Abstract

Objectives

Scleroderma renal crisis (SRC) is a rare vascular complication of systemic sclerosis with substantial risks for end-stage renal disease and premature death. Activating autoantibodies (Abs) targeting the angiotensin II type 1 (AT1R) and the endothelin-1 type A receptor (ETAR) have been identified as predictors for SRC. Here, we sought to determine their pathogenic significance for acute renal vascular injury potentially triggering kidney failure and malignant hypertension.

Methods

IgG from patients with SRC was studied for AT1R and ETAR dependent biologic effects on isolated rat renal interlobar arteries and vascular cells including contraction, signalling and mechanisms of receptor activation.

Results

In myography experiments, patient IgG exerted vasoconstriction sensitive to inhibition of AT1R and ETAR. This relied on MEK-ERK signalling indicating functional relevance of anti-AT1R and anti-ETAR Abs. The contractile response to angiotensin II and endothelin-1 was amplified by patient IgG containing anti-AT1R and anti-ETAR Abs with substantial crosstalk between both receptors implicating autoimmune receptor hypersensitization. Co-immunoprecipitation experiments indicated heterodimerization between both receptor types which may enable the observed functional interrelation by direct structural interactions.

Conclusion

We provide experimental evidence that agonistic Abs may contribute to SRC. This effect is presumably related to direct receptor stimulation and additional allosteric effects, at least in heterodimeric receptor constellations. Novel therapies targeted at autoimmune hyperactivation of AT1R and ETAR might improve outcomes in severe cases of SRC.

Rheumatology key messages
  • IgG from SRC patients containing anti-AT1R and anti-ETAR autoantibodies exerts MEK-ERK dependent contraction of renal resistance arteries.

  • Crosstalk between AT1R and ETAR and autoantibody mediated hypersensitization may be enabled by receptor heterodimerization.

Introduction

SSc is a rheumatic disease of yet unknown pathogenesis characterized by autoantibodies directed against various cellular targets, fibrosis of the skin and internal organs, and severe obliterative vasculopathy [1, 2].

A potentially life-threatening vascular complication is scleroderma renal crisis (SRC) occurring in 4.9% of patients [3]. An acute decline of renal function is the hallmark of SRC. Hypertension occurs in about 90% of cases [4], often with blood pressure-related end-organ damage such as hypertensive retinopathy, encephalopathy with seizures and thrombotic microangiopathy (TMA) with haemolytic anaemia (MAHA) and thrombocytopenia [5]. In kidney biopsies from patients with SRC, acute and chronic vascular lesions are predominantly found in small vessels rather than in glomeruli [6]. Early findings are endothelial swelling, thrombosis and fibrinoid necrosis while chronic changes include onion-skin lesions, fibrointimal sclerosis and glomerulosclerosis [6]. Recurring or continuous endothelial injury in the arcuate and interlobular arteries results in intimal thickening and progressive luminal narrowing leading to decreased cortical blood flow [4]. This triggers renin production and activation of the renin-angiotensin-aldosterone system (RAAS). Expression of ET-1 [7] and its receptors [8] is enhanced and systemic ET-1 levels are elevated [9], whereas endothelial synthesis of prostacyclin is decreased [4]. This imbalance of vasoconstrictive and vasodilative factors culminates in endothelial damage and SRC.

Introduction of angiotensin-converting enzyme inhibitors (ACEI) in the late 1970s reduced the very high 1-year mortality rate from 76% to <15% and restored kidney function in >50% of patients initially requiring dialysis treatment in a US single-centre cohort [10]. However, despite routine use of ACEI in SRC, patient survival is still limited to 50–60% after 5 years [11] and a history of SRC is an independent predictor of 3-year mortality in patients with SSc [12]. Furthermore, 25–50% of SRC patients progress to end-stage renal disease [12].

Our group has described agonistic autoantibodies (Abs) directed against AT1R and ETAR in patients with SSc. High Abs levels were associated with an increased risk for vascular complications including SRC, pulmonary arterial hypertension and SSc-related death [13, 14]. Anti-AT1R and anti-ETAR Abs can activate microvascular endothelial cells [14], stimulate neutrophil migration [15], diminish endothelial wound repair [15] and have been attributed a pathogenic role in SSc-related pulmonary vasculopathy [13]. Recently, we demonstrated that these antibodies activate an Extracellular signal-Regulated Kinases 1/2 (ERK1/2)-Erythroblastosis virus E26 homologue transcription factor-1 (Ets-1)-Tissue Factor (TF) signalling cascade leading to endothelial cell proliferation [16].

Here, we hypothesized that agonistic anti-AT1R and anti-ETAR Abs found in patients with SSc contribute to the development of SRC not only by activation of endothelial cells but also by increasing contractility of renal interlobar arteries. Enhanced responsiveness to the natural ligands AngII and ET-1 due to Abs effects as well as synergistic responses were explored.

Patients and methods

Extended methods are available in the supplementary material available at Rheumatology online.

Isolation of human IgG

Endotoxin-free IgG was isolated from SSc patients with SRC and healthy controls as described previously [17]. The institutional review board of Charité Universitätsmedizin Berlin approved the study and all participants provided written informed consent.

Measurement of AT1R and ETAR abs levels

Levels of anti-AT1R and anti-ETAR Abs in IgG preparations were measured with a solid-phase sandwich ELISA using membrane extracts from Chinese Hamster Ovary cells overexpressing human AT1R or ETAR in their native conformation (CellTrend GmbH, Luckenwalde, Germany). Conformational epitopes were maintained by addition of 1 mM calcium.

Small resistance artery myography

All animal studies were conducted in accordance with local animal care guidelines. Arteries were harvested as previously described [18]. Ring segments from third to fourth-generation interlobar renal arteries were isolated from male Lewis rats and were mounted on a small vessel wire myograph (Multimyograph 610, Danish Myo Technology, Aarhus, Denmark). Contraction was normalized to the maximal response induced by 60 mM KCl set to 100%. Numbers of individual artery rings tested in each experiment are indicated in the figure legends.

