc patients (66 of 70 patients), 48.5%
gene stimulation. of healthy controls (63 of 130 subjects), and 42.3% of
Detection of anti-PDGFRa autoantibodies in patients with primary RP (11 of 26 patients) were posi-
the serum of SSc patients. Three independent assays tive for anti-PDGFRa autoantibodies (P , 0.0001
were established to detect anti-PDGFRa autoantibodies between SSc patients and the other groups) (Figure
in the serum of SSc patients. The first was a direct 5A). Similar results were obtained using a commercial
ELISA with PDGFRa-His immobilized onto 96-well PDGFRa-His (results available upon request from the
plates. We tested serum samples from 70 consecutive corresponding author), thus ruling out the possibility of
EPITOPE SPECIFICITY OF ANTI-PDGFRa AUTOANTIBODIES IN SSc 1899

Table 1. Comparison of findings between the ELISAs and ROS assay in serum samples from SSc
patients and healthy control subjects*
ELISAs ROS assay, SI
Inhibition of serum
Serum sample Serum binding, OD binding by peptide 2, % IgG-induced ROS ROS 1 AG1296

SSc (n 5 28)
1 0.583 51.1 1.03 –
2 0.456 49.8 0.40 –
3 0.382 46.1 0.73 –
4 0.333 44.4 0.59 –
5 0.281 29.9 1.15 –
6 0.269 35.3 1.15 –
7 0.261 49.3 0.74 –
8 0.259 34.1 0.50 –
9 0.229 30.6 1.13 –
10 0.194 34.2 1.60 –
11 0.178 34.5 0.87 –
12 0.171 53.1 1.21 –
13 0.165 43.2 1.42 –
14 0.146 36.8 1.68 –
15 0.141 39.2 1.23 –
16 0.129 32.9 1.23 –
17 0.179 4.8 1.51 0.17
18 0.175 10.1 1.80 0.00
19 0.169 29.5 1.51 0.00
20 0.155 216.2 1.31 0.10
21 0.151 12.2 0.81 0.00
22 0.143 21.6 0.85 0.18
23 0.186 13.8 0.53 0.36
24 0.170 11.6 0.00 –
25 0.114 – 0.11 –
26 0.100 – 0.08 –
27 0.084 – 0.00 –
28 0.080 – 0.28 –
Healthy controls (n 5 26)
1 0.000 – 0.07 –
2 0.091 – 0.15 –
3 0.220 – 0.00 –
4 0.227 – 0.00 –
5 0.164 – 0.00 –
6 0.210 – 0.00 –
7 0.178 – 0.00 –
8 0.201 – 0.00 –
9 0.140 – 0.00 –
10 0.129 – 0.00 –
11 0.266 – 0.00 –
12 0.130 – 0.00 –
13 0.446 – 0.00 –
14 0.225 – 0.00 –
15 0.266 – 0.04 –
16 0.350 – 0.10 –
17 0.139 – 0.13 –
18 0.164 – 0.17 –
19 0.168 – 0.27 –
20 0.147 – 0.29 –
21 0.266 – 0.30 –
22 0.190 – 0.33 –
23 0.191 – 0.34 –
24 0.174 – 0.40 –
25 0.190 – 0.61 0.63
26 0.304 – 0.72 0.80

* Human IgG (200 mg/ml) was purified from the serum of 24 patients with systemic sclerosis (SSc) tested for
binding to platelet-derived growth factor receptor a (PDGFRa) on both direct and competitive enzyme-
linked immunosorbent assays (ELISAs), the serum of 4 SSc patients testing negative for anti-PDGFRa auto-
antibodies on direct ELISA, and the serum of 26 healthy control subjects, of which 24 were positive and 2
were negative for anti-PDGFRa on direct ELISA. All IgG samples were tested for their ability to induce pro-
duction of reactive oxygen species (ROS) in mouse embryonic fibroblasts transfected with full-length human
PDGFRa. Except where indicated in boldface, values were above the cutoff for positivity. In direct ELISA,
the cutoff for binding was an OD value of 0.129. In competitive ELISA, the cutoff for inhibition was 29.8%.
