Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 80
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nucleic Acids Res ; 52(8): 4215-4233, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38364861

ABSTRACT

The limited regenerative capacity of the human heart contributes to high morbidity and mortality worldwide. In contrast, zebrafish exhibit robust regenerative capacity, providing a powerful model for studying how to overcome intrinsic epigenetic barriers maintaining cardiac homeostasis and initiate regeneration. Here, we present a comprehensive analysis of the histone modifications H3K4me1, H3K4me3, H3K27me3 and H3K27ac during various stages of zebrafish heart regeneration. We found a vast gain of repressive chromatin marks one day after myocardial injury, followed by the acquisition of active chromatin characteristics on day four and a transition to a repressive state on day 14, and identified distinct transcription factor ensembles associated with these events. The rapid transcriptional response involves the engagement of super-enhancers at genes implicated in extracellular matrix reorganization and TOR signaling, while H3K4me3 breadth highly correlates with transcriptional activity and dynamic changes at genes involved in proteolysis, cell cycle activity, and cell differentiation. Using loss- and gain-of-function approaches, we identified transcription factors in cardiomyocytes and endothelial cells influencing cardiomyocyte dedifferentiation or proliferation. Finally, we detected significant evolutionary conservation between regulatory regions that drive zebrafish and neonatal mouse heart regeneration, suggesting that reactivating transcriptional and epigenetic networks converging on these regulatory elements might unlock the regenerative potential of adult human hearts.


Subject(s)
Chromatin , Gene Regulatory Networks , Heart , Animals , Humans , Mice , Cell Differentiation , Chromatin/metabolism , Chromatin/genetics , Epigenesis, Genetic , Histone Code , Histones/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/cytology , Regeneration/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Zebrafish/genetics
2.
Eur Heart J ; 45(21): 1904-1916, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38554125

ABSTRACT

BACKGROUND AND AIMS: There is significant potential to streamline the clinical pathway for patients undergoing transcatheter aortic valve implantation (TAVI). The purpose of this study was to evaluate the effect of implementing BENCHMARK best practices on the efficiency and safety of TAVI in 28 sites in 7 European countries. METHODS: This was a study of patients with severe symptomatic aortic stenosis (AS) undergoing TAVI with balloon-expandable valves before and after implementation of BENCHMARK best practices. Principal objectives were to reduce hospital length of stay (LoS) and duration of intensive care stay. Secondary objective was to document patient safety. RESULTS: Between January 2020 and March 2023, 897 patients were documented prior to and 1491 patients after the implementation of BENCHMARK practices. Patient characteristics were consistent with a known older TAVI population and only minor differences. Mean LoS was reduced from 7.7 ± 7.0 to 5.8 ± 5.6 days (median 6 vs. 4 days; P < .001). Duration of intensive care was reduced from 1.8 to 1.3 days (median 1.1 vs. 0.9 days; P < .001). Adoption of peri-procedure best practices led to increased use of local anaesthesia (96.1% vs. 84.3%; P < .001) and decreased procedure (median 47 vs. 60 min; P < .001) and intervention times (85 vs. 95 min; P < .001). Thirty-day patient safety did not appear to be compromised with no differences in all-cause mortality (0.6% in both groups combined), stroke/transient ischaemic attack (1.4%), life-threatening bleeding (1.3%), stage 2/3 acute kidney injury (0.7%), and valve-related readmission (1.2%). CONCLUSIONS: Broad implementation of BENCHMARK practices contributes to improving efficiency of TAVI pathway reducing LoS and costs without compromising patient safety.


Subject(s)
Aortic Valve Stenosis , Benchmarking , Length of Stay , Transcatheter Aortic Valve Replacement , Humans , Transcatheter Aortic Valve Replacement/methods , Aortic Valve Stenosis/surgery , Male , Female , Aged, 80 and over , Length of Stay/statistics & numerical data , Aged , Critical Pathways , Europe/epidemiology , Postoperative Complications/epidemiology , Postoperative Complications/prevention & control , Patient Safety
3.
Basic Res Cardiol ; 119(3): 453-479, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38491291

ABSTRACT

Though myocardial infarction (MI) in pigs is a well-established translational large animal model, it has not yet been widely used for immunotherapy studies, and a comprehensive description of the immune response to MI in this species is lacking. We induced MI in Landrace pigs by balloon occlusion of the left anterior descending artery over 90 min. Within 14 days, the necrotic myocardium was progressively replaced by scar tissue with involvement of myofibroblasts. We characterized the immune response in the heart ex vivo by (immuno)histology, flow cytometry, and RNA sequencing of myocardial tissue on days 3, 7, and 14 after MI. Besides a clear predominance of myeloid cells among heart-infiltrating leukocytes, we detected activated T cells and an increasing proportion of CD4+ Foxp3+ regulatory T cells (Treg), especially in the infarct core-findings that closely mirror what has been observed in mice and humans after MI. Transcriptome data indicated inflammatory activity that was persistent but markedly changing in character over time and linked to extracellular matrix biology. Analysis of lymphocytes in heart-draining lymph nodes revealed significantly higher proliferation rates of T helper cell subsets, including Treg on day 7 after MI, compared to sham controls. Elevated frequencies of myeloid progenitors in the spleen suggest that it might be a site of emergency myelopoiesis after MI in pigs, as previously shown in mice. We thus provide a first description of the immune response to MI in pigs, and our results can aid future research using the species for preclinical immunotherapy studies.


