Skip to main content

Identification of target antigens of anti-endothelial cell and anti-vascular smooth muscle cell antibodies in patients with giant cell arteritis: a proteomic approach

Abstract

Introduction

Immunological studies of giant cell arteritis (GCA) suggest that a triggering antigen of unknown nature could generate a specific immune response. We thus decided to detect autoantibodies directed against endothelial cells (ECs) and vascular smooth muscle cells (VSMCs) in the serum of GCA patients and to identify their target antigens.

Methods

Sera from 15 GCA patients were tested in 5 pools of 3 patients' sera and compared to a sera pool from 12 healthy controls (HCs). Serum immunoglobulin G (IgG) reactivity was analysed by 2-D electrophoresis and immunoblotting with antigens from human umbilical vein ECs (HUVECs) and mammary artery VSMCs. Target antigens were identified by mass spectrometry.

Results

Serum IgG from GCA patients recognised 162 ± 3 (mean ± SD) and 100 ± 17 (mean ± SD) protein spots from HUVECs and VSMCs, respectively, and that from HCs recognised 79 and 94 protein spots, respectively. In total, 30 spots from HUVECs and 19 from VSMCs were recognised by at least two-thirds and three-fifths, respectively, of the pools of sera from GCA patients and not by sera from HCs. Among identified proteins, we found vinculin, lamin A/C, voltage-dependent anion-selective channel protein 2, annexin V and other proteins involved in cell energy metabolism and key cellular pathways. Ingenuity pathway analysis revealed that most identified target antigens interacted with growth factor receptor-bound protein 2.

Conclusions

IgG antibodies to proteins in the proteome of ECs and VSMCs are present in the sera of GCA patients and recognise cellular targets that play key roles in cell biology and maintenance of homeostasis. Their potential pathogenic role remains to be determined.

Introduction

Giant cell arteritis (GCA), also known as temporal arteritis, is a primary systemic vasculitis involving large- and medium-sized vessels. GCA commonly causes bitemporal headaches, jaw claudication, scalp tenderness and/or abnormal temporal arteries (tender, nodular, swollen and thickened arteries with decreased pulses) detected during physical examinations. GCA does not occur in people younger than 50 years old, and its incidence increases with age and peaks in Caucasians older than 70 years of age [1, 2]. Ocular ischaemic complications occur in 25% of the patients and leads to irreversible visual loss in 15% [3]. No definite immunological marker has been identified in GCA, and patients usually present with increased erythrocyte sedimentation rates and/or C-reactive protein levels. Diagnosing GCA can be difficult, and temporal artery biopsy is the gold standard for making the diagnosis [4]. However, in 10% to 20% of patients with GCA, the biopsy shows no specific change [5].

GCA is an inflammatory condition of unknown origin characterised by the presence of giant cells and a remodelling process in the arterial wall [6]. In patients with GCA, an immune-mediated reaction is suspected to be triggered by an antigen of unknown origin, either microbial or a self-antigen, that could be presented to T cells by dendritic cells [7]. Thus, macrophages and giant cells stimulated by interferon-γ (IFN-γ) play a major role in the disruption of the elastic lamina and the remodelling of vessel walls. In addition, in the adventitia, macrophages produce proinflammatory cytokines such as interleukin 1 (IL-1) and IL-6, whereas in the media and intima they contribute to arterial injury by producing metalloproteinases and nitric oxide [6, 8, 9].

Anti-endothelial cell (anti-EC) antibodies (AECAs) have been detected in a wide range of systemic inflammatory and/or autoimmune diseases, including primary and/or secondary systemic vasculitis [10]. Although the pathogenic role of AECAs remains controversial [11, 12], these antibodies may be responsible for EC activation [13] and induction of antibody-dependent, cell-mediated cytotoxicity and apoptosis [14]. In GCA, AECAs were detected in 33% of sera by performing ELISA on fixed human umbilical vein ECs (HUVECs) [15], but their presence was not confirmed by indirect immunofluorescence [16]. Anti-vascular smooth muscle cell (anti-VSMC) antibodies have been detected in an experimental rat model of vasculitis [17]; however, to our knowledge, these antibodies have not been investigated in patients with primary systemic vasculitis.

We used 1-D and 2-D immunoblotting, followed by mass spectrometry (MS), to investigate the presence of autoantibodies directed against ECs and VSMCs and identify their target antigens in patients with GCA.

Materials and methods

Patients

Serum samples were obtained from 15 patients who fulfilled the American College of Rheumatology (ACR) criteria for GCA [4] and 33 patients with anti-neutrophil cytoplasm antibody (ANCA)-associated vasculitis who fulfilled the ACR and the Chapel Hill criteria used as vasculitis controls, with the control group comprising 15 patients with Wegener's granulomatosis (WG), 9 with Churg-Strauss syndrome (CSS) and 9 with microscopic polyangiitis (MPA) [18]. In each group of patients with ANCA-associated vasculitis, two-thirds of the patients had active disease as assessed by a Birmingham Vasculitis Activity Score (BVAS) >3 in the absence of treatment, and one-third of the patients had inactive disease as assessed by a BVAS <3. Some patients in both groups either received corticosteroids and/or immunsuppressants at the time of blood sampling. Sera from 12 healthy blood donors were used as healthy controls (HCs). Serum samples were collected from patients and HCs, aliquoted and stored at -80°C until use. Serum samples were used individually for 1-D immunoblotting and pooled for 2-D immunoblotting (five pools of sera from three patients with GCA each, and one pool of sera from twelve HCs). All patients and healthy controls gave their written informed consent to participate in the study. Serum samples were collected with the approval of the ethics committee of the groupe hospitalier Pitié-Salpêtrière, and the study conformed to the principles outlined in the Declaration of Helsinki.

Cell culture

Human internal mammary artery VSMCs were obtained from patients undergoing aortocoronary bypass surgery. All patients gave their written consent, and the protocol for waste surgical tissue was approved by the ethics committee of groupe hospitalier Pitié-Salpêtrière. These cells were immortalised by transduction of a lentiviral vector incorporating the catalytic subunit of the human holoenzyme telomerase RT and T antigen of simian virus 40 in a primary culture of VSMCs as previously described [19]. Immortalised VSMCs were cultured in Smooth Muscle Cell Basal Medium (PromoCell, Heidelberg, Germany) supplemented with decomplemented FCS (5%), insulin (5 μg/mL), basic fibroblast growth factor (bFGF) (2 ng/mL), epidermal growth factor (EGF) (0.5 ng/ml), streptomycin/penicillin (1%) and ciprofloxacin (1%) at 37°C in 5% CO2. The VSMC phenotype was confirmed by using smooth muscle myosin heavy chains 1 and 2 and sm22α antibodies (Abcam, Cambridge, UK) (data not shown).

