Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 Mar 14.
Published in final edited form as: Hypertension. 2011 May 16;58(1):77–84. doi: 10.1161/HYPERTENSIONAHA.111.171348

Angiotensin II Type 1 Receptor Antibodies and Increased Angiotensin II Sensitivity in Pregnant Rats

Katrin Wenzel 1, Augustine Rajakumar 1, Hannelore Haase 1, Nele Geusens 1, Norbert Hubner 1, Herbert Schulz 1, Justin Brewer 1, Lyndsay Roberts 1, Carl A Hubel 1, Florian Herse 1, Lydia Hering 1, Fatimunnisa Qadri 1, Carsten Lindschau 1, Gerd Wallukat 1, Robert Pijnenborg 1, Harald Heidecke 1, Gabriela Riemekasten 1, Friedrich C Luft 1, Dominik N Muller 1, Babette Lamarca 1, Ralf Dechend 1
PMCID: PMC3597080  NIHMSID: NIHMS443612  PMID: 21576625

Abstract

Pregnant women who subsequently develop preeclampsia are highly sensitive to infused angiotensin (Ang) II; the sensitivity persists postpartum. Activating autoantibodies against the Ang II type 1 (AT1) receptor are present in preeclampsia. In vitro and in vivo data suggest that they could be involved in the disease process. We generated and purified activating antibodies against the AT1 receptor (AT1-AB) by immunizing rabbits against the AFHYESQ epitope of the second extracellular loop, which is the binding epitope of endogenous activating autoantibodies against AT1 from patients with preeclampsia. We then purified AT1-AB using affinity chromatography with the AFHYESQ peptide. We were able to detect AT1-AB both by ELISA and a functional bioassay. We then passively transferred AT1-AB into pregnant rats, alone or combined with Ang II. AT1-AB activated protein kinase C-α and extracellular-related kinase 1/2. Passive transfer of AT1-AB alone or Ang II (435 ng/kg per minute) infused alone did not induce a preeclampsia-like syndrome in pregnant rats. However, the combination (AT1-AB plus Ang II) induced hypertension, proteinuria, intrauterine growth retardation, and arteriolosclerosis in the uteroplacental unit. We next performed gene-array profiling of the uteroplacental unit and found that hypoxia- inducible factor 1α was upregulated by Ang II plus AT1-AB, which we then confirmed by Western blotting in villous explants. Furthermore, endothelin 1 was upregulated in endothelial cells by Ang II plus AT1-AB. We show that AT1-AB induces Ang II sensitivity. Our mechanistic study supports the existence of an “autoimmune-activating receptor” that could contribute to Ang II sensitivity and possible to preeclampsia.

Keywords: preeclampsia, angiotensin II, immunology, autoimmune disease


Preeclampsia, namely hypertension and proteinuria after >20 weeks of pregnancy,1 affects 3% to 5% of all pregnancies and is the major cause of fetal and maternal morbidity and mortality.2 Children and mothers after a preeclamptic pregnancy are at long-term cardiovascular risk.3,4 A dysregulated renin-angiotensin (Ang) system is implicated.5,6 Pregnant women who subsequently develop preeclampsia are highly sensitive to infused Ang II,7,8 whereas pregnant women without preeclampsia are resistant.8 The increased Ang II sensitivity in preeclamptic patients persists postpartum.9 Activating autoantibodies against Ang II receptor 1 (AT1-AA) occur in preeclamptic patients.10,11 AT1-AAs induce several signaling mechanisms, including nuclear factor-κB, JAK-STAT (Janus kinase-signal transducer and activator of transcription), and the Nuclear factor of activated T-cell/calcineurin pathways.12,13 AT1-AAs from preeclamptic patients increase intracellular Ca2+, NADPH oxidase, and tumor necrosis factor-α.12 They also activate AT1 receptors on human trophoblasts, induce soluble vascular endothelial growth factor receptor 1, and soluble endoglin.13,14 Zhou et al13 showed that passive transfer of either total IgG or purified AT1-AAs induced a preeclamptic-like syndrome in pregnant mice. The disease was prevented by losartan or by a neutralizing 7-amino acid epitope peptide. LaMarca et al15 suggested that AT1-AAs increase blood pressure via endothelin 1 (ET-1). These studies together suggest that preeclampsia may result in part from autoantibody-induced AT1 receptor activation.11,13 Active immunization should be able to elicit such antibodies and cause a similar syndrome.16 Jahns et al17 demonstrated that generation of antibodies against the β-adrenergic receptor induced dilatative cardiomyopathy. Similar long-term active immunization experiments have also been performed for other G protein– coupled receptors.18,19 However, such experiments have not been done in pregnant rats. We generated and isolated AT1 antibodies (AT1-AB) in rabbits using the peptide sequence AFHYESQ of the second extracellular loop detected as a binding epitope of AT1-AAs from preeclamptic patients. We then characterized the AT1-ABs and investigated their effects in pregnant rats alone and in combination with infused Ang II.

Materials and Methods

AT1-AB Generation, Purification, and Functional Testing

We immunized rabbits with the peptide sequence AFHYESQ (Biosyntan GmbH, Berlin, Germany) to generate AT1-ABs. To purify the AT1-ABs from sera, the corresponding peptides were covalently bound to ε-aminocapryl agarose (Sigma-Aldrich, Munich, Germany) to yield epitope-specific affinity beads. The preparation of antibodies and the cardiomyocyte contraction assay were carried out as earlier described.20 AT1-ABs were detected by an AT1-AB ELISA. ELISA for α1-adrenergic and β1-adrenergic receptor autoantibodies were used as negative controls (CellTrend, Luckenwalde, Germany). Because AT1-ABs were raised in rabbits, we detected them with a peroxidase-labeled antirabbit IgG antibody (Johnson & Johnson).