Subject(s)
Disease Models, Animal , Myocardial Infarction , Myocardium , T-Lymphocytes, Regulatory , Animals , Myocardial Infarction/immunology , Myocardial Infarction/pathology , T-Lymphocytes, Regulatory/immunology , Myocardium/pathology , Myocardium/immunology , Sus scrofa , Swine , Lymphocyte Activation , Male , Transcriptome , Female , Time Factors
4.
Circulation ; 145(10): 765-782, 2022 03 08.
Article in English | MEDLINE | ID: mdl-35113652

ABSTRACT

BACKGROUND: Recent studies have established that CCR2 (C-C chemokine receptor type 2) marks proinflammatory subsets of monocytes, macrophages, and dendritic cells that contribute to adverse left ventricle (LV) remodeling and heart failure progression. Elucidation of the effector mechanisms that mediate adverse effects of CCR2+ monocytes, macrophages, and dendritic cells will yield important insights into therapeutic strategies to suppress myocardial inflammation. METHODS: We used mouse models of reperfused myocardial infarction, angiotensin II and phenylephrine infusion, and diphtheria toxin cardiomyocyte ablation to investigate CCL17 (C-C chemokine ligand 17). We used Ccl17 knockout mice, flow cytometry, RNA sequencing, biochemical assays, cell trafficking studies, and in vivo cell depletion to identify the cell types that generate CCL17, define signaling pathways that controlled its expression, delineate the functional importance of CCL17 in adverse LV remodeling and heart failure progression, and determine the mechanistic basis by which CCL17 exerts its effects. RESULTS: We demonstrated that CCL17 is expressed in CCR2+ macrophages and cluster of differentiation 11b+ conventional dendritic cells after myocardial infarction, angiotensin II and phenylephrine infusion, and diphtheria toxin cardiomyocyte ablation. We clarified the transcriptional signature of CCL17+ macrophages and dendritic cells and identified granulocyte-macrophage colony-stimulating factor (GM-CSF) signaling as a key regulator of CCL17 expression through cooperative activation of STAT5 (signal transducer and activator of transcription 5) and canonical NF-κB (nuclear factor κ-light-chain-enhancer of activated B cells) signaling. Ccl17 deletion resulted in reduced LV remodeling, decreased myocardial fibrosis and cardiomyocyte hypertrophy, and improved LV systolic function after myocardial infarction and angiotensin II and phenylephrine infusion. We observed increased abundance of regulatory T cells (Tregs) in the myocardium of injured Ccl17 knockout mice. CCL17 inhibited Treg recruitment through biased activation of CCR4. CCL17 activated Gq signaling and CCL22 (C-C chemokine ligand 22) activated both Gq and ARRB (ß-arrestin) signaling downstream of CCR4. CCL17 competitively inhibited CCL22 stimulated ARRB signaling and Treg migration. We provide evidence that Tregs mediated the protective effects of Ccl17 deletion on myocardial inflammation and adverse LV remodeling. CONCLUSIONS: These findings identify CCL17 as a proinflammatory mediator of CCR2+ macrophages and dendritic cells and suggest that inhibition of CCL17 may serve as an effective strategy to promote Treg recruitment and suppress myocardial inflammation.


Subject(s)
Heart Failure , Myocardial Infarction , Angiotensin II/pharmacology , Animals , Chemokine CCL17/metabolism , Chemokine CCL17/pharmacology , Diphtheria Toxin/metabolism , Diphtheria Toxin/pharmacology , Heart Failure/genetics , Heart Failure/metabolism , Humans , Inflammation/metabolism , Ligands , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenylephrine/metabolism , Phenylephrine/pharmacology , T-Lymphocytes, Regulatory/metabolism , Ventricular Remodeling
5.
Basic Res Cardiol ; 118(1): 25, 2023 06 28.
Article in English | MEDLINE | ID: mdl-37378715

