Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Curr Opin Pharmacol ; 76: 102458, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38636195

ABSTRACT

ß-blockers are a solid pillar in the treatment of cardiovascular diseases. However, they are highly discussed regarding effectiveness for certain indications and side-effects. Even though there are up to 20 licensed compounds, only four are used for heart failure (HF) therapy. On the receptor level several key characteristics seem to influence the clinical outcome: subtype selectivity, antagonistic vs (inverse/biased) agonistic properties and -in particular- ancillary capacities. On a molecular level, divergent and novel signaling patterns are being identified and extra-cardiac effects on e.g. inflammation, metabolism and oxidative stress are highlighted. This review discusses different well-known and newly discovered characteristics that need to be considered for HF therapy and in the context of co-morbidities.


Subject(s)
Adrenergic beta-Antagonists , Heart Failure , Receptors, Adrenergic, beta , Signal Transduction , Humans , Signal Transduction/drug effects , Animals , Heart Failure/drug therapy , Heart Failure/metabolism , Receptors, Adrenergic, beta/metabolism , Adrenergic beta-Antagonists/pharmacology , Adrenergic beta-Antagonists/therapeutic use
2.
Chemistry ; 30(11): e202303506, 2024 Feb 21.
Article in English | MEDLINE | ID: mdl-38212242

ABSTRACT

ß2 -adrenergic receptor (ß2 -AR) agonists are used for the treatment of asthma and chronic obstructive pulmonary disease, but also play a role in other complex disorders including cancer, diabetes and heart diseases. As the cellular and molecular mechanisms in various cells and tissues of the ß2 -AR remain vastly elusive, we developed tools for this investigation with high temporal and spatial resolution. Several photoswitchable ß2 -AR agonists with nanomolar activity were synthesized. The most potent agonist for ß2 -AR with reasonable switching is a one-digit nanomolar active, trans-on arylazopyrazole-based adrenaline derivative and comprises valuable photopharmacological properties for further biological studies with high structural accordance to the native ligand adrenaline.


Subject(s)
Adrenergic Agents , Adrenergic beta-2 Receptor Agonists , Adrenergic beta-2 Receptor Agonists/pharmacology , Molecular Probes , Receptors, Adrenergic, beta-2/chemistry , Epinephrine/pharmacology , Signal Transduction
3.
ESC Heart Fail ; 10(5): 3227-3231, 2023 10.
Article in English | MEDLINE | ID: mdl-37688355

ABSTRACT

AIMS: Agonistic antibodies against neurohumoral receptors can induce cardio-noxious effects by altering the baseline receptor activity. To estimate the prevalence of autoantibodies directed against the beta-1 receptor (b1-AAB) in patients admitted to the hospital for acute heart failure (HF) at (i) baseline and (ii) after 6 months of follow-up (F6) and (iii) after another 12 months of follow-up (i.e. 18 months after index hospitalization), to estimate their prognostic impact on clinical outcome (death or first hospitalization for HF). METHODS AND RESULTS: In 47 patients, b1-AAB were serially determined in serum samples collected at index hospitalization and at 6 months of follow-up (F6) with a flow cytometry-based assay: median age 71 years (quartiles 60, 80), 23 (49%) women, 24 (51%) HF with preserved ejection fraction. Beta1-AAB were detected in three subjects at index hospitalization (6%), and in eight subjects at F6 (17%). There were no differences apparent between patients with and without b1-AAB at F6 with regard to age, sex, type, duration, or main cause of HF. During the 12 month period following F6 (i.e. up to month 18), eight events occurred. Event-free survival was associated with prevalence of b1-AAB at F6. Compared with patients without b1-AAB at F6, age-adjusted Cox regression indicated a higher event risk in patients harbouring b1-AAB, with a hazard ratio of 8.96 (95% confidence interval 1.81-44.50, P = 0.007). CONCLUSIONS: Our results suggest a possible adverse prognostic relevance of b1-AAB in patients with acute HF, but this observation needs to be confirmed in larger patient collectives.


