Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2015 Dec 1.
Published in final edited form as: Curr Heart Fail Rep. 2014 Dec;11(4):500–508. doi: 10.1007/s11897-014-0217-5

AUTOANTIBODIES AND CARDIOVASCULAR DYSFUNCTION: CAUSE OR CONSEQUENCE?

Yuji Nagatomo 1, W H Wilson Tang 1,2
PMCID: PMC4224601  NIHMSID: NIHMS622561  PMID: 25142807

Abstract

There has been a long history of the exploration into autoimmunity as possible pathogenic factor of cardiovascular diseases from unknown cause represented by dilated cardiomyopathy (DCM). Auto-antibodies (AAbs) have emerged either as humoral responses provoked by release of "self-antigens" due to tissue damage or dysregulated humoral immunity itself. The pathogenic roles of some AAbs have been suggested by the findings from basic research using in vitro and in vivo disease model as well as clinical studies including immunoadsorption studies removing AAbs from patients with DCM. In this context, the importance of AAbs belonging to IgG3 subclass has also been implicated. In this review article we summarize the findings accumulated to date regarding AAbs which have been considered to be involved in the pathology of DCM or pregnancy-related cardiovascular disease. Furthermore, we discuss the significance of AAbs as a possible cause of DCM and the potential roles as novel therapeutic target.

Keywords: autoantibody, myosin, β1 adrenergic receptor, muscarinic M2 acetylcholine receptor, troponin I, dilated cardiomyopathy, peripartum cardiomyopathy, preeclampsia

Introduction

Dilated cardiomyopathy (DCM) is one of the most common causes of heart failure without an ischemic etiology and the most common reason for heart transplantation. In many cases, the underlying causes of DCM are unknown or "idiopathic" and may affect individuals across all ages. There has been a long-standing interest in exploring the contribution of autoimmunity towards the pathogenesis of DCM. This is based on the presence of anti-cardiac auto-antibodies (AAbs) in a subset of patients with DCM. It has been postulated that myocardial tissue damage may lead to the release of intracellular proteins that can serve as "self-antigens" in order to provoke humoral responses leading to the generation of AAbs. On the other hand, dysregulated humoral immunity itself can serve as the primary driver of AAb production, directly contributing to progressive myocyte damage as observed in some systemic autoimmune disorders associated with cardiac complications (e.g. systemic lupus erythematosus).

Over the past decades, researchers have investigated the direct physiological role of AAbs via basic in vitro or in vivo experiments, as well as via modulation of their effects by removal or neutralization. In this review article, we critically examine the contemporary understanding of specific AAbs that have been mechanistically linked to the pathogenesis of DCM with emphasis on the discussion of how quantitative AAb measurements may lead to potential therapeutic implications.

Dilated Cardiomyopathy: A Possible Autoimmune Origin

Several cardiac AAbs have been consistently reported to be present in sera from patients with DCM [13]. However, such associations do not necessarily establish causality, especially when the acuity, time course, and localization of autoimmune responses are largely unknown. Earlier research focus was based upon establishing the association between introduction of AAbs and induction of DCM phenotypes. Indeed, immunization with non-cardiac peptides such as β1-adrenergic receptor (β1AR) second extracellular loop [4,5] or muscarinic M2 acetylcholine receptor (M2R) [6,7], as well as cardiac-specific peptides such as myosin [8] or troponin I [9] can directly lead to the generation of AAbs and myocarditis- or DCM-like phenotype in experimental animals. These findings support the development of AAbs upon exposure to •self-antigens•, thereby establishing the first step for specific AAbs as contributors in the development of DCM. Clinical and translational research studies regarding these specific AAbs are illustrated in Table 1.

Table 1.

Summary of Studies of AAbs in DCM

Epitope
of AAb
Physiological effect
on
pathology
Reproduction of
disease
by
passive
transfer
Induction
of
disease
by
immunization of
epitope
Association with
phenotype
or
outcomes
in human
Improvement of
pathology
after
removal
Comments
Myosin +[12] ±[10,11]* +[8] +[13,15] ? *Transfer of AAbs causes myocarditis only in certain strains of mice [10,11]
β1AR +++[4,2027,29]
−[32,33]*
+[5] +[4] ++[28,83]†
[30]‡
±[57]
+[31,55]
*AAbs from some patients are functionally inactive [32,33].
†Associated with ventricular arrhythmia, sudden death [83] and cardiovascular death[28].
‡Associated with more favorable response to β-blocker[30]
M2R ++[35,3739] *(See comments) +[6,7] +[38,41]† ? *Reproduced by adoptive transfer into Rag2 knockout mice of splenocytes from M2R knockout mice immunized with M2R protein.
†Association with Af comorbid with DCM and recurrent after ablation[38,41]
Troponin I +[9] +[9] +[42] −?[46]
+[44]*
−?[58] *Associated with higher LVEF and more favorable outcomes in DCM[44]
Na-K-ATPase +[84] ? +[85] +[84]* ? *Associated with ventricular arrhythmia and sudden death [84]
“Cardio-depressant AAb” +[59,60,61] ? ? ? +[60,62] Epitope unknown

AAb, autoantibody; DCM, dilated cardiomyopathy; β1AR, β1-adrenergic receptor; M2R, muscarinic M2 acetylcholine receptor; Af, atrial fibrillation; LVEF, left ventricular ejection fraction.

Anti-myosin Autoantibody

The anti-myosin AAb has long been studied for its causal roles in the pathology of myocarditis or DCM. In 1987, immunization with cardiac myosin was found to induce anti-myosin AAbs and myocarditis in certain strains of mice [8]. However, since transfer of serum with high titer anti-myosin AAbs from C.B-17 mice to SCID (severe combined immune deficiency) mice failed to cause myocarditis [10], the pathogenic role of anti-myosin AAbs was questioned by some researchers. It has been proposed that myosin or a similar protein was present in the extracellular matrix of susceptible mouse strains [11]. And the pathogenic effects of anti-myosin AAbs was mediated at least partly by reacting with β-adrenergic receptor and activating downstream protein kinase A pathway [12]. In human, anti-myosin AAbs are detected in 20–30% of patients with DCM and 4–30% in those with ICM [13,14]. However, the clinical findings regarding the significance of anti-myosin AAbs are inconsistent. One study showed that persistence of anti-myosin AAb was associated with milder symptoms at presentation and stable disease [13]. Whereas anti-myosin AAbs were also shown to associate with deterioration of left ventricular function in patients with biopsy-proven chronic myocarditis [15].

