Abstract Collection from the 10th Annual Meeting of the European Section of the International Academy of Cardiovascular Sciences
Article Category: Special Issue - Book of abstracts
Published Online: Dec 21, 2024
Page range: 1 - 138
Received: Oct 12, 2024
Accepted: Oct 29, 2024
DOI: https://doi.org/10.2478/afpuc-2024-0013
Keywords
© 2024 Csaba Horvath et al., published by Sciendo
This work is licensed under the Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License.
Dear Esteemed Readers,
With great pleasure, we present you with an abstract collection – a special issue of the
The International Academy of Cardiovascular Sciences is a non-profit organisation that provides a unique platform for the worldwide sharing of research and education information in the field of cardiovascular health. The main mission of this organisation is to promote cardiovascular education for professionals and laypeople and to recognise major cardiovascular achievements by the young and established investigators globally. In line with this, the motto of the jubilee IACS-ES conference was ‘Know the heart: novel insights into the prevention and treatment of heart disease to live and promote a healthy lifestyle’. The scientific program was scheduled in 15 symposia running in two parallel sections and referring to many exciting topics, including (1) cardiac inflammation and immunology, (2) women's heart: when gender matters, (3) cardiac ageing, (4) inter-organ communication in heart disease, (5) extracellular vesicles and the cardiovascular system, (6) cardiac electrophysiology and arrhythmias, (7) heart failure, (8) bioengineering in heart research, (9) myocardial adaptation and novel targets of cardioprotection, (10) healthy diet for a healthy heart, (11) cardiac toxicity of non-cardiovascular drugs and (12) efficacy and safety of novel pharmacological interventions for the treatment of cardiovascular diseases. In addition to the lectures, a special focus was given to scientific networking among the young delegates and their scientific promotion.
The abstracts are listed alphabetically, rather than referring to the above-mentioned related topics. Furthermore, they are divided into two particular subgroups – (1) abstracts of the distinguished speakers and delegates and (2) abstracts of the regular participants. These abstracts provide the latest findings in heart research and give us new ideas and inspiration for further scientific projects as well as opportunities for further international collaborations in the field of cardiovascular sciences.
Adriana Duris Adameova, Prof. PharmD., DSc, FIACS – Chair of the Meeting
Csaba Horvath, PharmDr., PhD. and Tomas Rajtik, Assoc. Prof. PharmDr., PhD. – Local Scientific Committee Members
Comenius University Bratislava, Faculty of Pharmacy, Department of Pharmacology and Toxicology, Bratislava, Slovak Republic
Devendra Agrawal
Prevention of atherosclerotic plaque formation, reduction in plaque burden and stabilization of a vulnerable plaque are of therapeutic significance in decreasing the incidence of acute events. Initially, we found an imbalance between the inflammation and reparative process, leading to plaque instability. The findings supported the critical role of TLR4 and TREM-1 in underlying pathophysiology of chronic inflammation, atherosclerotic plaque formation and plaque instability. Thus, we examined the effect of the selective inhibitors of TREM-1 (LR-12) and TLR4 (TAK242) in a microswine model of carotid artery atherosclerosis. Hypercholesterolemic Yucatan microswine were treated with a control vehicle and TREM-1 or TLR4 inhibitor at the time of intimal injury and were sacrificed after 5–6 months. Optical coherence tomography (OCT) showed plaques with thick fibrous cap and occluded arteries in the controls. Radiological evaluation of carotid arteries revealed decreased neointimal hyperplasia and plaque formation with treatment using the TREM-1 inhibitor compared to the scrambled peptide group. There was no significant effect of TAK242 when dissolved in 30% ethanol. However, TAK242 dissolved in a clinically relevant vehicle significantly decreased the plaque size and enhanced plaque stabilization. Histomorphologically, there was neointimal hyperplasia with significantly increased inflammation and elastin degradation in the controls compared to the inhibitor groups on H&E and Movat pentachrome staining. The gene and protein expression for the markers of plaque vulnerability, including MMP-7, IL-6, IL-12/23, and CD36, were significantly decreased with treatment using the TREM and TLR4 inhibitors (dissolved in clinically relevant vehicle) compared to individual vehicle control. The findings of this study support the therapeutic efficacy of inhibiting either TLR4 or TREM-1 signalling alone or in combination in attenuating atherosclerotic plaque vulnerability, reducing plaque burden and thus preventing the occurrence of transient ischaemic attack and stroke.
This work was supported by the research grant R01 HL144125 from the National Institutes of Health, USA.
David Astapenko
Hydrogen has the potential to be a valuable agent in clinical medicine due to its antioxidant and anti-inflammatory properties. This gas easily passes through membranes and quickly reaches the tissues, where it acts by quenching the hydroxyl radical, irrespective of other reactive oxygen species. The current interest in this gas is reflected in studies of ischaemia-reperfusion injury, cardiovascular disease, transplantation surgery, haemorrhagic shock and post-cardiac arrest syndrome. After successful resuscitation of cardiac arrest by achieving the return of spontaneous circulation, the whole body is subject to an ischaemia-reperfusion injury that involves a massive ROS generation, which is believed to subsequently damage the endothelial cells and the endothelial glycocalyx (EG) layer. The latter is a delicate sugar-based gel-like structure covering the endoluminal aspect of endothelial cells and is ascribed to have a multitude of functions, including endothelium protection, regulation of vasomotion, antioxidation, anti-thrombosis and regulation of capillary permeability, hence preventing the free movement of water and electrolytes. Preserving the EG layer is considered a clinically relevant study objective, as damage to this layer results in the loss of its protective functions. Positive effects of hydrogen gas have previously been reported in cardiac arrest settings in animals and humans. Moreover, a preventive effect was observed on syndecan-1 release in a rat model of haemorrhagic shock. The lecture describes the author's experience in experimental research with hydrogen gas and the protection of EG in a rat model of cardiac arrest.
This work was supported by the University Hospital Hradec Kralove, Czech Republic: MH CZ-DRO (UHHK, 00179906).