In the ROS assay, the cutoff for positivity was a stimulation index (SI) of 0.4, calculated as SI 5 (S 2 C) 4 (P
2 C), where S is the dihydrodichlorofluorescein diacetate (DCFH-DA) fluorescence intensity of the test IgG,
C is the DCFH-DA fluorescence intensity of a negative control obtained by culturing cells without IgG, and P
is the DCFH-DA fluorescence intensity of a positive control obtained by incubating cells with PDGF-BB (15
ng/ml for 15 minutes). Samples able to induce ROS were tested in the presence of the PDGFRa inhibitor
AG1296 (2 mM).
1900 MORONCINI ET AL

any artificial binding of serum IgG to the PDGFRa-His ses indicated that in the 2 competitive ELISAs with the
that was produced in-house. None of the serum samples SSc serum (P , 0.0001, r 5 0.678), but not those with
showed nonspecific binding to uncoated plates (results the healthy control serum (P 5 0.403, r 5 20.147) or
not shown). primary RP serum (P 5 0.668, r 5 20.108), the results
The second assay was a competitive ELISA were statistically significantly associated.
between immobilized PDGFRa-His and peptides cover- Finally, direct and competitive ELISAs were per-
ing the VHPAM–Vk16F4 epitope. To evaluate the pres- formed with serum samples from 29 consecutive patients
ence of serum anti-PDGFRa IgG recognizing the same with SLE (SLE Disease Activity Index range 2–20). The
epitope of the agonistic recombinant human mAb distribution of PDGFRa binding in SLE serum was
VHPAM–Vk16F4, sera from patients with SSc and comparable to that in healthy control serum, as deter-
healthy controls (each n 5 34) and from patients with mined by direct ELISA with PDGFRa-His. Moreover,
primary RP (n 5 18), chosen on the basis of homogene- this was not influenced by the extent of disease activity or
ous frequency distribution of OD values (as determined peptide 2 competition (Figure 5D).
in the first assay), were incubated with peptide 2 (the Comparison between ELISA and ROS assays. IgG
most effective inhibitor of the stimulatory effects of purified from the serum of 24 of 34 SSc patients ana-
VHPAM–Vk16F4 and SSc IgG [Figures 3B–D]) before lyzed for PDGFRa binding by the direct and competi-
binding to immobilized PDGFRa. Based on the optimal tive ELISAs (Figures 5A and B) and from 4 SSc serum
cutoff value for inhibition determined on the ROC samples found negative for anti-PDGFRa on the direct
curve, preincubation with peptide 2 significantly inhib- ELISA (Figure 5A) were tested by ROS assay (3)
ited PDGFRa binding of 70.6% of SSc serum samples (Table 1). All sera found positive for anti-PDGFRa
(24 of 34 patients) compared with 11.7% of healthy con- autoantibodies by direct ELISA, including 16 samples
trol serum samples (4 of 34 subjects; P , 0.001) and found positive and 8 found negative by the competitive
none of the primary RP serum samples (P , 0.0001) ELISA, contained ROS-inducing IgG, except SSc serum
(Figure 5B). Similar results were obtained when the sample 24, which was negative on the competitive
assay was repeated with peptide 1, whereas peptide 3 ELISA. Conversely, the 4 sera (samples 25–28) found
was less efficient, inhibiting binding of 50% of SSc sera negative on the direct ELISA did not contain ROS-
(17 of 34 patients), 8.8% of healthy control sera (3 of 34 inducing IgG.
subjects), and none of the primary RP sera (results The ROS-inducing activity of the 7 samples neg-
available upon request from the corresponding author). ative for anti-PDGFRa on the competitive ELISA (i.e.,
To confirm that inhibition was due to specific not containing antibodies with the same specificity of
binding of peptide 2 to serum IgG, the assay was VHPAM–Vk16F4) was abolished by the PDGFRa tyro-
repeated using IgG purified from 8 of 24 autoantibody- sine kinase inhibitor AG1296, further suggesting that
positive SSc serum samples and from all 4 autoantibody- VHPAM–Vk16F4-like IgG is not the only agonistic anti-
positive healthy control serum samples. While binding of PDGFRa autoantibody species. Healthy control serum
SSc IgG was inhibited by peptide 2, binding of IgG from positive for IgG binding to PDGFRa on the direct
healthy control serum was no longer inhibited (results ELISA did not induce ROS, except samples 25 and 26,
available upon request from the corresponding author). which induced ROS through a PDGFR-independent
These findings suggest that there are serum components mechanism, since ROS production was unaffected by the
mediating the binding of healthy control serum to PDGFRa inhibitor AG1296 (Table 1).
peptide 2.