Autoantibody against β1-Adrenergic Receptor (β1AR-AAb)

Detectable circulating AAbs against β1AR have been observed in approximately 30–40% of patients with chronic heart failure due to DCM [4,1619]. β1AR-AAb shows agonist-like effects [2023], inducing receptor uncoupling [4,24,25], myocyte apoptosis [26], sustained calcium influx resulting in electric instability of the heart [27], and persistent myocardial damage [5]. These effects were abolished by β-blockers in vitro[23,28] and in vivo [4]. There have also been prior reports demonstrating the association between detectable β1AR-AAb and increased mortality [28] as well as the occurrence of fatal ventricular arrhythmias and sudden death [4,29] in patients with DCM. However, a majority of the subjects in these association studies were not receiving anti-adrenergic therapy at the time. Interestingly, more favorable recovery of cardiac performance in response to β-blocker therapy was observed in β1AR-AAb-positive patients compared to β1AR-AAb-negative patients [30]. These findings are consistent with the hypothesis that heightened adrenergic drive associated with β1AR-AAb generation in DCM can lead to adverse cardiac consequences. Besides anti-adrenergic therapy, studies are currently ongoing to determine if an aptamer for neutralizing β1AR-AAbs may curtail disease progression and perhaps even facilitate recovery [31].

It is important to note that not all of the detectable β1AR-AAbs uniformly exerts their adverse physiological effects across the spectrum of clinical conditions. While β1AR-AAbs were also detectable in patients with valvular or hypertensive heart disease (or even in some healthy subjects), AAbs in these non-DCM individuals were functionally inactive [32,33]. Furthermore, the presence of β1AR-AAb was consistently associated with increased mortality risk largely in DCM and not in ischemic cardiomyopathy [28]. Evidence also suggests that some β1AR-AAbs can be generated, at least partly, by cardiac loading or damage. In an analysis of weaned DCM patients who tested positive for β1AR-AAb prior to implantation of left ventricular assist device (LVAD), β1AR-AAb disappeared after 3 to 31 weeks following LV unloading by LVAD support in 33 of 34 patients [34]. The potential mechanism of these inconsistencies has not yet been elucidated, and further studies are needed.

Autoantibody against Muscarinic M2 Acetylcholine Receptor

Circulating AAbs against the second extracellular loop of M2R (M2R-AAb) have been detected in a large number of cardiovascular diseases such as DCM and chronic Chagas’ disease [3539]. M2R-AAbs extracted from DCM patients induced a significant decrease in Ca2+ currents [39], negative chronotropic effect in vitro [37] and supraventricular arrhythmia ex vivo [38] which were blocked by M2 antagonist [38,39] or a synthetic peptide derived from the second extracellular loop of M2R [38]. M2R-AAbs were also detected along with the induction of DCM-like morphology in the heart of mice receiving adoptive transfer of splenocytes from M2R null mice immunized with synthetic M2R peptide [40]. In addition, AAbs from these mice induced a significant decrease in Ca2+ currents in ventricular cardiomyocytes, which was blocked by M2R antagonists or synthetic M2R peptides [40]. Clinically the presence of M2R-AAbs was strongly and independently associated with the comorbidity of atrial fibrillation [38], and may serve as an independent predictor for the recurrence of atrial fibrillation after catheter ablation [41].

Autoantibody against Troponin I

Monoclonal antibodies against cardiac troponin I (cTnI) induce chamber dilatation and contractile dysfunction of hearts of wild-type mice [9]. Interestingly, the immunization of cTnI, but not cardiac troponin T, induced severe autoimmune inflammation in the mouse myocardium [42]. Monoclonal cTnI-AAbs extracted from mice following immunization with recombinant cTnI are able to increase the voltage-dependent L-type calcium current in cardiomyocytes of wild-type mice [9].

Despite promising data regarding the effects of cTnI-AAbs in animal models, the pathogenic role of cTnI-AAbs in humans and their possible involvement in the pathogenesis of DCM remains controversial. Although the cTnI-AAbs were reported to be detected in DCM and ICM patients [43,44], IgG preparations reactive towards cTnI did not exhibit measurable effects on Ca2+ transients in cultured neonatal rat ventricular cardiomyocytes, nor did they bind the respective cells by direct immunofluorescence [43]. When examining cellular autoimmune responses from peripheral blood mononuclear cells, there were no significant differences between DCM patients with versus without detectable cTnI-AAb levels [45]. Furthermore, in the analysis of 95 patients with DCM, circulating cTnI-AAbs was not associated with patients’ clinical status or outcome, while the presence of cTnI was associated with worse cardiac function, morphology, and worse clinical outcome [46]. In contrast, another study showed that cTnI-AAb positive patients had significantly higher left ventricular ejection fraction and more favorable outcomes than negative patients with DCM, but not ICM [44].

Data from Immunoadsorption Studies

Like other autoimmune disorders, the removal of pathogenic AAbs by immunoadsorption (IA) therapy can be considered as a therapeutic option for DCM patients. Clinical studies have consistently demonstrated that non-specific IA can lead to improved hemodynamics and cardiac function, better survival and quality of life in DCM patients [4754] (Table 2). Specifically, IA using the column specific for β1AR-AAbs was effective in alleviating the cardiac dysfunction in an observational series of patients with DCM [55,56]. However, many of these studies are small in sample size and few have blinding or control group comparisons. Moreover, it is also important to point out that non-specific IA was effective in both patients with β1AR-AAbs and those without [57]. These findings suggest the possibility that the causal mechanism in the pathology of DCM cannot be explained solely by β1AR-AAbs, and that other immune or non-immune factors may influence the effectiveness of IA in this population.

Table 2.

Summary of IA Studies for Human DCM

IA Column IV/IG Number of
Patients
Main Findings / Comments References

Anti-human IgG + 9 Induced acute hemodynamic benefit.
Five alive patients showed the increase of LVEF at 3 years.
Two deteriorated patients showed the re-increase in β1AR-AAb titer.
[47,51]
+ 34 (control 17) Improved LVEF at 12 months
No significant reincrease in β1AR-AAb was seen during the 1-year follow-up
[49]
+ 18 (control 9) Improved cardiac function and hemodynamics at 3 months [48]
+ 45 CI and LVEF improved significantly only in patients with “cardio-depressant AAb” [60]
+ 22 The similar effect between patients with β1AR-AAb and those without it [57]

Anti-human IgG or Protein A + 18 (anti-human IgG 9, protein A 9) Anti-human IgG was superior to standard protein A column in terms of elimination IgG3 subclass and improvement of LVEF [70]

Protein A + 18 Improved treatment regimen for IgG3 subclass elimination induced hemodynamic benefit. [71]
+ 22 Improved cardiac function and hemodynamics at 6 months [52]
4 Improved QOL
Insignificant improvement of cardiac function at 6 months
[53]

Tryptophan 16 Improved cardiac function at 3 month.
The increase in LVEF correlated with “cardio-depressant AAbs” and anti-cardiac AAbs belonging to IgG3 subclass.
No control group
[54,62]

Column specific for β1AR-AAb 8 Improved cardiac function at 1 year
No control group
[55,56]

IA, immunoadsorption; IV/IG, intravenous immunoglobulin administration; LVEF, left ventricular ejection fraction; β1AR-AAb, autoantibody against β1-adrenergic receptor; CI, cardiac index; QOL, quality of life.