Istvan Baczko
The antiarrhythmic and cardiac electrophysiological effects of SZV-2649, a new amiodarone-like antiarrhythmic compound that lacks the benzofuran moiety, were studied in a mammalian cell line as well as in rat and dog cardiac preparations. SZV-2649 exerted antiarrhythmic effects against coronary artery occlusion/reperfusion-induced ventricular arrhythmias in rats and in acetylcholine- and burst stimulation–induced atrial fibrillation in dogs. SZV-2649 inhibited hERG and GIRK currents in HEK cells with IC50 values of 342 nM and 529 nM, respectively. In canine cardiac ventricular myocytes, SZV-2649 (10 μM) decreased the densities of IKr, Ito, INaL and ICaL currents. The compound (2.5–10 μM) elicited Vaughan Williams Class IB type
This work was supported by the National Research Development and Innovation Office (NKFIH-K147212).
Monika Barteková1,2, Lucia Kindernay1, Barbora Kaločayová1,3, Linda Bartošová3, Ulrika Duľová1, Jakub Strapec1, Tomáš Rajtík1,3, Kristína Ferenczyová1,3
Quercetin (QCT) is a natural polyphenolic substance that has been shown to exert several beneficial effects in the cardiovascular system, including antioxidant, anti-inflammatory and antihypertensive effects. In our experimental studies, we focused on potential the cardioprotective effects of QCT in myocardial (I/R) injury in different experimental settings, including the presence of selected comorbidities, co-medications and ageing.
The current presentation aims to summarize data obtained in our studies documenting the effects of QCT in several animal models of myocardial I/R injury and to uncover the role of age, metabolic comorbidities including diabetes and obesity, and co-medication with the anthracycline doxorubicin in its cardiac effects.
QCT in the dose of 20 mg/kg/day was administered orally for 4–6 weeks to rats of different age and rats with selected comorbidities/co-medications. After the end of treatment, hearts were isolated and ex vivo exposed to I/R (30-min global ischaemia/2-h reperfusion). Recovery of cardiac function and infarct size were assessed as the physiological outputs of the experiments. Molecular mechanisms of QCT action were evaluated as well.
The results showed that QCT exerts cardioprotective effects in I/R injury in healthy young and doxorubicin-treated rats but is inefficient in preventing I/R injury in rats with comorbidities (hypertensive/type 2 diabetic). Moreover, the cardioprotective efficiency of QCT is negatively influenced by increasing age. In conclusion, QCT might be potentially cardioprotective in preventing myocardial I/R injury; however, ageing and/or the presence of comorbidities may decrease or even abolish the anti-ischaemic effects of QCT.
Supported by VEGA 2/0104/20 and APVV-21-0194.
Jerzy Beltowski
Perivascular adipose tissue (PVAT) plays an important role in the regulation of vascular homeostasis. Under physiological conditions, PVAT produces vasodilating and anti-inflammatory factors. However, in obesity/metabolic syndrome, PVAT phenotype changes to a pro-inflammatory one. Recently, it has been demonstrated that green tea polyphenols, such as (−)-epigallocatechin (EGC), oxidize the gasotransmitter hydrogen sulphide (H2S) to polysulphides (H2Sn), the important signalling molecules (
Expression/secretion of leptin, resistin, TNF-alpha, IL-6 and MCP-1 was higher and adiponectin was lower in PAT of obese rats, which was accompanied by increased H2S production. The H2S and H2Sn donors, Na2S and Na2S4, had pro- and anti-inflammatory effects on PVAT, respectively. Administration of EGC in HFD rats reduced the expression of leptin, resistin, TNF-alpha, IL-6 and MCP-1 and increased the expression of adiponectin. In addition, EGC reduced the M1 macrophage marker, inducible NO synthase (iNOS), and increased the expression of M2 markers, IL-10 and arginase-1. The effects of EGC were mimicked by Na2S4. EGC decreased H2S and increased H2Sn in PAT of obese rats. Similar results were observed ex vivo in EGC-treated PVAT explants.
EGC has the anti-inflammatory effect on PVAT mediated by oxidation of H2S to H2Sn. This effect may contribute to the anti-atherosclerotic properties of green tea polyphenols and may explain, at least in part, the beneficial vascular effect of green tea in the metabolic syndrome.
This study was supported by the Medical University of Lublin, Lublin, Poland, under the grant DS476.
Péter Bencsik1,2, Tamara Szabados1,2, András Makkos2,3, Bettina Benczik2,3, Barnabás Váradi2,3, Bence Ágg2,3, Zoltán V Varga3, Anikó Görbe1,2,3, Péter Ferdinandy2,3
This study was supported by the 2020-1.1.5-GYORSÍTÓSÁV call programme (2020-1.1.5-GYORSÍTÓSÁV-2021-00011; Project no. RRF-2.3.1-21-2022-00003 ‘National Heart Laboratory, Hungary’; Hungarian National Scientific Research Fund (OTKA-138223).
Ghassan Bkaily, Danielle Jacques
Taurine is a well-known non-essential amino acid that is very important for cell function. This circulating factor is produced mainly by the brain and the heart. Nutrients, particularly seafood, ensure its supplementation. Taurine's beneficial effects are numerous, particularly at the cardiovascular level. A prolonged diet rich in sodium salt can lead to salt-sensitive hypertension. Until recently, it was accepted that there are sexual differences between men and women in salt-sensitive hypertension. Indeed, knowledge of the process behind the development of female-specific salt sensitivity remains to be elucidated. Recently, in humans, it has been shown that sodium-sensitive hypertension is due in part to morphological remodelling and ion homeostasis of human contractile vascular smooth muscle cells (hcVSMCs) induced by glycocalyx destruction. Our study shows that chronic exposure to extracellular sodium is associated with greater remodelling of ionic and morphological homeostasis in male compared to female hcVSMCs. Long-term treatment with taurine prevents hypernatraemia-induced remodelling of hcVSMCs in a sex-independent manner.
This work was supported by a grant from CIHR and NSERC.