The third assay was a competitive ELISA
DISCUSSION
between serum and VHPAM–Vk16F4. To confirm that
SSc sera contain autoantibodies similar to VHPAM– Human PDGFRa has been implicated in a variety
Vk16F4, the same SSc patient serum samples as tested of diseases, including cancer, atherosclerosis, and fibrosis
in the second assay were incubated onto immobilized (31–33); however, the PDGFRa domains critical for dis-
PDGFRa-His before biotinylated VHPAM–Vk16F4 was ease pathogenesis are not completely defined. In vitro
added. The 24 of 34 SSc serum samples previously mutagenesis (26) and crystallographic studies (34) have
shown to be inhibited by peptide 2 significantly dimin- indicated that the amino acid residues involved in recep-
ished VHPAM–Vk16F4 binding to PDGFRa. Only 2 tor binding by PDGF are located in the Ig-like extracellu-
(5.9%) of 34 healthy control serum samples and 2 lar domains 1, 2, and 3. The recombinant human mAb
(11.1%) of 18 primary RP serum samples inhibited generated from PDGFRa-specific memory B cells ob-
binding (Figure 5C). Spearman’s rank correlation analy- tained from 1 patient with SSc extend this information by
EPITOPE SPECIFICITY OF ANTI-PDGFRa AUTOANTIBODIES IN SSc 1901

identifying regions of the receptor that transduce signals ings, and indicate that complete discrimination between
to NADPH oxidase (35,36) and to collagen genes (37). nonagonistic and agonistic autoantibodies can be
These regions can either largely overlap the canonical achieved only by a combination of epitope-based bind-
PDGF binding site (epitope of VHPAM–Vk16F4) or be ing assays and functional assays. Of note, IgG purified
distinct from it (epitope of VHPAM–Vl16F4). The find- from the serum of 2 healthy control subjects and 2
ings suggest that these different PDGFRa motifs, when patients with primary RP that inhibited binding of bio-
stimulated, modulate local and/or long-distance propaga- tinylated VHPAM–Vk16F4 to PDGFRa did not induce
tion of signals of the activated receptor that selectively ROS (results available upon request from the corre-
govern complex phenotypes, such as fibrosis and oxida- sponding author).
tive stress (38). The assessment of the diagnostic relevance of
This study reconciles the controversial results stimulatory anti-PDGFRa antibodies, i.e., their efficacy
concerning the existence of stimulatory anti-PDGFR in discriminating patients affected by early-stage SSc
antibodies in SSc (4,5). First, our findings demonstrate from patients affected by related conditions such as pri-
that stimulatory and nonstimulatory anti-PDGFRa mary RP, will require the prospective screening of large
autoantibodies may coexist in the same SSc patient. Sec- cohorts of patients suspected of having SSc (39), whose
ond, stimulatory anti-PDGFRa autoantibodies recog- serum should be assessed by optimized peptide-based
nize specific conformational epitopes, which must be assays. Moreover, the longitudinal analysis of agonistic
preserved in binding assays to discriminate agonistic anti-PDGFRa antibodies during disease progression
from total antibodies, and can be mimicked by specific will indicate their potential usefulness as a biomarker of
peptides. Third, stimulatory anti-PDGFRa autoanti- SSc (40). Notably, the 24 patients with SSc whose serum
bodies specific for these conformational epitopes are tested positive for anti-PDGFRa on both competitive
enriched in the serum of SSc patients. Moreover, the ELISAs had a more severe disease phenotype compared
comparison between the ROS bioassay (3) and the with the 10 SSc patients whose serum tested negative,
peptide-based competitive ELISA described herein, but this observation must be confirmed in larger cohorts
both performed with the same SSc and healthy control of patients.
sera used in this study (although performed with a lim- Finally, recent reports suggest that PDGFR inhibi-
ited number of samples), indicated a remarkable tion may be a therapeutic option for SSc and other fibrotic
concordance. conditions (41). Abrogation of VHPAM–Vk16F4-induced
The discordant results, i.e., the 7 SSc serum sam- collagen gene overexpression by selective mutation of the
ples testing negative by competitive ELISA but positive minimal epitope motif, and inhibition of SSc IgG–induced
by ROS bioassay, can also be meaningfully interpreted. ROS production and collagen overexpression by use of
Indeed, these samples became unable to induce ROS in peptides mimicking the epitope itself, is further corrobo-
the presence of a specific PDGFR tyrosine kinase inhib- rating evidence to support the possibility of PDGFR inhi-
itor, which demonstrates that they contain agonistic bition as a promising new therapeutic strategy against
PDGFRa-specific antibodies. These antibodies differ SSc and other pathologic conditions characterized by oxi-
from VHPAM–Vk16F4, since they were not inhibited by dative stress, fibrosis, and abnormal PDGFR signaling.
peptide 2. It is tempting to speculate that they are simi-
lar to VHPAM–Vl16F4, the other agonistic antibody ACKNOWLEDGMENTS
cloned from the PAM B cells.