The potential contribution of cTnI-AAbs is less certain. In a small study, the presence of cTnI-AAbs was not associated with the effect of IA therapy which removes such AAbs. Despite being statistically insignificant, the study even showed a weaker effect of IA on the absolute change of LVEF after 6 months in cTnI-AAb positive patients [58].

“Cardio-depressant Autoantibody”

Some groups have shown that IgG antibodies purified from patients with DCM may induce a negative inotropic effect or reduction of calcium transient in adult rat cardiomyocytes [59,60] or chick embryo ex vivo systems [61,62]. Often referred to as the “cardio-depressant” AAb, the epitope(s) of such AAbs have not been identified. Interestingly, patients with cardio-depressant AAbs demonstrated an acute increase in cardiac index and improvement in left ventricular ejection fraction following IA therapy, while those without such AAbs did not show significant changes [60,62]. Nevertheless, cardio-depressant effects of these AAbs are unlikely to be induced by either the F(ab’)2 or Fc fragments alone. Reconstitution of the Fc part by incubation of cardiomyocytes with DCM-F(ab’)2 fragments followed by goat-anti-human-F(ab’)-IgG again induced reduction of cell shortening and of calcium transients [63]. Taken together, these findings suggest that AAbs require Fc fragment binding to Fc receptor as well as F(ab’)2 fragments binding to the epitope peptide in order to exert its pathological effect, and hence the effects of the AAb vary depending on the structure of the Fc fragment.

The Significance of IgG3 Subclass AAbs

IgG has 4 subclasses and they differ from one another both immunologically and functionally. Antibodies capable of triggering effector functions and the most likely to be involved in immunoregulatory activities are IgG3 and IgG1. Complement activation through binding of C1q is most effective with IgG3 [64]. Affinity-purified IgG fractions positive for IgG3 anti-myosin AAb significantly inhibited the contraction of cardiomyocytes to a greater extent than those with non-IgG3 AAb in vitro [65]. In 1999, Warraich et al reported anti-myosin AAbs belonging to IgG3 subclass were selectively raised in patients with DCM compared with HF patients of an ischemic etiology [66]. Indices of hemodynamic dysfunction also significantly correlated with anti-myosin AAb titer belonging to IgG3, but not total IgG in patients with DCM [67]. AAbs against some other cardiac components belonging to the IgG3 subclass were also detected in patients with DCM and their presence were an independent predictor of the presence of cardio-depressant AAb [68]. The importance of IgG3 AAbs was supported when cardiac function was improved with IA via anti-human IgG columns (high affinity for all IgG subclasses) compared to using protein A (high affinity for IgG1, 2, and 4, but low affinity for IgG3) [69]. Column eluent from an anti-IgG column, but not from a protein A column exerted cardio-depressant effects on rat cardiomyocytes, whereas such cardio-depressant effects were abolished after removal of IgG3 subclass from eluent of an anti-IgG column [70]. Furthermore, IA using a protein A column with a modified protocol to effectively eliminate IgG3 subclass demonstrated better hemodynamic improvement in patients with DCM [71]. Previously, we performed an IA study for patients with refractory DCM by utilizing a tryptophan column. IA therapy removed IgG3 efficiently and to a greater extent than the other subclasses [54]. Left ventricular ejection fraction significantly increased after IA and such improvement in cardiac function correlated with the AAb titer belonging to IgG3 subclass more than total IgG, which also suggested that the removal of IgG3-AAb is important to maximize the effect of IA for the patients with DCM [62]. Therefore, AAb belonging to the IgG3 subclass may play a pivotal role in the onset and progression of cardiac dysfunction in new-onset DCM and the therapeutic efficacy of IA. From these findings, anti-cardiac AAbs belonging to IgG3 subclass might be involved in the pathogenesis of DCM, but further research is needed to elucidate this issue.

Autoimmune Dysregulation as a Possible Cause of Pregnancy-related Cardiovascular Diseases

While most research has been focused on deciphering the cause versus consequence theories of AAb generation in the setting of DCM, the potential role of autoimmune dysregulation has also been extended to other at-risk clinical states. Recently, detection of AAbs in pregnancy-related cardiovascular diseases point to extension of this concept beyond dilated cardiomyopathy.

Peripartum Cardiomyopathy

Peripartum cardiomyopathy (PPCM) is a serious disease during pregnancy with unknown etiology. Because of its rarity the cause or pathology of the disease has not yet been fully investigated. A literature search only revealed a few published investigations which examined the correlation between autoimmunity and PPCM. However, the hypothesis of autoimmunity as a possible cause of PPCM is supported by the findings that AAbs against adenine nucleotide translocator, branched chain α-keto acid dehydrogenase and cardiac myosin were found to be detected in patients with PPCM [72]. Furthermore, the serum titer of these three antibodies was observed to be higher in patients with versus without PPCM [72]. Whereas the IgG3 subclass anti-myosin AAbs was predominant in DCM patients, AAbs detected in PPCM patients were subclass nonspecific. However, among these PPCM patients, the presence of AAbs belonging to IgG3 was still associated with higher NYHA functional class and heart rate [73]. Although some potential correlation between IgG3-antimyosin AAbs and the pathology of PPCM would be suspected, this study was cross-sectional in design and more longitudinal data will be needed to elucidate this issue. On the other hand, functionally active β1AR-AAb was detected in PPCM patients as well [74]. Recently it was reported that β1AR-AAb and M2R-AAbs were detected in 60% and 46% of PPCM patients, respectively. The frequency and titer of both β1AR-AAb and M2R-AAbs had a positive correlation with left ventricular dimension, plasma NT-proBNP level, and NYHA functional class and a negative correlation with LVEF. After 12 months the titer and frequency of both AAbs significantly decreased [75]. Taken together, we can consider that the onset and course of PPCM accompanies some autoimmune activation, and it subsides in accordance with the amelioration of disease activity. However, further examination will be needed to elucidate the causal relationship between anti-cardiac AAbs and the pathology of PPCM.