Chinese hamster ovary (CHO) cells stably transfected with human AT1-receptor (CHO/AT1R) were cultured in F12 HAM medium supplemented with glutamine, 10% FCS, and 1% penicillin/streptomycin. Protein kinase C-α activity in CHO/AT1R cells was detected with AT1-ABs (2.5 and 25.0 µg/mL of medium) using an MRC 1024 confocal imaging system (Bio-Rad, Munich, Germany) with an argon/krypton laser.20 As positive control, the AT1-receptor agonist Ang II (100 nmol/L to 1 µmol/L) was used, and for inhibition experiments, irbesartan (1 µmol/L; Sanofi-Aventis, Paris, France) was used. For extracellular-regulated kinase 1/2 phosphorylation, CHO/AT1R cells were maintained in serum-free medium for 4 hours, respectively, and treated with AT1-ABs (25 and 50 µg/mL of medium) for 15 minutes, respectively. For inhibition experiments, 1 µmol/L of irbesartan was added. Western blot experiments were carried out as described earlier.20

Rats

Sprague-Dawley rats (R. Janvier Breeding Center, Le Genest St Isle, France) were outfitted with radiotelemetry pressure transducers (TA11PA-C20).18 Animals were then mated and pregnancy monitored. For Ang II infusion, osmotic pumps (ALZET, Cupertino, CA) were implanted 9 days later. The animals received 435 ng of Ang II per kilogram of body weight per minute. AT1-AB or rabbit control IgG were IV injected on days 9 and 11 of pregnancy (1 µg per gram of body weight, 6 animals per group). Rat serum was sampled on days 11 and 13 for rabbit IgG detection by Western blotting using a peroxidase-coupled antirabbit IgG antibody (Thermo Fisher Scientific, Bonn, Germany). We maintained the animals in metabolic cages on days 17 and 18 for 24-hour albumin excretion by ELISA (CellTrend GmbH). Animals were euthanized on day 20 of pregnancy. The quantitative analysis of depth of trophoblast invasion and staining for atherosis-like lesions has been described earlier.21,22 Briefly, all of the implantation sites (placenta with associated mesometrial triangle) were removed, fixed in JB-Fix fixative, and embedded. The whole mesometrial triangle was photographed, and the images were shade corrected. The lumen of each spiral artery cross-section was manually delineated, and stretches of endovascular trophoblast were traced separately over the lumen contour tracing. Local authorities (Berlin, Germany) approved the animal protocol that complied with criteria outlined by the American Physiological Society.

Gene Expression, Cell Culture, ELISA, and Data Analysis

We extracted total RNA from the mesometrial triangle and placenta using the RNeasy Purification kit (Qiagen). RNA was treated by deoxyribonuclease I (Qiagen). We compared the gene expression profile in the mesometrial triangle in nonpooled microarray experiments (RatRef-12 Expression BeadChip kit; 22 523 rat targets, Illumina). After quantile normalization without background correction (Illumina BeadStudio 1.0.2.20706 Platform), an offset addition16 and log-2 transformation were performed. The general expression profile over the 6 experimental conditions with respect to the 3 to 6 replicates was characterized using ANOVA statistic followed by false discovery rate multiple-testing correction. Consistently low-expressed transcripts were removed (BeadStudio detection P>0.05) before using test statistic. We found that 16 041 of 22 523 transcripts remained after filtering. A total of 998 transcripts were significantly differentially expressed at 5% false discovery rate level.

Human umbilical vein endothelial cells were pretreated with serum-free medium, Ang II (10−7 mol/L), AT1-AB, or Ang II + T1-AB for 6 hours. Endothelin 1 (ET-1) was measured using ELISA and normalized to total cellular protein per flask. For detection of soluble fms-like tyrosine kinase 1 (sFLT-1) and placenta-like growth factor (PLGF) protein concentration, soluble vascular endothelial growth factor R1 Immunoassay (R&D Systems GmbH, Wiesbaden-Nordenstadt, Germany) and placenta-like growth factor Immunoassay (R&D Systems GmbH) were used, respectively. Villous explant cultures, treatment, and Western blot analysis were performed as described earlier.23 We relied on the Mann-Whitney test at P<0.05 and expressed the data as mean±SD.

Results

AT1-AB Assays and Characteristics

AT1-ABs increased the beating rate of neonatal rat cardiomyocytes, similar to isolated autoantibodies from preeclamptic patients (AT1-AA). The positive chronotropic effect was blocked by the AT1 receptor antagonist irbesartan and by the AFHYESQ peptide, respectively (Figure 1A). Using the purified AT1-AB as capture antibody, we recovered a positive signal in a cell-based ELISA after CHO cells were transfected with AT1 receptor (CHO/AT1R), but not when CHO cells remained untransfected or were transfected with α1- or β1-adrenergic receptor (Figure 1B). We next examined the effects of AT1-AB on signal transduction in stably AT1 receptor–transfected cells (CHO/AT1R). We concentrated on protein kinase C-α and extracellular-regulated kinase 1/2, because both are important in AT1 receptor signaling. We found that AT1-AB exposure to CHO/AT1R resulted in protein kinase C-α activation, as did the positive Ang II control. Irbesartan blocked these effects (Figure 1C). Incubation of CHO/AT1R cells with AT1-ABs resulted in transient extracellular-regulated kinase 1/2 phosphorylation. The specificity of phosphorylation was shown by inhibition with irbesartan (Figure 1D).