Subject(s)
Heart Failure , Aged , Female , Humans , Male , Heart Failure/epidemiology , Hospitalization , Prevalence , Prognosis , Middle Aged , Aged, 80 and over
4.
ESC Heart Fail ; 10(1): 432-441, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36271665

ABSTRACT

AIMS: Cellular communication network factor 1 (CCN1) is an independent predictor of MACE after ACS and elevated levels correlated with infarct size after STEMI. We compared the prognostic accuracy of baseline levels of CCN1, NT-proBNP, hsTnT, and ST2 and changes in levels over time to predict the development of structural and functional alterations typical of LV remodelling. METHODS: Serial 3-T cMRI scans were performed to determine LVEF, LVEDV, LVESV, infarct size, and relative infarct size, which were correlated with serial measurements of the four biomarkers. The prognostic significance of these biomarkers was assessed by multiple logistic regression analysis by examining their performance in predicting dichotomized cardiac MRI values 12 months after STEMI based on their median. For each biomarker three models were created using baseline (BL), the Δ value (BL to 6 months), and the two values together as predictors. All models were adjusted for age and renal function. Receiver operator curves were plotted with area under the curve (AUC) to discriminate the prognostic accuracy of individual biomarkers for MRI-based structural or functional changes. RESULTS: A total of 44 predominantly male patients (88.6%) from the ETiCS (Etiology, Titre-Course, and Survival) study were identified at a mean age of 55.5 ± 11.5 (SD) years treated by successful percutaneous coronary intervention (97.7%) at a rate of 95.5% stent implantation within a median pain-to-balloon time of 260 min (IQR 124-591). Biomarkers hsTnT and ST2 were identified as strong predictors (AUC > 0.7) of LVEDV and LVEF. BL measurement to predict LVEF [hsTnT: AUC 0.870 (95% CI: 0.756-0.983), ST2: AUC 0.763 (95% CI: 0.615-0.911)] and the Δ value BL-6M [hsTnT: AUC 0.870 (95% CI: 0.756-0.983), ST2: AUC 0.809 (95% CI: 0.679-0.939)] showed a high prognostic value without a significant difference for the comparison of the BL model vs. the Δ-value model (BL-6M) for hsTnT (P = 1) and ST2 (P = 0.304). The combined model that included baseline and Δ value as predictors was not able to improve the ability to predict LVEF [hsTnT: AUC 0.891 (0.791-0.992), P = 0.444; ST2: AUC 0.778 (0.638-0.918), P = 0.799]. Baseline levels of CCN1 were closely associated with LVEDV at 12 months [AUC 0.708 (95% CI: 0.551-0.865)] and infarct size [AUC 0.703 (95% CI: 0.534-0.872)]. CONCLUSIONS: Baseline biomarker levels of hsTnT and ST2 were the strongest predictors of LVEF and LVEDV at 12 months after STEMI. The association of CCN1 with LVEDV and infarct size warrants further study into the underlying pathophysiology of this novel biomarker.


Subject(s)
ST Elevation Myocardial Infarction , Humans , Male , Adult , Middle Aged , Aged , Female , Ventricular Remodeling/physiology , Interleukin-1 Receptor-Like 1 Protein , Stroke Volume , Biomarkers
5.
Int J Cardiol ; 370: 43-50, 2023 01 01.
Article in English | MEDLINE | ID: mdl-36306955

ABSTRACT

BACKGROUND: A high body mass index (BMI) is often associated with metabolic syndrome, which is accompanied by systemic low-grade chronic inflammation. Here, we analyzed whether BMI, other components of metabolic syndrome, and/or inflammatory markers correlate with left ventricular geometry, function, and infarct size as assessed by serial cardiac magnetic resonance imaging (MRI) after a first (clinically evident) ST-elevation MI (STEMI). METHODS: Within the Etiology, Titre-Course, and effect on Survival (ETiCS) study, cardiac MRI conducted 7-9 days and 12 months after MI enabled longitudinal characterization of patients with a first STEMI along with serial routine blood counts and multiplex cytokine measurements. RESULTS: Of 91 locally included STEMI patients, 47% were overweight (25 kg/m2 < BMI < 30 kg/m2) and 24% were obese (BMI ≥ 30 kg/m2). No patient died during 12 months of follow-up. Left ventricular ejection fraction (LVEF), measured 7-9 days after STEMI, was significantly lower in overweight (49.5 ± 7.1%) and obese (45.8 ± 12.0%) patients than in the normal weight group (56.2 ± 7.7%). Along with BMI (T = -3.8; p < 0.001), hemoglobin A1c (HbA1c; T = -3.1; p = 0.004) and peak C-reactive protein (T = -2.6; p = 0.013) emerged as independent predictors of worse LVEF 7-9 days post MI (R2 = 0.45). Only peak C-reactive protein (T = -4.4; p < 0.001), but not parameters of the metabolic syndrome, predicted worse LVEF 12 months after STEMI (R2 = 0.20). CONCLUSION: Both BMI and HbA1c correlated negatively with LVEF only early, but not late after STEMI. Peak CRP evolved as strongest predictor of cardiac function at all time points independent of the metabolic syndrome.


