American Journal of Respiratory and Critical Care Medicine

Rationale: Systemic sclerosis (SSc)-associated pulmonary arterial hypertension (PAH) portends worse outcome than other forms of PAH. Vasoconstrictive and vascular remodeling actions of endothelin (ET) 1 and angiotensin (Ang) II via endothelin receptor type A (ETAR) and Ang receptor type-1 (AT1R) activation are implicated in PAH pathogenesis.

Objectives: We hypothesized that stimulating autoantibodies (Abs) targeting and activating AT1R and ETAR may contribute to SSc-PAH pathogenesis, and tested their functional and biomarker relevance.

Methods: Anti-AT1R and -ETAR Abs were detected by ELISA in different cohorts of patients and tested in vitro and in an animal model for their pathophysiological effects.

Measurements and Main Results: The Abs were significantly higher and more prevalent in patients with SSc-PAH (n = 81) and connective tissue disease–associated PAH (n = 110) compared with other forms of PAH/pulmonary hypertension (n = 106). High anti-AT1R and anti-ETAR Abs predicted development of SSc-PAH and SSc-PAH–related mortality in a prospective analysis. Both Abs increased endothelial cytosolic Ca2+ concentrations in isolated perfused rat lungs, which could be blocked by respective specific receptor antagonists. Ab-mediated stimulation of intralobar pulmonary rat artery ring segments increased vasoconstrictive responses to Ang II and ET-1, and implicated cross-talk between both pathways demonstrated by reciprocal blockade with respective antagonists. Transfer of SSc-IgG containing both autoantibodies into healthy C57BL/6J mice led to more abundant vascular and airway α-smooth muscle actin expression and inflammatory pulmonary vasculopathy.

Conclusions: Anti-AT1R and -ETAR Abs are more frequent in SSc-PAH/connective tissue disease–PAH compared with other forms of pulmonary hypertension, and serve as predictive and prognostic biomarkers in SSc-PAH. Both antibodies may contribute to SSc-PAH via increased vascular endothelial reactivity and induction of pulmonary vasculopathy.

Scientific Knowledge on the Subject

Systemic sclerosis (SSc)-associated pulmonary arterial hypertension (PAH) portends worse outcome than other forms of PAH. Vasoconstrictive and vascular remodeling actions of endothelin (ET) 1 and angiotensin (Ang) II via endothelin receptor type A (ETAR) and Ang receptor type-1 (AT1R) activation are implicated in PAH pathogenesis.

What This Study Adds to the Field

Anti-AT1R and anti-ETAR antibodies are more frequent in SSc-PAH/connective tissue disease–PAH compared to other forms of pulmonary hypertension and serve as predictive and prognostic biomarkers in SSc-PAH. Both antibodies may contribute to SSc-PAH via increased vascular endothelial reactivity and induction of pulmonary vasculopathy.

Pulmonary arterial hypertension (PAH) is a rare yet frequently fatal clinical condition (1). Patients with connective tissue diseases (CTDs) account for 20–30% of patients with PAH in clinical trials. The majority suffers from systemic sclerosis (SSc; e.g., Ref. 2). Despite substantial therapeutic improvements, patients with CTD-PAH and patients with SSc-PAH have a reduced survival compared with patients with idiopathic PAH (IPAH) (3). The complex pathogenesis of PAH includes sustained vasoconstriction and vascular remodeling of small pulmonary arteries (4). Endothelin (ET) 1 or angiotensin II (Ang II) and their receptors have been implicated in disease pathogenesis. Pharmacologic targeting of ET-1 actions, mainly via activation of ET-1 receptor type A (ETAR), has become a mainstay of PAH therapy (5, 6). Increased Ang II type-1 receptor (AT1R) activation was linked to disease progression and mortality in patients with IPAH (7, 8). Pharmacologic targeting of AT1R with losartan alleviated experimental PAH (9).

In addition, dysregulated immunity and inflammation are also common in CTD-PAH and other PAH entities, as suggested by inflammatory infiltration, growth factor expression in the remodeled pulmonary vasculature, and increased cytokine and chemokine levels in the circulation (10). However, heterogeneity in therapeutic responses asks for a more precise definition of pathophysiological differences among PAH entities. Therefore new diagnostic and prognostic biomarkers will be critical to identify patient subsets that are likely to benefit from targeted therapies.

Recently, we have identified simultaneous presence of functional autoantibodies (Abs) against AT1R and ETAR in patients with SSc (11) and linked them to increased prevalence of SSc-related vascular and fibrotic complications and higher risk for SSc-related mortality. Both antibodies induce phosphorylation of extracellular signal–regulated kinase 1/2 and transforming growth factor-β expression in endothelial cells, and hence activate endothelial cells to secrete cytokines (11, 12). In addition, the antibodies mediate decreased wound repair by endothelial cells and act on immune cells (12, 13). Based on these data and inducible renal obliterative lesions after transfer of anti-AT1R Abs into animals (14), we hypothesized an involvement of anti-AT1R Abs and anti-ETAR Abs in the pathogenesis of CTD-associated PAH, especially SSc-related PAH. After investigating their presence in different PAH entities, we studied their potential relevance as predictive and prognostic biomarkers for SSc-PAH, and their contribution to its pathogenesis.

Some of the results of these studies have been previously reported in the form of abstracts (15, 16).