Michal Botek, Jakub Krejčí
The main aim of this study was to determine whether molecular hydrogen (H2) administration through the hydrogen-rich water (HRW) intake can affect cardiovascular system regulation and lactate response during and immediately after an experimental exercise protocol in well-trained soccer players. An experiment was designed as a randomized, double-blind, placebo-controlled crossover trial. The heart rate (HR) responses during and following a repeated sprint ability protocol (15 m ± 30 m with 20 s recovery between each sprint) were performed in 12 professional soccer players aged 18.8 ± 1.3 years. The HRW contained 0.9 ppm of dissolved H2. HR was monitored using the HR monitor during the repeated spring protocol (maximal achieved heart rate – HRmax), average HR and at 1 (HRR1) and 3 min (HRR3) post-exercise HR. Post-exercise blood lactate concentration was measured after the last sprint. A statistically significant difference was found between HRW and placebo in HRR1 (HRW = 46.8 ± 10.6 bpm, placebo = 41.6 ± 12.6 bpm,
Patrick Burgon
Muscular-enriched Lamin Interacting Protein (MLIP) is increasingly recognized as a pivotal component in maintaining cellular homeostasis across various tissues, particularly within muscle cells. MLIP interacts intimately with the nuclear lamina, influencing nuclear architecture, gene expression and mechanotransduction pathways. This protein's role extends to the regulation of cellular stress responses, differentiation and proliferation, highlighting its multifaceted involvement in cellular physiology. Recent studies underscore MLIP's significance in muscle development and function, where its dysregulation is linked to muscular dystrophies and other myopathies. Notably, MLIP modulates key signalling cascades such as the PI3K/AKT and MAPK pathways, which are critical for cell survival and growth. These insights suggest that MLIP not only contributes to the structural integrity and dynamic functions of the nucleus but also plays a crucial role in cellular adaptive mechanisms. Given its central role in cellular homeostasis, MLIP emerges as a promising therapeutic target. Strategies to modulate MLIP activity could offer novel interventions for muscle-related diseases and potentially other conditions characterized by nuclear architecture disruptions and impaired stress responses. This presentation aims to synthesize current knowledge on MLIP's biological functions and therapeutic prospects, proposing future research directions to harness its potential in clinical applications.
This work was supported by the Qatar University (QUCG-CAS-23/24-214) and Qatar National Research Fund (UREP30-000-3-001).
Roberto Bolli
Two decades and > 200 trials have shown that adult stem/progenitor cells are safe, and several Phase I/II studies suggest that they are efficacious in heart failure and refractory angina, although large Phase III trials are needed to establish efficacy conclusively. In contrast, embryonic stem cells have never been tested in a controlled trial in cardiovascular medicine and are plagued by major problems, including ethical concerns, arrhythmias, tumourigenicity, rejection requiring immunosuppression, genomic instability, prohibitive dose requirements, phenotypic heterogeneity and lack of long-term engraftment. The most reasonable interpretation of current data is that embryonic stem cell-based therapies are not likely to have clinical application for heart disease. At the preclinical level, there is overwhelming evidence that various stem/progenitor cells can improve cardiac function after myocardial infarction. The mechanism remains unclear, but it does not involve regeneration of new myocytes; rather, transplanted cells work via secretion of factors that act in a paracrine fashion. The use of repeated dosing is particularly important because mounting evidence shows that the efficacy of cell therapy is underestimated if only one dose is used. In the clinical arena, heart failure and refractory angina appear to be the main target populations. The use of the intravenous route for cell delivery may fundamentally transform the field of cell therapy. Regarding cardioprotection, this field has stalled, and no interventions have proven efficacious in ST-segment Elevation Myocardial Infarction despite myriad clinical trials. Gene therapy is a promising approach that has not yet been tested adequately in the clinical arena. This lecture critically examines the current state of clinical research on cell-based therapies, cardioprotection and gene therapy, highlighting the controversies in the field, improvements in clinical trial design and the application of new strategies.
Harpal Buttar
The global rise in diabetes, obesity, cardiovascular diseases (CVDs), and healthcare costs is alarming. Consequently, there is an urgent need to identify cost-effective strategies for the prevention of CVDs, diabetes and obesity. Consuming healthful foods rich in flavonoids, polyphenols and carotenoids—such as fresh fruits and vegetables—and probiotics/prebiotics, along with reducing intake of salt, sugar-sweetened beverages and saturated fats; quitting smoking; reducing excessive drinking and engaging in moderate exercise for 30 min a day, are collectively recognized as effective in preventing CVDs. This comprehensive approach is likely the most cost-efficient for promoting health and preventing chronic conditions such as cancer, diabetes, obesity, CVDs and neurodegenerative diseases. Strong evidence supports that a Mediterranean-type diet, which includes whole grains, omega-3 fatty acids, poultry, fish, nuts, seeds, olive oil, dairy products, limited red meat and moderate red wine consumption, is associated with lower mortality and morbidity from CVDs. Additionally, recent studies indicate that dietary supplements containing flavonoids, carotenoids and antioxidants can influence gene and protein expression, altering metabolic pathways and maintaining homeostasis, thus reducing the risk of chronic diseases with complex causes. The positive effects of plant-derived bioactive compounds and their metabolites are due to their combined antioxidant and anti-inflammatory properties. Probiotics, prebiotics and synbiotics contribute to health by fostering healthy gut microbiota, enhancing gut endothelial integrity and strengthening the body's immune function.
Michael Czubryt
Cardiac fibrosis is a clinically important contributor to heart failure and arrhythmias, leading to an increased risk of death, typically in response to stressors such as myocardial infarction or pressure overload. Despite many years of research, no therapies currently exist to directly attenuate cardiac fibrosis, and new approaches are required. For fibrosis to occur, resident cardiac fibroblasts undergo a phenotypic conversion to myofibroblasts, which secrete large quantities of extracellular matrix proteins and contribute to cardiac remodelling. Blocking fibroblast activation is thus a promising means to stop, and possibly reverse, cardiac fibrosis. We have shown that scleraxis is required for cardiac fibroblast activation and that, as a transcription factor, it works by transactivating a suite of pro-fibrotic genes that are responsible for the myofibroblast phenotype, including alpha-smooth muscle actin, periostin and collagen 1 alpha 2. In addition, scleraxis controls the expression of glutaminase 1, the rate-limiting enzyme of glutaminolysis, which appears to be a required energy-producing pathway for myofibroblasts. Our data have shown that blockade of scleraxis by knockout or knockdown prevents the activation of pro-fibrotic genes as well as glutaminase 1, interfering with fibroblast activation. Drug-mediated blockade of glutaminolysis has also shown promising impacts on fibroblast activation.
Supported by Canadian Institutes of Health Research.