Figure 1.

Figure 1

A, Autoantibodies against angiotensin II receptor 1 (AT1-AAs) from preeclamptic patients and antibodies against the AT1 receptor (AT1-AB) increased the cardiomyocyte spontaneous beating rate. Irbesartan (Irb; 1 µmol/L) and the AFHYESQ peptide inhibited. B, AT1-ABs (1 µg) were used in different concentrations to detect AT1, α-1, or β-1 receptor autoantibodies (α1-AB, β1-AB, and AT1-AB). A strong dose-dependent signal for AT1-AB was only present in the AT1-receptor autoantibody ELISA; α- and β-adrenergic receptors were not recognized. C, AT1-AB activated protein kinase C (PKC)-α in Chinese hamster ovary (CHO)/AT1 receptor (AT1R) cells; irbesartan (1 µmol/L) inhibited. Yellow shows PKC-α activation; red is more intense activation. D, Extracellular-regulated kinase (ERK) 1/2 was activated by AT1-AB. CHO/AT1R cells were treated for 15 minutes with 25 and 50 µg of AT1-AB, respectively. Lanes 1 and 2 represent untreated cells. Eukaryotic initiation factor 4E (elF4E) and ERK1/2 antibody were loading controls.

AT1-AB and Ang II in Pregnant Rats

We performed passive transfer experiments in pregnant rats. We verified the presence of the rabbit AT1-AB in the rat sera on days 11 and 13 by rabbit IgG detection in an immunoblot (Figure 2A). On day 20, AT1-ABs were detectable by ELISA (Figure 2B). AT1-AB or control rabbit IgG injection in pregnant rats did not influence blood pressure. Ang II at a high dose (435 ng/kg per minute) also did not increase blood pressure. However, AT1-AB plus Ang II resulted in a significant increase in mean arterial blood pressure in our pregnant rats (Figure 2C). We next performed acute Ang II infusions. Ang II (100 ng/mg per minute) was administered via the jugular vein to controls and chronic AT1-AB–infused pregnant rats on day 19 of gestation as a bolus. Blood pressure increased to 141±4 mm Hg in normal pregnant rats but markedly increased to 168±7 mmHg in AT1 -AB pretreated pregnant rats (Figure 2D). AT1-AB transfer did not induce albuminuria (317.3 µg/mL per 24 hours), and Ang II alone induced modest albuminuria in the rats compared with no treatment (2014.50 versus 352.33 µg/mL per 24 hours). However, Ang II plus AT1-AB produced albuminuria (4388.2 µg/mL/24 hours; P=0.0071; please see http://hyper.ahajournals.org, online Data Supplement Figure S1). Renal fibrosis was not present in any group (Figures S2 and S3).

Figure 2.

Figure 2

A, Immunoblot for rabbit IgG. Rabbit IgG was detected in the serum of antibodies against the angiotensin II (Ang II) type 1 receptor (AT1-ABs)–treated rats on day 11 and day 13 of pregnancy. B, ELISA for AT1-AB shows that detection was possible on day 20 in the serum of AT1-AB–treated rats. C, Telemetric blood pressures show that mean arterial blood pressure increased with Ang II infusion only when AT1-ABs were present. D, Acute blood pressure response to Ang II (100 ng/kg per minute) infusion as bolus is shown. Ang II induced a higher blood pressure response in AT1-AB pretreated rats vs control rats.

We determined the body, kidney, and placental and mesometrial triangle weights. There were no significant differences between the groups (data not shown). Ang II, IgG, or AT1-AB treatment did not influence the fetal weights, but fetal weights were significantly decreased in the Ang II plus AT1-AB–treated group. Fetal weights of the Ang II plus IgG-treated group were also lower compared with control (P≤0.05; Figure 3A). We also looked for intrauterine growth restriction. An increased brain:liver ratio is an important indicator for fetal stress resulting in intrauterine growth restriction. The brain:liver ratio was significantly increased only in the Ang II plus AT1-AB group. All of the other groups showed no increased brain:liver ratio (Figure 3B). In an earlier study involving an Ang II– dependent transgenic rat model for preeclampsia, we identified focal necrosis in the walls of spiral arteries. We termed these changes “arteriolosclerosis” because they resembled the “atherosis” of human preeclampsia.21 This atherosis-like vasculopathy of the spiral arteries was located in the mesometrial triangle, which represents the maternal part of the uteroplacental unit and has strong homologies to the human placenta.24 Signs for such arteriolosclerotic lesions were significantly higher in Ang II+AT1-AB–treated rats compared with the other groups (Figure 3C). We then investigated the capacity to further remodel the spiral arteries in the uteroplacental unit. As in our earlier study, we calculated the amount of trophoblast cells per vessel contour.24 In this study, the complete mesometrial triangle trophoblast invasion into spiral arteries was reduced in the Ang II plus AT1-AB group compared with the AT1-AB plus IgG group. Surprisingly, Ang II alone had no effect, but AT1-AB alone also reduced trophoblast-induced spiral artery remodeling (Figure 3D).

Figure 3.