Preeclampsia

Preeclampsia is characterized by proteinuria and hypertension during pregnancy. It is characterized by the presence of AAbs that bind to and activate the angiotensin II type1 receptor (AT1R) [76]. AAbs against AT1R (AT1R-AAb) isolated from sera of preeclamptic women increased reactive oxygen species (ROS) production, the NADPH oxidase components [77], and intracellular Ca2+ concentration in vitro [78]. Interestingly, the procedure of clipping ovarian arteries in pregnant rats to induce placental ischemia produced hypertension and proteinuria, representing the clinical features of preeclampsia [79]. A follow-up study found that AT1R-AAb was produced in this rat preeclampsia model [80]. AT1R-AAbs also were found to be detected in TNF-α infused pregnant rats, while non-pregnant rats did not produce AT1R-AAbs [80]. Passive transfer of AT1R-AAb extracted from preeclampsia patients induced hypertension and proteinuria in pregnant mice, but not in non-pregnant ones [81]. Collectively, AT1R-AAb works as one of the pathogenic factors in preeclampsia. However, the detailed mechanism by which AT1R-AAb is produced is still unknown.

Challenges in Clinical Translation

Despite these promising experimental findings, the applicability of AAbs testing and translation into clinical practice remains challenging. The first challenge is the issue of generalizability of findings, and the lack of broad clinical availability of testing. AAbs are largely measured in single-center, cross-sectional research studies (and often at a single-time point), in which autoimmune responses may differ among individuals or among geographic locations. In other words, their biological variability is largely unknown. The second challenge is the issue of availability and harmonization of laboratory assays. None of the AAb assays are clinically available; even measurements of AAbs against the same protein may vary widely, as they were developed by individual research laboratories without standardization. Finally, it has become increasingly clear that not all affected individuals have detectable AAbs. In addition, those with detectable AAbs may already have other established etiologies of heart failure, such as genetic/inherited or ischemic cardiomyopathies. It is therefore conceivable that pathogenic contributions of AAbs may only be applicable to a subset of susceptible individuals [32,33,82]. Whether targeting these subgroups may alter the natural history of the disease warrants further investigations.

Conclusions

AAbs can be produced in various diseases and conditions. Initially it had been considered to act as a diagnostic marker for DCM of autoimmune origin. However, some of these AAbs are physiologically active and can function as causal factors in the pathogenesis of multiple cardiovascular diseases (including DCM, PPCM, and preeclampsia), particularly of the IgG3 subclass. Hence, detection of AAbs might identify those who have autoimmune origins that may warrant the early initiation of conventional antagonists against their pathogenic effects (e.g. β-blockers for β1AR-AAb). Promising novel therapies such as IA which removes AAbs or AAb-neutralizing agents are also under investigation, albeit only providing temporary relief through a more invasive mechanism. We believe these therapeutic tools targeting pathogenic AAbs can potentially benefit selected individuals suffering from their intractable conditions. Still, further examination is necessary in order to elucidate the roles of AAbs as diagnostic tool in order to establish AAb-targeted personalized medicine for the population.

Funding

Dr. Nagatomo received research grant funding from the Myocarditis Foundation. Dr. Tang is supported by a grant from the National Institutes of Health (R01HL103931).

Footnotes

Compliance with Ethics Guidelines

Conflict of Interest

Yuji Nagatomo and W. H. Wilson Tang declare that they have no conflict of interest.

Human and Animal Rights and Informed Consent

This article does not contain any studies with human or animal subjects performed by any of the authors.

References

Papers of particular interest, published recently, have been highlighted as:

