Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 214
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Circ Res ; 130(12): 1763-1779, 2022 06 10.
Article in English | MEDLINE | ID: mdl-35679359

ABSTRACT

Cardiovascular complications of pregnancy have risen substantially over the past decades, and now account for the majority of pregnancy-induced maternal deaths, as well as having substantial long-term consequences on maternal cardiovascular health. The causes and pathophysiology of these complications remain poorly understood, and therapeutic options are limited. Preclinical models represent a crucial tool for understanding human disease. We review here advances made in preclinical models of cardiovascular complications of pregnancy, including preeclampsia and peripartum cardiomyopathy, with a focus on pathological mechanisms elicited by the models and on relevance to human disease.


Subject(s)
Cardiomyopathies , Pre-Eclampsia , Pregnancy Complications, Cardiovascular , Animals , Cardiomyopathies/therapy , Female , Humans , Models, Animal , Pregnancy , Pregnancy Complications, Cardiovascular/therapy , Risk Factors
2.
Int J Mol Sci ; 25(8)2024 Apr 20.
Article in English | MEDLINE | ID: mdl-38674105

ABSTRACT

Here, we target the high-density lipoprotein (HDL) proteome in a case series of 16 patients with post-COVID-19 symptoms treated with HMG-Co-A reductase inhibitors (statin) plus angiotensin II type 1 receptor blockers (ARBs) for 6 weeks. Patients suffering from persistent symptoms (post-acute sequelae) after serologically confirmed SARS-CoV-2 infection (post-COVID-19 syndrome, PCS, n = 8) or following SARS-CoV-2 vaccination (PVS, n = 8) were included. Asymptomatic subjects with corresponding serological findings served as healthy controls (n = 8/8). HDL was isolated using dextran sulfate precipitation and the HDL proteome of all study participants was analyzed quantitatively by mass spectrometry. Clinical symptoms were assessed using questionnaires before and after therapy. The inflammatory potential of the patients' HDL proteome was addressed in human endothelial cells. The HDL proteome of patients with PCS and PVS showed no significant differences; however, compared to controls, the HDL from PVS/PCS patients displayed significant alterations involving hemoglobin, cytoskeletal proteins (MYL6, TLN1, PARVB, TPM4, FLNA), and amyloid precursor protein. Gene Ontology Biological Process (GOBP) enrichment analysis identified hemostasis, peptidase, and lipoprotein regulation pathways to be involved. Treatment of PVS/PCS patients with statins plus ARBs improved the patients' clinical symptoms. After therapy, three proteins were significantly increased (FAM3C, AT6AP2, ADAM10; FDR < 0.05) in the HDL proteome from patients with PVS/PCS. Exposure of human endothelial cells with the HDL proteome from treated PVS/PCS patients revealed reduced inflammatory cytokine and adhesion molecule expression. Thus, HDL proteome analysis from PVS/PCS patients enables a deeper insight into the underlying disease mechanisms, pointing to significant involvement in metabolic and signaling disturbances. Treatment with statins plus ARBs improved clinical symptoms and reduced the inflammatory potential of the HDL proteome. These observations may guide future therapeutic strategies for PVS/PCS patients.


Subject(s)
COVID-19 , Hydroxymethylglutaryl-CoA Reductase Inhibitors , Lipoproteins, HDL , Proteome , SARS-CoV-2 , Humans , Proteome/metabolism , Male , COVID-19/blood , COVID-19/virology , COVID-19/complications , Female , Lipoproteins, HDL/blood , Lipoproteins, HDL/metabolism , Middle Aged , SARS-CoV-2/drug effects , Aged , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Post-Acute COVID-19 Syndrome , Angiotensin II Type 1 Receptor Blockers/therapeutic use , COVID-19 Drug Treatment , Adult
3.
J Mol Cell Cardiol ; 185: 26-37, 2023 12.
Article in English | MEDLINE | ID: mdl-37797718

ABSTRACT

Hypertrophic cardiomyopathy (HCM) is the most prevalent inherited cardiac disease. Up to 40% of cases are associated with heterozygous mutations in myosin binding protein C (cMyBP-C, MYBPC3). Most of these mutations lead to premature termination codons (PTC) and patients show reduction of functional cMyBP-C. This so-called haploinsufficiency most likely contributes to disease development. We analyzed mechanisms underlying haploinsufficiency using cardiac tissue from HCM-patients with truncation mutations in MYBPC3 (MYBPC3trunc). We compared transcriptional activity, mRNA and protein expression to donor controls. To differentiate between HCM-specific and general hypertrophy-induced mechanisms we used patients with left ventricular hypertrophy due to aortic stenosis (AS) as an additional control. We show that cMyBP-C haploinsufficiency starts at the mRNA level, despite hypertrophy-induced increased transcriptional activity. Gene set enrichment analysis (GSEA) of RNA-sequencing data revealed an increased expression of NMD-components. Among them, Up-frameshift protein UPF3B, a regulator of NMD was upregulated in MYBPC3trunc patients and not in AS-patients. Strikingly, we show that in sarcomeres UPF3B but not UPF1 and UPF2 are localized to the Z-discs, the presumed location of sarcomeric protein translation. Our data suggest that cMyBP-C haploinsufficiency in HCM-patients is established by UPF3B-dependent NMD during the initial translation round at the Z-disc.