Naranjan S. Dhalla
Although different antiplatelet agents are used for the prevention of thrombosis and treatment of ischaemic heart disease, very little information regarding the therapeutic potential of these agents in heart failure is available. We have investigated the effects of some antiplatelet agents, such as sarpogrelate (SAR) and cilostazol (CIL) treatments, on cardiac dysfunction, cardiac remodelling and subcellular defects in heart failure due to myocardial infarction. Heart failure in rats was induced by occluding the coronary artery for 8 weeks, and the drug treatment was started 4 weeks after inducing myocardial infarction. Marked depression in cardiac output and ejection fraction and increases in heart rate, left ventricle (LV) thickness and LV volume in the infarcted animals were attenuated by SAR and CIL. Alterations in myofibril Ca2+-ATPase, as well as myosin isozyme contents and gene expression in the failing heart were reduced by SAR and CIL. Likewise, changes in sarcoplasmic reticular Ca2+ uptake and release activities, Ca2+ pump and Ca2+ release protein content as well as their mRNA levels were attenuated by both drug treatments. These results provide evidence that both SAR and CIL delay the progression of heart failure and improve cardiac function by attenuating cardiac remodelling, subcellular defects and abnormalities in cardiac gene expression. It is suggested that antiplatelet agents may prove to be a viable therapy for the treatment of heart failure. (Infrastructure support for this study was provided by the St. Boniface Hospital Foundation.)
Sanjiv Dhingra
Allogeneic (unrelated donor) mesenchymal stem cells (MSCs) and induced pluripotent stem cells (iPSCs) are being tested in animal studies and clinical trials for cardiac regeneration and repair. The outcome of initial studies was very encouraging, and transplanted stem cells were safe after transplantation in the recipient heart. However, poor survival of transplanted stem cells in the infarcted heart has impaired the clinical translation of stem cell-based therapies. We have performed investigations to understand the mechanisms of poor survival of transplanted stem cells in the heart. We found that allogeneic stem cells after transplantation in the ischaemic heart turned immunogenic and were subsequently rejected by the host immune system. In our ongoing studies, we are developing biomaterial-based strategies to prevent rejection of transplanted stem cells in the heart. We synthesized and characterized titanium carbide (Ti3C2) MXene quantum dots (MQDs). The immunomodulatory MQDs selectively reduce activation of CD4+IFN-γ+ T-lymphocytes and promote the expansion of immunosuppressive CD4+CD25+FoxP3+ regulatory T-cells in an activated lymphocyte population. We also incorporated MQDs into a chitosan-based hydrogel to create a 3D platform for stem cell delivery to the heart. This composite immunomodulatory hydrogel-based platform improved survival of stem cells and mitigated allo-immune responses. We also found that MQDs have the potential to mitigate allograft vasculopathy and prevent rejection of transplanted organs. These studies highlight the potential of MXene-based next-generation biomaterials for cardiac regenerative medicine.
Transverse (t)-tubules are important for the rapid and synchronous rise in calcium, which brings about the contraction of cardiac myocytes. In the atrial, t-tubules are almost completely lost at the point of heart failure. We set out to determine if it is possible to restore t-tubules to cardiac myocytes following their loss in heart failure and if their restoration can restore the calcium transient. Heart failure was induced by rapid ventricular pacing in sheep. Atrial t-tubules were almost entirely lost, and the calcium transient amplitude decreased. In a subset of sheep, rapid pacing was terminated, and the animals were allowed to recover for 5 weeks. Following recovery from heart failure, atrial t-tubule density was restored, but t-tubules were structurally disordered. Despite their disorder, we show recovered t-tubules were able to trigger calcium release. The recovery of atrial t-tubules restored the L-type calcium current, the calcium content of the sarcoplasmic reticulum and the calcium transient. Our data suggest that myotubularin, a lipid phosphatase, is important for t-tubule restoration.
Funded by British Heart Foundation.
Dragan Djuric
Folic acid, a synthetic form of folate (vitamin B9), has been associated with several cardioprotective effects, primarily in reducing non-proteinogenic amino acid homocysteine level (epigenetic biomarker linked to an increased risk of cardiovascular, neurodegenerative disease and cancer when elevated) and metabolizing into other amino acids. Moreover, folate is involved in methylation reactions, which are crucial for regulating gene expression, involved in DNA synthesis and repair, and maintaining cardiovascular health. Folic acid effects have been studied in homocysteine-dependent (induced hyperhomocysteinaemia, isoprenaline-induced myocardial injury) and homocysteine-independent (monocrotaline-induced heart failure, streptozotocin-induced diabetes mellitus) rat models of cardiometabolic diseases for their potential cardioprotective effects on coronary flow, endothelial function, oxidative stress, inflammation and activities of cardiometabolic enzymes. Folic acid has been found to reduce homocysteine and glucose levels when elevated, exhibit vasodilator and antioxidant properties, improve endothelial dysfunction and affect cardiac, hemostatic and hepatorenal biomarkers; cardiac oxidative stress and cardiometabolic enzyme activities. Individual responses to folic acid may vary, and these effects also depend on the applied dose of folic acid, the method of application and the used experimental model of cardiometabolic disease. It is important to note that while folic acid supplementation may help lower homocysteine levels, its direct impact on reducing the risk of cardiovascular events like heart attack, and stroke is still a subject of ongoing research and debate. However, research on its direct cardioprotective effects is ongoing, and it is often recommended as part of a balanced diet rather than as a standalone treatment for heart disease.
Supported by The Ministry of Science, Technological Development and Innovation of the Republic of Serbia.