Cell culture and Western blot analysis

Vascular smooth muscle cells (VSMC) from rat aorta, human microvascular endothelial cells (HMEC-1) and human umbilical vein endothelial cells (HUVEC) were cultured as described previously [19]. Cells were stimulated with IgG from healthy controls (Control IgG) or SRC patients (SRC IgG) for 15 min with or without pre-treatment with the AT1R blocker (AT1RB) valsartan or the ETA/BR blocker (ETA/BRB) bosentan at 10 µM for 60 min. Protein extraction, SDS–PAGE, transfer and membrane treatment were performed following standard procedures [20].

Luciferase reporter assay

HMEC-1 were transfected with human wild type AT1R and wild type ETAR cloned into pcDNA3 and a serum response element luciferase reporter plasmid (Promega, Madison, WI, USA). Cells were incubated with indicated combinations of Control IgG or SRC IgG (1.0 mg/ml), AngII (1000 nM) or ET-1 (100 nM), and bosentan or valsartan (10 µM). Luciferase production was measured with the Luciferase Assay System (Promega).

Immunoprecipitation

Three different clonal lines of transfected human embryonic kidney 293 cells (HEK293) stably overexpressing Flag-tagged ETAR and His-tagged AT1R (clones 7, 22 and 32) were generated. Cell lysates were incubated with either an anti-AT1R antibody (Santa Cruz Biotechnology, Dallas, TX, USA) or isotype control (Sigma-Aldrich, Saint Louis, Missouri, USA) coupled to agarose beads overnight at 4°C. Pelleted beads were then analysed by western blotting using anti-ETAR (BD Biosciences, Franklin Lakes, NJ, USA) or anti-FLAG antibodies (Sigma-Aldrich).

Statistical analysis

Results are expressed as means (s.e.m.). Multiple groups were compared using one-way or two-way analysis of variance (ANOVA). Post-testing was performed with Bonferroni’s multiple comparisons test. Correlations were calculated with the Pearson correlation coefficient. A two-sided P-value <0.05 was considered statistically significant.

Results

IgG from patients with SRC induces contraction in renal interlobar arteries via AT1R and ETAR

To test our hypothesis that agonistic Abs targeting the G-protein coupled receptors (GPCR) with vasoconstrictive activity AT1R and ETAR are pathophysiologically relevant in SRC, we studied contractile responses of small renal resistance arteries ex vivo. mRNA transcripts for AT1R and ETAR were more abundant in rat renal interlobar arteries than in total kidney extracts (Supplementary Fig. S1, available at Rheumatology online). Expression levels in interlobar arteries were comparable to those found in rat VSMC (Supplementary Fig. S1, available at Rheumatology online).

IgG purified from SRC patients who were ELISA-positive for anti-AT1R and anti-ETAR Abs (SRC IgG) was compared with IgG from healthy controls (Control IgG). SRC IgG dose-dependently induced contraction of renal interlobar arteries whereas Control IgG did not (Fig. 1A).

Contraction of isolated rat renal interlobar arteries in response to IgG from patients with SRC. Small vessel myography of artery rings exposed to (A) different concentrations of Control IgG or SRC IgG. n =12. *P <0.001 for SRC IgG versus Control IgG and for comparison with 0.06 mg/mL and 0.125 mg/mL, #P <0.01 for 1.0 mg/mL versus 0.25 mg/mL, P <0.05 for 1.0 mg/mL versus 0.5 mg/mL. (B) Myography of vessels exposed to 1.0 mg/mL Control IgG or SRC IgG after pre-treatment with an AT1RB (valsartan), an ETARB (sitaxsentan) or an ETA/BRB (bosentan). n =12. **P <0.01, ***P <0.001. Contraction is expressed as % of the maximal contraction in response to 60 mM KCl of each individual vessel. Mean±SEM. (C) Correlation of the contractile response of artery rings to concentrations of anti-AT1R Abs and (D) anti-ETAR Abs in IgG preparations from SRC patients. n =12. Abs: agonistic autoantibodies; AT1RB: angiotensin II type 1 receptor blocker; Control IgG: IgG isolated from healthy controls; ETARB: ET-1 type A receptor blocker; ETA/BRB: dual ET-1 type A and type B receptor blocker; SRC: scleroderma renal crisis; SRC IgG: IgG isolated from patients with SRC
Figure 1.

Contraction of isolated rat renal interlobar arteries in response to IgG from patients with SRC. Small vessel myography of artery rings exposed to (A) different concentrations of Control IgG or SRC IgG. n =12. *P <0.001 for SRC IgG versus Control IgG and for comparison with 0.06 mg/mL and 0.125 mg/mL, #P <0.01 for 1.0 mg/mL versus 0.25 mg/mL, P <0.05 for 1.0 mg/mL versus 0.5 mg/mL. (B) Myography of vessels exposed to 1.0 mg/mL Control IgG or SRC IgG after pre-treatment with an AT1RB (valsartan), an ETARB (sitaxsentan) or an ETA/BRB (bosentan). n =12. **P <0.01, ***P <0.001. Contraction is expressed as % of the maximal contraction in response to 60 mM KCl of each individual vessel. Mean±SEM. (C) Correlation of the contractile response of artery rings to concentrations of anti-AT1R Abs and (D) anti-ETAR Abs in IgG preparations from SRC patients. n =12. Abs: agonistic autoantibodies; AT1RB: angiotensin II type 1 receptor blocker; Control IgG: IgG isolated from healthy controls; ETARB: ET-1 type A receptor blocker; ETA/BRB: dual ET-1 type A and type B receptor blocker; SRC: scleroderma renal crisis; SRC IgG: IgG isolated from patients with SRC

Pre-treatment of the artery segments with the AT1RB valsartan partly abolished the contractile response to SRC IgG (Fig. 1B). Both the ETAR blocker (ETARB) sitaxsentan and the dual ETA/BRB bosentan completely prevented SRC IgG induced vasoconstriction (Fig. 1B). Contractile responses of the artery rings were positively correlated to the concentrations of anti-AT1R and anti-ETAR Abs in the individual IgG preparations from SRC patients (r = 0.596, P < 0.001 for anti-AT1R Abs, r = 0.790, P = 0.002 for anti-ETAR Abs; Fig. 1C and D). These results indicate that contraction of renal resistance arteries induced by anti-AT1R and anti-ETAR Abs-positive IgG from SRC patients is mediated by AT1R and ETAR.