Subject(s)
Cardiomyopathy, Hypertrophic , Myocytes, Cardiac , Humans , Cardiomyopathy, Hypertrophic/metabolism , Haploinsufficiency , Hypertrophy/metabolism , Mutation , Myocytes, Cardiac/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism
4.
Blood ; 137(19): 2657-2661, 2021 05 13.
Article in English | MEDLINE | ID: mdl-33512436

ABSTRACT

Adult patients with relapsed B-cell precursor acute lymphoblastic leukemia (BCP-ALL) have a dismal prognosis. To improve pharmacotherapy, we analyzed induction of apoptosis by venetoclax and inotuzumab ozogamicin in terms of cytotoxicity and mode of action. Flow cytometry-based analyses of mitochondrial outer membrane permeabilization (MOMP) and ataxia telangiectasia mutated activation demonstrate rapid induction of MOMP by venetoclax and DNA damage signaling by inotuzumab ozogamicin, respectively. In primary ALL samples and patient-derived xenograft (PDX) models, venetoclax and inotuzumab ozogamicin cooperated and synergized in combination with dexamethasone in vitro in all tested samples of ALL. In murine PDX models, inotuzumab ozogamicin, but not venetoclax, induced complete remission in a dose-dependent manner but constantly failed to achieve relapse-free survival. In contrast, combination therapy with venetoclax, dexamethasone, and inotuzumab ozogamicin induced long-term leukemia-free survival and treatment-free survival in all 3 ALL-PDX models tested. These data demonstrate synergistic and highly efficient pharmacotherapy in preclinical models that qualify for evaluation in clinical trials.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , DNA Damage , DNA, Neoplasm/drug effects , Dexamethasone/pharmacology , Inotuzumab Ozogamicin/pharmacology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Sulfonamides/pharmacology , Adolescent , Adult , Aged , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Bridged Bicyclo Compounds, Heterocyclic/administration & dosage , Calicheamicins/pharmacology , DNA Breaks, Double-Stranded , Dexamethasone/administration & dosage , Drug Synergism , Female , Humans , Inotuzumab Ozogamicin/administration & dosage , Male , Mice , Mice, Inbred NOD , Mice, SCID , Middle Aged , Mitochondrial Membranes/drug effects , Recurrence , Sulfonamides/administration & dosage , Xenograft Model Antitumor Assays
5.
PLoS Biol ; 18(12): e3000739, 2020 12.
Article in English | MEDLINE | ID: mdl-33370269

ABSTRACT

Cardiac levels of the signal transducer and activator of transcription factor-3 (STAT3) decline with age, and male but not female mice with a cardiomyocyte-specific STAT3 deficiency conditional knockout (CKO) display premature age-related heart failure associated with reduced cardiac capillary density. In the present study, isolated male and female CKO-cardiomyocytes exhibit increased prostaglandin (PG)-generating cyclooxygenase-2 (COX-2) expression. The PG-degrading hydroxyprostaglandin-dehydrogenase-15 (HPGD) expression is only reduced in male cardiomyocytes, which is associated with increased prostaglandin D2 (PGD2) secretion from isolated male but not female CKO-cardiomyocytes. Reduced HPGD expression in male cardiomyocytes derive from impaired androgen receptor (AR)-signaling due to loss of its cofactor STAT3. Elevated PGD2 secretion in males is associated with increased white adipocyte accumulation in aged male but not female hearts. Adipocyte differentiation is enhanced in isolated stem cell antigen-1 (SCA-1)+ cardiac progenitor cells (CPC) from young male CKO-mice compared with the adipocyte differentiation of male wild-type (WT)-CPC and CPC isolated from female mice. Epigenetic analysis in freshly isolated male CKO-CPC display hypermethylation in pro-angiogenic genes (Fgfr2, Epas1) and hypomethylation in the white adipocyte differentiation gene Zfp423 associated with up-regulated ZFP423 expression and a shift from endothelial to white adipocyte differentiation compared with WT-CPC. The expression of the histone-methyltransferase EZH2 is reduced in male CKO-CPC compared with male WT-CPC, whereas no differences in the EZH2 expression in female CPC were observed. Clonally expanded CPC can differentiate into endothelial cells or into adipocytes depending on the differentiation conditions. ZFP423 overexpression is sufficient to induce white adipocyte differentiation of clonal CPC. In isolated WT-CPC, PGD2 stimulation reduces the expression of EZH2, thereby up-regulating ZFP423 expression and promoting white adipocyte differentiation. The treatment of young male CKO mice with the COX inhibitor Ibuprofen or the PGD2 receptor (DP)2 receptor antagonist BAY-u 3405 in vivo increased EZH2 expression and reduced ZFP423 expression and adipocyte differentiation in CKO-CPC. Thus, cardiomyocyte STAT3 deficiency leads to age-related and sex-specific cardiac remodeling and failure in part due to sex-specific alterations in PGD2 secretion and subsequent epigenetic impairment of the differentiation potential of CPC. Causally involved is the impaired AR signaling in absence of STAT3, which reduces the expression of the PG-degrading enzyme HPGD.