Đorđe Đukanović1,2, Relja Suručić2, Milica Gajić Bojić1,3, Ranko Škrbić1,3,4,5, Žarko Gagić2
The prevalence of hypertension has been steadily increasing over the past three decades, necessitating the constant development of novel treatments for this condition. Plants from the Mediterranean cuisine, such as oregano, have a history of traditional use in hypertension treatment. Carvacrol, a major constituent of essential oils from the Lamiaceae family, is known for its vasorelaxant properties and potential hypotensive effects. Therefore, this study aims to investigate the role of Transient Receptor Potential Ankyrin 1 (TRPA1) channels in carvacrol-induced vasodilation and to design novel compounds based on the structure of carvacrol with enhanced activity. An isolated tissue bath methodology was used to investigate the role of TRPA1 in carvacrol-induced vasodilation. Different in silico approaches were combined in the design and evaluation of novel TRPA1 agonists. The selective TRPA1 antagonist A967079 significantly reduced carvacrol-induced vasodilation of isolated human mesenteric arteries. A reliable 3D-QSAR model with good prediction accuracy (R2 = 0.83; Q2 = 0.59; R_pred^2 = 0.84) was created, and the obtained results were used to design novel TRPA1 agonists. Predicted pEC50 activities of these compounds were higher (4.996–5.235) compared to the experimental pEC50 of 4.77 for carvacrol. All created molecules had satisfying predicted Absorption, Distribution, Metabolism, Excretion, Toxicity (ADMET) properties, with six of them having better characteristics than carvacrol. Molecular docking studies demonstrated that the designed molecules bind to the same amino acid residues of the TRPA1 channel as carvacrol, with eight compounds showing lower binding energies. Molecular dynamics simulations revealed the good stability of carvacrol-TRPA1 and D27-TRPA1 complexes. Conclusion: Activation of TRPA1 channels is partly responsible for the vasorelaxant effect of human mesenteric arteries caused by carvacrol. Based on the results from various in silico studies, compound D27 exhibits the most promising predicted characteristics as a potential novel TRPA1 agonist regarding its activity, ADMET profile, interactions with residues and stability in complex with the TRPA1 channel.
This work was supported by the Ministry of Scientific and Technological Development and Higher Education of Republic of Srpska: 19.032/961-28/23.
Alicia D'Souza
There is a circadian rhythm in the normal heart rhythm (in heart rate and PR, QRS and QT intervals) as well as in the incidence of bradyarrhythmias, atrial fibrillation, atrioventricular block, ventricular fibrillation and sudden cardiac death. This lecture summarizes our current understanding of the underlying mechanisms: The oldest explanation is that cardiac circadian rhythms are the result of an acute post-translational regulation of cardiac ion channels by the autonomic nervous system under the control of the suprachiasmatic nucleus, but, paradoxically, there is little or no evidence of this. Instead, there is a plausible suggestion that rhythms in the transcription of cardiac ion channels driven by local circadian clocks in the heart and circadian rhythms in plasma catecholamines and glucocorticoid (all ultimately under the control of the suprachiasmatic nucleus) are involved. Understanding these fundamental processes could potentially highlight new ways of treating cardiac arrhythmias and novel ‘chronotherapeutic’ strategies – for example, we have recently shown that a glucocorticoid receptor blocker can prevent the morning increase in ventricular arrhythmia susceptibility.
This work was support by the British Heart Foundation Intermediate Fellowship (FS/19/1/34035), British Heart Foundation Project grant (PG/22/10919), British Heart Foundation Programme Grant (RG/18/2/33392) and a Fondation Leducq TNE award (‘FANTASY’ 19CV03).
Péter Ferdinandy1,2,3
Despite nearly four decades of intensive research, drug treatment for myocardial infarction and subsequent post-infarction heart failure remains an unmet clinical need, possibly due to its complex molecular mechanism. Therefore, targeting a molecular network rather than a single molecular drug target may be a more effective strategy for cardioprotective therapy. An unbiased, comprehensive analysis of gene expression patterns in normal and protected ischaemic myocardium may lead to the exploration of transcriptomic and proteomic molecular networks to identify novel molecular targets for cardioprotection. The non-coding oligonucleotides microRNAs (miRNAs) targeting multiple mRNAs form a dynamic transcriptomic molecular network. Targeted perturbation of the molecular network by miRNA oligonucleotide compounds is a promising approach for the treatment of diseases of complex molecular mechanisms. By a systematic unbiased analysis of the transcriptomic network of healthy, infarcted, as well as protected by ischaemic conditioning heart samples, we have discovered endogenous cardioprotective miRNAs (e.g. miR125b-3p, miR-450a-3p) and validated them
Rodolphe Fischmeister1, Rima Kamel1, Aurélia Bourcier1, Jean Piero Margaria2, Valentin Jin1, Audrey Varin1, Agnès Hivonnait3, Françoise Mercier-Nome4, Delphine Mika1, Alessandra Ghigo2, Flavien Charpentier3, Vincent Algalarrondo1, Emilio Hirsch2, Grégoire Vandecasteele1, Jérôme Leroy1
Intracellular cAMP levels produced upon β-AR stimulation are counterbalanced by the degradation of the cyclic nucleotide by enzymes called phosphodiesterases (PDEs). Our past studies showed that PDEs not only terminate the activation of cAMP effectors but also compartmentalize this second messenger in discrete subcellular compartments. Among the six PDE families expressed in cardiomyocytes to degrade cAMP, we recently focused our interest on PDE2 for several reasons. First, PDE2 hydrolytic activity is stimulated 10 to 30-fold by cGMP via binding to the regulatory N-terminal GAF-B domain, making PDE2 a unique integrator of cAMP/cGMP signals. Second, using selective PDE2 inhibitors, we showed that PDE2 inhibits cardiac L-type Ca2 channel activity in various species, including humans, and controls excitation-contraction coupling. Third, in contrast to PDE3 and PDE4, the two main enzymes degrading cAMP in the heart, whose expression and activities are generally decreased in pathological hypertrophy and heart failure (HF), PDE2 is increased in HF to blunt β-AR/cAMP signals, suggesting a protective mechanism against increased circulating catecholamines. Fourth, transgenic (TG) mice with constitutive cardiac overexpression of PDE2 exhibit improved cardiac function and are protected against catecholamine-induced arrhythmias after ischaemic injury.
In an attempt to further delineate the impact of PDE2 upregulation on cardiac function and its putative beneficial effects in HF, we performed gene therapy with adeno-associated virus serotype 9 (AAV9) allowing preferential cardiac overexpression of PDE2. This strategy allows expression from a definite time point rather than constitutive cardiac expression as previously employed. With this strategy, we demonstrate
Thus, our results demonstrate that increasing PDE2 activity is beneficial in HF. They suggest that a gene therapy with PDE2 in cardiac cells, or PDE2 activators yet to be discovered, could be of therapeutic value.
This work was supported by the Leducq Foundation for Cardiovascular Research (19CVD02), ERA-CVD ‘PDE4HEART’, ANR-23-CE14-0051-01, Fédération Française de Cardiologie, and Fondation Lefoulon-Delalande.