ABSTRACT

RNA-protein interactions are central to cardiac function, but how activity of individual RNA-binding protein is regulated through signaling cascades in cardiomyocytes during heart failure development is largely unknown. The mechanistic target of rapamycin kinase is a central signaling hub that controls mRNA translation in cardiomyocytes; however, a direct link between mTOR signaling and RNA-binding proteins in the heart has not been established. Integrative transcriptome and translatome analysis revealed mTOR dependent translational upregulation of the RNA binding protein Ybx1 during early pathological remodeling independent of mRNA levels. Ybx1 is necessary for pathological cardiomyocyte growth by regulating protein synthesis. To identify the molecular mechanisms how Ybx1 regulates cellular growth and protein synthesis, we identified mRNAs bound to Ybx1. We discovered that eucaryotic elongation factor 2 (Eef2) mRNA is bound to Ybx1, and its translation is upregulated during cardiac hypertrophy dependent on Ybx1 expression. Eef2 itself is sufficient to drive pathological growth by increasing global protein translation. Finally, Ybx1 depletion in vivo preserved heart function during pathological cardiac hypertrophy. Thus, activation of mTORC1 links pathological signaling cascades to altered gene expression regulation by activation of Ybx1 which in turn promotes translation through increased expression of Eef2.


Subject(s)
Heart Failure , TOR Serine-Threonine Kinases , Cardiomegaly/metabolism , Heart Failure/metabolism , Myocytes, Cardiac/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/physiology , TOR Serine-Threonine Kinases/metabolism , Animals , Mice , Rats
6.
Basic Res Cardiol ; 117(1): 32, 2022 06 23.
Article in English | MEDLINE | ID: mdl-35737129

ABSTRACT

Alterations of RNA editing that affect the secondary structure of RNAs can cause human diseases. We therefore studied RNA editing in failing human hearts. Transcriptome sequencing showed that adenosine-to-inosine (A-to-I) RNA editing was responsible for 80% of the editing events in the myocardium. Failing human hearts were characterized by reduced RNA editing. This was primarily attributable to Alu elements in introns of protein-coding genes. In the failing left ventricle, 166 circRNAs were upregulated and 7 circRNAs were downregulated compared to non-failing controls. Most of the upregulated circRNAs were associated with reduced RNA editing in the host gene. ADAR2, which binds to RNA regions that are edited from A-to-I, was decreased in failing human hearts. In vitro, reduction of ADAR2 increased circRNA levels suggesting a causal effect of reduced ADAR2 levels on increased circRNAs in the failing human heart. To gain mechanistic insight, one of the identified upregulated circRNAs with a high reduction of editing in heart failure, AKAP13, was further characterized. ADAR2 reduced the formation of double-stranded structures in AKAP13 pre-mRNA, thereby reducing the stability of Alu elements and the circularization of the resulting circRNA. Overexpression of circAKAP13 impaired the sarcomere regularity of human induced pluripotent stem cell-derived cardiomyocytes. These data show that ADAR2 mediates A-to-I RNA editing in the human heart. A-to-I RNA editing represses the formation of dsRNA structures of Alu elements favoring canonical linear mRNA splicing and inhibiting the formation of circRNAs. The findings are relevant to diseases with reduced RNA editing and increased circRNA levels and provide insights into the human-specific regulation of circRNA formation.


Subject(s)
Induced Pluripotent Stem Cells , RNA Editing , Humans , Induced Pluripotent Stem Cells/metabolism , RNA/chemistry , RNA/genetics , RNA/metabolism , RNA, Circular/genetics , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
7.
Basic Res Cardiol ; 117(1): 61, 2022 11 16.
Article in English | MEDLINE | ID: mdl-36383299

ABSTRACT

AIMS: P-selectin is an activatable adhesion molecule on platelets promoting platelet aggregation, and platelet-leukocyte complex (PLC) formation. Increased numbers of PLC are circulating in the blood of patients shortly after acute myocardial infarction and predict adverse outcomes. These correlations led to speculations about whether PLC may represent novel therapeutic targets. We therefore set out to elucidate the pathomechanistic relevance of PLC in myocardial ischemia and reperfusion injury. METHODS AND RESULTS: By generating P-selectin deficient bone marrow chimeric mice, the post-myocardial infarction surge in PLC numbers in blood was prevented. Yet, intravital microscopy, flow cytometry and immunohistochemical staining, echocardiography, and gene expression profiling showed unequivocally that leukocyte adhesion to the vessel wall, leukocyte infiltration, and myocardial damage post-infarction were not altered in response to the lack in PLC. CONCLUSION: We conclude that myocardial infarction associated sterile inflammation triggers PLC formation, reminiscent of conserved immunothrombotic responses, but without PLC influencing myocardial ischemia and reperfusion injury in return. Our experimental data do not support a therapeutic concept of selectively targeting PLC formation in myocardial infarction.