Patients

Anti-AT1R/ETAR Ab levels were first measured in serum samples from 62 consecutive patients with SSc-PAH admitted to the Rheumatology Department of the Charité University Hospital Berlin (Berlin, Germany). Additional serum samples were obtained from 182 patients with different pulmonary hypertension (PH)/PAH entities, followed at the Department of Respiratory Medicine at the University Hannover (Hannover, Germany; n = 115), and at the Justus-Liebig University (Giessen, Germany; n = 67). Of these 182 patients, 62 had IPAH, 31 were classified as having chronic thromboembolic PH (CTEPH), 14 had PH due to congenital heart disease (CHD), 19 had SSc-PAH, and 56 were patients with PAH associated with CTD, which was not further specified. Combining all cases from the three centers, the cohort included 110 patients with CTD-PAH, of which 81 had SSc-PAH. Patients with SSc fulfilled the new European League against Rheumatism/American College of Rheumatology criteria for SSc (17). In addition, 253 consecutive patients with SSc entering our out-patient clinic between 2006 and 2011, without right heart catheter (RHC)-proven PAH and naive to the use of ETAR or AT1R blockers, were tested for anti-AT1R and anti-ETAR Ab levels, and were prospectively observed. The epidemiologic data of the patients are shown in Table 1. Under clinical routine conditions, patients were screened for PAH at least in 1-year intervals by assessment of World Health Organization functional class, echocardiography, lung function with detection of predicted single-breath diffusion capacity for carbon monoxide (DlCO-SB), and, during the last years, also by detection of N-terminal pro-brain natriuretic peptide (NT-proBNP) levels. Only if PAH was suspected was RHC performed, using a mean pulmonary arterial pressure (mPAP) of 25 mm Hg or greater and a pulmonary capillary wedge pressure of 15 mm Hg or less at rest for PAH diagnosis (18). All patients were examined in experienced PAH centers. Pulmonary vascular resistance (PVR) and cardiac index (CaI) were determined according to the local guidelines, yet not reported for all patients. The study was approved by the local ethics committee of each contributing center, and included written consent of each patient.

Table 1. Epidemiologic and Hemodynamic Data of Patients from Different Pulmonary Hypertension Cohorts and of Patients with Systemic Sclerosis without PAH

 SSc-PAH (n = 67)SSc Non-PAH (n = 217)IPAH (n = 62)CTEPH (n = 31)CHD (n = 14)CTD-PAH (n = 110)
Female sex, %71.683.556.551.657.181.8
Age, yr61.9 (37.7–81.9)54.2 (29.0–71.0)51 (46.8–55.2)61.6 (57–66.1)59.4 (50.4–68.4)58.9 (56.4–61.3)
Duration of PAH, yr3.5 (1.30.6–8.7)n.a.3.6 (2.8–4.5)3.1 (2–4.1)2.2 (0.4–4.1)2.9 (1.9–4.0)
Duration of SSc, yr8.81 (0.85–25.2)*6.0 (0–20.0)*n.a.n.a.n.a.n.a.
mPAP, mm Hg40.5 (36.6–44.4)n.a.53.4 (48.7–58.4)43.6 (40.8–47.2)36.9 (22.3–51.5)40.8 (37.7–44)
PVR, dyn ⋅ s/cm5557.6 (457.1–658.0)n.a.874 (747–1001)747 (621–737)555 (117–993.6)608.7 (512–706)
Cardiac index, L/min/m22.6 (2.4–2.8)n.a.2.3 (2.1–2.6)2.2 (2.1–2.4)2.5 (2.1–2.9)2.8 (2.5–3.0)
Anti-AT1R Ab levels, U17.9 (14.4–21.4)19.5 (17.7–21.2)8.1 (5.5–10.7)5.5 (4.3–6.7)6 (3.1–8.8)16.9 (14.3–19.6)
Anti-ETAR Ab levels, U17.2 (13.7–20.8)21.1 (19.2–22.9)6.4 (3.5–9.3)3.5 (2.8–4.1)4.9 (1.5–8.2)15.8 (13.2–18.4)

Definition of abbreviations: Ab = autoantibody; AT1R = angiotensin receptor type-1; CHD = congenital heart disease; CTD = connective tissue disease; CTEPH = chronic thromboembolic pulmonary hypertension; ETAR = endothelin receptor type A; IPAH = idiopathic pulmonary arterial hypertension; mPAP = mean pulmonary arterial pressure; n.a. = not applicable; PAH = pulmonary arterial hypertension; PVR = pulmonary vascular resistance; SSc = systemic sclerosis.

Mean values are given and the 95% quantile (95% confidence interval).

* Only identified in the Charité SSc cohort.

Missing values between 16 and 22% in all cohorts.

Detection of Anti-AT1R and Anti-ETAR Abs

Serum antibody levels were measured by an ELISA (CellTrend GmbH, Luckenwalde, Germany [since 2012, One Lambda, Inc., Canoga Park, CA) as previously described (11). Briefly, polystyrene plates were coated with extracts from cells overexpressing the human AT1R or the human ETAR in native conformation, respectively. Detection of the Ab levels was performed by staff unaware of patient data.

Isolation of Human Anti-AT1R and Anti-ETAR Abs Containing IgG and Control IgG

All ex vivo and in vivo experiments were conducted with endotoxin-free IgG isolated from serum samples as previously described (11, 12). For in vitro experiments, final concentration was 1 mg/ml if not stated otherwise. Purified pooled IgG from 14 patients with SSc with high levels of anti-AT1R Abs (mean concentration, 21.8 U) and of anti-ETAR Abs (mean concentration, 17.91 U) was used for animal experiments. IgG from 15 healthy donors served as control (mean concentration, 3.85 U for anti-AT1R Abs and 2.5 U for anti-ETAR Abs, respectively).