• Of importance

•• Of major importance

  • 1.Caforio AL, Grazzini M, Mann JM, Keeling PJ, Bottazzo GF, McKenna WJ, et al. Identification of alpha- and beta-cardiac myosin heavy chain isoforms as major autoantigens in dilated cardiomyopathy. Circulation. 1992;85:1734–1742. doi: 10.1161/01.cir.85.5.1734. [DOI] [PubMed] [Google Scholar]
  • 2.Schultheiss HP, Schulze K, Schauer R, Witzenbichler B, Strauer BE. Antibody-mediated imbalance of myocardial energy metabolism. A causal factor of cardiac failure? Circ Res. 1995;76:64–72. doi: 10.1161/01.res.76.1.64. [DOI] [PubMed] [Google Scholar]
  • 3.Limas CJ, Goldenberg IF, Limas C. Autoantibodies against beta-adrenoceptors in human idiopathic dilated cardiomyopathy. Circ Res. 1989;64:97–103. doi: 10.1161/01.res.64.1.97. [DOI] [PubMed] [Google Scholar]
  • 4.Iwata M, Yoshikawa T, Baba A, Anzai T, Nakamura I, Wainai Y, et al. Autoimmunity against the second extracellular loop of beta(1)-adrenergic receptors induces beta-adrenergic receptor desensitization and myocardial hypertrophy in vivo. Circ Res. 2001;88:578–586. doi: 10.1161/01.res.88.6.578. [DOI] [PubMed] [Google Scholar]
  • 5.Jahns R, Boivin V, Hein L, Triebel S, Angermann CE, Ertl G, et al. Direct evidence for a beta 1-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]
  • 6.Matsui S, Fu ML, Katsuda S, Hayase M, Yamaguchi N, Teraoka K, et al. Peptides derived from cardiovascular G-protein-coupled receptors induce morphological cardiomyopathic changes in immunized rabbits. J Mol Cell Cardiol. 1997;29:641–655. doi: 10.1006/jmcc.1996.0307. [DOI] [PubMed] [Google Scholar]
  • 7.Matsui S, Fu ML, Hayase M, Katsuda S, Yamaguchi N, Teraoka K, et al. Beneficial effect of muscarinic-2 antagonist on dilated cardiomyopathy induced by autoimmune mechanism against muscarinic-2 receptor. J Cardiovasc Pharmacol. 2001;38(Suppl 1):S43–S49. doi: 10.1097/00005344-200110001-00010. [DOI] [PubMed] [Google Scholar]
  • 8.Neu N, Rose NR, Beisel KW, Herskowitz A, Gurri-Glass G, Craig SW. Cardiac myosin induces myocarditis in genetically predisposed mice. J Immunol. 1987;139:3630–3636. [PubMed] [Google Scholar]
  • 9.Okazaki T, Tanaka Y, Nishio R, Mitsuiye T, Mizoguchi A, Wang J, et al. Autoantibodies against cardiac troponin I are responsible for dilated cardiomyopathy in PD-1-deficient mice. Nat Med. 2003;9:1477–1483. doi: 10.1038/nm955. [DOI] [PubMed] [Google Scholar]
  • 10.Smith SC, Allen PM. Myosin-induced acute myocarditis is a T cell-mediated disease. J Immunol. 1991;147:2141–2147. [PubMed] [Google Scholar]
  • 11.Liao L, Sindhwani R, Rojkind M, Factor S, Leinwand L, Diamond B. Antibody-mediated autoimmune myocarditis depends on genetically determined target organ sensitivity. J Exp Med. 1995;181:1123–1131. doi: 10.1084/jem.181.3.1123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Li Y, Heuser JS, Cunningham LC, Kosanke SD, Cunningham MW. Mimicry and antibody-mediated cell signaling in autoimmune myocarditis. J Immunol. 2006;177:8234–8240. doi: 10.4049/jimmunol.177.11.8234. [DOI] [PubMed] [Google Scholar]
  • 13.Caforio AL, Goldman JH, Baig MK, Haven AJ, Dalla Libera L, Keeling PJ, et al. Cardiac autoantibodies in dilated cardiomyopathy become undetectable with disease progression. Heart. 1997;77:62–67. doi: 10.1136/hrt.77.1.62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Goldman JH, Keeling PJ, Warraich RS, Baig MK, Redwood SR, Dalla Libera L, et al. Autoimmunity to alpha myosin in a subset of patients with idiopathic dilated cardiomyopathy. Br Heart J. 1995;74:598–603. doi: 10.1136/hrt.74.6.598. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Lauer B, Schannwell M, Kuhl U, Strauer BE, Schultheiss HP. Antimyosin autoantibodies are associated with deterioration of systolic and diastolic left ventricular function in patients with chronic myocarditis. J Am Coll Cardiol. 2000;35:11–18. doi: 10.1016/s0735-1097(99)00485-4. [DOI] [PubMed] [Google Scholar]
  • 16.Magnusson Y, Marullo S, Hoyer S, Waagstein F, Andersson B, Vahlne A, et al. Mapping of a functional autoimmune epitope on the beta 1-adrenergic receptor in patients with idiopathic dilated cardiomyopathy. J Clin Invest. 1990;86:1658–1663. doi: 10.1172/JCI114888. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Magnusson Y, Wallukat G, Waagstein F, Hjalmarson A, Hoebeke J. Autoimmunity in idiopathic dilated cardiomyopathy. Characterization of antibodies against the beta 1-adrenoceptor with positive chronotropic effect. Circulation. 1994;89:2760–2767. doi: 10.1161/01.cir.89.6.2760. [DOI] [PubMed] [Google Scholar]
  • 18.Magnusson Y, Hjalmarson A, Hoebeke J. Beta 1-adrenoceptor autoimmunity in cardiomyopathy. Int J Cardiol. 1996;54:137–141. doi: 10.1016/0167-5273(96)02590-9. [DOI] [PubMed] [Google Scholar]
  • 19.Jahns R, Boivin V, Siegmund C, Inselmann G, Lohse MJ, Boege F. Autoantibodies activating human beta1-adrenergic receptors are associated with reduced cardiac function in chronic heart failure. Circulation. 1999;99:649–654. doi: 10.1161/01.cir.99.5.649. [DOI] [PubMed] [Google Scholar]
  • 20.Wallukat G, Wollenberger A, Morwinski R, Pitschner HF. Anti-beta 1-adrenoceptor autoantibodies with chronotropic activity from the serum of patients with dilated cardiomyopathy: mapping of epitopes in the first and second extracellular loops. J Mol Cell Cardiol. 1995;27:397–406. doi: 10.1016/s0022-2828(08)80036-3. [DOI] [PubMed] [Google Scholar]
  • 21.Mobini R, Magnusson Y, Wallukat G, Viguier M, Hjalmarson A, Hoebeke J. Probing the immunological properties of the extracellular domains of the human beta(1)-adrenoceptor. J Autoimmun. 1999;13:179–186. doi: 10.1006/jaut.1999.0310. [DOI] [PubMed] [Google Scholar]
  • 22.Mobini R, Fu M, Wallukat G, Magnusson Y, Hjalmarson A, Hoebeke J. A monoclonal antibody directed against an autoimmune epitope on the human beta1-adrenergic receptor recognized in idiopathic dilated cardiomyopathy. Hybridoma. 2000;19:135–142. doi: 10.1089/02724570050031176. [DOI] [PubMed] [Google Scholar]