Subject(s)
Myocytes, Cardiac/metabolism , Prostaglandin D2/metabolism , STAT3 Transcription Factor/metabolism , Adipocytes, White/metabolism , Animals , Cell Differentiation/genetics , Cells, Cultured , Cyclooxygenase 2/metabolism , Endothelial Cells/metabolism , Female , Heart Failure/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Multipotent Stem Cells/metabolism , Prostaglandin D2/physiology , STAT3 Transcription Factor/genetics , Signal Transduction/genetics , Stem Cells/metabolism
6.
Artif Organs ; 47(2): 417-424, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36113950

ABSTRACT

BACKGROUND: Peripartum cardiomyopathy (PPCM) is a rare cause of heart failure (HF), presenting with left ventricular (LV) systolic dysfunction either at the end of pregnancy or in the months following delivery. In rare cases, PPCM leads to severe impairment of LV function, refractory cardiogenic shock or advanced HF. LV assist devices (LVAD) have been shown to be a feasible treatment option in advanced HF. However, little is known about long-term outcomes and prognosis of PPCM patients undergoing LVAD implantation. METHODS: A retrospective analysis of data from PPCM patients undergoing LVAD implantation in two tertiary centers with respect to long-term outcomes was performed. RESULTS: Twelve patients of median age 30 (18-39) years were included. Eight patients were experiencing cardiogenic shock (INTERMACS 1) at implantation. Seven patients were implanted within 1 month of their PPCM diagnosis. Median duration of LVAD support was 19 (2-92) months with median follow up of 67 (18-136) months (100% complete). In-hospital and 1-year mortality were 0% and 8.3%, respectively. Two patients died on LVAD support, four patients were successfully bridged to transplantation, two patients are still on LVAD, and four were successfully weaned due to sufficient LV recovery (one died after LV function deteriorated again). CONCLUSION: LVAD treatment of decompensated end-stage PPCM is feasible. Early LVAD provision led to hemodynamic stabilization in our cohort and facilitated safe LV recovery in one third of these young female patients.


Subject(s)
Cardiomyopathies , Heart Failure , Heart-Assist Devices , Ventricular Dysfunction, Left , Pregnancy , Humans , Female , Adult , Shock, Cardiogenic/therapy , Retrospective Studies , Heart-Assist Devices/adverse effects , Peripartum Period , Treatment Outcome , Cardiomyopathies/complications , Cardiomyopathies/surgery , Heart Failure/surgery , Heart Failure/complications , Ventricular Dysfunction, Left/complications , Ventricular Dysfunction, Left/therapy
7.
Circulation ; 143(19): 1852-1862, 2021 05 11.
Article in English | MEDLINE | ID: mdl-33874732

ABSTRACT

BACKGROUND: Peripartum cardiomyopathy (PPCM) occurs in ≈1:2000 deliveries in the United States and worldwide. The genetic underpinnings of PPCM remain poorly defined. Approximately 10% of women with PPCM harbor truncating variants in TTN (TTNtvs). Whether mutations in other genes can predispose to PPCM is not known. It is also not known if the presence of TTNtvs predicts clinical presentation or outcomes. Nor is it known if the prevalence of TTNtvs differs in women with PPCM and preeclampsia, the strongest risk factor for PPCM. METHODS: Women with PPCM were retrospectively identified from several US and international academic centers, and clinical information and DNA samples were acquired. Next-generation sequencing was performed on 67 genes, including TTN, and evaluated for burden of truncating and missense variants. The impact of TTNtvs on the severity of clinical presentation, and on clinical outcomes, was evaluated. RESULTS: Four hundred sixty-nine women met inclusion criteria. Of the women with PPCM, 10.4% bore TTNtvs (odds ratio=9.4 compared with 1.2% in the reference population; Bonferroni-corrected P [P*]=1.2×10-46). We additionally identified overrepresentation of truncating variants in FLNC (odds ratio=24.8, P*=7.0×10-8), DSP (odds ratio=14.9, P*=1.0×10-8), and BAG3 (odds ratio=53.1, P*=0.02), genes not previously associated with PPCM. This profile is highly similar to that found in nonischemic dilated cardiomyopathy. Women with TTNtvs had lower left ventricular ejection fraction on presentation than did women without TTNtvs (23.5% versus 29%, P=2.5×10-4), but did not differ significantly in timing of presentation after delivery, in prevalence of preeclampsia, or in rates of clinical recovery. CONCLUSIONS: This study provides the first extensive genetic and phenotypic landscape of PPCM and demonstrates that predisposition to heart failure is an important risk factor for PPCM. The work reveals a degree of genetic similarity between PPCM and dilated cardiomyopathy, suggesting that gene-specific therapeutic approaches being developed for dilated cardiomyopathy may also apply to PPCM, and that approaches to genetic testing in PPCM should mirror those taken in dilated cardiomyopathy. Last, the clarification of genotype/phenotype associations has important implications for genetic counseling.


Subject(s)
Cardiomyopathies/genetics , Peripartum Period/genetics , Adult , Cardiomyopathies/physiopathology , Female , Humans , Phenotype , Pregnancy , Retrospective Studies
8.
Eur Heart J ; 42(32): 3094-3102, 2021 08 21.
Article in English | MEDLINE | ID: mdl-34322694

ABSTRACT

Peripartum cardiomyopathy (PPCM) is a disease that occurs globally in all ethnic groups and should be suspected in any peripartum women presenting with symptoms and signs of heart failure, towards the end of pregnancy or in the months following delivery, with confirmed left ventricular dysfunction. After good history taking, all women should be thoroughly assessed, and alternative causes should be excluded. Urgent cardiac investigations with electrocardiogram and natriuretic peptide measurement (if available) should be performed. Echocardiography follows as the next step in investigation. Patients with abnormal cardiac investigations should be urgently referred to a cardiology team for expert management. Referral for genetic work-up should be considered if there is a family history of cardiomyopathy or sudden death. PPCM is a disease with substantial maternal and neonatal morbidity and mortality. Maternal mortality rates range widely, from 0% to 30%, depending on the ethnic background and geographic region. Just under half of women experience myocardial recovery. Remarkable advances in the comprehension of the pathogenesis and in patient management and therapy have been achieved, largely due to team efforts and close collaboration between basic scientists, cardiologists, intensive care specialists, and obstetricians. This review summarizes current knowledge of PPCM genetics, pathophysiology, diagnostic approach, management, and outcome.