Subject(s)
Metabolic Syndrome , Myocardial Infarction , Percutaneous Coronary Intervention , ST Elevation Myocardial Infarction , Humans , Ventricular Function, Left , Stroke Volume , C-Reactive Protein , Metabolic Syndrome/complications , Percutaneous Coronary Intervention/methods , Inflammation/complications
6.
Int J Mol Sci ; 23(18)2022 Sep 07.
Article in English | MEDLINE | ID: mdl-36142218

ABSTRACT

Acute ischemic cardiac injury predisposes one to cognitive impairment, dementia, and depression. Pathophysiologically, recent positron emission tomography data suggest astroglial activation after experimental myocardial infarction (MI). We analyzed peripheral surrogate markers of glial (and neuronal) damage serially within 12 months after the first ST-elevation MI (STEMI). Serum levels of glial fibrillary acidic protein (GFAP) and neurofilament light chain (NfL) were quantified using ultra-sensitive molecular immunoassays. Sufficient biomaterial was available from 45 STEMI patients (aged 28 to 78 years, median 56 years, 11% female). The median (quartiles) of GFAP was 63.8 (47.0, 89.9) pg/mL and of NfL 10.6 (7.2, 14.8) pg/mL at study entry 0-4 days after STEMI. GFAP after STEMI increased in the first 3 months, with a median change of +7.8 (0.4, 19.4) pg/mL (p = 0.007). It remained elevated without further relevant increases after 6 months (+11.7 (0.6, 23.5) pg/mL; p = 0.015), and 12 months (+10.3 (1.5, 22.7) pg/mL; p = 0.010) compared to the baseline. Larger relative infarction size was associated with a higher increase in GFAP (ρ = 0.41; p = 0.009). In contrast, NfL remained unaltered in the course of one year. Our findings support the idea of central nervous system involvement after MI, with GFAP as a potential peripheral biomarker of chronic glial damage as one pathophysiologic pathway.


Subject(s)
Myocardial Infarction , ST Elevation Myocardial Infarction , Biocompatible Materials , Biomarkers , Female , Glial Fibrillary Acidic Protein , Humans , Intermediate Filaments , Male , Myocardial Infarction/diagnostic imaging , Neurofilament Proteins
7.
ESC Heart Fail ; 7(5): 2354-2364, 2020 10.
Article in English | MEDLINE | ID: mdl-32548915

ABSTRACT

AIMS: Acute myocardial infarction (MI) is the major cause of chronic heart failure. The activity of blood coagulation factor XIII (FXIIIa) plays an important role in rodents as a healing factor after MI, whereas its role in healing and remodelling processes in humans remains unclear. We prospectively evaluated the relevance of FXIIIa after acute MI as a potential early prognostic marker for adequate healing. METHODS AND RESULTS: This monocentric prospective cohort study investigated cardiac remodelling in patients with ST-elevation MI and followed them up for 1 year. Serum FXIIIa was serially assessed during the first 9 days after MI and after 2, 6, and 12 months. Cardiac magnetic resonance imaging was performed within 4 days after MI (Scan 1), after 7 to 9 days (Scan 2), and after 12 months (Scan 3). The FXIII valine-to-leucine (V34L) single-nucleotide polymorphism rs5985 was genotyped. One hundred forty-six patients were investigated (mean age 58 ± 11 years, 13% women). Median FXIIIa was 118% (quartiles, 102-132%) and dropped to a trough on the second day after MI: 109% (98-109%; P < 0.001). FXIIIa recovered slowly over time, reaching the baseline level after 2 to 6 months and surpassed baseline levels only after 12 months: 124% (110-142%). The development of FXIIIa after MI was independent of the genotype. FXIIIa on Day 2 was strongly and inversely associated with the relative size of MI in Scan 1 (Spearman's ρ = -0.31; P = 0.01) and Scan 3 (ρ = -0.39; P < 0.01) and positively associated with left ventricular ejection fraction: ρ = 0.32 (P < 0.01) and ρ = 0.24 (P = 0.04), respectively. CONCLUSIONS: FXIII activity after MI is highly dynamic, exhibiting a significant decline in the early healing period, with reconstitution 6 months later. Depressed FXIIIa early after MI predicted a greater size of MI and lower left ventricular ejection fraction after 1 year. The clinical relevance of these findings awaits to be tested in a randomized trial.