HUVECs were isolated from sterile, freshly obtained umbilical cords at the time of a normal delivery by using 15 mg/mL collagenase type I digestion as previously described [20, 21]. All donors gave their written consent. HUVECs were cultured with EC medium (PromoCell) supplemented with decomplemented FCS (2%), bFGF (1 ng/mL), EGF (0.1 ng/mL), EC growth supplement/heparin (0.4%), hydrocortisone (1 μg/mL), streptomycin/penicillin (1%) and ciprofloxacin (1%) at 37°C in 5% CO2. HUVECs from four donors were harvested after the third passage to perform protein extraction.

One-dimensional immunoblotting

Confluent VSMCs were detached with the use of 0.05% trypsin and 0.53 mM ethylenediaminetetraacetic acid. Protein extract was obtained by use of a 125 mM Tris, pH 6.8, solution containing 4% SDS, 1.45 M β-mercaptoethanol, 1 μg/mL aprotinin, 1 μg/mL leupeptin, 1 μg/mL pepstatin and 1 mM phenylmethylsulphonyl fluoride (PMSF). Protein extract was then sonicated four times for 30 seconds each and boiled. In total, 120 μL of solubilised proteins were separated by electrophoresis on 10% SDS-PAGE gels (Bio-Rad Laboratories, Hercules, CA, USA), transferred onto nitrocellulose membranes by using a semidry electroblotter (model A; Ancos, Hojby, Denmark) and incubated with sera from patients with GCA, WG, CSS and MPA or from healthy donors at a 1:100 dilution overnight at 4°C with the Cassette Miniblot System (Immunetics Inc., Cambridge, MA, USA). Detection of IgG reactivity was carried out as previously reported [2224] (Additional file 1) with the use of a γ-chain-specific secondary rabbit anti-human IgG antibody coupled to alkaline phosphatase. Immunoreactivity was revealed by Nitro Blue Tetrazolium/5-bromo-4-chloro-3-indolyl phosphate staining (Sigma-Aldrich, St. Louis, MO, USA) as previously reported [23, 24] (Additional file 1) and quantified by densitometry in reflective mode (Epson Perfection 1200 S densitometer; Seiko Epson Corp., Nagano-ken, Japan) and scanned again to quantify transferred proteins [23, 24].

Two-dimensional immunoblotting

Protein extracts

HUVECs and VSMCs were stored at -80°C in 1 mM PMSF and protease inhibitors (Complete Mini; Roche Diagnostics, Meylan, France). Protein extraction was performed as described previously [25] (Additional file 1). Briefly, cells were suspended at 1 × 106/mL in a sample solution extraction kit (Kit 3; Bio-Rad Laboratories). Cell samples were sonicated, and the supernatant was collected after ultracentrifugation (Optima L90-K ultracentrifuge; Beckman Coulter, Fullerton, CA, USA) at 150,000 × g for 25 minutes at 4°C. Protein quantification was carried out using the Lowry method [26]. The supernatant was aliquoted and stored at -80°C.

Two-dimensional electrophoresis

Two-dimensional electrophoresis (2-DE), 2-D immunoblotting and protein identification by MS were performed as previously reported [27] and are detailed in Additional file 1.

Modelling with the use of ingenuity pathway analysis software

To gain insight into the biological pathways and networks that were significantly represented in our proteomic data sets, we used ingenuity pathway analysis software (IPA; Ingenuity Systems, Redwood City, CA, USA). IPA selects 'focus proteins' to be used for generating biological networks. Focus proteins are the proteins from data sets that are mapped to corresponding gene objects in the Ingenuity Pathway Knowledgebase (IPKB) and are known to interact with other proteins on the basis of published, peer-reviewed content in the IPKB. From these interactions, IPA builds networks with a size of no more than 35 genes or proteins. A P value for each network is calculated according to the fit of the user's set of significant genes and/or proteins. IPA computes a score for each network from the P value that indicates the likelihood of the focus proteins in a network being found together by chance. We selected only networks scoring ≥ 2 with P < 0.01 of not being generated by chance. Biological functions were assigned to each network by use of annotations from the scientific literature and stored in the IPKB. Fisher's exact test was used to calculate the P value to determine the probability of each biological function and/or disease or pathway being assigned by chance. We used P ≤ 0.05 to select highly significant biological functions and pathways represented in our proteomic data sets. The build function of IPA allows the generation of pathways that can complete the data analysis by showing interactions of identified proteins with a specific group of molecules [28, 29].

Results

The clinical and histological characteristics of patients with GCA are summarised in Additional file 2, Supplemental Table S1. The mean age (± SD) of the patients with GCA was 74.8 ± 8.15 years. Among the 15 patients (5 men), 13 had histological evidence of GCA. All the 15 patients had active disease at the time of blood sampling: twelve were included at the time of diagnosis, two experienced a disease relapse and another one had an acute flare while being treated with prednisone. None of the other 14 patients were taking corticosteroids at the time of blood sampling.

One-dimensional immunoblotting of IgG reactivity against VSMC protein extracts

One-dimensional immunoblots of IgG reactivity were analysed with VSMC protein extracts in sera from patients with GCA; control patients with ANCA-associated vasculitis, including those with WG, MPA and CSS; and HCs. All subjects tested expressed an IgG reactivity band directed against a 45-kDa protein. In patients with GCA, a number of IgG reactivities were expressed that were not identified in patients with ANCA-associated vasculitis or in HCs, including reactivities directed against protein bands of 85 kDa (Additional file 3).

Two-dimensional immunoblotting of IgG reactivity against VSMC protein extracts

The proteome of VSMCs contained 1,427 different proteins ranging from 3 to 10 isoelectrofocalisation points (IPs) and from 10 to 250 kDa. Among those, a mean (± SD) of 679 ± 258 protein spots were detected after being transferred onto polyvinylidene fluoride (PVDF) membranes. Serum IgG from the HC pool recognised 94 protein spots, whereas IgG from the 5 pools from GCA patients recognised a mean (± SD) of 100 ± 17 protein spots corresponding to a total of 268 different protein spots. Most of these 268 protein spots were recognised from only 1 or 2 pools from patients with GCA and/or from the HC pool. Among these protein spots, 29 were recognised by at least three-fifths of the pools from GCA patients, including 19 not recognised by the HC pool (Additional file 4, Supplemental Table S2). These 19 protein spots were identified by MS as detailed in Table 1 and Additional file 5. The localisations of identified protein spots in the analytical gel are depicted in Figure 1. Among these proteins, only one, the far upstream element-binding protein 2 (FUBP2) (Figure 2), was recognised in all five pools of sera from GCA patients, whereas three different proteins were identified in four pools of sera from GCA patients: actin cytoplasmic 1, actin cytoplasmic 2 and ANKRD26-like family C member 1A (Additional file 4, Supplemental Table S2). Interestingly, IgG from pools of sera from each of three GCA patients recognised lamin A/C (Figure 3) and vinculin (Additional file 6).