Subject(s)
Cardiomyopathies , Pregnancy Complications, Cardiovascular , Puerperal Disorders , Ventricular Dysfunction, Left , Cardiomyopathies/diagnosis , Cardiomyopathies/genetics , Cardiomyopathies/therapy , Female , Humans , Infant, Newborn , Peripartum Period , Pregnancy , Pregnancy Complications, Cardiovascular/diagnosis , Pregnancy Complications, Cardiovascular/genetics , Pregnancy Complications, Cardiovascular/therapy , Puerperal Disorders/diagnosis , Puerperal Disorders/genetics , Puerperal Disorders/therapy
9.
J Biol Chem ; 295(5): 1261-1270, 2020 01 31.
Article in English | MEDLINE | ID: mdl-31882540

ABSTRACT

MicroRNA 199 (miR-199) negatively impacts pancreatic ß-cell function and its expression is highly increased in islets from diabetic mice as well as in plasma of diabetic patients. Here we investigated how miR-199 expression is regulated in ß-cells by assessing expression of miR-199 precursors (primiR-199a1, primiR-199a2, and primiR-199b) and mature miR-199 (miR-199-3p and miR-199-5p) and promoter transcriptional activity assays in mouse islets and mouse insulinoma cells (MIN6) under different stimuli. We found that mouse islets equally express miR-199-3p and miR-199-5p. However, the primiRNA expression levels differed; although primiR-199a1 expression was about 30% greater than that of primiR-199a2, primiR-199b is barely detected in islets. We observed a 2-fold increase in primiR-199a1 and primiR-199a2 mRNA levels in mouse islets cultured in 10 mm glucose compared with 5.5 mm glucose. Similar responses to glucose were observed in MIN6 cells. Exposure to 30 mm KCl to induce membrane depolarization and calcium influx increased expression of primiR-199a2 but not of primiR-199a1 in MIN6 cells, indicating that calcium influx was involved. Transcriptional activity studies in MIN6 cells also revealed that primiR-199a2 promoter activity was enhanced by glucose and reduced by 2-deoxy-D-glucose-induced starvation. KCl and the potassium channel blocker tolbutamide also stimulated primiR-199a2 promoter activity. Calcium channel blockade by nifedipine reduced primiR-199a2 promoter activity in MIN6 cells, and diazoxide-mediated calcium influx inhibition blunted glucose up-regulation of miR-199-3p in islets. In conclusion, we uncover that glucose acutely up-regulates miR-199 family expression in ß-cells. Glucose metabolism and calcium influx are involved in primiR-199a2 expression but not primiR-199a1 expression.


Subject(s)
Glucose/metabolism , Insulin-Secreting Cells/metabolism , MicroRNAs/metabolism , Animals , Calcium/metabolism , Cell Line , Cell Membrane/metabolism , Female , Male , Mice , MicroRNAs/genetics , Up-Regulation
10.
Basic Res Cardiol ; 116(1): 61, 2021 10 20.
Article in English | MEDLINE | ID: mdl-34669013

ABSTRACT

Systemic effects of advanced cancer impact on the heart leading to cardiac atrophy and functional impairment. Using a murine melanoma cancer model (B16F10 melanoma cells stably transduced with a Ganciclovir (GCV)-inducible suicide gene), the present study analysed the recovery potential of cancer-induced cardiomyopathy with or without use of doxorubicin (Dox). After Dox-free tumor elimination and recovery for 70 ± 5 days, cancer-induced morphologic, functional, metabolic and molecular changes were largely reversible in mice previously bearing tumors. Moreover, grip strength and cardiac response to angiotensin II-induced high blood pressure were comparable with healthy control mice. In turn, addition of Dox (12 mg/kg BW) to melanoma-bearing mice reduced survival in the acute phase compared to GCV-alone induced recovery, while long-term effects on cardiac morphologic and functional recovery were similar. However, Dox treatment was associated with permanent changes in the cardiac gene expression pattern, especially the circadian rhythm pathway associated with the DNA damage repair system. Thus, the heart can recover from cancer-induced damage after chemotherapy-free tumor elimination. In contrast, treatment with the cardiotoxic drug Dox induces, besides well-known adverse acute effects, long-term subclinical changes in the heart, especially of circadian clock genes. Since the circadian clock is known to impact on cardiac repair mechanisms, these changes may render the heart more sensitive to additional stress during lifetime, which, at least in part, could contribute to late cardiac toxicity.