Subject(s)
Myocardial Infarction , Ventricular Remodeling , Factor XIII/genetics , Humans , Myocardial Infarction/diagnosis , Prospective Studies , Stroke Volume , Ventricular Function, Left
8.
ESC Heart Fail ; 7(4): 1830-1841, 2020 08.
Article in English | MEDLINE | ID: mdl-32436653

ABSTRACT

AIMS: Chronic heart failure (CHF) can be caused by autoantibodies stimulating the heart via binding to first and/or second extracellular loops of cardiac ß1 -adrenoceptors. Allosteric receptor activation depends on conformational features of the autoantibody binding site. Elucidating these features will pave the way for the development of specific diagnostics and therapeutics. Our aim was (i) to fine-map the conformational epitope within the second extracellular loop of the human ß1 -adrenoceptor (ß1 ECII ) that is targeted by stimulating ß1 -receptor (auto)antibodies and (ii) to generate competitive cyclopeptide inhibitors of allosteric receptor activation, which faithfully conserve the conformational auto-epitope. METHODS AND RESULTS: Non-conserved amino acids within the ß1 ECII loop (compared with the amino acids constituting the ECII loop of the ß2 -adrenoceptor) were one by one replaced with alanine; potential intra-loop disulfide bridges were probed by cysteine-serine exchanges. Effects on antibody binding and allosteric receptor activation were assessed (i) by (auto)antibody neutralization using cyclopeptides mimicking ß1 ECII  ± the above replacements, and (ii) by (auto)antibody stimulation of human ß1 -adrenoceptors bearing corresponding point mutations. With the use of stimulating ß1 -receptor (auto)antibodies raised in mice, rats, or rabbits and isolated from exemplary dilated cardiomyopathy patients, our series of experiments unmasked two features of the ß1 ECII loop essential for (auto)antibody binding and allosteric receptor activation: (i) the NDPK211-214 motif and (ii) the intra-loop disulfide bond C209 ↔C215 . Of note, aberrant intra-loop disulfide bond C209 ↔C216 almost fully disrupted the functional auto-epitope in cyclopeptides. CONCLUSIONS: The conformational auto-epitope targeted by cardio-pathogenic ß1 -receptor autoantibodies is faithfully conserved in cyclopeptide homologues of the ß1 ECII loop bearing the NDPK211-214 motif and the C209 ↔C215 bridge while lacking cysteine C216 . Such molecules provide promising tools for novel diagnostic and therapeutic approaches in ß1 -autoantibody-positive CHF.


Subject(s)
Cardiomyopathy, Dilated , Heart Failure , Animals , Autoantibodies , Cardiomyopathy, Dilated/diagnosis , Epitopes , Heart Failure/diagnosis , Humans , Mice , Rabbits , Rats , Receptors, Adrenergic, beta-1/genetics
9.
Analyst ; 145(2): 486-496, 2020 Jan 21.
Article in English | MEDLINE | ID: mdl-31781708

ABSTRACT

Dilated cardiomyopathy (DCM) is a leading cardiomyopathy condition and is the leading reason for heart transplantation. Due to high etiologic and genetic heterogeneity of the pathologies, different therapeutic treatment strategies are available and have been successful for different treatments. Immunoadsorption (IA) therapy removes the circulating anticardiac antibodies and improves the left ventricular function in substantial proportion of DCM patients. Powerful, non-invasive analytical tools are highly desired to investigate the efficiency and success of IA therapy. In this contribution, we followed the changes of a female DCM patient undergoing IA therapy at different treatment time points in a label-free, non-invasive manner from blood samples (plasma and serum) on the basis of vibrational spectroscopy (Raman scattering and IR absorption). Chemometric methods, including dimension reduction and statistical modeling, were used to interpret spectral data. The impact of different time points of the IA treatment can be identified in both the plasma and serum, using both techniques, with high accuracy. The removal of antibodies of immunoglobulin G (IgG) group during IA therapy and their restoration was reflected in both Raman and FTIR spectra. Relative changes in the spectral bands assigned to IgG agreed well with the immunoturbidimetry measurement of total IgG. Successful clinical treatment was accompanied by spectral differences between vibrational spectra obtained at initial disease state and 11 months after the IA treatment. The long-term follow-up of the patient reveals the stabilization of the health state after therapy. It is noteworthy that the treatment time points were distinguished with a better accuracy using spectra from plasma compared to those from serum samples, which might indicate the involvement of corresponding proteins in the coagulation. Vibrational spectroscopy is a powerful tool for personalized medicine to follow-up the treatment success of IA therapy for the DCM disorder.