Ferenc Gallyas1, Robert Halmosi2, Kalman Toth2
Cardiovascular diseases are the leading cause of morbidity and mortality worldwide, imposing significant health and economic burdens on individuals, families and societies. Accordingly, a substantial effort is exerted to find novel, effective therapeutics for these diseases. Recently, inhibitors of the poly(ADP-ribose) polymerase (PARPi), originally developed for cancer therapy, have shown promise for repurposing in cardiovascular diseases. These drugs' cytoprotective, antioxidant and anti-inflammatory properties can be pivotal in treating heart conditions characterized by tissue damage and inflammation. Studies suggest that PARPi could protect cardiac cells from ischaemia-reperfusion injury and reduce adverse remodelling post-myocardial infarction. Additionally, their anti-inflammatory properties might mitigate atherosclerosis progression. Although prolonged administration of PARPi could lead to malignant transformation due to DNA damage accumulation, the short-term application of these drugs may be beneficial in post-infarct cardiac remodelling. We are presenting experimental evidence for the use of PARPi in the aforementioned conditions and providing mechanistic background for the protective effects of PARPi, including the effects of these drugs on protein kinase B/Akt and mitogen-activated protein kinase signalling.
Ricardo J. Gelpi1,2, Pablo Evelson2,3
Environmental pollution is one of the biggest problems facing the modern world because it affects the health of virtually all living beings on the planet, increasing morbidity and mortality. In this chapter, we have described the effects of acute and chronic pollution on the lungs and heart, as well as three hypotheses that could explain how polluting particles reach different organs, especially the lungs and heart. Polluting particles reach the lungs through the respiration of living beings, where they activate pro-inflammatory cytokines, which reach the heart, producing damage to the mitochondria that increases the formation of free radicals and decreases the synthesis of ATP. This mechanism leads to a situation of myocardial ischaemia whose end point is myocardial infarction. This can occur with or without coronary obstruction; obviously, the presence of the latter aggravates the situation. Acute pollution produces cellular and molecular changes that lead to alterations in important cellular structures. Chronic pollution over time produces structural remodelling of the lungs and heart, leading to cardiac pathologies that, due to pressure and volume overload, modify the geometry of the organs. In summary, whether due to acute or chronic exposure to polluting particles, we are in the presence of an important risk factor for cardiovascular health that increases the incidence of myocardial infarction and heart failure in patients exposed to breathing severely polluted air. Although there is no specific treatment to reduce the effects of pollution, experimental evidence suggests that chronic exercise could be beneficial and improve the living conditions of these patients.
This work was supported by the University of Buenos Aires - CONICET – ANPCYT.
Eva Goncalvesova
Heart failure with reduced ejection fraction (HFrEF) remains a serious and highly relevant issue in cardiology. Due to advances in pharmacological and device-based treatments, a growing number of patients are experiencing favourable changes in left ventricular (LV) geometry, accompanied by improvements in LV ejection fraction (LVEF). This process is referred to as LV reverse remodelling (LVRR), and patients meeting certain criteria are classified as having heart failure with improved ejection fraction (HFimpEF). If the symptoms and signs of heart failure disappear, we may speak of heart failure remission. An important fact is that these favourable structural changes in the myocardium are associated with better clinical outcomes. In patients with HFimpEF, an improved quality of life, better exercise tolerance, lower risk of heart failure-related hospitalizations and a reduced risk of sudden cardiac death have been observed. Given these positive clinical indicators, LVRR has become a key therapeutic goal in patients with HFrEF. The occurrence of reverse remodelling has been observed in approximately 22% of HFrEF cases. The likelihood of LVRR depends on heart failure aetiology, clinical characteristics, laboratory tests and imaging findings. Generally, patients with shorter disease duration, non-ischaemic aetiology, smaller LV end-diastolic dimensions, a lower percentage of late gadolinium enhancement (LGE) on cardiac MRI, higher global longitudinal strain (GLS), and good tolerance to pharmacological therapy are more likely to experience LVRR. Personalized prediction offers an opportunity for individualized patient management, including decisions on the timing and implementation of device therapy. In conclusion, LVRR not only becomes one of the main therapeutic targets in patients with HFrEF, but investigating the mechanisms and predictors of LVRR also opens up possibilities for potential new treatment options for these patients.
Robert Hatala1,2
Structural heart disease is widely recognized as the cause of most abnormalities in the electrical activity of the heart. However, persistent cardiac electrical abnormalities are frequently not the consequence but rather the ignored cause or an important additional etiological factor in the development of chronic heart failure (HF). Such electrical disturbances comprise persistent arrhythmias and/or left bundle branch block (LBBB). Therefore, electrically induced cardiomyopathy (EI-CMP) can be defined as a potentially reversible myocardial disease in which persistent arrhythmia and/or LBBB causes left ventricular systolic dysfunction (LVSD), which is phenotypically manifested as HF with reduced LV ejection fraction (HFrEF).
Experimental studies on animals exposed to rapid atrial or ventricular pacing were instrumental in gaining insight into the pathophysiological mechanisms of LVSD and, ultimately, HF. Tissue, myocyte, and electrical remodelling are observed in both animal models and human hearts with EI-CMPs. A decline in LV function is linked to impaired calcium handling dynamics and basal ATP activity. The remodelling process is characterized by a number of key features, including fibrosis, inflammation, apoptosis, expansion of the extracellular matrix, myocyte disarray, mitochondrial dysfunction and prolonged ventricular repolarization.
In humans, potential etiological factors for EI-CMP include:
Atrial fibrillation/flutter (irrespective of ventricular rate) Persistent tachycardia (supraventricular/ventricular), Ventricular ectopy with a burden >15%, Spontaneous or iatrogenic (as with RV pacing) LBBB with subsequent LV dyssynchrony.
The majority of electrical abnormalities with the potential to induce or exacerbate heart failure with reduced ejection fraction (HFrEF) are treatable by catheter ablation or resynchronization therapy (CRT) utilizing biventricular pacing or pacing of the conduction system. When applied in a timely manner, these interventions can result in a complete or partial reversal of LVSD. Therefore, ensuring an accurate and timely diagnosis, followed by appropriate therapy, is crucial for the prognosis of patients with EI-CMP.