Figure 3

A, Fetal weights were significantly decreased in the angiotensin II (Ang II) plus antibodies against the Ang II type 1 receptor (AT1-ABs) treated group. B, Embryo brain:liver ratio on day 20 showed that Ang II plus AT1-ABs gave higher values than all of the other groups, indicating intrauterine growth restriction (IUGR) in this group. C, Trophoblast invasion into the spiral arteries in the Ang II plus AT1-AB group was reduced compared with control. AT1-AB alone also reduced the spiral artery trophoblast/vessel contour. D, Atherosis (arteriolosclerosis) lesions were increased in the Ang II plus AT1-AB group. Left and middle show a cross-section of spiral arteries containing endovascular trophoblast. Between both lumens, atherotic lesions are seen with necrotic cells surrounded by interstitial trophoblasts. D, Atherosis was increased in the Ang II plus AT1-AB group. Right is shown a different cross-section of a spiral artery with advanced lesions (*).

Molecular Mechanisms

We measured the serum concentration of sFLT-1 and PLGF by ELISA. There were no differences in serum concentration of sFLT-1 or PLGF comparing all of the treated groups with the untreated controls. We then tested the hypothesis that binding of the AT1-AB to the AT-1 receptor increases endothelial cell ET-1 production. Basal ET-1 production increased modestly in response to AT1-AB and to Ang II. In sharp contrast, ET-1 production increased markedly in response to Ang II in the presence of AT1-AB. These data support the hypothesis that AT1-ABs increase endothelial cell sensitivity to Ang II (Figure 4A). To find novel pathways induced by AT1-AB and Ang II, we analyzed the transcriptome of mesometrial triangle by gene expression profiling using Illumina Arrays. We found that the transcription factor hypoxia-inducible factor (HIF) 1α was robustly induced by AT1-AB plus Ang II (Figure 4B). The results were confirmed in villous explants. Ang II and AT1-AB both induced HIF-1α protein expression in villous explants compared with controls, however, to a lesser degree than the combined treatment (Figure 4C). We encountered other less prominently differentially expressed genes in the mesometrial triangle (Table S1).

Figure 4.

Figure 4

A, Endothelin 1 (ET-1) protein levels in the endothelial cell supernatant are shown. Angiotensin II (Ang II) and antibodies against the Ang II type 1 receptor (AT1-AB) induced ET-1 production. Ang II plus AT1-AB resulted in a strong ET-1 increase. B, Dot plots of log-2 signal values of rat hypoxia-inducible factor (HIF) 1α are shown. HIF-1α was differentially expressed at false discovery rate 3.02*10−4. HIF-1α was induced in the mesometrial triangle (maternal part of the rat placenta) from rats treated with Ang II plus AT1-AB. C, HIF-1α was expressed in human placental villous explants. Ang II or AT1-AB induced protein expression of HIF-1α. However, Ang II plus AT1-AB increased HIF-1α further.

Discussion

Our novel finding is that AT1-ABs increase Ang II sensitivity, possibly explaining the increased Ang II sensitivity observed in preeclamptic patients. We report that AT1-ABs can be generated by immunization and that purified AT1-ABs induced a chronotropic effect in the cardiomyocyte contraction assay, initiated extracellular-regulated kinase 1/2 phosphorylation, and stimulated protein kinase C-α activation. We were able to detect AT1-ABs with ELISA. Passive transfer of AT1-ABs alone into pregnant rats did not induce a preeclamptic syndrome. However, the combination of AT1-ABs and Ang II produced hypertension, proteinuria, abnormal placental vascular remodeling, and intrauterine growth restriction. We also found that AT1-AB plus Ang II induced HIF-1α, which was associated with arteriolosclerotic lesions in the spiral arteries and altered trophoblast invasion. Finally, AT1-AB plus Ang II induced ET-1 expression.

Gant et al8 infused Ang II into pregnant patients from week 10 of pregnancy onward and showed that those who later developed preeclampsia required diminishing amounts of Ang II to obtain a similar pressor response. Baker et al7 later confirmed these findings.7 They observed increased Ang II–induced calcium signaling in platelets of pregnant women who subsequently developed preeclampsia. Recently Siddiqui et al10 showed that AT1-AAs occur in 95% of preeclamptic women. They also found AT1-AAs in women with gestational hypertension. AbdAlla et al25 postulated that AT1-bradykinin 2 receptor heterodimerization mediates an increased Ang II response in preeclampsia.6,25 Systemic alterations in AT1 receptor expression did not correlate with increased Ang II sensitivity.25 We and others showed that the AT1 receptor is indeed upregulated in the uteroplacental unit of preeclamptic women.6,26 However, whether a locally upregulated AT1 receptor in the uteroplacental unit is sufficient to induce a systemic blood pressure response is unknown.

We do not know how AT1-AAs activate the AT1 receptor. They could alter the receptor conformation so that the receptor’s ability to bind circulating Ang II is enhanced.11 The activation may involve a cross-linking between receptors, thereby holding receptors in their activated conformation and preventing tachyphylaxis.27 Cruz et al28 reported an increase in fetoplacental vascular responsiveness to Ang II in isolated perfused placentas from preterm pregnancies with absent tachyphylaxis, whereas marked tachyphylaxis was present in all of the preparations from term pregnancies. Saxena et al9 showed recently that increased sensitivity to Ang II persists postpartum in preeclamptic women. In line with these data, we found that AT1-AAs persisted in 20% of former preeclamptic patients 1 year postpartum.29 Although blood pressure returns to normal after delivery, women with preeclampsia have an increased risk of cardiovascular disease later in life.4 Several reports postulate that the abnormalities in vascular function persist, which predisposes these women to the increased risk.4 AT1-AAs could conceivably exercise longer-term effects.