Subject(s)
Cardiomyopathy, Dilated/therapy , Immunoglobulin G/blood , Plasmapheresis , Female , Follow-Up Studies , Humans , Middle Aged , Precision Medicine/methods , Principal Component Analysis , Spectrum Analysis/methods , Time Factors , Vibration
10.
Science ; 366(6467): 881-886, 2019 11 15.
Article in English | MEDLINE | ID: mdl-31727837

ABSTRACT

Myocarditis can develop into inflammatory cardiomyopathy through chronic stimulation of myosin heavy chain 6-specific T helper (TH)1 and TH17 cells. However, mechanisms governing the cardiotoxicity programming of heart-specific T cells have remained elusive. Using a mouse model of spontaneous autoimmune myocarditis, we show that progression of myocarditis to lethal heart disease depends on cardiac myosin-specific TH17 cells imprinted in the intestine by a commensal Bacteroides species peptide mimic. Both the successful prevention of lethal disease in mice by antibiotic therapy and the significantly elevated Bacteroides-specific CD4+ T cell and B cell responses observed in human myocarditis patients suggest that mimic peptides from commensal bacteria can promote inflammatory cardiomyopathy in genetically susceptible individuals. The ability to restrain cardiotoxic T cells through manipulation of the microbiome thereby transforms inflammatory cardiomyopathy into a targetable disease.


Subject(s)
Autoimmune Diseases/complications , Bacteroides/immunology , Cardiomyopathy, Dilated/immunology , Cardiomyopathy, Dilated/microbiology , Gastrointestinal Microbiome/immunology , Myocarditis/complications , Peptides/immunology , beta-Galactosidase/immunology , Animals , Autoimmune Diseases/immunology , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Disease Models, Animal , Humans , Intestines/microbiology , Mice , Mice, Inbred BALB C , Mice, Transgenic , Myocarditis/immunology , Myosin Heavy Chains/genetics , Myosin Heavy Chains/immunology , Th17 Cells/immunology
11.
Eur Heart J Cardiovasc Imaging ; 20(4): 467-474, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30102319

ABSTRACT

AIMS: Although mortality rate is very high, diagnosis of acute myocarditis remains challenging with conventional tests. We aimed to elucidate the potential role of longitudinal 2-Deoxy-2-18F-fluoro-D-glucose (18F-FDG) positron emission tomography (PET) inflammation monitoring in a rat model of experimental autoimmune myocarditis. METHODS AND RESULTS: Autoimmune myocarditis was induced in Lewis rats by immunizing with porcine cardiac myosin emulsified in complete Freund's adjuvant. Time course of disease was assessed by longitudinal 18F-FDG PET imaging. A correlative analysis between in- and ex vivo18F-FDG signalling and macrophage infiltration using CD68 staining was conducted. Finally, immunohistochemistry analysis of the cell-adhesion markers CD34 and CD44 was performed at different disease stages determined by longitudinal 18F-FDG PET imaging. After immunization, myocarditis rats revealed a temporal increase in 18F-FDG uptake (peaked at week 3), which was followed by a rapid decline thereafter. Localization of CD68 positive cells was well correlated with in vivo18F-FDG PET signalling (R2 = 0.92) as well as with ex vivo18F-FDG autoradiography (R2 = 0.9, P < 0.001, respectively). CD44 positivity was primarily observed at tissue samples obtained at acute phase (i.e. at peak 18F-FDG uptake), while CD34-positive staining areas were predominantly identified in samples harvested at both sub-acute and chronic phases (i.e. at 18F-FDG decrease). CONCLUSION: 18F-FDG PET imaging can provide non-invasive serial monitoring of cardiac inflammation in a rat model of acute myocarditis.