Autoantibody-induced vasoconstriction involves AT1R and ETA/BR-dependent activation of the MEK-ERK pathway

Since activation of ERK1/2 by agonistic anti-AT1R and anti-ETAR Abs from SSc patients has been described in HMEC-1 [14, 16] and this pathway has been implicated in AngII and ET-1 induced arterial vasoconstriction [21], we analysed its relevance for autoimmune modulation of renal vasoconstriction in SRC. In rat VSMC, we observed a 2–3-fold increase in ERK1/2 phosphorylation in response to SRC IgG compared with Control IgG (Fig. 2A and B). Inhibition of AT1R or ETA/BR prevented IgG-induced ERK1/2 phosphorylation (Fig. 2A and B). Similar effects were also found in HUVEC (Supplementary Fig. S2, available at Rheumatology online).

Involvement of the MEK-ERK pathway in autoantibody-induced constriction of isolated rat interlobar arteries. (A, B) Western blots for phosphorylated ERK1/2 from rat vascular smooth muscle cells stimulated with Control IgG or SRC IgG. Cells were pre-treated with (A) an AT1RB (valsartan) or (B) an ETA/BRB (bosentan). GAPDH served as a loading control. Blots of one representative experiment out of seven individual experiments are shown. Band intensities of ERK1/2 were divided by band intensities of GAPDH. Control IgG from each individual experiment was set to 1. n =7. (C) Contraction of rat renal interlobar artery rings upon exposure to Control IgG or SRC IgG ± MEKB expressed as % of the maximal contraction in response to 60 mM KCl. n =12. Mean±SEM. *P <0.05, **P <0.01, ***P <0.001. AT1RB: angiotensin II type 1 receptor blocker; Control IgG: IgG isolated from healthy controls; ETA/BRB: dual ET-1 type A and type B receptor blocker; GAPDH: Glyceraldehyde 3-phosphate dehydrogenase; MEKB: MEK blocker; MEK-ERK: mitogen-activated protein kinase-extracellular signal-regulated kinase; SRC IgG: IgG isolated from patients with scleroderma renal crisis
Figure 2.

Involvement of the MEK-ERK pathway in autoantibody-induced constriction of isolated rat interlobar arteries. (A, B) Western blots for phosphorylated ERK1/2 from rat vascular smooth muscle cells stimulated with Control IgG or SRC IgG. Cells were pre-treated with (A) an AT1RB (valsartan) or (B) an ETA/BRB (bosentan). GAPDH served as a loading control. Blots of one representative experiment out of seven individual experiments are shown. Band intensities of ERK1/2 were divided by band intensities of GAPDH. Control IgG from each individual experiment was set to 1. n =7. (C) Contraction of rat renal interlobar artery rings upon exposure to Control IgG or SRC IgG ± MEKB expressed as % of the maximal contraction in response to 60 mM KCl. n =12. Mean±SEM. *P <0.05, **P <0.01, ***P <0.001. AT1RB: angiotensin II type 1 receptor blocker; Control IgG: IgG isolated from healthy controls; ETA/BRB: dual ET-1 type A and type B receptor blocker; GAPDH: Glyceraldehyde 3-phosphate dehydrogenase; MEKB: MEK blocker; MEK-ERK: mitogen-activated protein kinase-extracellular signal-regulated kinase; SRC IgG: IgG isolated from patients with scleroderma renal crisis

To test ERK1/2 activation mediated by anti-AT1R and anti-ETAR Abs containing patient IgG translates into contraction of renal interlobar arteries, we pre-treated the artery segments with an inhibitor of the MEK-ERK pathway. The contractile response to SRC IgG was diminished by 50% (Fig. 2C). Thus, the MEK-ERK pathway constitutes a functional link between autoimmune AT1R and ETAR stimulation and renal vasoconstriction in SRC.

Anti-AT1R and anti-ETAR abs hypersensitize renal interlobar arteries to AngII and ET-1

AngII and ET-1 have been ascribed crucial roles in the pathophysiology of SRC [4, 11]. To explore putative functional interactions with the agonistic Abs, we tested the response of renal interlobar artery rings to AngII and ET-1 alone and in combination with SRC IgG. Both AngII and ET-1 elicited a dose-dependent contractile response that was unaltered when the vessels were pre-incubated with Control IgG (Fig. 3A and B). However, exposure to agonistic Abs containing SRC IgG enhanced the response to AngII by 48–85% (Fig. 3A) and to ET-1 by 28–83% (Fig. 3B). Because the responses to the natural ligands in combination with the autoantibodies were supra-additive in comparison to separate stimulation, this indicated hypersensitization of both receptors to their natural ligands by binding of anti-AT1R and anti-ETAR Abs.

Hypersensitization of isolated rat renal interlobar arteries by agonistic autoantibodies targeting AT1R and ETAR. Myography of artery rings exposed to AngII (A) or ET-1 (B) ± 1.0 mg/ml Control IgG or SRC IgG. Contraction is expressed as % of the maximal contraction in response to 60 mM KCl of each individual vessel. Mean±SEM. (A) AT1RB: valsartan. n =6–18. *P <0.05 for SRC IgG versus Control IgG and versus SRC IgG+ AT1RB, #P <0.05 for SRC IgG versus AngII, §P <0.05 for 1000 nM versus 1 nM AngII, $P <0.001 for 1000 nM versus 10 nM AngII, %P <0.01 for 1000 nM versus 100 nM AngII, &P <0.001 for 100 nM versus 1 nM AngII, +P <0.05 for 100 nM versus 10 nM AngII. (B) ETARB: sitaxsentan, ETA/BRB: bosentan. n =4–11. *P <0.001 for SRC IgG versus ET-1 and versus Control IgG and versus SRC IgG+ETARB and versus SRC IgG+ETA/BRB, &P <0.001 for 100 nM versus 1 nM ET-1, +P <0.001 for 100 nM versus 10 nM ET-1,/P <0.001 for 10 nM versus 1 nM ET-1. AngII: angiotensin II; AT1RB: angiotensin II type 1 receptor blocker; Control IgG: IgG isolated from healthy controls; ETARB: ET-1 type A receptor blocker; ETA/BRB: dual ET-1 type A and type B receptor blocker; SRC IgG: IgG isolated from patients with scleroderma renal crisis
Figure 3.