Subject(s)
Myocardial Infarction , Myocardial Ischemia , Myocardial Reperfusion Injury , Reperfusion Injury , Mice , Animals , P-Selectin/metabolism , Myocardial Reperfusion Injury/metabolism , Leukocytes , Myocardial Infarction/metabolism , Reperfusion Injury/metabolism , Myocardial Ischemia/metabolism
8.
J Immunol ; 205(8): 2276-2286, 2020 10 15.
Article in English | MEDLINE | ID: mdl-32938726

ABSTRACT

The number and activity of T cell subsets in the atherosclerotic plaques are critical for the prognosis of patients with acute coronary syndrome. ß2 Integrin activation is pivotal for T cell recruitment and correlates with future cardiac events. Despite this knowledge, differential regulation of adhesiveness in T cell subsets has not been explored yet. In this study, we show that in human T cells, SDF-1α-mediated ß2 integrin activation is driven by a, so far, not-described reactive oxidative species (ROS)-regulated calcium influx. Furthermore, we show that CD4+CD28null T cells represent a highly reactive subset showing 25-fold stronger ß2 integrin activation upon SDF-1α stimulation compared with CD28+ T cells. Interestingly, ROS-dependent Ca release was much more prevalent in the pathogenetically pivotal CD28null subset compared with the CD28+ T cells, whereas the established mediators of the classical pathways for ß2 integrin activation (PKC, PI3K, and PLC) were similarly activated in both T cell subsets. Thus, interference with the calcium flux attenuates spontaneous adhesion of CD28null T cells from acute coronary syndrome patients, and calcium ionophores abolished the observed differences in the adhesion properties between CD28+ and CD28null T cells. Likewise, the adhesion of these T cell subsets was indistinguishable in the presence of exogenous ROS/H2O2 Together, these data provide a molecular explanation of the role of ROS in pathogenesis of plaque destabilization.


Subject(s)
Acute Coronary Syndrome/immunology , CD18 Antigens/immunology , CD4-Positive T-Lymphocytes/immunology , Calcium Signaling/immunology , Reactive Oxygen Species/immunology , Acute Coronary Syndrome/pathology , CD28 Antigens/immunology , CD4-Positive T-Lymphocytes/pathology , Chemokine CXCL12/immunology , Female , Humans , Male
9.
Int J Mol Sci ; 23(20)2022 Oct 14.
Article in English | MEDLINE | ID: mdl-36293109

ABSTRACT

Wnt (a portmanteau of Wingless and Int-1) signaling in the adult heart is largely quiescent. However, there is accumulating evidence that it gets reactivated during the healing process after myocardial infarction (MI). We here tested the therapeutic potential of the Wnt secretion inhibitor LGK-974 on MI healing. Ischemia/reperfusion (I/R) injury was induced in mice and Wnt signaling was inhibited by oral administration of the porcupine inhibitor LGK-974. The transcriptome was analyzed from infarcted tissue by using RNA sequencing analysis. The inflammatory response after I/R was evaluated by flow cytometry. Heart function was assessed by echocardiography and fibrosis by Masson's trichrome staining. Transcriptome and gene set enrichment analysis revealed a modulation of the inflammatory response upon administration of the Wnt secretion inhibitor LGK-974 following I/R. In addition, LGK-974-treated animals showed an attenuated inflammatory response and improved heart function. In an in vitro model of hypoxic cardiomyocyte and monocyte/macrophage interaction, LGK974 inhibited the activation of Wnt signaling in monocytes/macrophages and reduced their pro-inflammatory phenotype. We here show that Wnt signaling affects inflammatory processes after MI. The Wnt secretion inhibitor LGK-974 appears to be a promising compound for future immunomodulatory approaches to improve cardiac remodeling after MI.


Subject(s)
Myocardial Infarction , Myocardial Reperfusion Injury , Mice , Animals , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/genetics , Myocardium , Macrophages , Myocardial Infarction/genetics , Myocytes, Cardiac , Wnt Signaling Pathway , Mice, Inbred C57BL , Ventricular Remodeling , Disease Models, Animal
10.
Circulation ; 141(20): 1628-1644, 2020 05 19.
Article in English | MEDLINE | ID: mdl-32100557