Subject(s)
Inflammation/diagnostic imaging , Myocarditis/diagnostic imaging , Positron-Emission Tomography , Acute Disease , Animals , Autoimmune Diseases/complications , Disease Models, Animal , Female , Fluorodeoxyglucose F18 , Myocarditis/etiology , Radiopharmaceuticals , Rats , Rats, Inbred Lew
12.
PLoS One ; 13(8): e0201160, 2018.
Article in English | MEDLINE | ID: mdl-30125285

ABSTRACT

RATIONALE: Despite advances in pharmacotherapy, heart failure still incurs significant morbidity and mortality. Stimulating antibodies directed against the secondextracellular loop of the human ß1-adrenergic receptor (anti-ß1EC2) cause myocyte damage and heart failure in rats. This receptor domain is 100% homologous between rats and humans. OBJECTIVE: ß1EC2-mimicking cyclopeptides (25-meric) markedly improved the development and/or course of anti-ß1EC2-mediated cardiomyopathy. Further developments should be investigated. METHODS AND RESULTS: The shortened 18-meric cyclic peptide COR-1, in which one of the two disulphide bonds was removed to enable reproducible GMP production, can also be used to treat cardiomyopathic rats. Echocardiography, catheterization and histopathology of the rat hearts revealed that monthly intravenous administrations of COR-1 almost fully reversed the cardiomyopathic phenotype within 6 months at doses of 1 to 4 mg/kg body weight. Administration of COR-1 resulted in markedly reduced anti-ß1EC2-expressing memory B lymphocytes in the spleen despite continued antigenic boosts, but did not significantly decrease overall peripheral anti-ß1EC2 titers. COR-1 did not induce any anti-ß1EC2 or other immune response in naïve rats (corresponding to findings in healthy human volunteers). It did not cause any toxic side effects in GLP studies in dogs, rats or mice, and the "no observed adverse effect level" (NOAEL) exceeded the therapeutic doses by 100-fold. CONCLUSION: The second generation immunomodulating epitope-mimicking cyclopeptide COR-1 (also termed JNJ-5442840) offers promise to treat immune-mediated cardiac diseases.


Subject(s)
Heart Failure/drug therapy , Peptides, Cyclic/therapeutic use , Receptors, Adrenergic, beta-1/immunology , Amino Acid Sequence , Animals , Antibodies, Monoclonal/administration & dosage , Antibody Specificity , Disease Models, Animal , Female , Guinea Pigs , Heart Failure/etiology , Heart Failure/physiopathology , Humans , Male , Molecular Mimicry/immunology , Myocardium/metabolism , Peptide Fragments/chemistry , Peptide Fragments/therapeutic use , Peptides, Cyclic/chemistry , Peptides, Cyclic/toxicity , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Inbred Lew , Rats, Wistar , Receptors, Adrenergic, beta-1/chemistry , Receptors, Adrenergic, beta-1/genetics
13.
Front Biosci (Landmark Ed) ; 23(11): 2065-2081, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29772546

ABSTRACT

Chronic heart failure (CHF) is a syndrome characterized by shortness of breath, fluid retention, and a progressive reduction in cardiac function. More than 60% of the cases are ischemic in origin (i.e., due to myo-cardial infarction) and about 30% are caused by non-ischemic myocardial damage (i.e., due to genetic or non-genetic causes like myocardial inflammation). Because of alterations in both cellular and humoral immunity patients with non-ischemic CHF often develop abnormal or misled immune responses, including cross-reacting antibodies and/or autoantibodies to various cardiac anti-gens. Non-ischemic myo-cardial damage was found to progress to CHF particularly, when associated (a) with the generation of autoantibodies directed against distinct myocyte membrane proteins critically involved in cardiac function - like G-protein coup-led membrane receptors (GPCRs), or (b) with virus persistence in the myocardium. This article will review current knowledge on the pathophysiological relevance of GPCR-autoreactivity in CHF by giving an overview on the so far available evidence from pre-clinical, clinical and epidemiological studies on the CHF-inducing potential of GPCR-autoantibodies and thereon based novel therapeutic approaches in GPCR autoantibody-associated CHF.


Subject(s)
Autoantibodies/immunology , Heart Failure/immunology , Myocytes, Cardiac/immunology , Receptors, G-Protein-Coupled/immunology , Animals , Autoimmunity/drug effects , Autoimmunity/immunology , Chronic Disease , Ghrelin/therapeutic use , Heart Failure/diagnosis , Heart Failure/drug therapy , Humans , Myocytes, Cardiac/drug effects
14.
Front Biosci (Landmark Ed) ; 23(11): 2146-2156, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29772551