We thank Dennis Burton, Ann Hessel, and Michael
The presence of different agonistic PDGFRa- Zwick (Department of Immunology, The Scripps Research
specific antibodies, binding distinct epitopes, may Institute, La Jolla, CA) for providing the pDR12 vector, Stefania
account for the lower sensitivity of the competitive Mancini (Dipartimento di Scienze Cliniche e Molecolari, Uni-
ELISA, which was based on one epitope, as compared versita Politecnica delle Marche, Ancona, Italy) for help with
to the ROS bioassay. Notably, the combined analysis of the fluorescence-activated cell sorting analysis, Hetian Lei (The
Schepens Eye Research Institute, Harvard Medical School,
antibody binding and ROS production highlighted the Boston, MA) for providing the retroviral vector pLHDCX2
existence of a subgroup of SSc patients in whom stimu- containing the full-length human PDGFRa cDNA, Patrizia
latory anti-PDGFRa IgG was absent. This group was Bagnarelli, Stefano Menzo, and Pietro Emanuele Varaldo (Dipar-
not previously identified in the small SSc cohort in timento di Scienze Biomediche e Sanita Pubblica, Universita
whom only ROS activity was measured (3). Nonstimula- Politecnica delle Marche, Ancona, Italy) for providing the BSL3
facility, Milena Maule and Erika Ortolan (Dipartimento di
tory anti-PDGFRa antibodies have been identified by Scienze Mediche, Universita di Torino, Turin, Italy) for help with
other investigators in studies of SSc and SLE patients statistical analysis of the ELISA data, and Eline Luning Prak
and healthy controls (5,6). Our data confirm these find- (Human Immunology Core Laboratory, Perelman School of
1902 MORONCINI ET AL

Medicine, University of Pennsylvania, Philadelphia) for critical 15. Moroncini G, Kanu N, Solforosi L, Abalos G, Telling GC, Head
reading of the manuscript. M, et al. Motif-grafted antibodies containing the replicative
interface of cellular PrP are specific for PrPSc. Proc Natl Acad
Sci U S A 2004;101:10404–9.
16. Svegliati S, Cancello R, Sambo P, Luchetti M, Paroncini P,
AUTHOR CONTRIBUTIONS Orlandini G, et al. Platelet-derived growth factor and reactive
oxygen species (ROS) regulate Ras protein levels in primary
All authors were involved in drafting the article or revising it human fibroblasts via ERK1/2: amplification of ROS and Ras
critically for important intellectual content, and all authors approved in systemic sclerosis fibroblasts. J Biol Chem 2005;280:36474–
the final version to be published. Drs. Moroncini and Gabrielli had 82.
full access to all of the data in the study and take responsibility for the 17. Schwede T, Kopp J, Guex N, Peitsch MC. SWISS-MODEL: an
integrity of the data and the accuracy of the data analysis. automated protein homology-modeling server. Nucleic Acids
Study conception and design. Moroncini, Funaro, Gabrielli. Res 2003;31:3381–5.
Acquisition of data. Grieco, Nacci, Paolini, Tonnini, Pozniak, Cuccioloni, 18. Oefner C, D’Arcy A, Winkler FK, Eggimann B, Hosang M.
Mozzicafreddo, Svegliati, Kazlauskas. Crystal structure of human platelet-derived growth factor BB.
Analysis and interpretation of data. Moroncini, Angeletti, Avvedimento, EMBO J 1992;11:3921–6.
Funaro, Gabrielli. 19. Comeau SR, Gatchell DW, Vajda S, Camacho CJ. ClusPro: an
automated docking and discrimination method for the prediction
of protein complexes. Bioinformatics 2004;20:45–50.