ABSTRACT

BACKGROUND: Acute occlusion of a coronary artery results in swift tissue necrosis. Bordering areas of the infarcted myocardium can also experience impaired blood supply and reduced oxygen delivery, leading to altered metabolic and mechanical processes. Although transcriptional changes in hypoxic cardiomyocytes are well studied, little is known about the proteins that are actively secreted from these cells. METHODS: We established a novel secretome analysis of cardiomyocytes by combining stable isotope labeling and click chemistry with subsequent mass spectrometry analysis. Further functional validation experiments included ELISA measurement of human samples, murine left anterior descending coronary artery ligation, and adeno-associated virus 9-mediated in vivo overexpression in mice. RESULTS: The presented approach is feasible for analysis of the secretome of primary cardiomyocytes without serum starvation. A total of 1026 proteins were identified to be secreted within 24 hours, indicating a 5-fold increase in detection compared with former approaches. Among them, a variety of proteins have not yet been explored in the context of cardiovascular pathologies. One of the secreted factors most strongly upregulated upon hypoxia was PCSK6 (proprotein convertase subtilisin/kexin type 6). Validation experiments revealed an increase of PCSK6 on mRNA and protein level in hypoxic cardiomyocytes. PCSK6 expression was elevated in hearts of mice after 3 days of ligation of the left anterior descending artery, a finding confirmed by immunohistochemistry. ELISA measurements in human serum also indicate distinct kinetics for PCSK6 in patients with acute myocardial infarction, with a peak on postinfarction day 3. Transfer of PCSK6-depleted cardiomyocyte secretome resulted in decreased expression of collagen I and III in fibroblasts compared with control treated cells, and small interfering RNA-mediated knockdown of PCSK6 in cardiomyocytes impacted transforming growth factor-ß activation and SMAD3 (mothers against decapentaplegic homolog 3) translocation in fibroblasts. An adeno-associated virus 9-mediated, cardiomyocyte-specific overexpression of PCSK6 in mice resulted in increased collagen expression and cardiac fibrosis, as well as decreased left ventricular function, after myocardial infarction. CONCLUSIONS: A novel mass spectrometry-based approach allows investigation of the secretome of primary cardiomyocytes. Analysis of hypoxia-induced secretion led to the identification of PCSK6 as being crucially involved in cardiac remodeling after acute myocardial infarction.


Subject(s)
Myocardial Infarction/enzymology , Myocytes, Cardiac/enzymology , Proprotein Convertase 9/metabolism , Ventricular Function, Left , Ventricular Remodeling , Animals , Animals, Newborn , Cells, Cultured , Disease Models, Animal , Humans , Male , Mice, Inbred C57BL , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocytes, Cardiac/pathology , Proprotein Convertase 9/genetics , Proteome , Rats, Wistar , Secretory Pathway , Signal Transduction
11.
Circ Res ; 125(3): 282-294, 2019 07 19.
Article in English | MEDLINE | ID: mdl-31213138

ABSTRACT

RATIONALE: Fluid shear stress (FSS) maintains NOS-3 (endothelial NO synthase) expression. Homozygosity for the C variant of the T-786C single-nucleotide polymorphism of the NOS3 gene, which solely exists in humans, renders the gene less sensitive to FSS, resulting in a reduced endothelial cell (EC) capacity to generate NO. Decreased bioavailability of NO in the arterial vessel wall facilitates atherosclerosis. Consequently, individuals homozygous for the C variant have an increased risk for coronary heart disease (CHD). OBJECTIVE: At least 2 compensatory mechanisms seem to minimize the deleterious effects of this single-nucleotide polymorphism in affected individuals, one of which is characterized herein. METHODS AND RESULTS: Human genotyped umbilical vein ECs and THP-1 monocytes were used to investigate the role of 15-deoxy-Δ12,14-prostaglandin J2 (15d-PGJ2) in vitro. Its concentration in plasma samples from genotyped patients with CHD and age-matched CHD-free controls was determined using quantitative ultraperformance LC-MS/MS. Exposure of human ECs to FSS effectively reduced monocyte transmigration particularly through monolayers of CC-genotype ECs. Primarily in CC-genotype ECs, FSS elicited a marked rise in COX (cyclooxygenase)-2 and L-PGDS (lipocalin-type prostaglandin D synthase) expression, which appeared to be NO sensitive, and provoked a significant release of 15d-PGJ2 over baseline. Exogenous 15d-PGJ2 significantly reduced monocyte transmigration and exerted a pronounced anti-inflammatory effect on the transmigrated monocytes by downregulating, for example, transcription of the IL (interleukin)-1ß gene (IL1B). Reporter gene analyses verified that this effect is due to binding of Nrf2 (nuclear factor [erythroid-derived 2]-like 2) to 2 AREs (antioxidant response elements) in the proximal IL1B promoter. In patients with CHD, 15d-PGJ2 plasma levels were significantly upregulated compared with age-matched CHD-free controls, suggesting that this powerful anti-inflammatory prostanoid is part of an endogenous defence mechanism to counteract CHD. CONCLUSIONS: Despite a reduced capacity to form NO, CC-genotype ECs maintain a robust anti-inflammatory phenotype through an enhanced FSS-dependent release of 15d-PGJ2.


Subject(s)
Endothelial Cells/metabolism , Nitric Oxide Synthase Type III/deficiency , Nitric Oxide/blood , Polymorphism, Single Nucleotide , Prostaglandin D2/analogs & derivatives , Adaptation, Physiological , Aged , Aged, 80 and over , Coronary Disease/blood , Coronary Disease/genetics , Cyclooxygenase 2/biosynthesis , Cyclooxygenase 2/genetics , Enzyme Induction , Female , Genes, Reporter , Genetic Predisposition to Disease , Hemorheology , Human Umbilical Vein Endothelial Cells , Humans , Inflammation , Intramolecular Oxidoreductases/biosynthesis , Intramolecular Oxidoreductases/genetics , Lipocalins/biosynthesis , Lipocalins/genetics , Male , Middle Aged , NF-E2-Related Factor 2/physiology , Nitric Oxide Synthase Type III/genetics , Prostaglandin D2/biosynthesis , Prostaglandin D2/blood , Prostaglandin D2/physiology , RNA Interference , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , THP-1 Cells
12.
Circ Res ; 124(6): 881-890, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30661445