Table 1 Mass spectrometry data of vascular smooth muscle cell protein spots identified as specific target antigena
Figure 1
figure 1

Two-dimensional silver-stained gel of total protein extracted from vascular smooth muscle cells. Localisation of the 19 IgG-reactive spots recognised by three-fifths of the pools of sera from giant cell arteritis patients. Numbers were arbitrarily assigned by a computer program. Inset: Enlarged area ranging from 6.5 to 8.2 isoelectric points and 50 to 110 kDa. MW: molecular weight, pI: isoelectric points.

Figure 2
figure 2

Serum IgG reactivity to far upstream element-binding protein 2 in sera of giant cell arteritis patients. Protein extract is from vascular smooth muscle cells (VSMCs). (A) IgG reactivity to far upstream element-binding protein 2 (FUBP2) in five different pools of sera from three giant cell arteritis patients each (P1 to P5) and one pool from twelve healthy controls (HC). (B) FUBP2 spots are expressed in 3-D view for one representative serum pool of patients (top) and the HC pool (bottom). (C) Proteome of VSMCs showing the localisation of FUBP2 spots displayed in (A).

Figure 3
figure 3

Serum IgG reactivity to lamin in serum of giant cell arteritis patients. Protein extract is from vascular smooth muscle cells (VSMCs). (A) IgG reactivity to lamin in five different pools of sera from three giant cell arteritis patients each (P1 to P5) and one pool from twelve healthy controls (HC). (B) Lamin spots are expressed in 3-D views for one representative sera pool of giant cell arteritis patients (top) and the HC pool (bottom). (C) Proteome of VSMCs showing the localisation of lamin spots displayed in (A).

IgG reactivity against HUVEC protein extracts

The proteome of HUVECs contains 820 different proteins ranging from 3 to 10 IP and from 10 to 250 kDa. Among these, a mean (± SD) of 515 ± 73 protein spots were successfully detected after transfer onto PVDF membranes. Serum IgG from the HC pool recognised 79 protein spots, whereas IgG from the 3 pools of GCA patients recognised a mean (± SD) of 162 ± 3 protein spots corresponding to 191 different protein spots. Most of these 191 protein spots were recognised in only 1 pool of IgG from GCA patients and/or were also recognised in the HC pool. Among these protein spots, 45 were recognised in at least two-thirds of pools from GCA patients, including 30 that were not recognised in the HC pool (Additional file 7, Supplemental Table S3). Of these 30 proteins, 22 were identified by matrix-assisted laser desorption ionization time-of-flight/time-of-flight MS. Complete MS data are shown in Table 2. Localisations of identified protein spots in the analytical gel are depicted in Figure 4. Overall, three proteins were recognised by IgG in sera from GCA patients in HUVEC and VSMC protein extracts: mitochondrial fumarate hydratase, lamin A/C and vinculin. IgG reactivity against vinculin and lamin A/C in sera from GCA patients and the HC pool are depicted in Figure 5 and Additional file 8 respectively.

Table 2 Mass spectrometry data of the endothelial cell protein spots identified as specific target antigensa
Figure 4
figure 4

Two-dimensional silver-stained protein pattern of total protein extracted from human umbilical vein endothelial cells. Localisation of the 30 reactive spots recognised by two-thirds of the pools of sera from giant cell arteritis patients. Numbers were arbitrarily assigned by a computer program. Inset: Enlarged area ranging from 5.5 to 8.2 isoelectric points and 45 to 90 kDa. MW: molecular weight, pI: isoelectric points.

Figure 5
figure 5

Serum IgG reactivity to vinculin in serum of patients with giant cell arteritis. (A) IgG reactivities to vinculin in three different pools of sera from three GCA patients each (P1 to P3) and one pool from twelve healthy controls (HC). (B) Vinculin spots are expressed in 3-D views for one representative sera pool of patients (top) and the pool of HCs (bottom). (C) Proteome of human umbilical vein endothelial cells showing the localisation of vinculin spots displayed in (A).

Biological network analysis of identified autoantibody specificities

Lists of VSMC and HUVEC proteins specifically recognised and/or recognised with high intensity by IgG in sera from GCA patients were analysed with IPA. Interestingly, most of the VSMC and HUVEC proteins specifically recognised and/or recognised with high intensity interacted with growth factor receptor-bound protein 2 (Grb2), a protein involved in VSMC proliferation. Therefore, we could depict the signalling network between HUVEC and VSMC proteins identified as major targets of autoantibodies in patients with GCA (Figure 6). Interestingly, TNF-α, IL-4 (Figure 6) and other molecules such as platelet-derived growth factor and IFN-γ (Additional file 9) were also involved in this signalling network.

Figure 6
figure 6

Pathway generated by ingenuity pathway analysis with proteins identified as specific target antigens. Solid lines indicate direct interactions. Dashed lines indicate indirect interactions. Arrows indicate stimulation. Mass spectrometry-identified proteins are depicted in grey. ACTB: actin, β; ACTG1: actin, γ1; ANXA2: annexin A2; ANXA5: annexin A5; CCT2: chaperonin containing TCP1, subunit 2 (β); DLD: dihydrolipoamide dehydrogenase; ENO1: enolase 1, α; EZR: ezrin; FABP5: fatty acid-binding protein 5; FH: fumarate hydratase; GRB2: growth factor receptor-bound protein 2; HNRNPD: heterogeneous nuclear ribonucleoprotein D; HSP90AA2: heat shock protein 90 α (cytosolic), class A member 2; HSPB1: HSPB1: heat shock 27-kDa protein 1; IL4: interleukin 4; IMPDH2: IMP (inosine 5'-monophosphate) dehydrogenase; IRS1: insulin receptor substrate 1; KHSRP: KH-type splicing regulatory protein = FUBP2; LMNA: lamin A/C; MSN: moesin; NPM1: nucleophosmin (nuclear phosphoprotein B23, numatrin); PDIA3: protein disulphide isomerase, family A, member 3; PPIA: peptidylpropyl isomerase A; PRDX3: peroxiredoxin 3; RDX: radixin; RPLPO: ribosomal protein, large, P0; SEMA4D: Sema domain, immunoglobulin domain (Ig), transmembrane domain (TM) and short cytosolic domain (semaphorin) 4D; TNF: tumour necrosis factor; TOMM40: translocase of outer mitochondrial membrane 40 homolog (yeast); TUFM: Tu translation elongation factor, mitochondrial; UGDH: UDP-glucose 6-dehydrogenase; VCL: vinculin; VDAC2: voltage-dependent anion channel 2.