  • 23.Staudt A, Mobini R, Fu M, Grosse Y, Stangl V, Stangl K, et al. beta(1)-Adrenoceptor antibodies induce positive inotropic response in isolated cardiomyocytes. Eur J Pharmacol. 2001;423:115–119. doi: 10.1016/s0014-2999(01)01113-x. [DOI] [PubMed] [Google Scholar]
  • 24.Podlowski S, Luther HP, Morwinski R, Muller J, Wallukat G. Agonistic anti-beta1-adrenergic receptor autoantibodies from cardiomyopathy patients reduce the beta1-adrenergic receptor expression in neonatal rat cardiomyocytes. Circulation. 1998;98:2470–2476. doi: 10.1161/01.cir.98.22.2470. [DOI] [PubMed] [Google Scholar]
  • 25.Jahns R, Boivin V, Krapf T, Wallukat G, Boege F, Lohse MJ. Modulation of beta1-adrenoceptor activity by domain-specific antibodies and heart failure-associated autoantibodies. J Am Coll Cardiol. 2000;36:1280–1287. doi: 10.1016/s0735-1097(00)00881-0. [DOI] [PubMed] [Google Scholar]
  • 26.Staudt Y, Mobini R, Fu M, Felix SB, Kuhn JP, Staudt A. Beta1-adrenoceptor antibodies induce apoptosis in adult isolated cardiomyocytes. Eur J Pharmacol. 2003;466:1–6. doi: 10.1016/s0014-2999(03)01431-6. [DOI] [PubMed] [Google Scholar]
  • 27.Christ T, Wettwer E, Dobrev D, Adolph E, Knaut M, Wallukat G, et al. Autoantibodies against the beta1 adrenoceptor from patients with dilated cardiomyopathy prolong action potential duration and enhance contractility in isolated cardiomyocytes. J Mol Cell Cardiol. 2001;33:1515–1525. doi: 10.1006/jmcc.2001.1414. [DOI] [PubMed] [Google Scholar]
  • 28.Stork S, Boivin V, Horf R, Hein L, Lohse MJ, Angermann CE, et al. Stimulating autoantibodies directed against the cardiac beta1-adrenergic receptor predict increased mortality in idiopathic cardiomyopathy. Am Heart J. 2006;152:697–704. doi: 10.1016/j.ahj.2006.05.004. [DOI] [PubMed] [Google Scholar]
  • 29.Fukuda Y, Miyoshi S, Tanimoto K, Oota K, Fujikura K, Iwata M, et al. Autoimmunity against the second extracellular loop of beta(1)-adrenergic receptors induces early afterdepolarization and decreases in K-channel density in rabbits. J Am Coll Cardiol. 2004;43:1090–1100. doi: 10.1016/j.jacc.2003.09.057. [DOI] [PubMed] [Google Scholar]
  • 30.Nagatomo Y, Yoshikawa T, Kohno T, Yoshizawa A, Baba A, Anzai T, et al. A pilot study on the role of autoantibody targeting the beta1-adrenergic receptor in the response to beta-blocker therapy for congestive heart failure. J Card Fail. 2009;15:224–232. doi: 10.1016/j.cardfail.2008.10.027. [DOI] [PubMed] [Google Scholar]
  • 31.Wallukat G, Haberland A, Berg S, Schulz A, Freyse EJ, Dahmen C, et al. The first aptamer-apheresis column specifically for clearing blood of beta1-receptor autoantibodies. Circ J. 2012;76:2449–2455. doi: 10.1253/circj.cj-12-0212. [DOI] [PubMed] [Google Scholar]
  • 32.Jahns R, Boivin V, Siegmund C, Boege F, Lohse MJ, Inselmann G. Activating beta-1-adrenoceptor antibodies are not associated with cardiomyopathies secondary to valvular or hypertensive heart disease. J Am Coll Cardiol. 1999;34:1545–1551. doi: 10.1016/s0735-1097(99)00381-2. [DOI] [PubMed] [Google Scholar]
  • 33.Bornholz B, Weidtkamp-Peters S, Schmitmeier S, Seidel CA, Herda LR, Felix SB, et al. Impact of human autoantibodies on beta1-adrenergic receptor conformation, activity, and internalization. Cardiovasc Res. 2013;97:472–480. doi: 10.1093/cvr/cvs350. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Dandel M, Weng Y, Siniawski H, Potapov E, Drews T, Lehmkuhl HB, et al. Prediction of cardiac stability after weaning from left ventricular assist devices in patients with idiopathic dilated cardiomyopathy. Circulation. 2008;118:S94–S105. doi: 10.1161/CIRCULATIONAHA.107.755983. [DOI] [PubMed] [Google Scholar]
  • 35.Sterin-Borda L, Gorelik G, Borda ES. Chagasic IgG binding with cardiac muscarinic cholinergic receptors modifies cholinergic-mediated cellular transmembrane signals. Clin Immunol Immunopathol. 1991;61:387–397. doi: 10.1016/s0090-1229(05)80010-8. [DOI] [PubMed] [Google Scholar]
  • 36.Fu LX, Magnusson Y, Bergh CH, Liljeqvist JA, Waagstein F, Hjalmarson A, et al. Localization of a functional autoimmune epitope on the muscarinic acetylcholine receptor-2 in patients with idiopathic dilated cardiomyopathy. J Clin Invest. 1993;91:1964–1968. doi: 10.1172/JCI116416. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Wallukat G, Fu HM, Matsui S, Hjalmarson A, Fu ML. Autoantibodies against M2 muscarinic receptors in patients with cardiomyopathy display non-desensitized agonist-like effects. Life Sci. 1999;64:465–469. doi: 10.1016/s0024-3205(98)00589-x. [DOI] [PubMed] [Google Scholar]
  • 38.Baba A, Yoshikawa T, Fukuda Y, Sugiyama T, Shimada M, Akaishi M, et al. Autoantibodies against M2-muscarinic acetylcholine receptors: new upstream targets in atrial fibrillation in patients with dilated cardiomyopathy. Eur Heart J. 2004;25:1108–1115. doi: 10.1016/j.ehj.2004.05.012. [DOI] [PubMed] [Google Scholar]
  • 39.Del Corsso C, de Carvalho AC, Martino HF, Varanda WA. Sera from patients with idiopathic dilated cardiomyopathy decrease ICa in cardiomyocytes isolated from rabbits. Am J Physiol Heart Circ Physiol. 2004;287:H1928–H936. doi: 10.1152/ajpheart.00044.2004. [DOI] [PubMed] [Google Scholar]
  • 40.Yoshizawa A, Nagai S, Baba Y, Yamada T, Matsui M, Tanaka H, et al. Autoimmunity against M(2)muscarinic acetylcholine receptor induces myocarditis and leads to a dilated cardiomyopathy-like phenotype. Eur J Immunol. 2012;42:1152–1163. doi: 10.1002/eji.201142104. [DOI] [PubMed] [Google Scholar]
  • 41.Zou C, Zhang Z, Zhao W, Li G, Ma G, Yang X, et al. Predictive value of pre-procedural autoantibodies against M2-muscarinic acetylcholine receptor for recurrence of atrial fibrillation one year after radiofrequency catheter ablation. J Transl Med. 2013;11:17. doi: 10.1186/1479-5876-11-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Goser S, Andrassy M, Buss SJ, Leuschner F, Volz CH, Ottl R, et al. Cardiac troponin I but not cardiac troponin T induces severe autoimmune inflammation in the myocardium. Circulation. 2006;114:1693–1702. doi: 10.1161/CIRCULATIONAHA.106.635664. [DOI] [PubMed] [Google Scholar]