ABSTRACT

RATIONALE: Paradigm shifting studies have revealed that the heart contains functionally diverse populations of macrophages derived from distinct embryonic and adult hematopoietic progenitors. Under steady-state conditions, the heart is largely populated by CCR2- (C-C chemokine receptor type 2) macrophages of embryonic descent. After tissue injury, a dramatic shift in macrophage composition occurs whereby CCR2+ monocytes are recruited to the heart and differentiate into inflammatory CCR2+ macrophages that contribute to heart failure progression. Currently, there are no techniques to noninvasively detect CCR2+ monocyte recruitment into the heart and thus identify patients who may be candidates for immunomodulatory therapy. OBJECTIVE: To develop a noninvasive molecular imaging strategy with high sensitivity and specificity to visualize inflammatory monocyte and macrophage accumulation in the heart. METHODS AND RESULTS: We synthesized and tested the performance of a positron emission tomography radiotracer (68Ga-DOTA [1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid]-ECL1i [extracellular loop 1 inverso]) that allosterically binds to CCR2. In naive mice, the radiotracer was quickly cleared from the blood and displayed minimal retention in major organs. In contrast, biodistribution and positron emission tomography demonstrated strong myocardial tracer uptake in 2 models of cardiac injury (diphtheria toxin induced cardiomyocyte ablation and reperfused myocardial infarction). 68Ga-DOTA-ECL1i signal localized to sites of tissue injury and was independent of blood pool activity as assessed by quantitative positron emission tomography and ex vivo autoradiography. 68Ga-DOTA-ECL1i uptake was associated with CCR2+ monocyte and CCR2+ macrophage infiltration into the heart and was abrogated in CCR2-/- mice, demonstrating target specificity. Autoradiography demonstrated that 68Ga-DOTA-ECL1i specifically binds human heart failure specimens and with signal intensity associated with CCR2+ macrophage abundance. CONCLUSIONS: These findings demonstrate the sensitivity and specificity of 68Ga-DOTA-ECL1i in the mouse heart and highlight the translational potential of this agent to noninvasively visualize CCR2+ monocyte recruitment and inflammatory macrophage accumulation in patients.


Subject(s)
Heart/diagnostic imaging , Macrophages/physiology , Monocytes/physiology , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/pathology , Animals , Cell Movement , Humans , Mice , Mice, Inbred C57BL , Molecular Imaging , Positron-Emission Tomography , Receptors, CCR2/analysis
13.
Herz ; 46(Suppl 1): 41-47, 2021 Apr.
Article in German | MEDLINE | ID: mdl-32313970

ABSTRACT

With increasing age valvular heart disease is among the most frequent diseases of the heart. Relevant valvular disease impairs not only the long-term prognosis but also physical resilience, activities of daily living and the quality of life. In cases of middle to high-grade symptomatic cardiac defects, valve replacement or valve reconstruction is still the surgical procedure of choice; however, in recent years the transcatheter percutaneous aortic valve replacement (TAVI) procedure has become more prominent for the most frequent defect, aortic valve stenosis. This article provides an overview of the aftercare and rehabilitation of patients following a TAVI intervention.


Subject(s)
Aortic Valve Stenosis , Heart Valve Prosthesis Implantation , Heart Valve Prosthesis , Transcatheter Aortic Valve Replacement , Activities of Daily Living , Aortic Valve/surgery , Aortic Valve Stenosis/diagnostic imaging , Aortic Valve Stenosis/surgery , Heart Valve Prosthesis Implantation/adverse effects , Humans , Quality of Life , Subacute Care , Transcatheter Aortic Valve Replacement/adverse effects , Treatment Outcome
14.
Proc Natl Acad Sci U S A ; 115(24): E5556-E5565, 2018 06 12.
Article in English | MEDLINE | ID: mdl-29793936