Discussion

In the present work, we detected IgG antibodies directed against the proteome of VSMCs and HUVECs in the sera of patients with GCA and identified their target antigens by using a 2-D immunoblotting technique and MS.

Few studies have focused on perturbations of the humoral immune system in patients with GCA. Few B lymphocytes are detected in temporal artery biopsies from patients with GCA [30]. When present, they are mainly found in the adventitial layer [31]. Moreover, plasma cells can be found in the adventitia in 7% to 24% of temporal artery biopsies from patients with GCA [32]. Plasma cells might localise in adventitia because of an infectious agent initiating vascular inflammation. However, a number of studies failed to identify an infectious agent, either a virus or bacteria, in the arterial wall by immunohistochemistry or PCR [33]. Alternatively, an autoantigen present in the arterial wall might trigger a specific immune response in GCA.

AECAs have been detected in healthy individuals [34] and in a number of systemic autoimmune diseases [10, 35]. AECAs have been associated with disease activity in patients with vasculitis, particularly in those with anti-ANCA-associated vasculitis, Takayasu's arteritis or GCA [15], although these data remain controversial [36]. In addition, AECA could induce EC apoptosis in patients with systemic sclerosis [37]. However, the pathogenic role of AECAs has not yet been documented in GCA, and further investigations are necessary in this clinical setting.

Although to our knowledge anti-VSMC antibodies have not yet been reported in a human disease, such antibodies have been identified in a mouse model of vasculitis. Baiu et al. [17] showed that splenic mouse lymphocytes cultured with syngenic VSMCs induced vasculitic lesions after adoptive transfer into these mice. Serum collected from mice with vasculitis contained antibodies directed against VSMCs. Both wild-type and B-cell-deficient mice showed vascular inflammation after serum transfer, but mice deficient in both B and T cells (Rag2-/-) Yes it should did not, which suggests that immunoglobulin and cell-mediated pathways, particularly T cells, work in concert to contribute to the vasculitis lesions in this model. Thus, autoantibodies targeting proteins in the proteome of VSMCs might play a role in the pathogenesis of GCA, and their function needs to be further explored.

Few studies have been conducted to identify the potential targets of autoantibodies in GCA. Screening antigens in a cDNA library derived from normal human testis revealed high-intensity serum IgG reactivity directed against a number of ubiquitous autoantigens, including human lamin C, cytokeratin and mitochondrial cytochrome oxidase subunit II in the sera of patients with GCA [38]. Interestingly, we identified vinculin, lamin A/C and mitochondrial fumarate hydratase as target antigens of antibodies to proteins in the proteome of VSMCs and HUVECs. Vinculin is a cytoskeleton protein involved in extracellular matrix adhesion and intercellular junctions by binding to actin filaments. This protein has several interaction sites with numerous binding partners, including α-actin [39]. Changes in the relative content of vinculin and α-actin have been reported in the human aortic intima of patients with atherosclerosis [40]. Because vascular remodelling may occur in atherosclerosis, this type of change could be associated with vascular remodelling in GCA. Lamin A and C are both encoded by the LMNA gene and represent major constituents of the inner nuclear membrane. Mutations in the LMNA gene have been identified in a number of conditions, including Hutchinson-Gilford progeria syndrome [41]. The most frequent mutation responsible for progeria creates a truncated progeria mutant lamin A (progerin), which accumulates within the nuclei of human vascular cells and may be directly responsible for vascular involvement in progeria [42]. Other LMNA gene mutations, such as Dunnigan-type familial partial lipodystrophy (FPLD2), can lead to proatherogenic metabolic disturbances such as dyslipidemia, hyperinsulinemia, hypertension and diabetes. Premature atherosclerosis-induced FPLD2 seems to be associated with monogenic insulin resistance syndrome [43]. Identification of lamin A/C as target antigens in sera from patients with GCA seems interesting. Further investigations are necessary to characterise the implications of lamin A/C in vascular remodelling in this condition.

We found another antigen, far upstream element-binding protein 2 (FUBP = KHSRP), to be recognised by all five pools of sera from GCA patients in VSMC protein extracts. This protein, which binds to a DNA region called far-upstream element (FUSE), is a transcriptional activator of c-myc, a proto-oncogene that plays a key role in the regulation of cell growth, proliferation and differentiation. The FUSE-binding protein (FUBP) regulates FUSE activity [44]. Anti-FUBP antibodies were identified in synoviocytes in patients with rheumatoid arthritis, a condition marked by a proliferation of synovial tissue [45]. However, the potential pathogenic role of these antibodies has not yet been identified.

By using IPA, we found that most of the VSMC and HUVEC proteins specifically recognised and/or recognised with high intensity by IgG in sera from GCA patients interacted with Grb2. Grb2 is an intracellular linker protein that facilitates the activation of the small GTPase Ras by receptor tyrosine kinases and is involved in VSMC proliferation. Zhang and colleagues [46] reported that Grb2 is required for the development of neointima in response to vascular injury. Thus, Grb2 might be overexpressed and/or activated in ECs and VSMCs of patients with GCA and might stimulate the remodelling process. Moreover, proteins overexpressed in the remodelling process in the presence of activated Grb2 might trigger a specific immune response, possibly through structural antigen modifications occurring in the presence of metalloproteases and/or reactive oxygen species. A number of the VSMC and HUVEC proteins specifically recognised and/or recognised with high intensity by IgG in sera from GCA patients interacted with TNF-α. This result is in agreement with the pathophysiology of GCA and the ongoing inflammatory process in the arterial wall.

The combined use of 2-DE and immunoblotting offers an interesting approach to the identification of target antigens of autoantibodies [25]. However, our work has several limitations. Fewer than 1,500 protein spots were stained in the reference gel of VSMC and HUVEC protein extracts, which is less than the total number of proteins contained in these cells. Therefore, a number of proteins were probably lost at each step of the technique, depending on their charge, molecular weight, subcellular localisation and/or abundance in the cell. In addition, as expected, none of the identified antigens represented cell surface proteins, because protein extraction for 2-DE does not permit identification of membrane proteins. Finally, our pools of sera were from three patients each because this number was sufficiently low to allow the detection of strong reactivity that would be present in the serum of a single individual [25]. However, we cannot rule out the possibility that a low-intensity reactivity specific to a given individual might not be detected by using this pooling approach.

Conclusions

We provide evidence that IgG antibodies directed toward the proteome of VSMCs and HUVECs are present in the sera of patients with GCA. These antibodies recognise cellular targets that play key roles in cell biology and the maintenance of homeostasis. The potential pathogenic role of these antibodies should be further investigated.