Real-Time Fluorescence Imaging of Endothelial Calcium

Isolated lungs from male Sprague-Dawley rats (350–450 g body weight) were prepared (n = 9 per group), perfused, and ventilated as described previously (19, 20). For imaging of endothelial cytosolic Ca2+ concentration ([Ca2+]i), lungs were positioned under an upright fluorescence microscope and superfused with normal saline at 37°C. Fura-2-acetoxymethylester (Fura-2 AM, 5 μmol/L), which is de-esterified intracellularly to the Ca2+-sensitive dye, fura-2, was loaded to lung endothelial cells for 20 minutes via a microcatheter. Fura-2 fluorescence in endothelial cells of subpleural lung single venular capillaries of 15–30 μm in diameter was excited as described previously (21). Endothelial [Ca2+]i was determined from the 340 nm:380 nm ratio based on a Kd of 224 nmol/L and appropriate calibration parameters (21). After baseline recordings, purified IgG from either patients with SSc or from healthy donor sera was directly infused in HEPES dextran buffer into the imaged microvessels via a microcatheter over 30 minutes. The AT1R blocker, valsartan (a gift of D. N. Müller, Max Delbrück Center for Molecular Medicine, Berlin, Germany), and the ETAR blocker, sitaxsentan (provided by Pfizer Deutschland GmbH, Berlin, Germany) were each administered 10 minutes before IgG infusion in respective experiments (1 μmol/L each). All animal studies were approved by the local government authorities.

Small Vessel Myography

Third- to fourth-generation intralobar pulmonary rat artery ring segments from Sprague-Dawley rats (external diameter, 100–300 μm; length, 2 mm) were freed from connective tissue and mounted in a small vessel myograph (DMTDanish Myotechnology, Aarhus, Denmark). Contractile responses were measured as described previously (22). As ET-1–induced contraction in rat small pulmonary arteries is insensitive to either BQ-123, a selective ETAR antagonist, or the selective ETBR antagonist, BQ-788 (22), we used the combined ETA/BR antagonist, bosentan (a gift from Actelion, Basel, Switzerland). The vessels were incubated with 10 μM bosentan for 20 minutes, after which IgG (0.1 mg/mL) was added for 30 minutes. Thereafter, vessels were exposed to cumulatively increasing concentrations of Ang II. After repeated washing, the vessels were treated following the same protocol substituting valsartan 10 μM for bosentan and ET-1 for Ang II. Contractile responses to Ang II and ET-1 were measured as a percentage of the response to 60 mM K+; the effects were reversible by the respective receptor blockers.

Antibody Transfer Experiments

For short-term experiments, female 7-week-old C57BL/6J mice received a single intravenous injection of 800 μg IgG dissolved in 100 μl NaCl 0.9%. Lungs were harvested for histological and immunological analyses at Day 7. For long-term experiments, mice received repeated IgG injections of 200 μg IgG dissolved in 100 μl NaCl 0.9% at Days 1, 17, 30, and 93. Organs were harvested at Day 100. Body weight (Days 30, 58, 78, and at readout day) and rectal temperature control (at read out day; BAT-12 Microprobe; Physitemp Instruments, Clifton, NJ) were monitored.

Histology and Immunohistochemistry

For immunohistochemistry, lungs were removed and immediately snap frozen in O.C.T. medium (Tissue-Tek, Sakura, Europe). Sections of 3 μm were prepared and stained with a polyclonal anti–α-smooth muscle actin (α-SMA) antibody (Abcam, Cambridge, UK) and with an anti-human IgG F(ab′)2-fragment (mouse absorbed; Dianova, Hamburg, Germany) to detect human IgG deposition. 4′,6-diamidino-2-phenylindole (Sigma Aldrich, Seelze, Germany) was used for nuclear DNA staining. For histological assessment, lungs were fixed in 4% paraformaldehyde at room temperature for 24 hours and embedded in paraffin using standard procedures. Anti-CD31 antibody was used (clone SZ31; Dianova) to stain endothelial cells. Hematoxylin and eosin staining was performed to visualize cellular organization. Signals were detected by light microscopy (Axioplan; Carl Zeiss MicroImaging GmbH, Jena, Germany) as well as by immunofluorescence (LSM710; Carl Zeiss Microscopy GmbH) and analyzed by ZEN 2009 Light Edition software (Carl Zeiss Microscopy GmbH).

Statistical Analysis

Comparisons between groups were analyzed by Mann-Whitney U test for continuous variables. Kaplan-Meier analyses were performed to investigate the onset of PAH development and mortality after antibody detection. Significance was determined by log-rank test. Receiver-operating characteristic (ROC) curves were plotted to determine area under the curve (AUC), sensitivity, and specificity using calculated distance to 0.1. Small-vessel myography data were analyzed by ANOVA. Results are expressed as means (±SEM); in myograph experiments, n stands for the number of the studied pulmonary artery ring segments. Differences were considered statistically significant at a P value of 0.05 or less. GraphPad Prism 5.0 (GraphPad Software Inc., San Diego, CA) as well as IBM SPSS 21.0 (Ehningen, Germany) were used for statistical analyses.