ABSTRACT

Monocyte extravasation into the vessel wall is a key step in atherogenesis. It is still elusive how monocytes transmigrate through the endothelial cell (EC) monolayer at atherosclerosis predilection sites. Platelets tethered to ultra-large von Willebrand factor (ULVWF) multimers deposited on the luminal EC surface following CD40 ligand (CD154) stimulation may facilitate monocyte diapedesis. Human ECs grown in a parallel plate flow chamber for live-cell imaging or Transwell permeable supports for transmigration assay were exposed to fluid or orbital shear stress and CD154. Human isolated platelets and/or monocytes were superfused over or added on top of the EC monolayer. Plasma levels and activity of the ULVWF multimer-cleaving protease ADAMTS13 were compared between coronary artery disease (CAD) patients and controls and were verified by the bioassay. Two-photon intravital microscopy was performed to monitor CD154-dependent leukocyte recruitment in the cremaster microcirculation of ADAMTS13-deficient versus wild-type mice. CD154-induced ULVWF multimer-platelet string formation on the EC surface trapped monocytes and facilitated transmigration through the EC monolayer despite high shear stress. Two-photon intravital microscopy revealed CD154-induced ULVWF multimer-platelet string formation preferentially in venules, due to strong EC expression of CD40, causing prominent downstream leukocyte extravasation. Plasma ADAMTS13 abundance and activity were significantly reduced in CAD patients and strongly facilitated both ULVWF multimer-platelet string formation and monocyte trapping in vitro. Moderate ADAMTS13 deficiency in CAD patients augments CD154-mediated deposition of platelet-decorated ULVWF multimers on the luminal EC surface, reinforcing the trapping of circulating monocytes at atherosclerosis predilection sites and promoting their diapedesis.


Subject(s)
ADAMTS13 Protein/metabolism , Blood Platelets/metabolism , CD40 Antigens/metabolism , Cell Communication/physiology , Endothelial Cells/metabolism , von Willebrand Factor/metabolism , Adolescent , Adult , Aged , Animals , Atherosclerosis/metabolism , Cells, Cultured , Endothelium, Vascular/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Monocytes/metabolism , Platelet Aggregation/physiology , Stress, Mechanical , Young Adult
15.
Eur Heart J ; 41(9): 989-994, 2020 03 01.
Article in English | MEDLINE | ID: mdl-30945736

ABSTRACT

Over a century ago, Élie Metchnikoff described the macrophages' ability to phagocytose. Propelled by advances in technology enabling phenotypic and functional analyses at unpreceded resolution, a recent renaissance in macrophage research has shed new light on these 'big eaters'. We here give an overview of cardiac macrophages' provenance in the contexts of cardiac homeostasis and stress. We highlight the recently identified mechanism by which these cells regulate electrical conduction in the atrioventricular node and discuss why we need a deeper understanding of monocytes and macrophages in systolic and diastolic dysfunctions.


Subject(s)
Heart , Macrophages , Homeostasis , Monocytes
16.
Curr Heart Fail Rep ; 17(5): 213-224, 2020 10.
Article in English | MEDLINE | ID: mdl-32783147

ABSTRACT

PURPOSE OF REVIEW: The goal of this review is to summarize the state of big data analyses in the study of heart failure (HF). We discuss the use of big data in the HF space, focusing on "omics" and clinical data. We address some limitations of this data, as well as their future potential. RECENT FINDINGS: Omics are providing insight into plasmal and myocardial molecular profiles in HF patients. The introduction of single cell and spatial technologies is a major advance that will reshape our understanding of cell heterogeneity and function as well as tissue architecture. Clinical data analysis focuses on HF phenotyping and prognostic modeling. Big data approaches are increasingly common in HF research. The use of methods designed for big data, such as machine learning, may help elucidate the biology underlying HF. However, important challenges remain in the translation of this knowledge into improvements in clinical care.


Subject(s)
Big Data , Biomedical Research/statistics & numerical data , Heart Failure/genetics , Machine Learning , Humans , Prognosis
17.
J Mol Cell Cardiol ; 126: 13-22, 2019 01.
Article in English | MEDLINE | ID: mdl-30445017

ABSTRACT

AIMS: Circulating immune cells have a significant impact on progression and outcome of heart failure. Long non-coding RNAs (lncRNAs) comprise novel epigenetic regulators which control cardiovascular diseases and inflammatory disorders. We aimed to identify lncRNAs regulated in circulating immune cells of the blood of heart failure patients. METHODS AND RESULTS: Next-generation sequencing revealed 110 potentially non-coding RNA transcripts differentially expressed in peripheral blood mononuclear cells of heart failure patients with reduced ejection fraction. The up-regulated lncRNA Heat2 was further functionally characterized. Heat2 expression was detected in whole blood, PBMNCs, eosinophil and basophil granulocytes. Heat2 regulates cell division, invasion, transmigration and immune cell adhesion on endothelial cells. CONCLUSION: Heat2 is an immune cell enriched lncRNA that is elevated in the blood of heart failure patients and controls cellular functions.