Hypersensitization of isolated rat renal interlobar arteries by agonistic autoantibodies targeting AT1R and ETAR. Myography of artery rings exposed to AngII (A) or ET-1 (B) ± 1.0 mg/ml Control IgG or SRC IgG. Contraction is expressed as % of the maximal contraction in response to 60 mM KCl of each individual vessel. Mean±SEM. (A) AT1RB: valsartan. n =6–18. *P <0.05 for SRC IgG versus Control IgG and versus SRC IgG+ AT1RB, #P <0.05 for SRC IgG versus AngII, §P <0.05 for 1000 nM versus 1 nM AngII, $P <0.001 for 1000 nM versus 10 nM AngII, %P <0.01 for 1000 nM versus 100 nM AngII, &P <0.001 for 100 nM versus 1 nM AngII, +P <0.05 for 100 nM versus 10 nM AngII. (B) ETARB: sitaxsentan, ETA/BRB: bosentan. n =4–11. *P <0.001 for SRC IgG versus ET-1 and versus Control IgG and versus SRC IgG+ETARB and versus SRC IgG+ETA/BRB, &P <0.001 for 100 nM versus 1 nM ET-1, +P <0.001 for 100 nM versus 10 nM ET-1,/P <0.001 for 10 nM versus 1 nM ET-1. AngII: angiotensin II; AT1RB: angiotensin II type 1 receptor blocker; Control IgG: IgG isolated from healthy controls; ETARB: ET-1 type A receptor blocker; ETA/BRB: dual ET-1 type A and type B receptor blocker; SRC IgG: IgG isolated from patients with scleroderma renal crisis

Inhibition of AT1R abolished AngII induced vasoconstriction (Fig. 3A). Similarly, blockade of ETAR alone or ETA/BR completely precluded contraction upon exposure to ET-1 (Fig. 3B).

Crosstalk between AT1R and ETAR in response to anti-AT1R and anti-ETAR abs and natural ligands

In order to decipher the synergy between autoimmunity (anti-AT1R and anti-ETAR Abs) and the natural vasoconstrictors as a novel concept for SRC pathogenesis, we evaluated the response to SRC IgG in crossover blocking experiments where one receptor was stimulated with the natural agonist while the other receptor was pharmacologically inhibited.

When vessels were pre-treated with ETA/BRB prior to stimulation with SRC IgG in combination with AngII, the total contractile response was reduced by almost 60% compared with treatment with SRC IgG and AngII without receptor blockade (Fig. 4A). Vice versa, pre-treatment with AT1RB reduced the contraction induced by SRC IgG in combination with ET-1 to a similar amount (Fig. 4B). Remarkably, the observed crossover blocking effects exceeded the extent that could be attributed to stimulation of ETAR (Fig. 4A) or AT1R (Fig. 4B) by SRC IgG containing agonistic Abs. This observation was most prominent in the experiment with ET-1 and AT1RB: AT1RB reduced the maximal contraction elicited by SRC IgG together with ET-1 by 113.9 percentage points, whereas SRC IgG induced an additional contraction of only 5.9 percentage points over control IgG (Fig. 4B). With control IgG, crossover receptor blockade had no effect on vessel contraction (Supplementary Fig. S3, available at Rheumatology online). This indicates that inhibition of one of the receptors that is activated only by the agonistic Abs (e.g. AT1R) also impairs activation of the other receptor type (e.g. ETAR) that is stimulated by the agonistic Abs in combination with the natural ligand (e.g. ET-1). This finding clearly indicates a crosstalk between both receptors in activity regulation by ligand interaction.

Interdependence of AT1R and ETA/BR in the response of renal interlobar arteries to activating autoantibodies. Small vessel myography of artery rings exposed to 1.0 mg/mL Control IgG or SRC IgG and natural ligands with and without pretreatment with receptor blockers as indicated. (A) Additional stimulation with 1000 nM AngII±ETA/BRB (bosentan). n =11–18. **P <0.01, ***P <0.001. (B) Additional stimulation with 100 nM ET-1±AT1RB (valsartan). n =6–12. ***P <0.001 for all comparisons. Contraction is expressed as % of the maximal contraction in response to 60 mM KCl of each individual vessel. Mean±SEM. Ang II: angiotensin II; AT1RB: Ang II type 1 receptor blocker; Control IgG: IgG isolated from healthy controls; ETA/BRB: dual ET-1 type A and type B receptor blocker; SRC IgG: IgG isolated from patients with scleroderma renal crisis
Figure 4.

Interdependence of AT1R and ETA/BR in the response of renal interlobar arteries to activating autoantibodies. Small vessel myography of artery rings exposed to 1.0 mg/mL Control IgG or SRC IgG and natural ligands with and without pretreatment with receptor blockers as indicated. (A) Additional stimulation with 1000 nM AngII±ETA/BRB (bosentan). n =11–18. **P <0.01, ***P <0.001. (B) Additional stimulation with 100 nM ET-1±AT1RB (valsartan). n =6–12. ***P <0.001 for all comparisons. Contraction is expressed as % of the maximal contraction in response to 60 mM KCl of each individual vessel. Mean±SEM. Ang II: angiotensin II; AT1RB: Ang II type 1 receptor blocker; Control IgG: IgG isolated from healthy controls; ETA/BRB: dual ET-1 type A and type B receptor blocker; SRC IgG: IgG isolated from patients with scleroderma renal crisis

To identify the molecular pathway operative in the observed interplay of both receptors, we studied ERK1/2 activity with a luciferase assay under one-sided crossover receptor inhibition and simultaneous stimulation with SRC IgG in combination with AngII or ET-1. These experiments support that ERK1/2 activation elicited by SRC IgG in synergy with one of the natural agonists can be reduced by inhibition of the other receptor that is not occupied by a natural ligand (Supplementary Fig. S4A and B, available at Rheumatology online). On the other hand, combined stimulation with both natural ligands was less than additive (Supplementary Fig. S4C, available at Rheumatology online). Additional blockade of one of the two receptors resulted in ERK1/2 activity of the same magnitude as stimulation with the unblocked natural ligand alone (Supplementary Fig. S4C, available at Rheumatology online). Thus, functional crosstalk between AT1R and ETAR was evident with SRC IgG containing anti-AT1R and anti-ETAR Abs in combination with AngII and ET-1 but not with the natural ligands alone.