Subject(s)
Anthracyclines , Neoplasms , Animals , Anthracyclines/therapeutic use , Antibiotics, Antineoplastic/therapeutic use , Antibiotics, Antineoplastic/toxicity , Cardiotoxicity , Doxorubicin/therapeutic use , Mice , Neoplasms/drug therapy
11.
J Nucl Cardiol ; 28(4): 1636-1645, 2021 Aug.
Article in English | MEDLINE | ID: mdl-31659697

ABSTRACT

BACKGROUND: Leukocyte subtypes bear distinct pro-inflammatory, reparative, and regulatory functions. Imaging inflammation provides information on disease prognosis and may guide therapy, but the cellular basis of the signal remains equivocal. We evaluated leukocyte subtype specificity of characterized clinically relevant inflammation-targeted radiotracers. METHODS AND RESULTS: Leukocyte populations were purified from blood- and THP-1-derived macrophages were polarized into M1-, reparative M2a-, or M2c-macrophages. In vitro uptake assays were conducted using tracers of enhanced glucose or amino acid metabolism and molecular markers of inflammatory cells. Both 18F-deoxyglucose (18F-FDG) and the labeled amino acid 11C-methionine (11C-MET) displayed higher uptake in neutrophils and monocytes compared to other leukocytes (P = 0.005), and markedly higher accumulation in pro-inflammatory M1-macrophages compared to reparative M2a-macrophages (P < 0.001). Molecular tracers 68Ga-DOTATATE targeting the somatostatin receptor type 2 and 68Ga-pentixafor targeting the chemokine receptor type 4 (CXCR4) exhibited broad uptake by leukocyte subpopulations and polarized macrophages with highest uptake in T-cells/natural killer cells and B-cells compared to neutrophils. Mitochondrial translocator protein (TSPO)-targeted 18F-flutriciclamide selectively accumulated in monocytes and pro-inflammatory M1 macrophages (P < 0.001). Uptake by myocytes and fibroblasts tended to be higher for metabolic radiotracers. CONCLUSIONS: The different in vitro cellular uptake profiles may allow isolation of distinct phases of the inflammatory pathway with specific inflammation-targeted radiotracers. The pathogenetic cell population in specific inflammatory diseases should be considered in the selection of an appropriate imaging agent.


Subject(s)
Leukocytes/metabolism , Macrophages/metabolism , Radiopharmaceuticals/pharmacokinetics , Animals , Cell Culture Techniques , Coordination Complexes/pharmacokinetics , Fibroblasts/metabolism , Fluorine Radioisotopes/pharmacokinetics , Fluorodeoxyglucose F18/pharmacokinetics , Humans , Indoles/pharmacokinetics , Myocytes, Cardiac/metabolism , Octreotide/analogs & derivatives , Octreotide/pharmacokinetics , Organometallic Compounds/pharmacokinetics , Peptides, Cyclic/pharmacokinetics , Rats
12.
Herz ; 46(2): 107-114, 2021 Mar.
Article in German | MEDLINE | ID: mdl-33394058

ABSTRACT

The coronavirus disease 2019 (COVID-19) pandemic is a challenge for our healthcare system but at the same time is one of the excellent catalyzers and promoters of successful translational research. The COVID-19 is not only a simple viral infection of the bronchial system but is also a pandemic hyperinflammatory multiorgan disease. The cardiovascular system plays a causal role in this context, as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) invades host cells via the angiotensin-converting enzyme 2 (ACE-2), an enzyme in the renin-angiotensin system. Furthermore, cardiovascular comorbidities and risk factors, such as hypertension, diabetes and obesity play an important role in the severity of the course of the disease. Additional risk factors, such as gender, age, genetics and air pollution modulate both the severity of the SARS-CoV­2 infection as well as cardiovascular diseases. As sequelae of COVID-19, increased thrombosis, myocardial infarction, myocardial inflammation and vasculitis occur, which directly damage the cardiovascular system and substantially contribute to the high morbidity and mortality. Knowledge gained from many studies on the course of the disease in patients infected with SARS-CoV­2 has led to improved treatment possibilities, which now in the second wave are partly standardized and were, and are, in particular adapted to complications of the cardiovascular system. In this review we provide a short overview on the pathophysiology of the SARS-CoV­2 in general and also specifically on the cardiovascular system. Furthermore, we summarize the current treatment approaches and their pathophysiological principles (status November 2020).


Subject(s)
COVID-19 , Cardiovascular Diseases , Cardiovascular Diseases/therapy , Humans , Pandemics , Peptidyl-Dipeptidase A/metabolism , Renin-Angiotensin System , SARS-CoV-2
13.
Eur Heart J ; 41(39): 3787-3797, 2020 10 14.
Article in English | MEDLINE | ID: mdl-32840318

ABSTRACT

AIMS: We sought to describe the clinical presentation, management, and 6-month outcomes in women with peripartum cardiomyopathy (PPCM) globally. METHODS AND RESULTS: In 2011, >100 national and affiliated member cardiac societies of the European Society of Cardiology (ESC) were contacted to contribute to a global registry on PPCM, under the auspices of the ESC EURObservational Research Programme. These societies were tasked with identifying centres who could participate in this registry. In low-income countries, e.g. Mozambique or Burkina Faso, where there are no national societies due to a shortage of cardiologists, we identified potential participants through abstracts and publications and encouraged participation into the study. Seven hundred and thirty-nine women were enrolled in 49 countries in Europe (33%), Africa (29%), Asia-Pacific (15%), and the Middle East (22%). Mean age was 31 ± 6 years, mean left ventricular ejection fraction (LVEF) was 31 ± 10%, and 10% had a previous pregnancy complicated by PPCM. Symptom-onset occurred most often within 1 month of delivery (44%). At diagnosis, 67% of patients had severe (NYHA III/IV) symptoms and 67% had a LVEF ≤35%. Fifteen percent received bromocriptine with significant regional variation (Europe 15%, Africa 26%, Asia-Pacific 8%, the Middle East 4%, P < 0.001). Follow-up was available for 598 (81%) women. Six-month mortality was 6% overall, lowest in Europe (4%), and highest in the Middle East (10%). Most deaths were due to heart failure (42%) or sudden (30%). Re-admission for any reason occurred in 10% (with just over half of these for heart failure) and thromboembolic events in 7%. Myocardial recovery (LVEF > 50%) occurred only in 46%, most commonly in Asia-Pacific (62%), and least commonly in the Middle East (25%). Neonatal death occurred in 5% with marked regional variation (Europe 2%, the Middle East 9%). CONCLUSION: Peripartum cardiomyopathy is a global disease, but clinical presentation and outcomes vary by region. Just under half of women experience myocardial recovery. Peripartum cardiomyopathy is a disease with substantial maternal and neonatal morbidity and mortality.