Abbreviations

ACR:

American College of Rheumatology

AECA:

anti-endothelial cell antibody

ANCA:

antineutrophil cytoplasm antibody

bFGF:

basic fibroblast growth factor

CSS:

Churg-Strauss syndrome

EC:

endothelial cell

EGF:

epidermal growth factor

ELISA:

enzyme-linked immunosorbent assay

FCS:

foetal calf serum

FUBP2:

far upstream element-binding protein 2

FUSE:

far upstream element

GCA:

giant cell arteritis

Grb2:

growth factor receptor-bound protein 2

HC:

healthy control

HUVEC:

human umbilical vein endothelial cell

IFN-γ:

interferon-γ

IL:

interleukin

IP:

isoelectrofocalisation point

IPA:

ingenuity pathway analysis

IPKB:

Ingenuity Pathway Knowledgebase

MPA:

microscopic polyangiitis

MS:

mass spectrometry

PMSF:

phenylmethylsulphonyl fluoride

PVDF:

polyvinylidene fluoride

RT:

reverse transcriptase

TNF-α:

tumour necrosis factor α

VSMC:

vascular smooth muscle cell

WG:

Wegener's granulomatosis.

References

  1. Hunder GG: Epidemiology of giant-cell arteritis. Cleve Clin J Med. 2002, 69 (Suppl 2): SII79-SII82.

    Google Scholar 

  2. Gonzalez-Gay MA, Vazquez-Rodriguez TR, Lopez-Diaz MJ, Miranda-Filloy JA, Gonzalez-Juanatey C, Martin J, Llorca J: Epidemiology of giant cell arteritis and polymyalgia rheumatica. Arthritis Rheum. 2009, 61: 1454-1461. 10.1002/art.24459.

    Article  Google Scholar 

  3. González-Gay MA, Blanco R, Rodríguez-Valverde V, Martínez-Taboada VM, Delgado-Rodriguez M, Figueroa M, Uriarte E: Permanent visual loss and cerebrovascular accidents in giant cell arteritis: predictors and response to treatment. Arthritis Rheum. 1998, 41: 1497-1504. 10.1002/1529-0131(199808)41:8<1497::AID-ART22>3.0.CO;2-Z.

    Article  Google Scholar 

  4. Hunder GG, Bloch DA, Michel BA, Stevens MB, Arend WP, Calabrese LH, Edworthy SM, Fauci AS, Leavitt RY, Lie JT, Lightfoot RW, Masi AT, McShane DJ, Mills JA, Wallace SL, Zvaifler NJ: The American College of Rheumatology 1990 criteria for the classification of giant cell arteritis. Arthritis Rheum. 1990, 33: 1122-1128.

    Article  CAS  Google Scholar 

  5. Hall S, Persellin S, Lie JT, O'Brien PC, Kurland LT, Hunder GG: The therapeutic impact of temporal artery biopsy. Lancet. 1983, 2: 1217-1220.

    Article  CAS  Google Scholar 

  6. Ly KH, Regent A, Tamby MC, Mouthon L: Pathogenesis of giant cell arteritis: more than just an inflammatory condition?. Autoimmun Rev. 2010, 9: 635-645. 10.1016/j.autrev.2010.05.002.

    Article  CAS  Google Scholar 

  7. Brack A, Geisler A, Martinez-Taboada VM, Younge BR, Goronzy JJ, Weyand CM: Giant cell vasculitis is a T cell-dependent disease. Mol Med. 1997, 3: 530-543.

    CAS  Google Scholar 

  8. Rittner HL, Kaiser M, Brack A, Szweda LI, Goronzy JJ, Weyand CM: Tissue-destructive macrophages in giant cell arteritis. Circ Res. 1999, 84: 1050-1058.

    Article  CAS  Google Scholar 

  9. Rodríguez-Pla A, Bosch-Gil JA, Rosselló-Urgell J, Huguet-Redecilla P, Stone JH, Vilardell-Tarres M: Metalloproteinase-2 and -9 in giant cell arteritis: involvement in vascular remodeling. Circulation. 2005, 112: 264-269. 10.1161/CIRCULATIONAHA.104.520114.

    Article  Google Scholar 

  10. Guilpain P, Mouthon L: Antiendothelial cells autoantibodies in vasculitis-associated systemic diseases. Clin Rev Allergy Immunol. 2008, 35: 59-65. 10.1007/s12016-007-8069-3.

    Article  CAS  Google Scholar 

  11. Hu N, Westra J, Huitema MG, Stegeman CA, Limburg PC, Kallenberg CG: Autoantibodies against glomerular endothelial cells in anti-neutrophil cytoplasmic autoantibody-associated systemic vasculitis. Nephrology (Carlton). 2009, 14: 11-15. 10.1111/j.1440-1797.2008.01068.x.

    Article  CAS  Google Scholar 

  12. Holmén C, Christensson M, Pettersson E, Bratt J, Stjärne P, Karrar A, Sumitran-Holgersson S: Wegener's granulomatosis is associated with organ-specific antiendothelial cell antibodies. Kidney Int. 2004, 66: 1049-1060. 10.1111/j.1523-1755.2004.00853.x.

    Article  Google Scholar 

  13. Holmén C, Elsheikh E, Christensson M, Liu J, Johansson AS, Qureshi AR, Jalkanen S, Sumitran-Holgersson S: Anti endothelial cell autoantibodies selectively activate SAPK/JNK signalling in Wegener's granulomatosis. J Am Soc Nephrol. 2007, 18: 2497-2508. 10.1681/ASN.2006111286.

    Article  Google Scholar 

  14. van Paassen P, Duijvestijn A, Debrus-Palmans L, Damoiseaux J, Vroomen M, Tervaert JW: Induction of endothelial cell apoptosis by IgG antibodies from SLE patients with nephropathy: a potential role for anti-endothelial cell antibodies. Ann N Y Acad Sci. 2007, 1108: 147-156. 10.1196/annals.1422.017.

    Article  CAS  Google Scholar 

  15. Navarro M, Cervera R, Font J, Reverter JC, Monteagudo J, Escolar G, López-Soto A, Ordinas A, Ingelmo M: Anti-endothelial cell antibodies in systemic autoimmune diseases: prevalence and clinical significance. Lupus. 1997, 6: 521-526. 10.1177/096120339700600608.

    Article  CAS  Google Scholar 

  16. Amor-Dorado JC, García-Porrua C, Gonzalez-Gay MA: Anti-endothelial cell antibodies and biopsy-proven temporal arteritis. Lupus. 2002, 11: 134-10.1191/0961203302lu168xx.

    Article  CAS  Google Scholar 

  17. Baiu DC, Barger B, Sandor M, Fabry Z, Hart MN: Autoantibodies to vascular smooth muscle are pathogenic for vasculitis. Am J Pathol. 2005, 166: 1851-1860. 10.1016/S0002-9440(10)62494-7.