This interdependency of AT1R and ETAR activation in the presence of agonistic Abs and natural ligands implies a direct contact between the two receptors, known as heterodimeric arrangement [18]. We conducted co-immunoprecipitation studies to detect putative heterodimers formed by AT1R and ETAR. Total lysates from HEK293 cells transfected with His-tagged AT1R and FLAG-tagged ETAR were precipitated with anti-AT1R-antibodies or isotype control. Results from three independently generated clones selected for robust expression of His-tagged AT1R and FLAG-tagged ETAR are shown to exclude clone dependent effects. Staining of immunoblots from the precipitates with anti-ETAR or anti-FLAG antibodies revealed a distinct band at 52 kDa consistent with ETAR that was co-precipitated with anti-AT1R-antibodies but not isotype control (Fig. 5A and B). This strongly supports a close spatial relation of both receptors and suggests the formation of heterodimers in line with the observed functional reciprocal dependency between AT1R and ETAR.

Co-immunoprecipitation of ETAR with AT1R. (A, B) IP from lysates of HEK293 cells overexpressing His-tagged AT1R and FLAG-tagged ETAR with anti-AT1R-antibodies (anti-AT1R) or isotype control (isotype). The IB were developed either with an anti-ETAR antibody (A) or an anti-FLAG antibody (B). Results from three independent experiments with individual clones (numbers 7, 22 and 32) are shown. AT1R: angiotensin II type 1 receptor; ETAR: ET-1 type A receptor; IB: immunoblots; IP: immunoprecipitation
Figure 5.

Co-immunoprecipitation of ETAR with AT1R. (A, B) IP from lysates of HEK293 cells overexpressing His-tagged AT1R and FLAG-tagged ETAR with anti-AT1R-antibodies (anti-AT1R) or isotype control (isotype). The IB were developed either with an anti-ETAR antibody (A) or an anti-FLAG antibody (B). Results from three independent experiments with individual clones (numbers 7, 22 and 32) are shown. AT1R: angiotensin II type 1 receptor; ETAR: ET-1 type A receptor; IB: immunoblots; IP: immunoprecipitation

Discussion

Based on previous reports implicating agonistic autoantibodies targeting AT1R and ETAR in the pathogenesis of SSc [13–16], we aimed to determine their role in SRC. In ex vivo experiments with rat renal interlobar arteries, we found that IgG containing agonistic anti-AT1R and anti-ETAR Abs isolated from patients with SRC induced vasoconstriction via AT1R and ETAR. Anti-AT1R and anti-ETAR Abs positive IgG also hypersensitized the arteries to their natural ligands AngII and ET-1 priming them for an excessive contractile response. Furthermore, we found evidence for a crosstalk between AT1R and ETAR in the context of autoimmune receptor stimulation that can be explained by allosteric effects of agonistic Abs on the ligand-receptor interplay.

Our findings suggest that anti-AT1R and anti-ETAR Abs contribute to the development of SRC not only due to direct vasoconstrictive properties but also by amplifying the response of target organs such as small kidney arteries to AngII and ET-1. This mechanism can be considered particularly relevant, as both neurohumoral systems are activated in SRC [4, 11] and seem to be major drivers of detrimental pathophysiologic changes resulting in loss of kidney function and malignant hypertension. Anti-AT1R and anti-ETAR Abs from SSc patients have also been shown to augment vasoconstrictive responses to AngII and ET-1 in resistance arteries from rat lungs [13]. On the contrary, antibodies isolated from kidney transplant recipients with acute vascular rejection who were negative for donor-specific anti-HLA antibodies but positive for anti-AT1R Abs only increased the contractile response of rat renal arteries to AngII by trend [18]. These variances might be related to functional heterogeneity of agonistic autoantibodies arising in differing pathological environments such as preeclampsia [22, 23], kidney transplantation [18, 20] or SSc [14] that is probably caused by antibody specificity directed against different epitopes of the receptors [18]. In line with this, a bioassay using AT1R overexpressing Huh-7-cells revealed that anti-AT1R Abs can feature either stimulatory or inhibitory effects [24].

Furthermore, we found evidence for a functional crosstalk of AT1R and ETAR in the presence of IgG from SRC patients positive for anti-AT1R and anti-ETAR Abs. This indicates that both potent vasoconstrictive systems are highly interrelated in this condition and therapeutic targeting of just one of them might be suboptimal. Although anti-AT1R and anti-ETAR Abs are not found exclusively in SSc patients with SRC, they might predispose for extreme intrarenal vasoconstriction and endothelial damage when a second hit strikes such as local RAAS and endothelin activation.

Interaction of AT1R and ETAR function might have resulted in some apparent inconsistencies in our experiments. For example, while blockade of ETA/BR but not of AT1R resulted in complete abolishment of SRC IgG-induced vasoconstriction as shown in Fig. 1B, phosphorylation of ERK1/2 was totally prevented by the AT1RB and not by the ETA/BRB (Fig. 2A and B). Moreover, as recently shown, direct agonistic effects of anti-AT1R Abs or allosteric modulation of the receptors by the anti-AT1R Abs may depend on cell type and cellular function [25]. Accordingly, the Abs may induce different effects in primary ring segments vs isolated VSMC. Involved mechanisms might include varying receptor expression, receptor co-expression or the presence of different allosteric modulators [26].