Subject(s)
Cardiology , Cardiomyopathies , Pregnancy Complications, Cardiovascular , Adult , Africa , Asia/epidemiology , Cardiomyopathies/epidemiology , Cardiomyopathies/therapy , Europe , Female , Humans , Infant, Newborn , Middle East/epidemiology , Peripartum Period , Pregnancy , Pregnancy Complications, Cardiovascular/epidemiology , Pregnancy Complications, Cardiovascular/therapy , Registries , Stroke Volume , Ventricular Function, Left
14.
Basic Res Cardiol ; 115(6): 62, 2020 09 25.
Article in English | MEDLINE | ID: mdl-32975669

ABSTRACT

Neuraminidase (NEU)1 forms a multienzyme complex with beta-galactosidase (ß-GAL) and protective-protein/cathepsin (PPC) A, which cleaves sialic-acids from cell surface glycoconjugates. We investigated the role of NEU1 in the myocardium after ischemia/reperfusion (I/R). Three days after inducing I/R, left ventricles (LV) of male mice (3 months-old) displayed upregulated neuraminidase activity and increased NEU1, ß-GAL and PPCA expression. Mice hypomorphic for neu1 (hNEU1) had less neuraminidase activity, fewer pro-inflammatory (Lin-CD11b+F4/80+Ly-6Chigh), and more anti-inflammatory macrophages (Lin-CD11b+F4/80+Ly-6Clow) 3 days after I/R, and less LV dysfunction 14 days after I/R. WT mice transplanted with hNEU1-bone marrow (BM) and hNEU1 mice with WT-BM showed significantly better LV function 14 days after I/R compared with WT mice with WT-BM. Mice with a cardiomyocyte-specific NEU1 overexpression displayed no difference in inflammation 3 days after I/R, but showed increased cardiomyocyte hypertrophy, reduced expression and mislocalization of Connexin-43 in gap junctions, and LV dysfunction despite a similar infarct scar size to WT mice 14 days after I/R. The upregulation of NEU1 after I/R contributes to heart failure by promoting inflammation in invading monocytes/macrophages, enhancing cardiomyocyte hypertrophy, and impairing gap junction function, suggesting that systemic NEU1 inhibition may reduce heart failure after I/R.


Subject(s)
Heart Failure/etiology , Hypertrophy, Left Ventricular/etiology , Macrophages/enzymology , Monocytes/enzymology , Myocardial Infarction/complications , Myocardial Reperfusion Injury/complications , Myocytes, Cardiac/enzymology , Neuraminidase/deficiency , Ventricular Dysfunction, Left/etiology , Animals , Cathepsin A/metabolism , Connexin 43/metabolism , Disease Models, Animal , Female , Gap Junctions/enzymology , Gap Junctions/pathology , Heart Failure/enzymology , Heart Failure/immunology , Heart Failure/physiopathology , Hypertrophy, Left Ventricular/enzymology , Hypertrophy, Left Ventricular/immunology , Hypertrophy, Left Ventricular/physiopathology , Macrophages/immunology , Male , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Monocytes/immunology , Myocardial Infarction/enzymology , Myocardial Infarction/immunology , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/enzymology , Myocardial Reperfusion Injury/immunology , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/pathology , Neuraminidase/genetics , Ventricular Dysfunction, Left/enzymology , Ventricular Dysfunction, Left/immunology , Ventricular Dysfunction, Left/physiopathology , Ventricular Function, Left , Ventricular Remodeling , beta-Galactosidase/metabolism
15.
Herz ; 45(7): 632-636, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32542459

ABSTRACT

Cardiovascular diseases and cancer are major causes of mortality in industrialized societies. They share common risk factors (e.g., genetics, lifestyle, age, infection, toxins, and pollution) and might also mutually promote the onset of the respective other disease. Cancer can affect cardiac function directly while antitumor therapies may have acute- and/or late-onset cardiotoxic effects. Recent studies suggest that heart failure might promote tumorigenesis and tumor progression. In both cancer and cardiovascular diseases, genetic predisposition is implicated in the disease onset and development. In this regard, genetic variants classically associated with cardiomyopathies increase the risk for toxic side effects on the cardiovascular system. Genetic variants associated with increased cancer risk are frequent in patients with peripartum cardiomyopathy complicated by cancer, pointing to a common genetic predisposition for both diseases. Common risk factors, cardiotoxic antitumor treatment, genetic variants (associated with cardiomyopathies and/or cancer), and increased cardiac stress lead us to propose the "multi-hit hypothesis" linking cancer and cardiovascular diseases. In the present review, we summarize the current knowledge on potential connecting factors between cancer and cardiovascular diseases with a major focus on the role of genetic predisposition and its implication for individual therapeutic strategies and risk assessment in the novel field of oncocardiology.