ABSTRACT

Patients suffering from chronic heart failure (CHF) caused or promoted by autoantibodies against cardiac beta1-adrenergic receptors (beta1AR) could benefit from specific therapies aimed at tolerance induction, removal or neutralisation of beta1AR autoantibodies, provided the patients can be selected for these therapies by reliable detection and quantitation of beta1AR autoantibodies in their circulation and by a valid assessment of the autoantibodies's putative cardio-pathogenic potential. Here, we discuss the current state of knowledge regarding the effects of CHF-associated (auto)antibodies on beta1AR function and beta1AR-mediated signal tranduction and discuss the presumed role of these effects in the development and progression of CHF. Identification of disease-relevant functional autoantibody effects and their specific assessment in medical diagnostic will be a prerequesite for the implementation of novel specific therapies not only for CHF caused or promoted by beta1AR autoantibodies but alos for most other diseases involving autoantibodies that target G-protein coupled receptors.


Subject(s)
Autoantibodies/immunology , Heart Failure/immunology , Receptors, Adrenergic, beta-1/immunology , Signal Transduction/immunology , Animals , Autoantibodies/blood , Chronic Disease , Disease Progression , Heart Failure/blood , Heart Failure/diagnosis , Humans , Myocytes, Cardiac/immunology , Myocytes, Cardiac/metabolism , Prognosis
15.
J Nucl Med ; 57(12): 1985-1990, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27390159

ABSTRACT

Myocarditis represents a major cause of dilated cardiomyopathy and sudden cardiac death in younger adults. Currently, definitive diagnosis of myocarditis requires endomyocardial biopsy, which is highly invasive and has the drawback of variable sensitivity due to inherent sampling error. Therefore, reliable noninvasive methods to detect and monitor cardiac inflammation are clinically relevant. In this study, we explored the potential of radiolabeled methionine to assess myocardial inflammatory activity in a rat model of experimental autoimmune myocarditis (EAM). METHODS: Autoimmune myocarditis was induced by immunizing Lewis rats twice with porcine cardiac myosin and Freund complete adjuvant. Control animals were treated with adjuvant alone. Dual-tracer autoradiography was performed to assess 14C-methionine uptake and to compare the distributions of 14C-methionine versus 18F-FDG. Hematoxylin and eosin staining and anti-CD68 macrophage staining were performed for histologic analysis. Additionally, cardiac 11C-methionine PET was performed to evaluate the feasibility of in vivo imaging. 18F-FDG PET was also conducted to compare the in vivo uptake of 11C-methionine and 18F-FDG. RESULTS: Multiple focal cardiac inflammatory lesions were histologically identified in myosin-immunized rats, whereas no cardiac lesions were observed in the controls. Autoradiographic images clearly showed a high-density accumulation of 14C-methionine in inflammatory lesions of EAM rats, whereas no significant uptake was observed in the control animals. 14C-methionine uptake was significantly higher in inflammatory lesions than in remote noninflammatory areas and control rat hearts. The distribution of 14C-methionine correlated well with that of 18F-FDG and with macrophage density. The contrast between inflammatory and noninflammatory areas was higher for 18F-FDG than for 14C-methionine (3.45 ± 0.68 vs. 2.07 ± 0.21, respectively; P < 0.05). In the PET imaging study, the regional 11C-methionine uptake (percentage injected dose per cubic centimeter) observed in EAM rats was significantly higher than the values obtained for control animals (0.64 ± 0.09 vs. 0.28 ± 0.02, respectively; P < 0.001). A good positive correlation between 11C-methionine and 18F-FDG uptake was found. CONCLUSION: In a rat model of autoimmune myocarditis, we demonstrated the colocalization of radiolabeled methionine accumulation with 18F-FDG uptake in histologically proven inflammatory lesions. These data suggest that 11C-methionine might represent a promising candidate for the noninvasive detection and monitoring of myocarditis.


Subject(s)
Autoimmune Diseases/diagnostic imaging , Carbon Radioisotopes , Methionine , Myocarditis/diagnostic imaging , Positron-Emission Tomography , Animals , Autoimmune Diseases/metabolism , Biological Transport , Disease Models, Animal , Female , Methionine/chemistry , Methionine/metabolism , Myocarditis/metabolism , Rats
17.
Circulation ; 128(17): 1878-88, 2013 Oct 22.
Article in English | MEDLINE | ID: mdl-24025595