Vasodilatation of the maternal systemic circulation is an important physiological adaptation of mammalian pregnancy. 2,30 We observed that a substantial Ang II infusion (435 ng/kg per minute) did not increase blood pressure in pregnant rats. This dose regularly increases blood pressure in nonpregnant rats. Yu and Khraibi31 showed that an IV Ang II infusion at 200 ng/kg per minute is required to increase blood pressure in anesthetized midpregnancy rats in a short-term experiment. Zhou et al13 reported that passive transfer of isolated IgG and purified AT1-AAs in pregnant mice on day 13 induced hypertension. Similarly, LaMarca et al15 reported that AT1-AAs infused IP into pregnant rats from day 12 to 19 gestation via miniosmotic pumps induced a hypertensive response. However, these studies and our work have important differences. Our aim was to produce activating antibodies in a biological system distinct from the patients, as a “proof of principle.” Zhou et al13 isolated AT1-AAs from patients directed against the second extracellular loop. We used an epitope-specific isolation procedure. LaMarca et al15 administered their AT1-AAs differently than in our study. Liu and colleagues19,32 found differences between AT1-AAs from preeclamptic women and AT1-ABs generated by long-term active immunization in male rats. They found that their AT1-ABs alone did not induce hypertension.19,32

Naturally occurring autoantibodies and antibodies generated by immunization behave differently, although they are directed against the same epitope.33 Posttranslational modifications, such as acetylation, lipidation, citrullination, glycosylation, and so forth, could also influence the antibodies.34 Although the generated AT1-ABs showed effects similar to AT1-AAs in in vitro assays, we cannot exclude the possibility that various autoantibodies have additional properties that are necessary to induce the preeclamptic phenotype in vivo. In addition, different affinity and avidity of the antibodies may also influence the effects as shown for autoantibodies to monosialotetrahexosylganglioside 1, a major ganglioside important to Guillain-Barreé syndrome.35 Thus, antibodies directed to the same epitope may exhibit different receptor responses.

Iizuka et al36 showed that mouse autoantibodies differ, even in mice with the same genetic background and major histocompatibility complex class, when the antigen is either a “foreign” or a “self” antigen. In mice deficient in the M3 muscarinic acetylcholine receptor, functional antibodies were induced by immunization with the M3 receptor. However, these antibodies were not present when studied from mice having tolerance to this autoantigen, namely, littermates that were also immunized with M3 receptor. Furthermore, major histocompatibility complex class I molecules are different among different species and determine the expression of peptides on T cells, positive and negative selection of the immune response, and antibody specificity in vivo.34 By studying the autoimmune response against certain autoantigens in different lupus models, Monneaux et al37 reported that major histocompatibility complex and nonmajor histocompatibility complex genes are responsible for the autoantibody response and the fine specificity of the autoantibodies. These responses could be completely different in an antibody generated by immunization. We do not believe that our results are specifically related to low antibody titers.17,38 We also dosed the pregnant rats twice. We detected the transferred AT1-ABs for ≥11 days after injection. Siddiqui et al10 concluded that, in healthy pregnant women, AT1-AA concentrations were not high enough or the AT1-AAs were not present for sufficient duration. Furthermore, Ang II levels are increased 2.2-fold in normal human pregnancy.39 The levels that we obtained in the rat did not reach the physiological human pregnancy levels. Although preeclampsia, scleroderma, and non-human leukocyte antigen (HLA)–triggered acute kidney transplant rejection share vasculopathy as a common feature,11,40,41 which permissive factors are responsible for the preference of the different vascular beds is unclear.

We identified HIF-1α by means of gene expression analysis. We then supported this finding with Western blotting. Placental ischemia-hypoxia is the major inducer for HIF-α and protein stabilization in preeclampsia.42 Whether Ang II plus AT1-ABs induced HIF-1α directly or whether the response was secondary to hypoxia cannot be discerned from our study. However, Rajakumar et al23 suggested earlier that HIF-1α contributes to preeclampsia. The arteriolosclerotic lesions in the spiral arteries occurring in our experiments could have contributed to uteroplacental hypoxia. Furthermore, Burton et al43 proposed recently that turbulent blood flow could damage villous architecture. Reactive oxygen and the redox-sensitive part of the oxygen-sensing pathway are also important inducers of HIF-1α.44 Under situations of increased oxidative stress, changes in the redox state within a cell may contribute to an efficient and fast-responding O2-sensing system.45

We also found that ET-1 might be involved in mediating the blood pressure increases that we observed. In the reduced uterine perfusion pressure preeclampsia-like model, a selective endothelin (endothelin A) receptor antagonist attenuated the blood pressure response.46 Furthermore, LaMarca et al15 showed that the hypertensive effects elicited by rat AT1-AAs were blocked by an endothelin A receptor antagonist. These results are in line with in vitro observations from Yang et al,32 who found that AT1-AAs from preeclamptic patients caused vascular constriction in conduit and small resistance arteries from male rats and mechanistically implicated ET-1.

Perspectives

We provide further evidence to support the “immune” theory of preeclampsia and suggest that interventions along the lines of renin-Ang-system attenuation could be therapeutically useful. We recognize the fact that our findings are still not definitive and that we will have to conduct further studies.