Subject(s)
Cardiomyopathies , Heart Failure , Neoplasms , Cardiotoxicity , Genetic Predisposition to Disease/genetics , Heart Failure/genetics , Humans , Neoplasms/genetics
16.
Eur Heart J ; 40(47): 3859-3868c, 2019 12 14.
Article in English | MEDLINE | ID: mdl-31800034

ABSTRACT

The overall lifetime risk of heart failure (HF) is similar between men and women, however, there are marked sex differences in the landscape of this condition that are both important and under-recognized. Men are predisposed to HF with reduced ejection fraction (HFrEF), whereas women predominate in HF with preserved ejection fraction (HFpEF). Sex differences are also notable in the penetrance of genetic cardiomyopathies, risk factors, e.g. breast cancer which may be associated with cancer treatment-induced cardiomyopathy, as well as sex-specific conditions such as peripartum cardiomyopathy (PPCM). This review outlines the key sex differences with respect to clinical characteristics, pathophysiology, and therapeutic responses to HF treatments. Finally, we address important differences in the prognosis of HF. A central hypothesis is that the higher risk of HFrEF in men compared to women may be attributable to their predisposition to macrovascular coronary artery disease and myocardial infarction, whereas coronary microvascular dysfunction/endothelial inflammation has been postulated to play a key role in HFpEF and maybe the common link among HF syndromes that women are predisposed to Takotsubo cardiomyopathy, PPCM, and breast cancer radiotherapy-induced cardiomyopathy. Under-pinning current sex disparities in HF, there is a paucity of women recruited to HF clinical trials (20-25% of cohorts) and thus treatment guidelines are predominantly based on male-derived data. Large gaps in knowledge exist in sex-specific mechanisms, optimal drug doses for women and sex-specific criteria for device therapy.


Subject(s)
Heart Failure/epidemiology , Risk Assessment/methods , Stroke Volume/physiology , Ventricular Function, Left/physiology , Female , Global Health , Heart Failure/physiopathology , Humans , Male , Morbidity/trends , Risk Factors , Sex Distribution , Sex Factors , Survival Rate/trends
17.
Eur Heart J ; 40(44): 3626-3644, 2019 11 21.
Article in English | MEDLINE | ID: mdl-30295807

ABSTRACT

Acute heart failure (HF) and in particular, cardiogenic shock are associated with high morbidity and mortality. A therapeutic dilemma is that the use of positive inotropic agents, such as catecholamines or phosphodiesterase-inhibitors, is associated with increased mortality. Newer drugs, such as levosimendan or omecamtiv mecarbil, target sarcomeres to improve systolic function putatively without elevating intracellular Ca2+. Although meta-analyses of smaller trials suggested that levosimendan is associated with a better outcome than dobutamine, larger comparative trials failed to confirm this observation. For omecamtiv mecarbil, Phase II clinical trials suggest a favourable haemodynamic profile in patients with acute and chronic HF, and a Phase III morbidity/mortality trial in patients with chronic HF has recently begun. Here, we review the pathophysiological basis of systolic dysfunction in patients with HF and the mechanisms through which different inotropic agents improve cardiac function. Since adenosine triphosphate and reactive oxygen species production in mitochondria are intimately linked to the processes of excitation-contraction coupling, we also discuss the impact of inotropic agents on mitochondrial bioenergetics and redox regulation. Therefore, this position paper should help identify novel targets for treatments that could not only safely improve systolic and diastolic function acutely, but potentially also myocardial structure and function over a longer-term.


Subject(s)
Cardiotonic Agents/therapeutic use , Excitation Contraction Coupling/drug effects , Heart Failure/drug therapy , Shock, Cardiogenic/drug therapy , Acute Disease , Animals , Antioxidants/adverse effects , Antioxidants/therapeutic use , Calcium/metabolism , Cardiotonic Agents/adverse effects , Case-Control Studies , Catecholamines/adverse effects , Catecholamines/therapeutic use , Clinical Trials as Topic , Diastole/drug effects , Dobutamine/adverse effects , Dobutamine/therapeutic use , Dogs , Energy Metabolism/drug effects , Heart Failure/mortality , Humans , Mitochondria/metabolism , Models, Animal , Myocardial Contraction/drug effects , Nitrogen Oxides/adverse effects , Nitrogen Oxides/therapeutic use , Oxidation-Reduction/drug effects , Phosphodiesterase Inhibitors/adverse effects , Phosphodiesterase Inhibitors/therapeutic use , Placebos/administration & dosage , Receptors, Adrenergic/drug effects , Sarcomeres/drug effects , Sarcomeres/metabolism , Shock, Cardiogenic/mortality , Simendan/adverse effects , Simendan/therapeutic use , Swine , Systole/drug effects , Urea/adverse effects , Urea/analogs & derivatives , Urea/therapeutic use
18.
J Cell Mol Med ; 23(3): 2125-2135, 2019 03.
Article in English | MEDLINE | ID: mdl-30618214