Anti-AT1R and Anti-ETAR Antibodies Are Higher in SSc-PAH and CTD-PAH Than in Other Forms of PH/PAH

Epidemiologic and hemodynamic data of the cohorts studied are shown in Table 1. PH/PAH groups did not differ significantly among centers. Higher percentages of female patients and shorter duration of PAH were observed in CTD-PAH and SSc-PAH. mPAP, as detected by RHC in these two groups, was significantly lower as compared with IPAH (P < 0.001) and CTEPH (P = 0.029). Accordingly, PVR was significantly lower in patients with SSc-PAH compared with IPAH (P < 0.0001) and patients with CTEPH (P = 0.002). The CaI was significantly higher in patients with SSc-PAH compared with patients with IPAH or CTEPH, and similar to patients with CHD.

The levels of the anti-AT1R and anti-ETAR Abs were significantly higher in CTD-PAH and SSc-PAH as compared with IPAH (P < 0.0001), CTEPH, and CHD groups (P < 0.001; Figures 1A and 1B), with highest levels in patients with SSc-PAH. The majority of the patients with SSc-PAH (69.1%) and patients with CTD (62.7%) were positive for anti-AT1R Abs (cut-off, 9.2 units) as compared with patients with IPAH (21%), CTEPH (8%), and CHD (21.4%) (11). In addition, 65.4% of the patients with SSc-PAH and 54.5% of the patients with CTD, yet only 11.3% of the patients with IPAH, none of the patients with CTEPH, and 14.3% of the patients with CHD, were positive for anti-ETAR Abs (cut-off, 10.4 units) (11). The antibody levels were overall equally high or even slightly higher in patients with SSc that did not develop PAH (Table 1). The Abs did not show significant correlations (r2 > 0.2) with hemodynamic parameters (PVR, CaI, or mPAP) or NT-proBNP levels (data not shown). However, a strong correlation between anti-AT1R and anti-ETAR Abs was observed in patients with SSc-PAH (r2 = 0.81, Figure 1C). Results of ROC curve analyses show sensitivity, specificity, and AUC values (Table 2). All differences of the groups shown were highly significant (P < 0.001, respectively).

Table 2. Capability of the Antibodies to Discriminate between Different Cohorts as Analyzed by Receiver-Operating Characteristic Analyses

Question AddressedAnti-AT1R Abs Sensitivity/Specificity in % (AUC)Anti-ETAR Abs Sensitivity/Specificity in % (AUC)
SSc-PAH vs. IPAH68.8/85.5 (0.772)72.5/85.5 (0.786)
Non–SSc-PAH vs. SSc-PAH68.8/78.0 (0.735)70.0/82.4 (0.754)
CTD-PAH vs. non–CTD-PAH62.4/82.5 (0.760)72.5/78.1 (0.786)

Definition of abbreviations: Abs = antibodies; AT1R = angiotensin receptor type-1; AUC = area under the curve; CTD = connective tissue disease; CTD-PAH = all patients with CTD including SSc; ETAR = endothelin receptor type A; IPAH = idiopathic pulmonary arterial hypertension; non–SSc-PAH = all patients with PAH without SSc; PAH = pulmonary arterial hypertension; SSc = systemic sclerosis.

Data are shown as: sensitivity/specificity (AUC).

Anti-AT1R Abs and Anti-ETAR Abs Predict SSc-PAH Development

During prospective follow-up of 253 consecutive patients with SSc at the department of Rheumatology, 36 patients were diagnosed with PAH (mean observation period: 73 mo). Epidemiologic and hemodynamic data of the prospective PAH-SSc cohort were not different from the cross-sectional SSc-PAH cohorts (data not shown). By comparison of various clinical parameters for the ability to predict PAH, AUC was best for systolic pulmonary arterial pressure (sPAP) and predicted values of DlCO-SB. The predictive capacity of anti-AT1R and anti-ETAR Ab levels was comparable to NT-proBNP values (Figure 2A). The best calculated cut-off from the ROC analysis was then used for a Kaplan-Meier analysis, shown in Figure 2B. Both anti-AT1R (cut-off, 19.0 units; P < 0.0001), as well as anti-ETAR Abs (cut-off, 23.0 units; P = 0.0056), predicted development of PAH in patients with SSc. The autoantibodies were equally predictive as a FVC:DlCO ratio greater than 1.6 (P = 0.046; Figure 2C) or DlCO less than 55% (P < 0.0001; Figure 2D), that are known risk factors for PAH (23). For anti-AT1R– and anti-ETAR–positive patients with SSc, there was an increased risk for PAH with a hazard ratio of 4.3 (95% confidence interval [CI] = 2.2–8.4) and of 3.5 (CI = 1.51–5.60), respectively. However, in a multivariate analysis, sPAP, as detected by echocardiography, was the only independent predictor for PAH proven by RHC (P < 0.001; odds ratio = 1.102; CI = 1.055–1.152).