    Article  CAS  Google Scholar 

  18. Jennette JC, Falk RJ, Andrassy K, Bacon PA, Churg J, Gross WL, Hagen EC, Hoffman GS, Hunder GG, Kallenberg CGM, McCluskey RT, Sinico RA, Rees AJ, Van Es LA, Waldherr R, Wiik A: Nomenclature of systemic vasculitides: proposal of an international consensus conference. Arthritis Rheum. 1994, 37: 187-192. 10.1002/art.1780370206.

    Article  CAS  Google Scholar 

  19. Weksler BB, Subileau EA, Perrière N, Charneau P, Holloway K, Leveque M, Tricoire-Leignel H, Nicotra A, Bourdoulous S, Turowski P, Male DK, Roux F, Greenwood J, Romero IA, Couraud PO: Blood-brain barrier-specific properties of a human adult brain endothelial cell line. FASEB J. 2005, 19: 1872-1874.

    CAS  Google Scholar 

  20. Ronda N, Leonardi S, Orlandini G, Gatti R, Bellosta S, Bernini F, Borghetti A: Natural anti-endothelial cell antibodies (AECA). J Autoimmun. 1999, 13: 121-127. 10.1016/S0896-8411(99)99999-7.

    Article  CAS  Google Scholar 

  21. Tamby MC, Chanseaud Y, Humbert M, Fermanian J, Guilpain P, Garcia-de-la-Peña-Lefebvre P, Brunet S, Servettaz A, Weill B, Simonneau G, Guillevin L, Boissier MC, Mouthon L: Anti-endothelial cell antibodies in idiopathic and systemic sclerosis associated pulmonary arterial hypertension. Thorax. 2005, 60: 765-772. 10.1136/thx.2004.029082.

    Article  CAS  Google Scholar 

  22. García de la Peña-Lefebvre P, Chanseaud Y, Tamby MC, Reinbolt J, Batteux F, Allanore Y, Kahan A, Meyer O, Benveniste O, Boyer O, Guillevin L, Boissier MC, Mouthon L: IgG reactivity with a 100-kDa tissue and endothelial cell antigen identified as topoisomerase 1 distinguishes between limited and diffuse systemic sclerosis patients. Clin Immunol. 2004, 111: 241-251. 10.1016/j.clim.2004.01.005.

    Article  Google Scholar 

  23. Tamby MC, Bussone G, Mouthon L: Antitopoisomerase 1 antibodies in systemic sclerosis: how to improve the detection?. Ann N Y Acad Sci. 2007, 1109: 221-228. 10.1196/annals.1398.026.

    Article  CAS  Google Scholar 

  24. Chanseaud Y, García de la Peña-Lefebvre P, Guilpain P, Mahr A, Tamby MC, Uzan M, Guillevin L, Boissier MC, Mouthon L: IgM and IgG autoantibodies from microscopic polyangiitis patients but not those with other small- and medium-sized vessel vasculitides recognize multiple endothelial cell antigens. Clin Immunol. 2003, 109: 165-178. 10.1016/S1521-6616(03)00170-0.

    Article  CAS  Google Scholar 

  25. Terrier B, Tamby MC, Camoin L, Guilpain P, Broussard C, Bussone G, Yaïci A, Hotellier F, Simonneau G, Guillevin L, Humbert M, Mouthon L: Identification of target antigens of antifibroblast antibodies in pulmonary arterial hypertension. Am J Respir Crit Care Med. 2008, 177: 1128-1134. 10.1164/rccm.200707-1015OC.

    Article  CAS  Google Scholar 

  26. Lowry OH, Rosebrough NJ, Farr AL, Randall RJ: Protein measurement with the Folin phenol reagent. J Biol Chem. 1951, 193: 265-275.

    CAS  Google Scholar 

  27. Guilpain P, Servettaz A, Tamby MC, Chanseaud Y, Tamas N, Garcia de la Pena-Lefebvre P, Broussard C, Guillevin L, Camoin L, Mouthon L: A combined SDS-PAGE and proteomics approach to identify target autoantigens in healthy individuals and patients with autoimmune diseases. Ann N Y Acad Sci. 2007, 1109: 538-549. 10.1196/annals.1398.060.

    Article  CAS  Google Scholar 

  28. Bussone G, Dib H, Tamby MC, Broussard C, Federici C, Woimant G, Camoin L, Guillevin L, Mouthon L: Identification of new autoantibody specificities directed at proteins involved in the transforming growth factor β pathway in patients with systemic sclerosis. Arthritis Res Ther. 2011, 13: R74-10.1186/ar3336.

    Article  CAS  Google Scholar 

  29. Qi YX, Jiang J, Jiang XH, Wang XD, Ji SY, Han Y, Long DK, Shen BR, Yan ZQ, Chien S, Jiang ZL: PDGF-BB and TGF-β1 on cross-talk between endothelial and smooth muscle cells in vascular remodeling induced by low shear stress. Proc Natl Acad Sci USA. 2011, 108: 1908-1913. 10.1073/pnas.1019219108.

    Article  CAS  Google Scholar 

  30. Banks PM, Cohen MD, Ginsburg WW, Hunder GG: Immunohistologic and cytochemical studies of temporal arteritis. Arthritis Rheum. 1983, 26: 1201-1207. 10.1002/art.1780261005.

    Article  CAS  Google Scholar 

  31. Lavignac C, Jauberteau-Marchan MO, Liozon E, Vidal E, Catanzano G, Liozon F: Immunohistochemical study of lesions in Horton's temporal arteritis before and during corticotherapy in French. Rev Med Interne. 1996, 17: 814-820. 10.1016/0248-8663(96)82684-1.

    Article  CAS  Google Scholar 

  32. Chatelain D, Duhaut P, Loire R, Bosshard S, Pellet H, Piette JC, Sevestre H, Ducroix JP: Small-vessel vasculitis surrounding an uninflamed temporal artery: a new diagnostic criterion for polymyalgia rheumatica?. Arthritis Rheum. 2008, 58: 2565-2573. 10.1002/art.23700.

    Article  Google Scholar 

  33. Duhaut P, Bosshard S, Ducroix JP: Is giant cell arteritis an infectious disease? Biological and epidemiological evidence. Presse Med. 2004, 33: 1403-1408. 10.1016/S0755-4982(04)98939-7.

    Article  Google Scholar 

  34. Servettaz A, Guilpain P, Camoin L, Mayeux P, Broussard C, Tamby MC, Tamas N, Kaveri SV, Guillevin L, Mouthon L: Identification of target antigens of antiendothelial cell antibodies in healthy individuals: a proteomic approach. Proteomics. 2008, 8: 1000-1008. 10.1002/pmic.200700794.