Gerd Heusch
The clinical outcome of patients with an acute myocardial infarction who undergo reperfusion therapy by primary percutaneous coronary intervention is determined by the final infarct size. However, coronary microvascular injury is also increasingly appreciated as a decisive determinant of clinical outcome, beyond that of infarct size. In fact, a recent definition and grading of infarct severity encompasses not only cardiomyocyte death but also coronary microvascular injury. The mechanisms of coronary microvascular injury in the perfusion territory of a coronary artery with plaque rupture or plaque erosion are multifold: (a) microembolization of plaque debris with superimposed thrombotic material from the atherosclerotic lesion; (b) release of vasoconstrictor, pro-thrombotic and pro-inflammatory substances from the atherosclerotic lesion; (c) increased vascular permeability and oedema formation, which acts back by compression on the microcirculation; (d) platelet and platelet-leukocyte aggregates; (e) erythrocyte stasis; (f ) capillary constriction by pericytes; (g) endothelial shedding and capillary obstruction; (h) capillary rupture and intramyocardial haemorrhage. Many interventions that protect from myocardial ischaemia/reperfusion and reduce infarct size also attenuate coronary microvascular injury, including ischaemic conditioning protocols and cardioprotective drugs. The only intervention that appears to specifically protect the coronary microcirculation is angiopoietin-like 4.
This work was supported by the German Research Foundation (CRC 1116 B8, RTG 2989), EU Cost Action CARDIOPROTECTION and METAHEART.
Marketa Hlavackova1, Daniel Benak1,2, Blanka Holendova3, Frantisek Papousek1, Kristyna Holzerova1, Miloslava Chalupova1,2, Frantisek Kolar1, Stepanka Benakova3,4, Lydie Plecita3, Gabriela Pavlinkova5
Diabetes and diabetic cardiomyopathy are among the leading causes of morbidity worldwide, and their prevalence is still increasing. Diabetes is associated with tissue hypoxia and also results in the dysregulation of the hypoxia-inducible factor 1α (HIF-1α) signalling pathway, potentially contributing to the development of cardiomyopathy. The pathogenesis of diabetic cardiomyopathy involves changes in gene expression profiles, in which epigenetic modifications of RNA (epitranscriptomics) may play a role. In this study, we aimed to investigate the role of HIF-1α in the alteration of proteomic profiles in pancreatic islets and myocardial tissue, with a particular focus on epitranscriptomic regulators, following the induction of type 2 diabetes mellitus and the progression of diabetic cardiomyopathy. To achieve this, we utilized a Hif1a ± mouse model, where diabetic cardiomyopathy was induced through a combination of metabolic (high-fat diet; HFD) and hypertensive stress (L-NAME-induced inhibition of NO synthase) over 8 weeks. Diabetic animals developed cardiac hypertrophy and showed a reduction in cardiac output. Glucose tolerance tests, along with insulin assays, demonstrated the expected increases in glycaemia and insulin levels following HFD + L-NAME treatment in both wild-type and Hif1a ± mice. However, these levels were significantly higher in Hif1a ± mice compared to wild-type mice. Proteomic analysis revealed that the cardiac proteomic profile was influenced by both HFD + L-NAME treatment and HIF-1α deficiency, while pancreatic islets exhibited relatively homogeneous proteomic profiles across the experimental groups. Epitranscriptomic regulators were altered in a distinct pattern in both the heart and pancreatic islets in response to HFD + L-NAME treatment in both wild-type and Hif1a ± mice. This research contributes to the complex mosaic of diabetes research, offering new insights into the molecular basis of diabetic cardiomyopathy and highlighting the critical role of HIF-1α and epitranscriptomic regulation in the pathogenesis of this disease.
This work was supported by the National Institute for Research of Metabolic and Cardiovascular Diseases (Programme EXCELES, ID Project No. LX22NPO5104) – Funded by the European Union – Next Generation EU and the Grant Agency of the Czech Republic, grant No. 24-10497S.
Loredana N. Ionica1,2, Darius G. Buriman2,3, Adina V. Linta2,3, Raluca Sosdean4,5, Ana Lascu2,3,5, Caius G. Streian5,6, Horea B. Feier5,6, Adrian Sturza2,3, Danina M. Muntean2,3
Sodium-glucose co-transporter 2 inhibitors (SGLT2i), a novel class of glucose-lowering medications, have revolutionized the management of heart failure (HF) across the ejection fraction (EF) spectrum, regardless of the presence of diabetes. Extensive experimental and clinical evidence has unequivocally documented the cardiovascular benefits of SGLT2i, which extend far beyond glycaemia control and are supported by several ‘off-target’ effects. Oxidative stress is a central pathophysiological mechanism that disrupts cardiomyocyte homeostasis in the vast majority of cardiometabolic diseases, but the sources of reactive oxygen species (ROS) are far from being fully elucidated. Monoamine oxidase (MAO), with two isoforms MAO-A and B, contributes to cardiovascular oxidative stress by continuously producing ROS at the outer mitochondrial membrane. This study, performed in overweight, non-diabetic patients with chronic HF and the entire range of EF with indication of elective cardiac surgery, was aimed at assessing whether the SGLT2i included in HF guidelines, empagliflozin and dapagliflozin, decrease atrial MAO expression and alleviate oxidative stress elicited by exposure to angiotensin II (AII) and high glucose (HG). Right atrial appendages were incubated ex vivo with either empagliflozin or dapagliflozin (1, 10 μM, 12 h) in the presence or absence of AII (100 nM) and HG (400 mg/dL) and used for the assessment of MAO-A and B gene and protein expression (by qPCR and immunofluorescence) and oxidative stress (by FOX and dihydroethidium assays). Stimulation with AII and HG increased atrial expression of both MAOs and augmented ROS production. The effects were mitigated by both concentrations of the SGLT2i. Atrial oxidative stress is positively correlated with the echocardiographic parameters of heart cavities and negatively with left ventricular EF. In conclusion, in non-diabetic overweight patients with HF, MAO contributes to the cardiac oxidative stress in conditions that mimic the renin-angiotensin system activation and hyperglycaemia. MAO expression was mitigated with empagliflozin and dapagliflozin, as a novel ‘off-target’ class effect of the SGLT2i.