Supplementary Material

01

Acknowledgments

We thank Sabine Bartel, Jutta Meisel, Reika Langanki, Ralph Plehm, Ute Gerhard, Jana Czychi, and Astrid Schiche for the excellent technical assistance.

Sources of Funding

The Deutsche Forschungsgemeinschaft supported R.D. (DE 631/1). The National Institutes of Health P01-HD30367 supported A.R. and C.A.H.

Footnotes

This paper was sent to Nancy J. Brown, associate editor, for review by expert referees, editorial decision, and final disposition.

Disclosures

None.

References

  • 1.ACOG Committee on Practice Bulletins—Obstetrics. ACOG practice bulletin: diagnosis and management of preeclampsia and eclampsia. Obstet Gynecol. 2002;99:159–167. doi: 10.1016/s0029-7844(01)01747-1. [DOI] [PubMed] [Google Scholar]
  • 2.Redman CW, Sargent IL. Latest advances in understanding preeclampsia. Science. 2005;308:1592–1594. doi: 10.1126/science.1111726. [DOI] [PubMed] [Google Scholar]
  • 3.Bellamy L, Casas JP, Hingorani AD, Williams DJ. Pre-eclampsia and risk of cardiovascular disease and cancer in later life: systematic review and meta-analysis. BMJ. 2007;335:974. doi: 10.1136/bmj.39335.385301.BE. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Staff AC, Dechend R, Pijnenborg R. Learning from the placenta: acute atherosis and vascular remodeling in preeclampsia-novel aspects for atherosclerosis and future cardiovascular health. Hypertension. 2010;56:1026–1034. doi: 10.1161/HYPERTENSIONAHA.110.157743. [DOI] [PubMed] [Google Scholar]
  • 5.Herse F, Staff AC, Hering L, Muller DN, Luft FC, Dechend R. AT1- receptor autoantibodies and uteroplacental RAS in pregnancy and preeclampsia. J Mol Med. 2008;86:697–703. doi: 10.1007/s00109-008-0332-4. [DOI] [PubMed] [Google Scholar]
  • 6.Shah DM. Role of the renin-angiotensin system in the pathogenesis of preeclampsia. Am J Physiol Renal Physiol. 2005;288:F614–F625. doi: 10.1152/ajprenal.00410.2003. [DOI] [PubMed] [Google Scholar]
  • 7.Baker PN, Kilby MD, Broughton Pipkin F. The effect of angiotensin II on platelet intracellular free calcium concentration in human pregnancy. J Hypertens. 1992;10:55–60. doi: 10.1097/00004872-199201000-00009. [DOI] [PubMed] [Google Scholar]
  • 8.Gant NF, Daley GL, Chand S, Whalley PJ, MacDonald PC. A study of angiotensin II pressor response throughout primigravid pregnancy. J Clin Invest. 1973;52:2682–2689. doi: 10.1172/JCI107462. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Saxena AR, Karumanchi SA, Brown NJ, Royle CM, McElrath TF, Seely EW. Increased sensitivity to angiotensin II is present postpartum in women with a history of hypertensive pregnancy. Hypertension. 2010;55:1239–1245. doi: 10.1161/HYPERTENSIONAHA.109.147595. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Siddiqui AH, Irani RA, Blackwell SC, Ramin SM, Kellems RE, Xia Y. Angiotensin receptor agonistic autoantibody is highly prevalent in preeclampsia: correlation with disease severity. Hypertension. 2010;55:386–393. doi: 10.1161/HYPERTENSIONAHA.109.140061. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Wallukat G, Homuth V, Fischer T, Lindschau C, Horstkamp B, Jupner A, Baur E, Nissen E, Vetter K, Neichel D, Dudenhausen JW, Haller H, Luft FC. Patients with preeclampsia develop agonistic autoantibodies against the angiotensin AT1 receptor. J Clin Invest. 1999;103:945–952. doi: 10.1172/JCI4106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Dechend R, Viedt C, Muller DN, Ugele B, Brandes RP, Wallukat G, Park JK, Janke J, Barta P, Theuer J, Fiebeler A, Homuth V, Dietz R, Haller H, Kreuzer J, Luft FC. AT1 receptor agonistic antibodies from preeclamptic patients stimulate NADPH oxidase. Circulation. 2003;107:1632–1639. doi: 10.1161/01.CIR.0000058200.90059.B1. [DOI] [PubMed] [Google Scholar]
  • 13.Zhou CC, Zhang Y, Irani RA, Zhang H, Mi T, Popek EJ, Hicks MJ, Ramin SM, Kellems RE, Xia Y. Angiotensin receptor agonistic autoantibodies induce pre-eclampsia in pregnant mice. Nat Med. 2008;14:855–862. doi: 10.1038/nm.1856. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Xia Y, Ramin SM, Kellems RE. Potential roles of angiotensin receptor-activating autoantibody in the pathophysiology of preeclampsia. Hypertension. 2007;50:269–275. doi: 10.1161/HYPERTENSIONAHA.107.091322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.LaMarca B, Parrish M, Ray LF, Murphy SR, Roberts L, Glover P, Wallukat G, Wenzel K, Cockrell K, Martin JN, Jr, Ryan MJ, Dechend R. Hypertension in response to autoantibodies to the angiotensin II type I receptor (AT1-AA) in pregnant rats: role of endothelin-1. Hypertension. 