Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
2.
Cardiovasc Res ; 116(11): 1875-1886, 2020 09 01.
Article in English | MEDLINE | ID: mdl-31711127

ABSTRACT

AIMS: Peripartum cardiomyopathy (PPCM) is a life-threatening heart disease occurring in previously heart-healthy women. A common pathomechanism in PPCM involves the angiostatic 16 kDa-prolactin (16 kDa-PRL) fragment, which via NF-κB-mediated up-regulation of microRNA-(miR)-146a induces vascular damage and heart failure. We analyse whether the plasminogen activator inhibitor-1 (PAI-1) is involved in the pathophysiology of PPCM. METHODS AND RESULTS: In healthy age-matched postpartum women (PP-Ctrl, n = 53, left ventricular ejection fraction, LVEF > 55%), PAI-1 plasma levels were within the normal range (21 ± 10 ng/mL), but significantly elevated (64 ± 38 ng/mL, P < 0.01) in postpartum PPCM patients at baseline (BL, n = 64, mean LVEF: 23 ± 8%). At 6-month follow-up (n = 23), PAI-1 levels decreased (36 ± 14 ng/mL, P < 0.01 vs. BL) and LVEF (49 ± 11%) improved. Increased N-terminal pro-brain natriuretic peptide and Troponin T did not correlate with PAI-1. C-reactive protein, interleukin (IL)-6 and IL-1ß did not differ between PPCM patients and PP-Ctrl. MiR-146a was 3.6-fold (P < 0.001) higher in BL-PPCM plasma compared with PP-Ctrl and correlated positively with PAI-1. In BL-PPCM serum, 16 kDa-PRL coprecipitated with PAI-1, which was associated with higher (P < 0.05) uPAR-mediated NF-κB activation in endothelial cells compared with PP-Ctrl serum. Cardiac biopsies and dermal fibroblasts from PPCM patients displayed higher PAI-1 mRNA levels (P < 0.05) than healthy controls. In PPCM mice (due to a cardiomyocyte-specific-knockout for STAT3, CKO), cardiac PAI-1 expression was higher than in postpartum wild-type controls, whereas a systemic PAI-1-knockout in CKO mice accelerated peripartum cardiac fibrosis, inflammation, heart failure, and mortality. CONCLUSION: In PPCM patients, circulating and cardiac PAI-1 expression are up-regulated. While circulating PAI-1 may add 16 kDa-PRL to induce vascular impairment via the uPAR/NF-κB/miR-146a pathway, experimental data suggest that cardiac PAI-1 expression seems to protect the PPCM heart from fibrosis. Thus, measuring circulating PAI-1 and miR-146a, together with an uPAR/NF-κB-activity assay could be developed into a specific diagnostic marker assay for PPCM, but unrestricted reduction of PAI-1 for therapy may not be advised.


Subject(s)
Cardiomyopathies/blood , Peripartum Period/blood , Plasminogen Activator Inhibitor 1/blood , Puerperal Disorders/blood , Adult , Animals , Biomarkers/blood , Cardiomyopathies/diagnostic imaging , Cardiomyopathies/physiopathology , Case-Control Studies , Disease Models, Animal , Female , Humans , Mice, Knockout , Myocytes, Cardiac/metabolism , Parity , Plasminogen Activator Inhibitor 1/genetics , Pregnancy , Prognosis , Puerperal Disorders/diagnostic imaging , Puerperal Disorders/physiopathology , Recovery of Function , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Stroke Volume , Time Factors , Up-Regulation , Ventricular Function, Left
3.
Cardiovasc Res ; 113(6): 598-608, 2017 May 01.
Article in English | MEDLINE | ID: mdl-28453725

ABSTRACT

AIMS: Peripartum cardiomyopathy (PPCM) is a systolic left ventricular dysfunction developing in the peripartum phase in previously healthy women. Relaxin-2 is a pregnancy hormone with potential beneficial effects in heart failure patients. We evaluated Relaxin-2 as a potential diagnostic marker and/or a therapeutic agent in PPCM. METHODS AND RESULTS: In healthy peripartum women, serum Relaxin-2 levels (measured by ELISA in the second half of pregnancy) were elevated showing a decreasing trend in the first postpartum week and returned to non-pregnant levels thereafter. In PPCM patients diagnosed in the first postpartum week, serum Relaxin-2 levels were lower compared to healthy postpartum stage-matched controls. In PPCM patients diagnosed later (0.5-10 months postpartum) Relaxin-2 levels were in the range of non-pregnant controls and not different from healthy postpartum stage-matched controls. In mice, serum Relaxin-1 (functional equivalent of human Relaxin-2) was increased late in pregnancy and rapidly cleared in the first postpartum week. In mice with PPCM due to a cardiomyocyte-specific knockout of STAT3 (CKO) neither low nor high dose of recombinant Relaxin-2 (serelaxin, sRlx-LD: 30 µg/kg/day; sRlx-HD: 300 µg/kg/day) affected cardiac fibrosis, inflammation and heart failure but sRlx-HD increased capillary/cardiomyocyte ratio. sRlx-HD significantly increased heart/body weight ratio and cardiomyocyte cross-sectional area in postpartum CKO and wild-type mice without changing the foetal gene expression program (ANP or ß-MHC). sRlx-HD augmented plasma Prolactin levels in both genotypes, which induced cardiac activation of STAT5. In vitro analyses showed that Prolactin induces cardiomyocyte hypertrophy via activation of STAT5. CONCLUSION: Although Relaxin-2 levels seemed lower in PPCM patients diagnosed early postpartum, we observed a high pregnancy-related variance of serum Relaxin-2 levels peripartum making it unsuitable as a biomarker for this condition. Supplementation with sRlx may contribute to angiogenesis and compensatory hypertrophy in the diseased heart, but the effects are not sufficient to prevent heart failure in an experimental PPCM model.