  • 43.Shmilovich H, Danon A, Binah O, Roth A, Chen G, Wexler D, et al. Autoantibodies to cardiac troponin I in patients with idiopathic dilated and ischemic cardiomyopathy. Int J Cardiol. 2007;117:198–203. doi: 10.1016/j.ijcard.2006.04.077. [DOI] [PubMed] [Google Scholar]
  • 44.Doesch AO, Mueller S, Nelles M, Konstandin M, Celik S, Frankenstein L, et al. Impact of troponin I-autoantibodies in chronic dilated and ischemic cardiomyopathy. Basic Res Cardiol. 2011;106:25–35. doi: 10.1007/s00395-010-0126-z. [DOI] [PubMed] [Google Scholar]
  • 45.Halley CM, Lappe JM, Cotleur AC, Shrestha K, Borowski AG, Pelfrey C, et al. Antiinflammatory autoimmune cellular responses to cardiac troponin I in idiopathic dilated cardiomyopathy. J Card Fail. 2011;17:359–365. doi: 10.1016/j.cardfail.2011.01.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Miettinen KH, Eriksson S, Magga J, Tuomainen P, Kuusisto J, Vanninen EJ, et al. Clinical significance of troponin I efflux and troponin autoantibodies in patients with dilated cardiomyopathy. J Card Fail. 2008;14:481–488. doi: 10.1016/j.cardfail.2008.02.009. [DOI] [PubMed] [Google Scholar]
  • 47.Dorffel WV, Felix SB, Wallukat G, Brehme S, Bestvater K, Hofmann T, et al. Short-term hemodynamic effects of immunoadsorption in dilated cardiomyopathy. Circulation. 1997;95:1994–1997. doi: 10.1161/01.cir.95.8.1994. [DOI] [PubMed] [Google Scholar]
  • 48.Felix SB, Staudt A, Dorffel WV, Stangl V, Merkel K, Pohl M, et al. Hemodynamic effects of immunoadsorption and subsequent immunoglobulin substitution in dilated cardiomyopathy: three-month results from a randomized study. J Am Coll Cardiol. 2000;35:1590–1598. doi: 10.1016/s0735-1097(00)00568-4. [DOI] [PubMed] [Google Scholar]
  • 49.Muller J, Wallukat G, Dandel M, Bieda H, Brandes K, Spiegelsberger S, et al. Immunoglobulin adsorption in patients with idiopathic dilated cardiomyopathy. Circulation. 2000;101:385–3891. doi: 10.1161/01.cir.101.4.385. [DOI] [PubMed] [Google Scholar]
  • 50.Knebel F, Bohm M, Staudt A, Borges AC, Tepper M, Jochmann N, et al. Reduction of morbidity by immunoadsorption therapy in patients with dilated cardiomyopathy. Int J Cardiol. 2004;97:517–520. doi: 10.1016/j.ijcard.2003.12.003. [DOI] [PubMed] [Google Scholar]
  • 51.Dorffel WV, Wallukat G, Dorffel Y, Felix SB, Baumann G. Immunoadsorption in idiopathic dilated cardiomyopathy, a 3-year follow-up. Int J Cardiol. 2004;97:529–534. doi: 10.1016/j.ijcard.2004.03.001. [DOI] [PubMed] [Google Scholar]
  • 52.Staudt A, Hummel A, Ruppert J, Dorr M, Trimpert C, Birkenmeier K, et al. Immunoadsorption in dilated cardiomyopathy: 6-month results from a randomized study. Am Heart J. 2006;152:712, e1–e6. doi: 10.1016/j.ahj.2006.06.027. [DOI] [PubMed] [Google Scholar]
  • 53.Cooper LT, Belohlavek M, Korinek J, Yoshifuku S, Sengupta PP, Burgstaler EA, et al. A pilot study to assess the use of protein a immunoadsorption for chronic dilated cardiomyopathy. J Clin Apher. 2007;22:210–214. doi: 10.1002/jca.20130. [DOI] [PubMed] [Google Scholar]
  • 54.Nagatomo Y, Baba A, Ito H, Naito K, Yoshizawa A, Kurita Y, et al. Specific immunoadsorption therapy using a tryptophan column in patients with refractory heart failure due to dilated cardiomyopathy. J Clin Apher. 2011;26:1–8. doi: 10.1002/jca.20268. [DOI] [PubMed] [Google Scholar]
  • 55.Wallukat G, Muller J, Hetzer R. Specific removal of beta1-adrenergic autoantibodies from patients with idiopathic dilated cardiomyopathy. N Engl J Med. 2002;347:1806. doi: 10.1056/NEJM200211283472220. [DOI] [PubMed] [Google Scholar]
  • 56.Schimke I, Muller J, Dandel M, Gremmels HD, Bayer W, Wallukat B, et al. Reduced oxidative stress in parallel to improved cardiac performance one year after selective removal of anti-beta 1-adrenoreceptor autoantibodies in patients with idiopathic dilated cardiomyopathy: data of a preliminary study. J Clin Apher. 2005;20:137–142. doi: 10.1002/jca.20050. [DOI] [PubMed] [Google Scholar]
  • 57.Mobini R, Staudt A, Felix SB, Baumann G, Wallukat G, Deinum J, et al. Hemodynamic improvement and removal of autoantibodies against beta1-adrenergic receptor by immunoadsorption therapy in dilated cardiomyopathy. J Autoimmun. 2003;20:345–350. doi: 10.1016/s0896-8411(03)00042-8. [DOI] [PubMed] [Google Scholar]
  • 58.Doesch AO, Konstandin M, Celik S, Kristen A, Frankenstein L, Hardt S, et al. Effects of protein A immunoadsorption in patients with advanced chronic dilated cardiomyopathy. J Clin Apher. 2009;24:141–149. doi: 10.1002/jca.20204. [DOI] [PubMed] [Google Scholar]
  • 59.Felix SB, Staudt A, Landsberger M, Grosse Y, Stangl V, Spielhagen T, et al. Removal of cardiodepressant antibodies in dilated cardiomyopathy by immunoadsorption. J Am Coll Cardiol. 2002;39:646–652. doi: 10.1016/s0735-1097(01)01794-6. [DOI] [PubMed] [Google Scholar]
  • 60. Staudt A, Staudt Y, Dorr M, Bohm M, Knebel F, Hummel A, et al. Potential role of humoral immunity in cardiac dysfunction of patients suffering from dilated cardiomyopathy. J Am Coll Cardiol. 2004;44:829–836. doi: 10.1016/j.jacc.2004.04.055. This article clearly showed the pathogenic significance of cardio-depressant AAb.
  • 61.Baba A. Autoantigen estimation and simple screening assay against cardiodepressant autoantibodies in patients with dilated cardiomyopathy. Ther Apher Dial. 2008;12:109–116. doi: 10.1111/j.1744-9987.2008.00555.x. [DOI] [PubMed] [Google Scholar]
  • 62. Baba A, Akaishi M, Shimada M, Monkawa T, Wakabayashi Y, Takahashi M, et al. Complete elimination of cardiodepressant IgG3 autoantibodies by immunoadsorption in patients with severe heart failure. Circ J. 2010;74:1372–1378. doi: 10.1253/circj.cj-09-0748. This article showed the pathogenic significance of cardio-depressant AAb and anti-cardiac AAb belonging to IgG3 subclass in human DCM.
  • 63. Staudt A, Eichler P, Trimpert C, Felix SB, Greinacher A. Fc(gamma) receptors IIa on cardiomyocytes and their potential functional relevance in dilated cardiomyopathy. J Am Coll Cardiol. 2007;49:1684–1692. doi: 10.1016/j.jacc.2006.11.051. This article showed the significance of Fc region in “cardio-depressant AAb” mediated pathogenesis in vitro.