Potential alterations of a GPCR adapting the response to binding partners that induce different conformational changes such as natural ligands, multiple types of agonistic autoantibodies, or different blocking molecules can be complex. They include differential recruitment of GPCR kinases, G-proteins or β-arrestins (biased signalling), surmountable and insurmountable antagonism, and short- or long-term desensitization [27, 28]. In addition, formation of homo- and/or heteromultimers of GPCRs has been associated with altered functionality in many cases [29] including enhanced signalling of AT1R in response to AngII [30, 31].

How to explain the mechanisms of interaction between both receptors and hypersensitization? Structural data from X-ray crystallography of the AT1/2 receptors and ETBR revealing details of ligand binding, antibody binding and putative dimeric arrangement [32] may help to understand the complex interplay of the different components.

First, AngII and ET-1 bind at their receptors deeply into a crevice between the extracellular loops 1–3 (EL1-3) as known from previously determined receptor-ligand complexes (Fig. 6A and reviewed by Speck et al. [32]). The transmembrane helices (H) and the N-terminus (Nt) cover the bound ligand from the extracellular site (Fig. 6A). In a homodimeric arrangement between two AT1R protomers, their contact surface is localized between H1-H2 and EL1 (Fig. 6B), one of the most observed dimeric interfaces in class A GPCR [33]. Additionally, for several GPCRs such as the melanocortin-4 receptor [34], it is known that dimer-arrangements can have allosteric effects on binding and signalling mutually between the protomers. This principal structural-functional relation in dimeric receptor units is also existent for heterodimeric GPCRs [35], as shown for the AT1R/prostaglandin F2α-receptor dimer with signalling variations for arrestin-recruitment [36]. Thus, a heterodimeric receptor arrangement can generally contribute to specific signalling effects in cells expressing both AT1R and ETAR.

Structures and structural models of Ang II type 1 receptor (AT1R) and ET-1 type A/B receptor (ETA/BR) complexes. Available structural information for AT1R and ETBR relevant for the current study are presented in A–C. (A) Left: the AT1R in complex with Ang II (PDB ID: 6os0 [40]); right: the active state ET-1/ETBR structure (orange) complex (PDB ID: 5glh [41]). (B) Homodimer of the active state AT1R bound with an Ang II analog (PDB ID: 6do1 [42]). (C) AT2R in complex with Fab and Ang II analog (PDB ID: 5xjm [37]). Receptors are presented as backbone cartoons, for ligands also side-chains are presented as sticks. (D) Complex model of a putative receptor heterodimer with bound agonistic ligands and Fab (surface). This model is based on structural information from A–C, whereby the Fab was oriented by superimposition to AT1R as observed in (C) for the AT2R complex. Moreover, an ETAR homology model together with the AT1R/Fab complex were superimposed with the AT1R homodimer as presented in (B). The translucent filled circle highlights putative contact regions between the Fab and both receptors. EL: extracellular loop; H8: helix 8; H1-7: transmembrane helices 1–7; IL: intracellular loop; Nt: N terminus
Figure 6.

Structures and structural models of Ang II type 1 receptor (AT1R) and ET-1 type A/B receptor (ETA/BR) complexes. Available structural information for AT1R and ETBR relevant for the current study are presented in A–C. (A) Left: the AT1R in complex with Ang II (PDB ID: 6os0 [40]); right: the active state ET-1/ETBR structure (orange) complex (PDB ID: 5glh [41]). (B) Homodimer of the active state AT1R bound with an Ang II analog (PDB ID: 6do1 [42]). (C) AT2R in complex with Fab and Ang II analog (PDB ID: 5xjm [37]). Receptors are presented as backbone cartoons, for ligands also side-chains are presented as sticks. (D) Complex model of a putative receptor heterodimer with bound agonistic ligands and Fab (surface). This model is based on structural information from AC, whereby the Fab was oriented by superimposition to AT1R as observed in (C) for the AT2R complex. Moreover, an ETAR homology model together with the AT1R/Fab complex were superimposed with the AT1R homodimer as presented in (B). The translucent filled circle highlights putative contact regions between the Fab and both receptors. EL: extracellular loop; H8: helix 8; H1-7: transmembrane helices 1–7; IL: intracellular loop; Nt: N terminus

Second, an AT2R-AngII analogue complex co-crystallized with an antibody-Fab has revealed a positive allosteric effect on signalling by the Fab [37]. Binding epitopes of this Fab fragment are located in the central part of EL2, the Nt and EL1 (Fig. 6C). Thus, the binding sites for Abs and endogenous ligand are not overlapping, but they are at the same structural entities like EL2 (ligand at the C-terminus of EL2, Abs at the central part of EL2). Based on these previous perspectives and insights, we suppose that the observed positive allosteric effects of agonistic Abs containing patient IgG can be explained by (i) Abs increasing the predisposition of the receptor to bind AngII by directly structurally altering the ligand binding site (e.g. at EL2), or (ii) increased signalling activity by bound Abs lowering the energetic barrier for the endogenous ligand to further stimulate the receptor thereby causing hyperstimulation ability. Such positive allosteric effect between the natural ligand and activating Abs [37] was also observed in this study for AT1R (Fig. 3A). We postulate that such mechanisms of positive allosterism may also exist between Abs and AT1R examined here because of high similarities in the sequences and structures of ligands and receptors involved. Moreover, heterodimerization of angiotensin and endothelin receptors is a widely known phenomenon [32].

Combining these insights on positive allosteric Abs effects with heterodimerization between AT1R and ETBR, we suggest the following scenario to explain hyperstimulating effects (Fig. 6A–D): agonistic ligands bind to their receptors, while activating Abs allosterically (laterally) support the activation at receptor protomers by structural modifications, such as close to or directly at EL2. In a heterodimeric receptor constellation, this effect should be accompanied by structural modifications at the dimeric interface (e.g. EL1) with potentially further allosteric (horizontal) positive effects on signalling. Interestingly, a heterodimeric complex (AT1R/ETAR)-ligand-Abs model (Fig. 6D) supposes a potentially extended scenario, where the Abs (Fab) bound at AT1R also contacts the second ETAR protomer. In this case, Abs trans-effects may occur at epitopes in EL1 and EL2, respectively. This idea is supported by findings of antibodies directly entering interleukin-4 receptor heterodimers [38] and by GPCR heterodimer selective Abs [39].