Anti-AT1R and Anti-ETAR Ab Predict Mortality in Patients with SSc-PAH

We analyzed the autoantibody levels in those 36 patients from our own cohort for their capacity to discriminate between patients that died and those who did not and the ROC AUC was 0.766 for the anti-AT1R Ab and 0.662 for the anti-ETAR Ab (data not shown). In addition, we had follow-up data from 34 patients with SSc-PAH from the Hannover cohort (70 patients with SSc-PAH in total). By using an ROC analysis and cut-off calculation, anti-AT1R Ab levels above 15.8 units predicted mortality with a sensitivity of 68.2% and a specificity of 62.2% (AUC = 0.669; P = 0.03). For the anti-ETAR antibodies, a cut-off of 18.3 units was associated with mortality, with a sensitivity of 68.2% and a specificity of 71.1% (AUC = 0.672; P = 0.02). As shown in Figure 3A, autoantibody levels against ETAR and AT1R predicted mortality better than the hemodynamic parameters mPAP, CaI, or PVR (each at PAH diagnosis). Kaplan-Meier survival analysis with respective cut-off levels (Figure 3A) showed a significantly different survival for anti-ETAR Ab–positive and –negative patients (P = 0.027; hazard ratio = 2.7; CI = 1.2–6.1; Figure 3B). This was not statistically significant for the anti-AT1R Abs in this cohort (P = 0.13). However, when the hemodynamic data (mPAP, CaI, and PVR) were compared with the anti-AT1R and anti-ETAR Ab levels in a multivariate analysis, anti-AT1R Abs were the only independent predictors of death (P = 0.03; odds ratio = 1.053; CI = 1.004–1.104). Autoantibody status in other PAH/PH entities (patients with CTD in general, patients without CTD, all patients with PH/PAH) did not reveal differences in survival.

Anti-AT1R and Anti-ETAR Abs Elicit Agonist Responses and Augment Vasoconstriction to Ang II and ET-1 in the Rat Pulmonary Vasculature

To evaluate the potential of these autoantibodies to elicit functional agonist effects in the intact pulmonary vasculature, we imaged endothelial [Ca2+]I as an established and assessable downstream signal of AT1R or ETAR activation during infusion of SSc and control IgG. SSc-IgG induced a marked increase in endothelial [Ca2+]i that was not detected after infusion of control IgG (Figures 4A–4C) and could be blocked by both valsartan and sitaxsentan (Figure 4D).

In small-vessel myography of intralobar pulmonary rat artery ring segments, anti-AT1R and anti-ETAR Abs amplified the vasoconstrictive responsiveness to ET-1 and Ang II (Figures 4E and 4F). Pretreatment of the vessels with bosentan abolished the response to Ang II (Figure 4E), whereas AT1R antagonist valsartan (Figure 4E) reduced responses to ET-1 (Figure 4F).

Transfer of PAH-SSc-IgG Induces Pulmonary Arteriopathy in Healthy Mice

For assessment of the pathophysiologic relevance of anti-AT1R and anti-ETAR Abs in vivo, SSc-IgG or healthy control (HC) IgG were injected into 7-week-old C57BL/6 mice (Figure 5). Human IgG was detected in frozen lung sections 7 days after a single injection of SSc-IgG, but not with HC-IgG (Figure 5A, shown in red), implicating Ab binding in murine lung tissue. In parallel, increased α-SMA expression was observed around the walls of small pulmonary vessels adjacent to airways in the SSc-IgG–treated group as compared with HC-IgG–treated mice (Figure 5B).

Repetitive injections of SSc-IgG (n = 8), HC-IgG (n = 7), and vehicle (n = 7) were administered in long-term pilot experiments over a period of 100 days. Obliterative pulmonary arteriopathy of various extent and increased cellular infiltrates were observed in mice injected with SSc-IgG, and were either not observed or observed to a lesser degree in controls (Figures 5C–5E). Fulton index (right ventricular weight over left ventricular plus interventricular septum weight ratio, RV/LV + S) and PVR were not different compared with controls.

We identified elevated autoantibodies against AT1R and ETAR in patients with SSc-PAH and CTD-PAH as compared with patients with IPAH or other PH entities. Simultaneously present anti-AT1R and anti-ETAR antibodies in patients with SSc predicted development of PAH and its associated mortality. Both receptor autoantibodies act as agonists by means of endothelial responses in isolated rat lungs and augmented vasoconstriction in response to natural ligands in rat pulmonary resistance vessels. Passive transfer of anti-AT1R and anti-ETAR antibody–positive human SSc-IgG increased α-SMA expression in healthy mice in concordance with human IgG detection within lung tissue 7 days after injection. Repetitive injections of SSc-IgG induced obliterative vasculopathy with perivascular lymphocyte infiltration. Our data unify autoimmune receptor activation processes with increased vascular reactivity and vascular remodeling. These data expand on our previous findings concerning the molecular and cellular effects of anti-AT1R and anti-ETAR autoantibodies on immune cells, endothelial cells, and fibroblasts, as well as their pathogenic role in mice after antibody transfer (12, 13).

Recently, IgG from both IPAH and patients with SSc-PAH was found to recognize antigens in vascular smooth muscle cells and fibroblasts (24, 25). The protein extraction and two-dimensional electrophoresis precluded isolation of membrane antigens, such as AT1R and ETAR (24, 25). Unlike intracellular antigens, functional consequences of autoimmune-mediated AT1R and ETAR activation are pleiotropic, and range from increase in endothelial [Ca2+]i influx, and enhanced vasoconstrictive responses to natural ligands, to the enhancement of chronic remodeling process. Vascular hyperreactivity, obliterative microvascular disease, and interstitial fibrosis are prominent features of SSc pulmonary involvement, which could all be influenced by anti-AT1R and anti-ETAR Abs. Autoantibody-mediated receptor cross-talk, together with enhanced reactivity to natural ligands, could represent an additional pathophysiological aspect of vascular dysregulation (22). The simultaneous occurrence of anti-AT1R and anti-ETAR Abs implies the importance of possible receptor heterodimerization, which could affect the influence of agonists and antagonists on signal transduction (26).