  • 64.Bruggemann M, Williams GT, Bindon CI, Clark MR, Walker MR, Jefferis R, et al. Comparison of the effector functions of human immunoglobulins using a matched set of chimeric antibodies. J Exp Med. 1987;166:1351–1361. doi: 10.1084/jem.166.5.1351. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Warraich RS, Griffiths E, Falconar A, Pabbathi V, Bell C, Angelini G, et al. Human cardiac myosin autoantibodies impair myocyte contractility: a cause-and-effect relationship. Faseb J. 2006;20:651–660. doi: 10.1096/fj.04-3001com. [DOI] [PubMed] [Google Scholar]
  • 66.Warraich RS, Dunn MJ, Yacoub MH. Subclass specificity of autoantibodies against myosin in patients with idiopathic dilated cardiomyopathy: pro-inflammatory antibodies in DCM patients. Biochem Biophys Res Commun. 1999;259:255–261. doi: 10.1006/bbrc.1999.0761. [DOI] [PubMed] [Google Scholar]
  • 67.Warraich RS, Noutsias M, Kazak I, Seeberg B, Dunn MJ, Schultheiss HP, et al. Immunoglobulin G3 cardiac myosin autoantibodies correlate with left ventricular dysfunction in patients with dilated cardiomyopathy: immunoglobulin G3 and clinical correlates. Am Heart J. 2002;143:1076–1084. doi: 10.1067/mhj.2002.124406. [DOI] [PubMed] [Google Scholar]
  • 68.Baba A. Targeted Autoantibodies in Apheresis Treatment against Severe Heart Failure. Japanese Journal of Apheresis. 2010;29:187–193. [Google Scholar]
  • 69.Braun N, Gutenberger S, Erley CM, Risler T. Immunoglobulin and circulating immune complex kinetics during immunoadsorption onto protein A sepharose. Transfus Sci. 1998;19(Suppl):25–31. doi: 10.1016/s0955-3886(97)00099-4. [DOI] [PubMed] [Google Scholar]
  • 70.Staudt A, Bohm M, Knebel F, Grosse Y, Bischoff C, Hummel A, et al. Potential role of autoantibodies belonging to the immunoglobulin G-3 subclass in cardiac dysfunction among patients with dilated cardiomyopathy. Circulation. 2002;106:2448–2453. doi: 10.1161/01.cir.0000036746.49449.64. [DOI] [PubMed] [Google Scholar]
  • 71. Staudt A, Dorr M, Staudt Y, Bohm M, Probst M, Empen K, et al. Role of immunoglobulin G3 subclass in dilated cardiomyopathy: results from protein A immunoadsorption. Am Heart J. 2005;150:729–736. doi: 10.1016/j.ahj.2004.11.002. This article showed the different efficacy of IA by the efficiency of IgG3 AAb removal.
  • 72.Pearson GD, Veille JC, Rahimtoola S, Hsia J, Oakley CM, Hosenpud JD, et al. Peripartum cardiomyopathy: National Heart, Lung, and Blood Institute and Office of Rare Diseases (National Institutes of Health) workshop recommendations and review. Jama. 2000;283:1183–1188. doi: 10.1001/jama.283.9.1183. [DOI] [PubMed] [Google Scholar]
  • 73.Warraich RS, Sliwa K, Damasceno A, Carraway R, Sundrom B, Arif G, et al. Impact of pregnancy-related heart failure on humoral immunity: clinical relevance of G3-subclass immunoglobulins in peripartum cardiomyopathy. Am Heart J. 2005;150:263–269. doi: 10.1016/j.ahj.2004.09.008. [DOI] [PubMed] [Google Scholar]
  • 74.Haberland A, Wallukat G, Dahmen C, Kage A, Schimke I. Aptamer neutralization of beta1-adrenoceptor autoantibodies isolated from patients with cardiomyopathies. Circ Res. 2011;109:986–992. doi: 10.1161/CIRCRESAHA.111.253849. [DOI] [PubMed] [Google Scholar]
  • 75.Liu J, Wang Y, Chen M, Zhao W, Wang X, Wang H, et al. The correlation between peripartum cardiomyopathy and autoantibodies against cardiovascular receptors. PLoS One. 2014;9:e86770. doi: 10.1371/journal.pone.0086770. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Wallukat G, Homuth V, Fischer T, Lindschau C, Horstkamp B, Jupner A, et al. 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]
  • 77.Dechend R, Viedt C, Muller DN, Ugele B, Brandes RP, Wallukat G, et al. 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]
  • 78.Thway TM, Shlykov SG, Day MC, Sanborn BM, Gilstrap LC, 3rd, Xia Y, et al. Antibodies from preeclamptic patients stimulate increased intracellular Ca2+ mobilization through angiotensin receptor activation. Circulation. 2004;110:1612–1619. doi: 10.1161/01.CIR.0000142855.68398.3A. [DOI] [PubMed] [Google Scholar]
  • 79.Gadonski G, LaMarca BB, Sullivan E, Bennett W, Chandler D, Granger JP. Hypertension produced by reductions in uterine perfusion in the pregnant rat: role of interleukin 6. Hypertension. 2006;48:711–716. doi: 10.1161/01.HYP.0000238442.33463.94. [DOI] [PubMed] [Google Scholar]
  • 80.LaMarca B, Wallukat G, Llinas M, Herse F, Dechend R, Granger JP. Autoantibodies to the angiotensin type I receptor in response to placental ischemia and tumor necrosis factor alpha in pregnant rats. Hypertension. 2008;52:1168–1172. doi: 10.1161/HYPERTENSIONAHA.108.120576. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Zhou CC, Zhang Y, Irani RA, Zhang H, Mi T, Popek EJ, et al. 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]
  • 82.Chen J, Larsson L, Haugen E, Fedorkova O, Angwald E, Waagstein F, et al. Effects of autoantibodies removed by immunoadsorption from patients with dilated cardiomyopathy on neonatal rat cardiomyocytes. Eur J Heart Fail. 2006;8:460–467. doi: 10.1016/j.ejheart.2005.10.019. [DOI] [PubMed] [Google Scholar]
  • 83.Iwata M, Yoshikawa T, Baba A, Anzai T, Mitamura H, Ogawa S. Autoantibodies against the second extracellular loop of beta1-adrenergic receptors predict ventricular tachycardia and sudden death in patients with idiopathic dilated cardiomyopathy. J Am Coll Cardiol. 2001;37:418–424. doi: 10.1016/s0735-1097(00)01109-8. [DOI] [PubMed] [Google Scholar]
  • 84.Baba A, Yoshikawa T, Ogawa S. Autoantibodies produced against sarcolemmal Na-K-ATPase: possible upstream targets of arrhythmias and sudden death in patients with dilated cardiomyopathy. J Am Coll Cardiol. 2002;40:1153–1159. doi: 10.1016/s0735-1097(02)02075-2. [DOI] [PubMed] [Google Scholar]
  • 85.Baba A, Yoshikawa T, Iwata M, Anzai T, Nakamura I, Wainai Y, et al. Antigen-specific effects of autoantibodies against sarcolemmal Na-K-ATPase pump in immunized cardiomyopathic rabbits. Int J Cardiol. 2006;112:15–20. doi: 10.1016/j.ijcard.2006.05.026. [DOI] [PubMed] [Google Scholar]

RESOURCES