ABSTRACT

Duchenne muscular dystrophy (DMD) is an X-linked progressive muscle degenerative disease, caused by mutations in the dystrophin gene and resulting in death because of respiratory or cardiac failure. To investigate the cardiac cellular manifestation of DMD, we generated induced pluripotent stem cells (iPSCs) and iPSC-derived cardiomyocytes (iPSC-CMs) from two DMD patients: a male and female manifesting heterozygous carrier. Dystrophin mRNA and protein expression were analysed by qRT-PCR, RNAseq, Western blot and immunofluorescence staining. For comprehensive electrophysiological analysis, current and voltage clamp were used to record transmembrane action potentials and ion currents, respectively. Microelectrode array was used to record extracellular electrograms. X-inactive specific transcript (XIST) and dystrophin expression analyses revealed that female iPSCs underwent X chromosome reactivation (XCR) or erosion of X chromosome inactivation, which was maintained in female iPSC-CMs displaying mixed X chromosome expression of wild type (WT) and mutated alleles. Both DMD female and male iPSC-CMs presented low spontaneous firing rate, arrhythmias and prolonged action potential duration. DMD female iPSC-CMs displayed increased beat rate variability (BRV). DMD male iPSC-CMs manifested decreased If density, and DMD female and male iPSC-CMs showed increased ICa,L density. Our findings demonstrate cellular mechanisms underlying electrophysiological abnormalities and cardiac arrhythmias in DMD.


Subject(s)
Heterozygote , Induced Pluripotent Stem Cells/physiology , Muscular Dystrophy, Duchenne/physiopathology , Myocytes, Cardiac/physiology , Action Potentials/genetics , Adult , Cell Differentiation/genetics , Dystrophin/genetics , Dystrophin/metabolism , Electrophysiological Phenomena , Female , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/ultrastructure , Male , Microscopy, Electron, Transmission , Middle Aged , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/ultrastructure
19.
N Engl J Med ; 374(3): 233-41, 2016 Jan 21.
Article in English | MEDLINE | ID: mdl-26735901

ABSTRACT

Background Peripartum cardiomyopathy shares some clinical features with idiopathic dilated cardiomyopathy, a disorder caused by mutations in more than 40 genes, including TTN, which encodes the sarcomere protein titin. Methods In 172 women with peripartum cardiomyopathy, we sequenced 43 genes with variants that have been associated with dilated cardiomyopathy. We compared the prevalence of different variant types (nonsense, frameshift, and splicing) in these women with the prevalence of such variants in persons with dilated cardiomyopathy and with population controls. Results We identified 26 distinct, rare truncating variants in eight genes among women with peripartum cardiomyopathy. The prevalence of truncating variants (26 in 172 [15%]) was significantly higher than that in a reference population of 60,706 persons (4.7%, P=1.3×10(-7)) but was similar to that in a cohort of patients with dilated cardiomyopathy (55 of 332 patients [17%], P=0.81). Two thirds of identified truncating variants were in TTN, as seen in 10% of the patients and in 1.4% of the reference population (P=2.7×10(-10)); almost all TTN variants were located in the titin A-band. Seven of the TTN truncating variants were previously reported in patients with idiopathic dilated cardiomyopathy. In a clinically well-characterized cohort of 83 women with peripartum cardiomyopathy, the presence of TTN truncating variants was significantly correlated with a lower ejection fraction at 1-year follow-up (P=0.005). Conclusions The distribution of truncating variants in a large series of women with peripartum cardiomyopathy was remarkably similar to that found in patients with idiopathic dilated cardiomyopathy. TTN truncating variants were the most prevalent genetic predisposition in each disorder.


Subject(s)
Cardiomyopathies/genetics , Cardiomyopathy, Dilated/genetics , Connectin/genetics , Genetic Predisposition to Disease , Mutation , Peripartum Period , Pregnancy Complications, Cardiovascular/genetics , Adult , Case-Control Studies , Connectin/chemistry , Female , Humans , Pregnancy , Protein Isoforms , Sequence Analysis, DNA , Stroke Volume
20.
Am J Pathol ; 187(12): 2645-2658, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28935576

ABSTRACT

Peripartum cardiomyopathy (PPCM) and dilated cardiomyopathy (DCM) show similarities in clinical presentation. However, although DCM patients do not recover and slowly deteriorate further, PPCM patients show either a fast cardiac deterioration or complete recovery. The aim of this study was to assess if underlying cellular changes can explain the clinical similarities and differences in the two diseases. We, therefore, assessed sarcomeric protein expression, modification, titin isoform shift, and contractile behavior of cardiomyocytes in heart tissue of PPCM and DCM patients and compared these with nonfailing controls. Heart samples from ischemic heart disease (ISHD) patients served as heart failure control samples. Passive force was only increased in PPCM samples compared with controls, whereas PPCM, DCM, and ISHD samples all showed increased myofilament Ca2+ sensitivity. Length-dependent activation was significantly impaired in PPCM compared with controls, no impairment was observed in ISHD samples, and DCM samples showed an intermediate response. Contractile impairments were caused by impaired protein kinase A (PKA)-mediated phosphorylation because exogenous PKA restored all parameters to control levels. Although DCM samples showed reexpression of EH-myomesin, an isoform usually only expressed in the heart before birth, PPCM and ISHD did not. The lack of EH-myomesin, combined with low PKA-mediated phosphorylation of myofilament proteins and increased compliant titin isoform, may explain the increase in passive force and blunted length-dependent activation of myofilaments in PPCM samples.


Subject(s)
Cardiomyopathies/physiopathology , Cardiomyopathy, Dilated/physiopathology , Myocytes, Cardiac/pathology , Myofibrils/pathology , Peripartum Period , Adult , Female , Humans , Male , Middle Aged , Myocardial Ischemia/physiopathology , Myocytes, Cardiac/metabolism , Myofibrils/metabolism , Pregnancy
SELECTION OF CITATIONS
SEARCH DETAIL