REFERENCES 20. Timmerman P, Puijk WC, Meloen RH. Functional reconstruc-
1. Gabrielli A, Avvedimento EV, Krieg T. Scleroderma. N Engl J tion and synthetic mimicry of a conformational epitope using
Med 2009;360:1989–2003. CLIPS technology. J Mol Recognit 2007;20:283–99.
2. Varga J, Abraham D. Systemic sclerosis: a prototypic multisys- 21. Timmerman P, Meloen RH, Puijk WC, Boshuizen RS, van
tem fibrotic disorder. J Clin Invest 2007;117:557–67. Dijken P, Slootstra JW, et al. Functional reconstruction of struc-
3. Baroni SS, Santillo M, Bevilacqua F, Luchetti M, Spadoni T, turally complex epitopes using CLIPSTM technology. Open Vac-
Mancini M, et al. Stimulatory autoantibodies to the PDGF cine J 2009;2:56–67.
receptor in systemic sclerosis. N Engl J Med 2006;354:2667–76. 22. Cui J, Lei H, Samad A, Basavanthappa S, Maberley D,
4. Classen JF, Henrohn D, Rorsman F, Lennartsson J, Lauwerys Matsubara J, et al. PDGF receptors are activated in human epi-
BR, Wikstrom G, et al. Lack of evidence of stimulatory auto- retinal membranes. Exp Eye Res 2009;88:438–44.
antibodies to platelet-derived growth factor receptor in patients 23. Miller AD, Buttimore C. Redesign of retrovirus packaging cell
with systemic sclerosis. Arthritis Rheum 2009;60:1137–44. lines to avoid recombination leading to helper virus production.
5. Loizos N, LaRiccia L, Weiner J, Griffith H, Boin F, Hummers L, Mol Cell Biol 1986;6:2895–902.
et al. Lack of detection of agonist activity by antibodies to platelet- 24. Lei H, Velez G, Hovland P, Hirose T, Gilbertson D, Kazlauskas
derived growth factor receptor a in a subset of normal and sys- A. Growth factors outside the PDGF family drive experimental
temic sclerosis patient sera. Arthritis Rheum 2009;60:1145–51. PVR. Invest Ophthalmol Vis Sci 2009;50:3394–403.
6. Kurasawa K, Arai S, Owada T, Maezawa R, Kumano K, Fukuda 25. Cuccioloni M, Moroncini G, Mozzicafreddo M, Pozniak KN,
T. Autoantibodies against platelet-derived growth factor receptor Nacci G, Grieco A, et al. Biosensor-based binding assay for
a in patients with systemic lupus erythematosus. Mod Rheuma- platelet-derived growth factor receptor-a autoantibodies in
tol 2010;20:458–65. human serum. J Anal Bioanal Tech 2013;S7:010. URL: http://
7. Dragun D, Distler JH, Riemekasten G, Distler O. Stimulatory omicsonline.org/biosensor-based-binding-assay-for-platelet-derived-
autoantibodies to platelet-derived growth factor receptors in sys- growth-factor-receptor-alpha-autoantibodies-in-human-serum-2155-
temic sclerosis: what functional autoimmunity could learn from 9872.S7-010.pdf.
receptor biology [editorial]. Arthritis Rheum 2009;60:907–11. 26. Yu JC, Mahadevan D, LaRochelle WJ, Pierce JH, Heidaran
8. LeRoy EC, Black C, Fleischmajer R, Jablonska S, Krieg T, MA. Structural coincidence of aPDGFR epitopes binding to
Medsger TA Jr, et al. Scleroderma (systemic sclerosis): classifi- platelet-derived growth factor-AA and a potent neutralizing
cation, subsets and pathogenesis. J Rheumatol 1988;15:202–5. monoclonal antibody. J Biol Chem 1994;269:10668–74.
9. Traggiai E, Becker S, Subbarao K, Kolesnikova L, Uematsu Y, 27. Lokker NA, O’Hare JP, Barsoumian A, Tomlinson JE,
Gismondo MR, et al. An efficient method to make human Ramakrishnan V, Fretto LJ, et al. Functional importance of
monoclonal antibodies from memory B cells: potent neutraliza- platelet-derived growth factor (PDGF) receptor extracellular
tion of SARS coronavirus. Nat Med 2004;10:871–5. immunoglobulin-like domains: identification of PDGF binding
10. Funaro A, Gribaudo G, Luganini A, Ortolan E, Lo Buono N, site and neutralizing monoclonal antibodies. J Biol Chem 1997;
Vicenzi E, et al. Generation of potent neutralizing human mono- 272:33037–44.