We are aware that our experimental data derived from mice injected with anti-AT1R and anti-ETAR containing IgG isolated from representative patients pose several limitations. In these preliminary experiments, we were not able to document increases in mPAP or PVR, or induction of right ventricular hypertrophy. However, endothelial cell activation, enhanced vasoconstriction, small-vessel obliteration, with increased α-SMA expression and perivascular infiltrates, indicate a vasculopathy that partly resembles changes seen in hypoxia and monocrotaline PAH models (27). Altered hemodynamics and right ventricular hypertrophy are also not commonly observed in early stages of other pulmonary hypertension models (27, 28). The utility and relevance of the Abs against AT1R and ETAR as predictive biomarkers of SSc-PAH development was underscored by similar sensitivity values as documented for NT-proBNP values, FVC:DlCO ratios, and DlCO values less than 55%, established predictors for SSc-PAH (2932). Concordant with this published work, DlCO and sPAP values were best predictors for PAH in our study. We suggest that the measurement of autoantibody levels could provide an additional important prognostic tool, together with echocardiography, with the calculation of sPAP and a detailed lung function analysis (33). We are aware of the putative limitation that our study focused on very specific patient cohorts treated at tertiary care centers. However; our SSc-PAH cohorts were representative of other cohorts, as reflected by lower mPAP and PVR values and higher CaIs in patients with SSc-PAH as compared with patients with IPAH (3032).

The clinical importance of the Abs against AT1R and ETAR in patients with SSc-PAH is further underscored by the fact that they were predictive of death. As hemodynamic parameters failed to predict death in other cohorts as well (30), our data indicate that the anti-AT1R and anti-ETAR Abs may have a role in risk stratification of SSc-PAH. In the era of personalized medicine, algorithms based on multiple parameters are increasingly valued for the identification of patients at risk, as recently demonstrated (33).

Although therapeutic implications of our findings remain to be established, the current study implies a contribution of functional autoimmunity to the development of PAH-associated vasculopathy. Immunosuppressive treatment with cyclophosphamide alone did not improve hemodynamic parameters in SSc-PAH (35). In the setting of heart and kidney transplantation, where AT1R autoantibodies are well established as diagnostic and prognostic biomarkers, intensified triple immunosuppression and AT1R blockade seem to be effective for the majority (36, 37), although some patients with fulminant phenotypes also required antibody removal by plasmapheresis (14).

In conclusion, our study suggests an involvement of functional autoantibody-mediated vascular receptor activation in SSc-PAH, and emphasizes anti-AT1R and anti-ETAR Abs as novel prognostic markers for CTD-PAH and SSc-PAH.