    Article  CAS  Google Scholar 

  35. Domiciano DS, Carvalho JF, Shoenfeld Y: Pathogenic role of anti-endothelial cell antibodies in autoimmune rheumatic diseases. Lupus. 2009, 18: 1233-1238. 10.1177/0961203309346654.

    Article  CAS  Google Scholar 

  36. Del Papa N, Guidali L, Sironi M, Shoenfeld Y, Mantovani A, Tincani A, Balestrieri G, Radice A, Sinico RA, Meroni PL: Anti-endothelial cell IgG antibodies from patients with Wegener's granulomatosis bind to human endothelial cells in vitro and induce adhesion molecule expression and cytokine secretion. Arthritis Rheum. 1996, 39: 758-766. 10.1002/art.1780390507.

    Article  CAS  Google Scholar 

  37. Bordron A, Dueymes M, Levy Y, Jamin C, Leroy JP, Piette JC, Shoenfeld Y, Youinou PY: The binding of some human antiendothelial cell antibodies induces endothelial cell apoptosis. J Clin Invest. 1998, 101: 2029-2035. 10.1172/JCI2261.

    Article  CAS  Google Scholar 

  38. Schmits R, Kubuschok B, Schuster S, Preuss KD, Pfreundschuh M: Analysis of the B cell repertoire against autoantigens in patients with giant cell arteritis and polymyalgia rheumatica. Clin Exp Immunol. 2002, 127: 379-385. 10.1046/j.1365-2249.2002.01751.x.

    Article  CAS  Google Scholar 

  39. Ziegler WH, Liddington RC, Critchley DR: The structure and regulation of vinculin. Trends Cell Biol. 2006, 16: 453-460. 10.1016/j.tcb.2006.07.004.

    Article  CAS  Google Scholar 

  40. Glukhova MA, Kabakov AE, Frid MG, Ornatsky OI, Belkin AM, Mukhin DN, Orekhov AN, Koteliansky VE, Smirnov VN: Modulation of human aorta smooth muscle cell phenotype: a study of muscle-specific variants of vinculin, caldesmon, and actin expression. Proc Natl Acad Sci USA. 1988, 85: 9542-9546. 10.1073/pnas.85.24.9542.

    Article  CAS  Google Scholar 

  41. Eriksson M, Brown WT, Gordon LB, Glynn MW, Singer J, Scott L, Erdos MR, Robbins CM, Moses TY, Berglund P, Dutra A, Pak E, Durkin S, Csoka AB, Boehnke M, Glover TW, Collins FS: Recurrent de novo point mutations in lamin A cause Hutchinson-Gilford progeria syndrome. Nature. 2003, 423: 293-298. 10.1038/nature01629.

    Article  CAS  Google Scholar 

  42. McClintock D, Gordon LB, Djabali K: Hutchinson-Gilford progeria mutant lamin A primarily targets human vascular cells as detected by an anti-lamin A G608G antibody. Proc Natl Acad Sci USA. 2006, 103: 2154-2159. 10.1073/pnas.0511133103.

    Article  CAS  Google Scholar 

  43. Hegele RA: Premature atherosclerosis associated with monogenic insulin resistance. Circulation. 2001, 103: 2225-2229.

    Article  CAS  Google Scholar 

  44. Duncan R, Bazar L, Michelotti G, Tomonaga T, Krutzsch H, Avigan M, Levens D: A sequence-specific, single-strand binding protein activates the far upstream element of c-myc and defines a new DNA-binding motif. Genes Dev. 1994, 8: 465-480. 10.1101/gad.8.4.465.

    Article  CAS  Google Scholar 

  45. Goëb V, Thomas-L'Otellier M, Daveau R, Charlionet R, Fardellone P, Le Loët X, Tron F, Gilbert D, Vittecoq O: Candidate autoantigens identified by mass spectrometry in early rheumatoid arthritis are chaperones and citrullinated glycolytic enzymes. Arthritis Res Ther. 2009, 11: R38-10.1186/ar2644.

    Article  Google Scholar 

  46. Zhang S, Ren J, Khan MF, Cheng AM, Abendschein D, Muslin AJ: Grb2 is required for the development of neointima in response to vascular injury. Arterioscler Thromb Vasc Biol. 2003, 23: 1788-1793. 10.1161/01.ATV.0000085015.49110.85.

    Article  CAS  Google Scholar 

  47. García de la Peña-Lefebvre P, Chanseaud Y, Tamby MC, Reinbolt J, Batteux F, Allanore Y, Kahan A, Meyer O, Benveniste O, Boyer O, Guillevin L, Boissier MC, Mouthon L: IgG reactivity with a 100-kDa tissue and endothelial cell antigen identified as topoisomerase 1 distinguishes between limited and diffuse systemic sclerosis patients. Clin Immunol. 2004, 111: 241-251. 10.1016/j.clim.2004.01.005.

    Article  Google Scholar 

  48. Shevchenko A, Wilm M, Vorm O, Mann M: Mass spectrometric sequencing of proteins silver-stained polyacrylamide gels. Anal Chem. 1996, 68: 850-858. 10.1021/ac950914h.

    Article  CAS  Google Scholar 

  49. Perkins DN, Pappin DJ, Creasy DM, Cottrell JS: Probability-based protein identification by searching sequence databases using mass spectrometry data. Electrophoresis. 1999, 20: 3551-3567. 10.1002/(SICI)1522-2683(19991201)20:18<3551::AID-ELPS3551>3.0.CO;2-2.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

AR received financial support from the Direction Régionale et départementale de Champagne-Ardennes et de la Marne and the Société Nationale Française de Médecine Interne (SNFMI). HD received financial support from Avenir Mutualiste des Professions Libérales & Indépendantes (AMPLI) and Association pour la Recherche en Médecine Interne et en Immunologie Clinique (ARMIIC). KHL received financial support from Limoges Hospital. MCT received a grant from Pfizer and the Direction de la Recherche Clinique (PHRC National Auto-HTAP). We also thank the Unité de Recherche Clinique Cochin-Necker. We also thank CSL Behring for financial support. None of the funding bodies had a role in the study design; the collection, analysis or interpretation of data; the writing of the manuscript; or the decision to submit the manuscript for publication.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Luc Mouthon.

Additional information

Competing interests

AR, HD and LM have applied for a patent related to the content of this article (Patent Procédé de diagnostic d'une vascularite FR0951205).