2009;54:905–909. doi: 10.1161/HYPERTENSIONAHA.109.137935. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Rose NR. Infection, mimics, and autoimmune disease. J Clin Invest. 2001;107:943–944. doi: 10.1172/JCI12673. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Jahns R, Boivin V, Hein L, Triebel S, Angermann CE, Ertl G, Lohse MJ. Direct evidence for a β1-adrenergic receptor-directed autoimmune attack as a cause of idiopathic dilated cardiomyopathy. J Clin Invest. 2004;113:1419–1429. doi: 10.1172/JCI20149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Wenzel K, Wallukat G, Qadri F, Hubner N, Schulz H, Hummel O, Herse F, Heuser A, Fischer R, Heidecke H, Luft FC, Muller DN, Dietz R, Dechend R. α1A-adrenergic receptor-directed autoimmunity induces left ventricular damage and diastolic dysfunction in rats. PLoS One. 2010;5:e9409. doi: 10.1371/journal.pone.0009409. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Zhang SL, Du YH, Wang J, Yang LH, Yang XL, Zheng RH, Wu Y, Wang K, Zhang MS, Liu HR. Endothelial dysfunction induced by antibodies against angiotensin AT receptor in immunized rats. Acta Pharmacol Sin. 2010;31:1381–1388. doi: 10.1038/aps.2010.144. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Wenzel K, Haase H, Wallukat G, Derer W, Bartel S, Homuth V, Herse F, Hubner N, Schulz H, Janczikowski M, Lindschau C, Schroeder C, Verlohren S, Morano I, Muller DN, Luft FC, Dietz R, Dechend R, Karczewski P. Potential relevance of α(1)-adrenergic receptor autoantibodies in refractory hypertension. PLoS One. 2008;3:e3742. doi: 10.1371/journal.pone.0003742. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Dechend R, Gratze P, Wallukat G, Shagdarsuren E, Plehm R, Brasen JH, Fiebeler A, Schneider W, Caluwaerts S, Vercruysse L, Pijnenborg R, Luft FC, Muller DN. Agonistic autoantibodies to the AT1 receptor in a transgenic rat model of preeclampsia. Hypertension. 2005;45:742–746. doi: 10.1161/01.HYP.0000154785.50570.63. [DOI] [PubMed] [Google Scholar]
  • 22.Geusens N, Hering L, Verlohren S, Luyten C, Drijkoningen K, Taube M, Vercruysse L, Hanssens M, Dechend R, Pijnenborg R. Changes in endovascular trophoblast invasion and spiral artery remodelling at term in a transgenic preeclamptic rat model. Placenta. 31:320–326. doi: 10.1016/j.placenta.2010.01.011. [DOI] [PubMed] [Google Scholar]
  • 23.Rajakumar A, Michael HM, Daftary A, Jeyabalan A, Gilmour C, Conrad KP. Proteasomal activity in placentas from women with preeclampsia and intrauterine growth restriction: implications for expression of HIF-α proteins. Placenta. 2008;29:290–299. doi: 10.1016/j.placenta.2007.11.015. [DOI] [PubMed] [Google Scholar]
  • 24.Geusens N, Verlohren S, Luyten C, Taube M, Hering L, Vercruysse L, Hanssens M, Dudenhausen JW, Dechend R, Pijnenborg R. Endovascular trophoblast invasion, spiral artery remodelling and uteroplacental haemodynamics in a transgenic rat model of pre-eclampsia. Placenta. 2008;29:614–623. doi: 10.1016/j.placenta.2008.04.005. [DOI] [PubMed] [Google Scholar]
  • 25.AbdAlla S, Lother H, el Massiery A, Quitterer U. Increased AT(1) receptor heterodimers in preeclampsia mediate enhanced angiotensin II responsiveness. Nat Med. 2001;7:1003–1009. doi: 10.1038/nm0901-1003. [DOI] [PubMed] [Google Scholar]
  • 26.Herse F, Dechend R, Harsem NK, Wallukat G, Janke J, Qadri F, Hering L, Muller DN, Luft FC, Staff AC. Dysregulation of the circulating and tissue-based renin-angiotensin system in preeclampsia. Hypertension. 2007;49:604–611. doi: 10.1161/01.HYP.0000257797.49289.71. [DOI] [PubMed] [Google Scholar]
  • 27.Elies R, Fu LX, Eftekhari P, Wallukat G, Schulze W, Granier C, Hjalmarson A, Hoebeke J. Immunochemical and functional characterization of an agonist-like monoclonal antibody against the M2 acetylcholine receptor. Eur J Biochem. 1998;251:659–666. doi: 10.1046/j.1432-1327.1998.2510659.x. [DOI] [PubMed] [Google Scholar]
  • 28.Cruz MA, Dominguez A, Gallardo V, Carrasco G, Miguel P, Gonzalez C. Vascular reactivity to angiotensin II and eicosanoid production in the human placenta from term and preterm pregnancy. Gynecol Obstet Invest. 2000;50:247–253. doi: 10.1159/000010325. [DOI] [PubMed] [Google Scholar]
  • 29.Hubel CA, Wallukat G, Wolf M, Herse F, Rajakumar A, Roberts JM, Markovic N, Thadhani R, Luft FC, Dechend R. Agonistic angiotensin II type 1 receptor autoantibodies in postpartum women with a history of preeclampsia. Hypertension. 2007;49:612–617. doi: 10.1161/01.HYP.0000256565.20983.d4. [DOI] [PubMed] [Google Scholar]