ABSTRACT

BACKGROUND: Monocytes and macrophages are indispensable in the healing process after myocardial infarction (MI); however, the spatiotemporal distribution of monocyte infiltration and its correlation to prognostic indicators of reperfused MI have not been well described. METHODS AND RESULTS: With combined fluorine 19/proton ((1)H) magnetic resonance imaging, we noninvasively visualized the spatiotemporal recruitment of monocytes in vivo in a rat model of reperfused MI. Blood monocytes were labeled by intravenous injection of (19)F-perfluorocarbon emulsion 1 day after MI. The distribution patterns of monocyte infiltration were correlated to the presence of microvascular obstruction (MVO) and intramyocardial hemorrhage. In vivo, (19)F/(1)H magnetic resonance imaging performed in series revealed that monocyte infiltration was spatially inhomogeneous in reperfused MI areas. In the absence of MVO, monocyte infiltration was more intense in MI regions with serious ischemia-reperfusion injuries, indicated by severe intramyocardial hemorrhage; however, monocyte recruitment was significantly impaired in MVO areas accompanied by severe intramyocardial hemorrhage. Compared with MI with isolated intramyocardial hemorrhage, MI with MVO resulted in significantly worse pump function of the left ventricle 28 days after MI. CONCLUSIONS: Monocyte recruitment was inhomogeneous in reperfused MI tissue. It was highly reduced in MVO areas defined by magnetic resonance imaging. The impaired monocyte infiltration in MVO regions could be related to delayed healing and worse functional outcomes in the long term. Therefore, monocyte recruitment in MI with MVO could be a potential diagnostic and therapeutic target that could be monitored noninvasively and longitudinally by (19)F/(1)H magnetic resonance imaging in vivo.


Subject(s)
Cell Movement/physiology , Coronary Circulation/physiology , Hemorrhage/physiopathology , Magnetic Resonance Imaging/methods , Monocytes/cytology , Myocardial Infarction/physiopathology , Myocardial Reperfusion , Animals , Disease Models, Animal , Female , Fluorine Radioisotopes , Hemorrhage/diagnostic imaging , Macrophages/cytology , Macrophages/physiology , Microcirculation/physiology , Monocytes/physiology , Myocardial Infarction/diagnostic imaging , Protons , Radionuclide Imaging , Rats , Rats, Wistar , Wound Healing/physiology
18.
Eur J Heart Fail ; 14(11): 1230-9, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22968742

ABSTRACT

AIMS: A novel concept for the treatment of heart failure is the neutralization of antibodies against the ß(1)-adrenergic receptor (anti-ß(1)AR-ab). In a rat model of autoimmune cardiomyopathy, the cyclic peptide COR-1 (given i.v. once monthly) neutralized anti-ß(1)AR-abs and prevented anti-ß(1)AR-ab-induced myocardial damage, and completely reverted cardiac dysfunction over 3-6 months. METHODS AND RESULTS: A clinical phase I trial was designed as a single-blinded, placebo-controlled study. Fifty human volunteers received COR-1 or matching placebo as a single i.v. administration with ascending doses (10-240 mg). Primary endpoints were safety and tolerability, while the pharmacokinetic profile of COR-1 was assessed as a secondary endpoint. All five investigated dose groups were well tolerated; no drug-related side effects occurred. Pharmacokinetics revealed a favourable profile with an almost complete plasma clearance within 60 min after administration. Pharmacodynamic investigation showed dose-dependent efficacy with almost complete scavenging of pathological anti-ß(1)AR-abs ex vivo at the two highest doses. No anti-COR-1 autoantibodies occurred. No other effects on the immune system (such as an increase of crucial cytokines) were observed up to 43 days after drug administration, nor upon incubation of anti-ß(1)AR-ab-positive patient blood samples with COR-1 ex vivo. CONCLUSIONS: COR-1 was shown to be safe after i.v. administration in vivo; no relevant side effects occurred. Efficacy was estimated from ex vivo investigation of the potency to neutralize specific anti-ß(1)-AR-abs. TRIAL REGISTRATION: NCT 01043146, Eudra CT 2008-007745-31.


Subject(s)
Adrenergic Antagonists/therapeutic use , Heart Failure/immunology , Immune System/drug effects , Peptides, Cyclic/therapeutic use , Receptors, Adrenergic/drug effects , Adrenergic Antagonists/administration & dosage , Adult , Analysis of Variance , Animals , Area Under Curve , Autoantibodies/immunology , Cytokines , Female , Heart Failure/drug therapy , Heart Failure/pathology , Hemodynamics , Humans , Immune System/immunology , Male , Peptides, Cyclic/administration & dosage , Rats , Single-Blind Method
SELECTION OF CITATIONS
SEARCH DETAIL
...