Authors' contributions

AR carried out the immunoblotting and proteomic experiments, analysed the results and drafted the manuscript. HD carried out immunoblotting and proteomic experiments with AR, participated in the analysis of the results and edited the manuscript. KHL carried out 1-D immunoblotting experiments and participated in the drafting of the manuscript. CA conducted the inclusion of patients into the study, analysed the results and edited the manuscript. MCT participated in the study design and the analysis of the results and also edited the manuscript. NT participated in immunoblotting and proteomic experiments, participated in the analysis of the results and edited the manuscript. CF performed ingenuity pathway analysis, participated in the analysis of the results and edited the manuscript. CB performed proteomic analysis, participated in the analysis of the results and edited the manuscript. BW provided immortalised VSMCs, participated in the analysis of the results and edited the manuscript. LG provided sera from patients, participated in the study design and analysis of the results and also edited the manuscript. LM provided sera from patients, designed the experiments, analysed the results and drafted the manuscript. All authors read and approved the final manuscript.

Kim H Ly, Christian Agard contributed equally to this work.

Electronic supplementary material

13075_2011_3150_MOESM1_ESM.DOC

Additional file 1: Supplemental file. Detailed data concerning 2-D electrophoresis technique and mass spectrometry identification [4749]. (DOC 56 KB)

13075_2011_3150_MOESM2_ESM.DOC

Additional file 2: Supplemental Table S1. Clinical and histological characteristics of 15 patients with giant cell arteritis. (DOC 30 KB)

13075_2011_3150_MOESM3_ESM.PPT

Additional file 3: Supplemental Figure S1. One-dimensional immunoblot IgG reactivity from giant cell arteritis patients with vascular smooth muscle cell proteins. Serum samples from three patients with GCA were tested, and sera from four patients with Wegener's granulomatosis (WG), two with Churg-Strauss syndrome (CSS) and two with microscopic polyangeitis (MPA) used as vasculitis controls, intravenous immunoglobulin (IVIg) as a positive control and PBS and sera from two healthy controls (HCs) as negative controls were immunoblotted at a dilution of 1:100 with a soluble extract of immortalised human mammary artery VSMCs. (PPT 168 KB)

13075_2011_3150_MOESM4_ESM.DOC

Additional file 4: Supplemental Table S2. Antigens specifically recognised by IgG of three-fifths of the pools of sera from giant cell arteritis patients. (DOC 49 KB)

Additional file 5: Mass spectrometry data of target antigens recognised by only one peptide. (PDF 540 KB)

13075_2011_3150_MOESM6_ESM.PPT

Additional file 6: Supplemental Figure S2. 2-D immunoblots of IgG reactivity to vinculin in sera from patients with giant cell arteritis. Protein extract is from vascular smooth muscle cells (VSMCs). (A) IgG reactivities of five different pools of sera from three giant cell arteritis patients each (P1 to P5) and one pool from twelve healthy controls (HCs). (B) Vinculin spots are expressed in 3-D views for one representative sera pool of patients (top) and the HC pool (bottom). (C) Proteome of VSMCs showing the localisation of vinculin spots displayed in (A). (PPT 784 KB)

13075_2011_3150_MOESM7_ESM.DOC

Additional file 7: Supplemental Table S3. Antigens specifically recognised by IgG of two-thirds of the pools of sera from giant cell arteritis patients. (DOC 52 KB)

13075_2011_3150_MOESM8_ESM.PPT

Additional file 8: Supplemental Figure S3. Two-dimensional immunoblots of IgG reactivity to lamin in sera of patients with giant cell arteritis. Protein extract is from human umbilical vein endothelial cells (HUVECs). (A) IgG reactivities to lamin of three different pools of sera from three giant cell arteritis patients each (P1 to P3) and one pool from twelve healthy controls (HCs). (B) Lamin spots are expressed in 3-D views for one representative sera pool from patients (top) and the HC pool (bottom). (C) Proteome of HUVECs showing the localisation of lamin spots displayed in (A). (PPT 1023 KB)

13075_2011_3150_MOESM9_ESM.PPT

Additional file 9: Supplement Figure S4. Protein network generated by merging the two pathways involved in target antigens. Protein extracts are from human umbilical vein endothelial cells and vascular smooth muscle cells. Solid lines indicate direct interactions. Dashed lines indicate indirect interactions. Arrows indicate stimulation. ABCA2: ATP-binding cassette, subfamily A, (ABC1), member 2; ACTB: actin, β; ACTG1: actin, γ1; ANXA2: annexin A2; ANXA5: annexin A5; CCND2: cyclin D2; CCT2: chaperonin containing TCP1, subunit 2 (β); COPA: coatomer protein complex, subunit α; CPOX: coproporphyrinogen oxidase; DLD: dihydrolipoamide dehydrogenase; ENO1: enolase 1, α; EZR: ezrin; FABP5: fatty acid-binding protein 5; FH: fumarate hydratase; FUBP1: far upstream element (FUSE)-binding protein 1; GRB2: growth factor receptor-bound protein 2; HNRNPD: heterogeneous nuclear ribonucleoprotein D; HSP90AA2: heat shock protein 90 kDa α (cytosolic), class A, member 2; HSPB1: heat shock 27-kDa protein 1; IFNG: interferon γ; IKBKG: inhibitor of κ light polypeptide gene enhancer in B cells, kinase γ; IL4: interleukin 4; IMPDH2: IMP (inosine 5'-monophosphate) dehydrogenase; IRS1: insulin receptor substrate 1; KHSRP: KH-type splicing regulatory protein; LMNA: lamin A/C; MSN: moesin; NDUFS3: NADH dehydrogenase (ubiquinone) iron-sulphur protein 3, 30 kDa (NADH coenzyme Q reductase); NPM1: nucleophosmin (nuclear phosphoprotein B23, numatrin); PARK7: Parkinson's disease (autosomal recessive early onset) 7; PDGF BB: platelet-derived growth factor B dimer; PDIA3: protein disulphide isomerase, family A, member 3; PHB: prohibitin; PPIA: peptidylpropyl-isomerase A; PRDX3: peroxiredoxin 3; PSMC5: proteasome 26S subunit, ATPase, 5; RDX: radixin; RPLPO: ribosomal protein, large, P0; SEMA4D: Sema domain, immunoglobulin domain (Ig), transmembrane domain (TM) and short cytosolic domain (semaphorin) 4D; TNF: tumour necrosis factor; TOMM40: translocase of outer mitochondrial membrane 40 homolog (yeast); TP53: tumour protein P53; TPP1: tripeptidyl 1 peptidase; TUFM: Tu translation elongation factor, mitochondrial; UGDH: UDP-glucose 6-dehydrogenase; VCL: vinculin; VDAC2: voltage-dependent anion channel 2. (PPT 497 KB)

Authors’ original submitted files for images

Rights and permissions

Reprints and permissions

About this article

Cite this article

Régent, A., Dib, H., Ly, K.H. et al. Identification of target antigens of anti-endothelial cell and anti-vascular smooth muscle cell antibodies in patients with giant cell arteritis: a proteomic approach. Arthritis Res Ther 13, R107 (2011). https://doi.org/10.1186/ar3388

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/ar3388

Keywords