Conclusions

In conclusion, we report evidence for a pathophysiologic involvement of agonistic Abs directed against AT1R and ETAR in SRC. Pathogenic mechanisms include a direct contractile effect on renal resistance arteries as well as hypersensitization to the natural ligands AngII and ET-1 and mediation of a crosstalk between the two receptors. Further research is needed to elucidate the molecular mechanisms linking binding of Abs to AT1R and ETAR to the observed biologic effects in order to develop precise novel therapeutic strategies.

Supplementary data

Supplementary data are available at Rheumatology online.

Data availability statement

The data underlying this article will be shared on reasonable request to the corresponding author.

Funding

This work was supported by the Bundesministerium für Wirtschaft und Energie (BMWi, Federal Ministry for Economic Affairs and Energy; ZIM project KF2257305AJ1) and the Deutsche Stiftung Sklerodermie (DSS, German Scleroderma Foundation). G.K., P.S., A.P. and R.C. are funded by the Deutsche Forschungsgemeinschaft (DFG, German Research Foundation) Project-ID 394046635—SFB 1365 subproject A03 and Project-ID 421152132—SFB 1423, subproject A01 (to P.S.).

Disclosure statement: The authors have declared no conflicts of interest.

Acknowledgements

This paper is dedicated to the memory of Duska Dragun, whose devotion to science and to understanding the biology of angiotensin II type 1 receptor activating autoantibodies was unsurpassed.

References

1

Denton
CP
,
Khanna
D.
Systemic sclerosis
.
Lancet
2017
;
390
:
1685
99
.

2

Gabrielli
A
,
Avvedimento
EV
,
Krieg
T.
Scleroderma
.
N Engl J Med
2009
;
360
:
1989
2003
.

3

Hughes
M
,
Zanatta
E
,
Sandler
RD
,
Avouac
J
,
Allanore
Y.
Improvement with time of vascular outcomes in systemic sclerosis: a systematic review and meta-analysis study
.
Rheumatology
2022
;
61
:
2755
69
.

4

Bose
N
,
Chiesa-Vottero
A
,
Chatterjee
S.
Scleroderma renal crisis
.
Semin Arthritis Rheum
2015
;
44
:
687
94
.

5

Hoa
S
,
Stern
EP
,
Denton
CP
,
Hudson
M
;
Scleroderma Clinical Trials Consortium Scleroderma Renal Crisis Working Group Investigators of the Scleroderma Clinical Trials Consortium Scleroderma Renal Crisis Working Group
.
Towards developing criteria for scleroderma renal crisis: a scoping review
.
Autoimmun Rev
2017
;
16
:
407
15
.

6

Batal
I
,
Domsic
RT
,
Medsger
TA
,
Bastacky
S.
Scleroderma renal crisis: a pathology perspective
.
Int J Rheumatol
2010
;
2010
:
543704
.

7

Mouthon
L
,
Mehrenberger
M
,
Teixeira
L
et al.
Endothelin-1 expression in scleroderma renal crisis
.
Hum Pathol
2011
;
42
:
95
102
.

8

Kobayashi
H
,
Nishimaki
T
,
Kaise
S
et al.
Immunohistological study endothelin-1 and endothelin-A and B receptors in two patients with scleroderma renal crisis
.
Clin Rheumatol
1999
;
18
:
425
7
.

9

Penn
H
,
Quillinan
N
,
Khan
K
et al.
Targeting the endothelin axis in scleroderma renal crisis: rationale and feasibility
.
QJM
2013
;
106
:
839
48
.

10

Steen
VD
,
Costantino
JP
,
Shapiro
AP
,
Medsger
TA
Jr
.
Outcome of renal crisis in systemic sclerosis: relation to availability of angiotensin converting enzyme (ACE) inhibitors
.
Ann Intern Med
1990
;
113
:
352
7
.

11

Woodworth
TG
,
Suliman
YA
,
Li
W
,
Furst
DE
,
Clements
P.
Scleroderma renal crisis and renal involvement in systemic sclerosis
.
Nat Rev Nephrol
2016
;
12
:
678
91
.

12

Nagaraja
V.
Management of scleroderma renal crisis
.
Curr Opin Rheumatol
2019
;
31
:
223
30
.

13

Becker
MO
,
Kill
A
,
Kutsche
M
et al.
Vascular receptor autoantibodies in pulmonary arterial hypertension associated with systemic sclerosis
.
Am J Respir Crit Care Med
2014
;
190
:
808
17
.

14

Riemekasten
G
,
Philippe
A
,
Nather
M
et al.
Involvement of functional autoantibodies against vascular receptors in systemic sclerosis
.
Ann Rheum Dis
2011
;
70
:
530
6
.

15

Kill
A
,
Tabeling
C
,
Undeutsch
R
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
.

16

Catar
R
,
Herse-Naether
M
,
Zhu
N
et al.
Autoantibodies targeting AT1- and ETA-receptors link endothelial proliferation and coagulation via Ets-1 transcription factor
.
Int J Mol Sci
2021
;
23
:
244
.

17

Simon
M
,
Lücht
C
,
Hosp
I
et al.
Autoantibodies from patients with scleroderma renal crisis promote PAR-1 receptor activation and IL-6 production in endothelial cells
.
Int J Mol Sci
2021
;
22
:
11793
.

18

Lukitsch
I
,
Kehr
J
,
Chaykovska
L
et al.
Renal ischemia and transplantation predispose to vascular constriction mediated by angiotensin II type 1 receptor-activating antibodies
.
Transplantation
2012
;
94
:
8
13
.