clonal antibodies against cytomegalovirus infection from immune 28. Corti D, Voss J, Gamblin SJ, Codoni G, Macagno A, Jarrossay D,
B cells. BMC Biotechnol 2008;8:85. et al. A neutralizing antibody selected from plasma cells that binds
11. Shannon-Lowe C, Baldwin G, Feederle R, Bell A, Rickinson A, to group 1 and group 2 influenza A hemagglutinins. Science 2011;
Delecluse HJ. Epstein-Barr virus-induced B-cell transformation: 333:850–6.
quantitating events from virus binding to cell outgrowth. J Gen 29. Tauriello DV, Jordens I, Kirchner K, Slootstra JW, Kruitwagen
Virol 2005;86:3009–19. T, Bouwman BA, et al. Wnt/b-catenin signaling requires interac-
12. Andrews A, Balciunaite E, Leong FL, Tallquist M, Soriano P, tion of the Dishevelled DEP domain and C terminus with a dis-
Refojo M, et al. Platelet-derived growth factor plays a key role continuous motif in Frizzled. Proc Natl Acad Sci U S A 2012;
in proliferative vitreoretinopathy. Invest Ophthalmol Vis Sci 109:E812–20.
1999;40:2683–9. 30. Goundry B, Bell L, Langtree M, Moorthy A. Diagnosis and
13. Welschof M, Terness P, Kolbinger F, Zewe M, Dubel S, Dorsam management of Raynaud’s phenomenon. BMJ 2012;344:e289.
H, et al. Amino acid sequence based PCR primers for amplifica- 31. Andrae J, Gallini R, Betsholtz C. Role of platelet-derived
tion of rearranged human heavy and light chain immunoglobulin growth factors in physiology and medicine. Genes Dev 2008;22:
variable region genes. J Immunol Methods 1995;179:203–14. 1276–312.
14. Burton DR, Pyati J, Koduri R, Sharp SJ, Thornton GB, Parren PW, 32. Ostman A. PDGF receptors: mediators of autocrine tumor
et al. Efficient neutralization of primary isolates of HIV-1 by a growth and regulators of tumor vasculature and stroma. Cyto-
recombinant human monoclonal antibody. Science 1994;266:1024–7. kine Growth Factor Rev 2004;15:275–86.
EPITOPE SPECIFICITY OF ANTI-PDGFRa AUTOANTIBODIES IN SSc 1903

33. Olson LE, Soriano P. Increased PDGFRa activation disrupts 38. Gabrielli A, Svegliati S, Moroncini G, Amico D. New insights
connective tissue development and drives systemic fibrosis. Dev into the role of oxidative stress in scleroderma fibrosis. Open
Cell 2009;16:303–13. Rheumatol J 2012;6:87–95.
34. Shim AH, Liu H, Focia PJ, Chen X, Lin PC, He X. Structures 39. Avouac J, Fransen J, Walker UA, Riccieri V, Smith V, Muller
of a platelet-derived growth factor/propeptide complex and a C, et al, EUSTAR Group. Preliminary criteria for the very early
platelet-derived growth factor/receptor complex. Proc Natl Acad diagnosis of systemic sclerosis: results of a Delphi Consensus
Sci U S A 2010;107:11307–12. Study from EULAR Scleroderma Trials and Research Group.
35. Heldin CH, Westermark B. Mechanism of action and in vivo Ann Rheum Dis 2011;70:476–81.
role of platelet-derived growth factor. Physiol Rev 1999;79: 40. Koenig M, Dieude M, Senecal JL. Predictive value of antinu-
1283–316. clear autoantibodies: the lessons of the systemic sclerosis auto-
36. Schlessinger J. Cell signaling by receptor tyrosine kinases. Cell antibodies. Autoimmun Rev 2008;7:588–93.
2000;103:211–25. 41. Bournia VK, Evangelou K, Sfikakis PP. Therapeutic inhibition
37. Smaldone S, Olivieri J, Gusella GL, Moroncini G, Gabrielli A, of tyrosine kinases in systemic sclerosis: a review of published
Ramirez F. Ha-Ras stabilization mediates pro-fibrotic signals in experience on the first 108 patients treated with imatinib. Semin
dermal fibroblasts. Fibrogenesis Tissue Repair 2011;4:8. Arthritis Rheum 2012;42:377–90.

You might also like