Subject(s)
Gene Expression Regulation , Heart Failure/genetics , RNA, Long Noncoding/genetics , Adult , Aged , Case-Control Studies , Cohort Studies , Eosinophils/metabolism , Female , Heart Failure/blood , Humans , Leukocytes, Mononuclear/metabolism , Male , Middle Aged , RNA, Long Noncoding/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
18.
Basic Res Cardiol ; 113(6): 44, 2018 10 16.
Article in English | MEDLINE | ID: mdl-30327885

ABSTRACT

Various cell types are involved in the healing process after myocardial infarction (MI). Besides cardiac resident cells (such as cardiomyocytes, fibroblasts and endothelial cells) already present at the lesion site, a massive influx of leukocytes (mainly monocytes and neutrophils) is observed within hours after the ischemic event. So far, little is known about modes of interaction of these cells. Wnt signaling is an evolutionary conserved signaling cassette known to play an important role in cell-cell communication. While the overall reactivation of Wnt signaling upon ischemic injury is well described, the precise expression pattern of Wnt proteins, however, is far from understood. We here describe known Wnt components that partake in MI healing and differentiate cell-specific aspects. The secretion of Wnt proteins and their antagonists in the context of cardiac inflammation after MI appear to be tightly regulated in a spatial-temporal manner. Overall, we aim to stress the importance of elucidating not only Wnt component-specific aspects, but also their sometimes contradicting effects in different target cells. A better understanding of Wnt signaling in MI healing may eventually lead to the development of successful therapeutic approaches in an often considered "un-druggable" pathway.


Subject(s)
Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardium/metabolism , Myocardium/pathology , Wnt Signaling Pathway/physiology , Animals , Humans
19.
J Interv Cardiol ; 31(6): 885-890, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30397939

ABSTRACT

BACKGROUND: To date, there are no guidelines recommending a specific prophylactic antibiotic treatment in transcatheter aortic valve replacement (TAVR). The aim of this study is to evaluate clinical data after TAVR with different periprocedural antibiotic regimens. METHODS: In May 2015 the institutional rules for periprocedural antibiotic prophylaxis were changed from 3 days to 1 day. Thus, a total of 450 consecutive TAVR patients between February 2014 and June 2016 were classified into two intention-to-treat groups: patients receiving a 1-day Cefuroxime prophylaxis (N = 225); patients receiving a 3-day Cefuroxime prophylaxis (N = 225). RESULTS: One-day Cefuroxime regimen was not associated with shorter hospitalization (3-day Cefuroxime 9 ± 4.7 vs 1-day Cefuroxime 8.9 ± 4.0; P = 0.87). Incidence of diarrhea (26.2% vs 18.2%; P = 0.04) and Clostridium difficile infections (4% vs 0.4%; P = 0.01) were significantly higher in the 3-day group. No endocarditis was registered after 1 year follow-up. There was no difference in 30-day overall mortality rate, major vascular complications, bleeding complications, pacemaker-implantation rate, paravalvular regurgitation, or acute kidney injury between patients groups. CONCLUSION: Three-day Cefuroxime prophylaxis does not seem to be advantageous compared to a shorter 1-day regimen, but even shows a significantly higher incidence of diarrhea and Clostridium difficile infection.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Antibiotic Prophylaxis/methods , Cefuroxime/administration & dosage , Transcatheter Aortic Valve Replacement/adverse effects , Aged , Aged, 80 and over , Anti-Bacterial Agents/adverse effects , Antibiotic Prophylaxis/adverse effects , Aortic Valve/surgery , Aortic Valve Stenosis/surgery , Cefuroxime/adverse effects , Female , Hospital Mortality , Humans , Intention to Treat Analysis , Length of Stay/statistics & numerical data , Male , Postoperative Complications/epidemiology , Postoperative Complications/etiology , Postoperative Complications/prevention & control , Retrospective Studies , Risk Factors , Treatment Outcome
20.
Nature ; 487(7407): 325-9, 2012 Jul 19.
Article in English | MEDLINE | ID: mdl-22763456

ABSTRACT

During progression of atherosclerosis, myeloid cells destabilize lipid-rich plaques in the arterial wall and cause their rupture, thus triggering myocardial infarction and stroke. Survivors of acute coronary syndromes have a high risk of recurrent events for unknown reasons. Here we show that the systemic response to ischaemic injury aggravates chronic atherosclerosis. After myocardial infarction or stroke, Apoe-/- mice developed larger atherosclerotic lesions with a more advanced morphology. This disease acceleration persisted over many weeks and was associated with markedly increased monocyte recruitment. Seeking the source of surplus monocytes in plaques, we found that myocardial infarction liberated haematopoietic stem and progenitor cells from bone marrow niches via sympathetic nervous system signalling. The progenitors then seeded the spleen, yielding a sustained boost in monocyte production. These observations provide new mechanistic insight into atherogenesis and provide a novel therapeutic opportunity to mitigate disease progression.


Subject(s)
Atherosclerosis/etiology , Atherosclerosis/pathology , Myocardial Infarction/complications , Myocardial Infarction/pathology , Animals , Apolipoproteins E/genetics , Hematopoietic Stem Cells/cytology , Inflammation/complications , Mice , Mice, Inbred C57BL , Monocytes/cytology , Spleen/cytology , Stem Cells/cytology
SELECTION OF CITATIONS
SEARCH DETAIL