Subject(s)
Cardiomegaly/pathology , Cardiomyopathies/drug therapy , Cardiovascular Agents/pharmacology , Heart Failure/prevention & control , Myocytes, Cardiac/drug effects , Postpartum Period/blood , Relaxin/pharmacology , Adult , Animals , Biomarkers/blood , Cardiomegaly/blood , Cardiomegaly/physiopathology , Cardiomyopathies/blood , Cardiomyopathies/pathology , Cardiomyopathies/physiopathology , Case-Control Studies , Disease Models, Animal , Female , Heart Failure/blood , Heart Failure/pathology , Heart Failure/physiopathology , Humans , Mice, Knockout , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Pregnancy , Prolactin/blood , Rats , Recombinant Proteins/pharmacology , Registries , Relaxin/blood , STAT3 Transcription Factor/deficiency , STAT3 Transcription Factor/genetics , STAT5 Transcription Factor/metabolism , Signal Transduction/drug effects , Stroke Volume , Ventricular Function, Left
4.
Eur J Heart Fail ; 19(12): 1723-1728, 2017 12.
Article in English | MEDLINE | ID: mdl-28345302

ABSTRACT

AIMS: Subsequent pregnancies (SSPs) in patients with peripartum cardiomyopathy (PPCM) have a high risk of heart failure relapse. We report on outcome of SSPs in PPCM patients in Germany, Scotland, and South Africa. METHODS AND RESULTS: Among 34 PPCM patients with a SSP, pregnancy ended prematurely in four patients while it was full-term in 30. Overall relapse rate [left ventricular ejection fraction, (LVEF) <50% or death after at least 6-month follow-up] was 56% with 12% (4/34) mortality. Relapse of PPCM after SSP was not associated with differences in parity, twin pregnancy, gestational hypertension, or smoking. Persistently reduced LVEF (<50%) before entering SSP was present in 47% of patients while full recovery (LVEF ≥50%) was present in 53%. The majority of patients entering SSP with persistently reduced LVEF were of African ethnicity (75%). Persistently reduced LVEF before SSP was associated with higher mortality (25% vs. 0%) and lower rate of full recovery at follow-up. Patients obtaining standard therapy for heart failure and bromocriptine immediately after delivery displayed significantly better LVEF at follow-up and a higher rate of full recovery with no patient dying compared with patients obtaining standard therapy for heart failure alone. This was independent of African or Caucasian race. CONCLUSION: Full recovery of LVEF before SSP was associated with lower mortality and better cardiac function at follow-up. Addition of bromocriptine to standard therapy for heart failure immediately after delivery was safe and seemed to be associated with a better outcome of SSP in African and Caucasian patients.


Subject(s)
Cardiomyopathies/epidemiology , Pregnancy Complications, Cardiovascular/epidemiology , Stroke Volume/physiology , Ventricular Function, Left/physiology , Adult , Cardiomyopathies/diagnosis , Cardiomyopathies/physiopathology , Female , Germany/epidemiology , Humans , Incidence , Infant, Newborn , Male , Peripartum Period , Pregnancy , Pregnancy Complications, Cardiovascular/diagnosis , Pregnancy Complications, Cardiovascular/physiopathology , Pregnancy Outcome , Scotland/epidemiology , South Africa/epidemiology , Survival Rate/trends
5.
Eur Heart J ; 36(18): 1090-7, 2015 May 07.
Article in English | MEDLINE | ID: mdl-25636745

ABSTRACT

Pregnancy is associated with marked physiological changes challenging the cardiovascular system. Among the more severe pregnancy associated cardiovascular complications, peripartum cardiomyopathy (PPCM) is a potentially life-threatening heart disease emerging towards the end of pregnancy or in the first postpartal months in previously healthy women. A major challenge is to distinguish the peripartum discomforts in healthy women (fatigue, shortness of breath, and oedema) from the pathological symptoms of PPCM. Moreover, pregnancy-related pathologies such as preeclampsia, myocarditis, or underlying genetic disease show overlapping symptoms with PPCM. Difficulties in diagnosis and the discrimination from other pathological conditions in pregnancy may explain why PPCM is still underestimated. Additionally, underlying pathophysiologies are poorly understood, biomarkers are scarce and treatment options in general limited. Experience in long-term prognosis and management including subsequent pregnancies is just beginning to emerge. This review focuses on novel aspects of physiological and pathophysiological changes of the maternal cardiovascular system by comparing normal conditions, hypertensive complications, genetic aspects, and infectious disease in PPCM-pregnancies. It also presents clinical and basic science data on the current state of knowledge on PPCM and brings them in context thereby highlighting promising new insights in diagnostic tools and therapeutic approaches and management.


Subject(s)
Cardiomyopathies/therapy , Pregnancy Complications, Cardiovascular/therapy , Biomarkers/metabolism , Biopsy/methods , Cardiomyopathies/diagnosis , Cardiomyopathies/etiology , Diagnosis, Differential , Echocardiography , Electrocardiography , Female , Heart Failure/diagnosis , Heart Failure/etiology , Heart Failure/therapy , Hemodynamics/physiology , Humans , Hypertension/complications , Magnetic Resonance Angiography , Preconception Care/methods , Pregnancy , Pregnancy Complications, Cardiovascular/diagnosis , Pregnancy Complications, Cardiovascular/physiopathology , Prenatal Diagnosis/methods , Risk Assessment/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...