1. Simonneau G, Robbins IM, Beghetti M, Channick RN, Delcroix M, Denton CP, Elliott CG, Gaine SP, Gladwin MT, Jing ZC, et al. Updated clinical classification of pulmonary hypertension. J Am Coll Cardiol 2009; 54(Suppl1):S43S54.
2. Badesch DB, Feldman J, Keogh A, Mathier MA, Oudiz RJ, Shapiro S, Farber HW, McGoon M, Frost A, Allard M, et al.; ARIES-3 Study Group. ARIES-3: ambrisentan therapy in a diverse population of patients with pulmonary hypertension. Cardiovasc Ther 2012;30:9399.
3. Chung L, Liu J, Parsons L, Hassoun PM, McGoon M, Badesch DB, Miller DP, Nicolls MR, Zamanian RT. Characterization of connective tissue disease–associated pulmonary arterial hypertension from REVEAL: identifying systemic sclerosis as a unique phenotype. Chest 2010;138:13831394.
4. Waxman AB, Zamanian RT. Pulmonary arterial hypertension: new insights into the optimal role of current and emerging prostacyclin therapies. Am J Cardiol 2013;111(Suppl5):1A16A, quiz 17A–19A.
5. Galié N, Manes A, Branzi A. The endothelin system in pulmonary arterial hypertension. Cardiovasc Res 2004;61:227237.
6. Davie NJ, Schermuly RT, Weissmann N, Grimminger F, Ghofrani HA. The science of endothelin-1 and endothelin receptor antagonists in the management of pulmonary arterial hypertension: current understanding and future studies. Eur J Clin Invest 2009;39:3849.
7. de Man FS, Handoko ML, Guignabert C, Bogaard HJ, Vonk-Noordegraaf A. Neurohormonal axis in patients with pulmonary arterial hypertension: friend or foe? Am J Respir Crit Care Med 2013;187:1419.
8. de Man FS, Tu L, Handoko ML, Rain S, Ruiter G, François C, Schalij I, Dorfmüller P, Simonneau G, Fadel E, et al. Dysregulated renin–angiotensin–aldosterone system contributes to pulmonary arterial hypertension. Am J Respir Crit Care Med 2012;186:780789.
9. Xie L, Lin P, Xie H, Xu C. Effects of atorvastatin and losartan on monocrotaline-induced pulmonary artery remodeling in rats. Clin Exp Hypertens 2010;32:547554.
10. El Chami H, Hassoun PM. Immune and inflammatory mechanisms in pulmonary arterial hypertension. Prog Cardiovasc Dis 2012;55:218228.
11. Riemekasten G, Philippe A, Näther M, Slowinski T, Müller DN, Heidecke H, Matucci-Cerinic M, Czirják L, Lukitsch I, Becker M, et al. Involvement of functional autoantibodies against vascular receptors in systemic sclerosis. Ann Rheum Dis 2011;70:530536.
12. Kill A, Tabeling C, Undeutsch R, Kühl AA, Günther J, Radic M, Becker MO, Heidecke H, Worm M, Witzenrath M, et al. Autoantibodies to angiotensin and endothelin receptors in systemic sclerosis induce cellular and systemic events associated with disease pathogenesis. Arthritis Res Ther 2014;16:R29.
13. Günther J, Kill A, Becker MO, Heidecke H, Rademacher J, Siegert E, Radić M, Burmester GR, Dragun D, Riemekasten G. Angiotensin receptor type 1 and endothelin receptor type A on immune cells mediate migration and the expression of IL-8 and CCL18 when stimulated by autoantibodies from systemic sclerosis patients. Arthritis Res Ther 2014;16:R65.
14. Dragun D, Müller DN, Bräsen JH, Fritsche L, Nieminen-Kelhä M, Dechend R, Kintscher U, Rudolph B, Hoebeke J, Eckert D, et al. Angiotensin II type 1-receptor activating antibodies in renal-allograft rejection. N Engl J Med 2005;352:558569.
15. Becker M, Kill A, Undeutsch R, Tabeling C, Witzenrath M, Kuebler W, Bock S, Sampati R, Heidecke H, Ghofrani H, et al. Pathogenic effects of autoantibodies against vascular receptors in patients with SSc [abstract]. Rheumatology 2012;51 (Suppl 2):1123.
16. Becker M, Kill A, Kutsche M, Günther J, Rose A, Tabeling C, Witzenrath M, Kühl A, Heidecke H, Ghofrani H, et al. Autoantibodies targeting angiotensin type 1 and endothelin type A receptors as biomarkers and mediators of systemic sclerosis associated pulmonary arterial hypertension [abstract]. Clin Exp Rheumatol 2014;32:2354.
17. van den Hoogen F, Khanna D, Fransen J, Johnson SR, Baron M, Tyndall A, Matucci-Cerinic M, Naden RP, Medsger TA Jr, Carreira PE, et al. 2013 classification criteria for systemic sclerosis: an American college of Rheumatology/European League against Rheumatism collaborative initiative. Ann Rheum Dis 2013;72:17471755.
18. Galiè N, Torbicki A, Barst R, Dartevelle P, Haworth S, Higenbottam T, Olschewski H, Peacock A, Pietra G, Rubin LJ, et al.; The Task Force on Diagnosis and Treatment of Pulmonary Arterial Hypertension of the European Society of Cardiology. Guidelines on diagnosis and treatment of pulmonary arterial hypertension. Eur Heart J 2004;25:22432278.
19. Samapati R, Yang Y, Yin J, Stoerger C, Arenz C, Dietrich A, Gudermann T, Adam D, Wu S, Freichel M, et al. Lung endothelial Ca2+ and permeability response to platelet-activating factor is mediated by acid sphingomyelinase and transient receptor potential classical 6. Am J Respir Crit Care Med 2012;185:160170.
20. Kerem A, Yin J, Kaestle SM, Hoffmann J, Schoene AM, Singh B, Kuppe H, Borst MM, Kuebler WM. Lung endothelial dysfunction in congestive heart failure: role of impaired Ca2+ signaling and cytoskeletal reorganization. Circ Res 2010;106:11031116.
21. Kuebler WM, Parthasarathi K, Lindert J, Bhattacharya J. Real-time lung microscopy. J Appl Physiol (1985) 2007;102:12551264.
22. Lukitsch I, Kehr J, Chaykovska L, Wallukat G, Nieminen-Kelhä M, Batuman V, Dragun D, Gollasch M. Renal ischemia and transplantation predispose to vascular constriction mediated by angiotensin II type 1 receptor-activating antibodies. Transplantation 2012;94:813.
23. Hsu VM, Chung L, Hummers LK, Wigley F, Simms R, Bolster M, Silver R, Fischer A, Hinchcliff ME, Varga J, et al. Development of pulmonary hypertension in a high-risk population with systemic sclerosis in the Pulmonary Hypertension Assessment and Recognition of Outcomes in Scleroderma (PHAROS) cohort study. Semin Arthritis Rheum 2014;44:5562.
24. Bussone G, Tamby MC, Calzas C, Kherbeck N, Sahbatou Y, Sanson C, Ghazal K, Dib H, Weksler BB, Broussard C, et al. IgG from patients with pulmonary arterial hypertension and/or systemic sclerosis binds to vascular smooth muscle cells and induces cell contraction. Ann Rheum Dis 2012;71:596605.
25. Terrier B, Tamby MC, Camoin L, Guilpain P, Broussard C, Bussone G, Yaïci A, Hotellier F, Simonneau G, Guillevin L, et al. Identification of target antigens of antifibroblast antibodies in pulmonary arterial hypertension. Am J Respir Crit Care Med 2008;177:11281134.
26. Barnes PJ. Receptor heterodimerization: a new level of cross-talk. J Clin Invest 2006;116:12101212.
27. Stenmark KR, Meyrick B, Galie N, Mooi WJ, McMurtry IF. Animal models of pulmonary arterial hypertension: the hope for etiological discovery and pharmacological cure. Am J Physiol Lung Cell Mol Physiol 2009;297:L1013L1032.
28. Daley E, Emson C, Guignabert C, de Waal Malefyt R, Louten J, Kurup VP, Hogaboam C, Taraseviciene-Stewart L, Voelkel NF, Rabinovitch M, et al. Pulmonary arterial remodeling induced by a Th2 immune response. J Exp Med 2008;205:361372.
29. Hesselstrand R, Wildt M, Ekmehag B, Wuttge DM, Scheja A. Survival in patients with pulmonary arterial hypertension associated with systemic sclerosis from a Swedish single centre: prognosis still poor and prediction difficult. Scand J Rheumatol 2011;40:127132.
30. Condliffe R, Kiely DG, Peacock AJ, Corris PA, Gibbs JS, Vrapi F, Das C, Elliot CA, Johnson M, DeSoyza J, et al. Connective tissue disease–associated pulmonary arterial hypertension in the modern treatment era. Am J Respir Crit Care Med 2009;179:151157.
31. Humbert M, Yaici A, de Groote P, Montani D, Sitbon O, Launay D, Gressin V, Guillevin L, Clerson P, Simonneau G, et al. Screening for pulmonary arterial hypertension in patients with systemic sclerosis: clinical characteristics at diagnosis and long-term survival. Arthritis Rheum 2011;63:35223530.
32. Allanore Y, Borderie D, Avouac J, Zerkak D, Meune C, Hachulla E, Mouthon L, Guillevin L, Meyer O, Ekindjian OG, et al. High N-terminal pro-brain natriuretic peptide levels and low diffusing capacity for carbon monoxide as independent predictors of the occurrence of precapillary pulmonary arterial hypertension in patients with systemic sclerosis. Arthritis Rheum 2008;58:284291.
33. Coghlan JG, Denton CP, Grünig E, Bonderman D, Distler O, Khanna D, Müller-Ladner U, Pope JE, Vonk MC, Doelberg M, et al.; DETECT study group. Evidence-based detection of pulmonary arterial hypertension in systemic sclerosis: the DETECT study. Ann Rheum Dis 2014;73:13401349.
34. Colvin KL, Dufva MJ, Delaney RP, Ivy DD, Stenmark KR, Yeager ME. Biomarkers for pediatric pulmonary arterial hypertension - a call to collaborate. Front Pediatr 2014;2:7.
35. Sanchez O, Sitbon O, Jaïs X, Simonneau G, Humbert M. Immunosuppressive therapy in connective tissue diseases–associated pulmonary arterial hypertension. Chest 2006;130:182189.
36. Giral M, Foucher Y, Dufay A, Van Huyen JP, Renaudin K, Moreau A, Philippe A, Hegner B, Dechend R, Heidecke H, et al. Pretransplant sensitization against angiotensin II type 1 receptor is a risk factor for acute rejection and graft loss. Am J Transplant 2013;13:25672576.
37. Hiemann NE, Meyer R, Wellnhofer E, Schoenemann C, Heidecke H, Lachmann N, Hetzer R, Dragun D. Non-HLA antibodies targeting vascular receptors enhance alloimmune response and microvasculopathy after heart transplantation. Transplantation 2012;94:919924.