  • 30.Roberts JM, Gammill HS. Preeclampsia: recent insights. Hypertension. 2005;46:1243–1249. doi: 10.1161/01.HYP.0000188408.49896.c5. [DOI] [PubMed] [Google Scholar]
  • 31.Yu T, Khraibi AA. Renal interstitial hydrostatic pressure and natriuretic response to high doses of angiotensin II in pregnant rats. Am J Hypertens. 2006;19:300–305. doi: 10.1016/j.amjhyper.2005.09.016. [DOI] [PubMed] [Google Scholar]
  • 32.Yang X, Wang F, Chang H, Zhang S, Yang L, Wang X, Cheng X, Zhang M, Ma XL, Liu H. Autoantibody against AT1 receptor from preeclamptic patients induces vasoconstriction through angiotensin receptor activation. J Hypertens. 2008;26:1629–1635. doi: 10.1097/HJH.0b013e328304dbff. [DOI] [PubMed] [Google Scholar]
  • 33.Doyle HA, Mamula MJ. Post-translational protein modifications in antigen recognition and autoimmunity. Trends Immunol. 2001;22:443–449. doi: 10.1016/s1471-4906(01)01976-7. [DOI] [PubMed] [Google Scholar]
  • 34.Chiu NM, Wang B, Kerksiek KM, Kurlander R, Pamer EG, Wang CR. The selection of M3-restricted T cells is dependent on M3 expression and presentation of N-formylated peptides in the thymus. J Exp Med. 1999;190:1869–1878. doi: 10.1084/jem.190.12.1869. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Lopez PH, Zhang G, Zhang J, Lehmann HC, Griffin JW, Schnaar RL, Sheikh KA. Passive transfer of IgG anti-GM1 antibodies impairs peripheral nerve repair. J Neurosci. 30:9533–9541. doi: 10.1523/JNEUROSCI.2281-10.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Iizuka M, Wakamatsu E, Tsuboi H, Nakamura Y, Hayashi T, Matsui M, Goto D, Ito S, Matsumoto I, Sumida T. Pathogenic role of immune response to M3 muscarinic acetylcholine receptor in Sjogren’s syndrome-like sialoadenitis. J Autoimmun. 2010;35:383–389. doi: 10.1016/j.jaut.2010.08.004. [DOI] [PubMed] [Google Scholar]
  • 37.Monneaux F, Dumortier H, Steiner G, Briand JP, Muller S. Murine models of systemic lupus erythematosus: B and T cell responses to spliceosomal ribonucleoproteins in MRL/Fas(lpr) and (NZB x NZW)F(1) lupus mice. Int Immunol. 2001;13:1155–1163. doi: 10.1093/intimm/13.9.1155. [DOI] [PubMed] [Google Scholar]
  • 38.Jacobson L, Polizzi A, Morriss-Kay G, Vincent A. Plasma from human mothers of fetuses with severe arthrogryposis multiplex congenita causes deformities in mice. J Clin Invest. 1999;103:1031–1038. doi: 10.1172/JCI5943. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Merrill DC, Karoly M, Chen K, Ferrario CM, Brosnihan KB. Angiotensin-( 1–7) in normal and preeclamptic pregnancy. Endocrine. 2002;18:239–245. doi: 10.1385/ENDO:18:3:239. [DOI] [PubMed] [Google Scholar]
  • 40.Dragun D, Scornik J, Meier-Kriesche HU. Kidney-transplant rejection and anti-MICA antibodies. N Engl J Med. 2008;358:196. author reply 196. [PubMed] [Google Scholar]
  • 41.Riemekasten G, Philippe A, Nather M, Slowinski T, Muller DN, Heidecke H, Matucci-Cerinic M, Czirjak L, Lukitsch I, Becker M, Kill A, van Laar JM, Catar R, Luft FC, Burmester GR, Hegner B, Dragun D. Involvement of functional autoantibodies against vascular receptors in systemic sclerosis. Ann Rheum Dis. 2011;70:530–536. doi: 10.1136/ard.2010.135772. [DOI] [PubMed] [Google Scholar]
  • 42.Schofield CJ, Ratcliffe PJ. Signalling hypoxia by HIF hydroxylases. Biochem Biophys Res Commun. 2005;338:617–626. doi: 10.1016/j.bbrc.2005.08.111. [DOI] [PubMed] [Google Scholar]
  • 43.Burton GJ, Woods AW, Jauniaux E, Kingdom JC. Rheological and physiological consequences of conversion of the maternal spiral arteries for uteroplacental blood flow during human pregnancy. Placenta. 2009;30:473–482. doi: 10.1016/j.placenta.2009.02.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Liu Q, Berchner-Pfannschmidt U, Moller U, Brecht M, Wotzlaw C, Acker H, Jungermann K, Kietzmann T. A Fenton reaction at the endoplasmic reticulum is involved in the redox control of hypoxia-inducible gene expression. Proc Natl Acad Sci U S A. 2004;101:4302–4307. doi: 10.1073/pnas.0400265101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Giordano FJ. Oxygen, oxidative stress, hypoxia, and heart failure. J Clin Invest. 2005;115:500–508. doi: 10.1172/JCI200524408. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Roberts L, LaMarca BB, Fournier L, Bain J, Cockrell K, Granger JP. Enhanced endothelin synthesis by endothelial cells exposed to sera from pregnant rats with decreased uterine perfusion. Hypertension. 2006;47:615–618. doi: 10.1161/01.HYP.0000197950.42301.dd. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

01

RESOURCES