19

Xu
S
,
Fu
J
,
Chen
J
et al.
Development of an optimized protocol for primary culture of smooth muscle cells from rat thoracic aortas
.
Cytotechnology
2009
;
61
:
65
72
.

20

Dragun
D
,
Muller
DN
,
Brasen
JH
et al.
Angiotensin II type 1-receptor activating antibodies in renal-allograft rejection
.
N Engl J Med
2005
;
352
:
558
69
.

21

Kobayashi
T
,
Nogami
T
,
Taguchi
K
,
Matsumoto
T
,
Kamata
K.
Diabetic state, high plasma insulin and angiotensin II combine to augment endothelin-1-induced vasoconstriction via ETA receptors and ERK
.
Br J Pharmacol
2008
;
155
:
974
83
.

22

Buttrup Larsen
S
,
Wallukat
G
,
Schimke
I
et al.
Functional autoantibodies against Endothelin-1 receptor type A and Angiotensin II receptor type 1 in patients with preeclampsia
.
Pregnancy Hypertens
2018
;
14
:
189
94
.

23

Wallukat
G
,
Homuth
V
,
Fischer
T
et al.
Patients with preeclampsia develop agonistic autoantibodies against the angiotensin AT1 receptor
.
J Clin Invest
1999
;
103
:
945
52
.

24

Bankamp
L
,
Preuß
B
,
Pecher
AC
et al.
Functionally active antibodies to the angiotensin II Type 1-receptor measured by a luminometric bioassay do not correlate with clinical manifestations in systemic sclerosis: a comparison with antibodies to vascular receptors and topoisomerase I detected by ELISA
.
Front Immunol
2021
;
12
:
786039
.

25

Yue
X
,
Yin
J
,
Wang
X
et al.
Induced antibodies directed to the angiotensin receptor type 1 provoke skin and lung inflammation, dermal fibrosis and act species overarching
.
Ann Rheum Dis
2022
;
81
:
1281
9
.

26

Gentry
PR
,
Sexton
PM
,
Christopoulos
A.
Novel allosteric modulators of G protein-coupled receptors
.
J Biol Chem
2015
;
290
:
19478
88
.

27

Rajagopal
S
,
Shenoy
SK.
GPCR desensitization: acute and prolonged phases
.
Cell Signal
2018
;
41
:
9
16
.

28

Rankovic
Z
,
Brust
TF
,
Bohn
LM.
Biased agonism: an emerging paradigm in GPCR drug discovery
.
Bioorg Med Chem Lett
2016
;
26
:
241
50
.

29

George
SR
,
O'Dowd
BF
,
Lee
SP.
G-protein-coupled receptor oligomerization and its potential for drug discovery
.
Nat Rev Drug Discov
2002
;
1
:
808
20
.

30

Quitterer
U
,
AbdAlla
S.
Discovery of pathologic GPCR aggregation
.
Front Med
2019
;
6
:
9
.

31

Quitterer
U
,
Fu
X
,
Pohl
A
et al.
Beta-arrestin1 prevents preeclampsia by downregulation of mechanosensitive AT1-B2 receptor heteromers
.
Cell
2019
;
176
:
318
33.e19. e319
.

32

Speck
D
,
Kleinau
G
,
Szczepek
M
et al.
Angiotensin and endothelin receptor structures with implications for signaling regulation and pharmacological targeting
.
Front Endocrinol
2022
;
13
:
880002
.

33

Audet
M
,
Bouvier
M.
Restructuring G-protein- coupled receptor activation
.
Cell
2012
;
151
:
14
23
.

34

Urizar
E
,
Montanelli
L
,
Loy
T
et al.
Glycoprotein hormone receptors: link between receptor homodimerization and negative cooperativity
.
EMBO J
2005
;
24
:
1954
64
.

35

Satake
H
,
Matsubara
S
,
Aoyama
M
,
Kawada
T
,
Sakai
T.
GPCR heterodimerization in the reproductive system: functional regulation and implication for biodiversity
.
Front Endocrinol
2013
;
4
:
100
.

36

Fillion
D
,
Devost
D
,
Sleno
R
,
Inoue
A
,
Hébert
TE.
Asymmetric recruitment of beta-arrestin1/2 by the angiotensin II Type I and prostaglandin F2alpha receptor dimer
.
Front Endocrinol
2019
;
10
:
162
.

37

Asada
H
,
Horita
S
,
Hirata
K
et al.
Crystal structure of the human angiotensin II type 2 receptor bound to an angiotensin II analog
.
Nat Struct Mol Biol
2018
;
25
:
570
6
.

38

Spangler
JB
,
Moraga
I
,
Jude
KM
,
Savvides
CS
,
Garcia
KC.
A strategy for the selection of monovalent antibodies that span protein dimer interfaces
.
J Biol Chem
2019
;
294
:
13876
86
.

39

Gomes
I
,
Gupta
A
,
Bushlin
I
,
Devi
LA.
Antibodies to probe endogenous G protein-coupled receptor heteromer expression, regulation, and function
.
Front Pharmacol
2014
;
5
:
268
.

40

Wingler
LM
,
Skiba
MA
,
McMahon
C
et al.
Angiotensin and biased analogs induce structurally distinct active conformations within a GPCR
.
Science
2020
;
367
:
888
92
.

41

Shihoya
W
,
Nishizawa
T
,
Okuta
A
et al.
Activation mechanism of endothelin ETB receptor by endothelin-1
.
Nature
2016
;
537
:
363
8
.

42

Wingler
LM
,
McMahon
C
,
Staus
DP
,
Lefkowitz
RJ
,
Kruse
AC.
Distinctive activation mechanism for angiotensin receptor revealed by a synthetic nanobody
.
Cell
2019
;
176
:
479
90.e12
.

Author notes

Aurélie Philippe and Rusan Catar contributed equally to this study.

This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://dbpia.nl.go.kr/pages/standard-publication-reuse-rights)

Supplementary data

Comments

0 Comments
Submit a comment
You have entered an invalid code
Thank you for submitting a comment on this article. Your comment will be reviewed and published at the journal's discretion. Please check for further notifications by email.