* These authors equally contributed to the manuscript.

Correspondence and requests for reprints should be addressed to Gabriela Riemekasten, M.D., Charité University Hospital, Rheumatology and Clinical Immunology, Charitéplatz 1, 10117 Berlin, Germany. E-mail: ; or Duska Dragun, M.D., Charité University Hospital, Nephrology and Intensive Care Medicine, Augustenburger Platz 1, 13353 Berlin, Germany. E-mail:

Supported by University Hospital Charité, Deutsche Forschungsgemeinschaft grant SFB-TR84 C3 and C6 (M.W.), European Community’s Seventh Framework Programme under grant agreement 241544–Systems Biology towards Novel Chronic Kidney Disease Diagnosis and Treatment (D.D.), Stiftung Sklerodermie e.V., Deutsches Netzwerk für systemische Sklerodermie (both G.R.), Heart & Stroke Foundation of Canada (W.M.K.), ARTICULUM Fellowship, Ministry of Industry grant KF2441006AJ3, CellTrend GmbH, and Actelion Pharmaceuticals Germany GmbH. The assay for measuring N-terminal pro-brain natriuretic peptide was kindly provided by Roche Germany.

Author Contributions: Conception and design—M.O.B., M.W., H.H., I.L., M.G., W.M.K., G.R.B., D.D., and G.R.; data acquisition—M.O.B., A.K., M.K., J.G., A.R., C.T., M.W., A.A.K., H.H., H.A.G., H.T., N.N., M.M.H., I.L., M.G., W.M.K., S.B., D.D., and G.R.; data analysis and interpretation—M.O.B., A.K., M.K., J.G., A.R., C.T., M.W., A.A.K., H.H., H.A.G., H.T., R.T.S., N.N., M.M.H., I.L., M.G., W.M.K., S.B., G.R.B., D.D., and G.R.; drafting or critically revising of the manuscript for important intellectual content—M.O.B., A.K., M.K., J.G., C.T., M.W., A.A.K., R.T.S., N.N., M.M.H., W.M.K., S.B., G.R.B., D.D., and G.R.

Originally Published in Press as DOI: 10.1164/rccm.201403-0442OC on September 2, 2014

Author disclosures are available with the text of this article at www.atsjournals.org.

Related

No related items
Comments Post a Comment




New User Registration

Not Yet Registered?
Benefits of Registration Include:
 •  A Unique User Profile that will allow you to manage your current subscriptions (including online access)
 •  The ability to create favorites lists down to the article level
 •  The ability to customize email alerts to receive specific notifications about the topics you care most about and special offers
American Journal of Respiratory and Critical Care Medicine
190
7

Click to see any corrections or updates and to confirm this is the authentic version of record