Vladimir Jakovljevic1,2,3
Usnic acid (UA), a natural dibenzofuran derivative found in numerous lichens, has gained considerable research interest due to its broad spectrum of biological activities. It holds promise for therapeutic use, including antimicrobial, anticancer and antioxidant properties. Despite its long-standing use in traditional medicine and well-established benefits, there is a distinct lack of research on the cardioprotective effects of UA. This study aims to evaluate the effects of UA on doxorubicin-induced cardiotoxicity in a rat model.
UA was isolated from the acetonic extract of the lichen
José Jalife
Sudden cardiac death causes around 300,000 adult deaths each year in Europe. Ventricular tachycardia/fibrillation (VT/VF) is an important immediate cause of sudden cardiac death. Electrical tornados (reentrant waves, rotors) spinning at high frequency and generating turbulent electrical activity in the ventricles are the origin of VT/VF. When these tornados occur, the heart is unable to effectively pump blood; the only treatment is a powerful electrical shock applied to the ventricles. Cellular and molecular studies may offer an alternative by explaining how tornados form, how they generate electrical turbulence and how they can be annihilated. We have identified a groundbreaking membrane macromolecular complex known as a ‘channelosome’, which contains NaV1.5, the channel responsible for the sodium inward current (INa), and Kir2.1, the strong inward rectifier potassium channel responsible for (IK1). Through their channelosome, these critical cardiac ion channels reciprocally modulate each other's cell surface expression and function, which enables them to rigorously control electrical excitability and impulse conduction velocity in the ventricles. The consequences of Kir2.1-NaV1.5 channelosome dysfunction are manifold: the resting membrane potential is depolarized, excitability is reduced, action potential upstroke velocity is reduced, and action potential duration and QT interval are prolonged. These proarrhythmic changes promote the formation of electrical tornados and are key in their maintenance. Such new knowledge provides strong evidence to consider macromolecular complexes and ion channel interplay in the development of antiarrhythmic therapeutic strategies to prevent rotor formation and sudden cardiac death in patients with heart failure and other cardiac diseases. Sudden cardiac death poses significant medical and economic challenges to society. These exciting new concepts offer a novel paradigm for the understanding of cardiac electrical function. Most importantly, they may lead to advanced targeted therapy based on the rational design of new small molecules or peptides that promote channelosome location and function at the cell membrane as a more effective alternative for preventing life-threatening ventricular arrhythmias in many patients.
This work was funded by the La Caixa Banking Foundation (project code LCF/PR/HR19/52160013); grants PI20/01220 and PI23/01039 by Instituto de Salud Carlos III (ISCIII); MCIU grants BFU2016-75144-R and PID2020-116935RB-I00, co-funded by FEDER; Fundación La Marató de TV3 (736/C/2020); European Union's Horizon 2020 [GA-965286]. CNIC is ISCIII, the Ministerio de Ciencia e Innovación and the Pro CNIC Foundation and is a Severo Ochoa Center of Excellence (grant CEX2020-001041-S funded by MICIN/AEI/10.13039/501100011033).
Nevena Jeremic1,2,3, Marina Nikolic2,4, Vladimir Jakovljevic2,3,4
This study examined the effect of combined valsartan/sacubitril administration on the browning promotion in healthy rats and rats with metabolic syndrome (MS). Using in silico studies, the signalling pathways by which the applied treatment induces browning were identified. The study included 24 Wistar Albino rats, grouped into healthy and MS-induced animals, further divided into two subgroups depending on the applied treatment. Animals from the experimental groups were treated orally with valsartan/sacubitril (Entresto) suspension (68 mg/kg) for 4 weeks. After the animals were sacrificed, biochemical parameters were determined, while three types of adipose tissue (retroperitoneal, epididymal and inguinal) were used for pathohistological and PCR analysis with adipocyte density and surface area assessment. Valsartan/sacubitril treatment had multiple positive effects on improving glycoregulation, insulin levels and lipid status. Besides antioxidant and anti-inflammatory effects, the applied treatment promotes browning by increasing UCP-1 expression as the main thermogenic biomolecule. The special contribution of this study is reflected through elucidation of the mechanisms by which valsartan/sacubitril induces browning and primarily refers to mTORC1 activation as a common target molecule through which this dual pharmacological therapy induces UCP-1 expression in adipose tissue of animals with MS.
Norbert Jost
Cardiovascular diseases, and in particular cardiac arrhythmias such as ventricular fibrillation, play a leading role in mortality statistics in developed countries. Accordingly, cardiac arrhythmias have become a major area of cardiovascular research. Drug therapy has traditionally been the main treatment for ventricular and supraventricular arrhythmias. It is therefore important to understand the mechanism of action of antiarrhythmic drugs at the organ, tissue, cellular and subcellular levels in order to develop new, more effective, less proarrhythmic agents. One such potential development direction is the development of a series of amiodarone-like antiarrhythmic agents in which repolarization-inducing (class III) activity is combined with other pharmacological properties (combined class III +I/B, II and/or IV activity) that are considered to be beneficial. Therefore, we investigated the antiarrhythmic and cardiac electrophysiological effects of a novel antiarrhythmic compound (SZV-2649), similar to amiodarone but lacking the benzofuran chemical structure, in rat and canine cardiac cell line cardiac preparations. SZV-2649 exerted antiarrhythmic effects on coronary occlusion reperfusion-induced ventricular fibrillation in rats and acetylcholine and burst stimulation-induced atrial fibrillation in dogs. SZV-2649 inhibited hERG/IKr and GIRK/IK,ACh currents in expressed HEK293 cell lines and
This work was supported by the National Research Development and Innovation Office (NKFIH K 135464, K 142738, K 147212, FK 138223 and TKP2021-EGA-32, as well as GINOP-2.3.2.-15-2016-00006 and GINOP-2.3.2.-15-2016-00040), the Ministry of Human Capacities Hungary (EFOP-3.6.2-16-2017-00006), the Albert Szent-Györgyi Medical School institutional grant (SZTE AOK-KKA 2021, SZGYA 2021 and SZTE AOK-KKA 2022), Hungarian Research Network (HUN-REN TKI project to A.V., N.J. and I.B.) and the Hungarian Academy of Sciences (bo_481_21).
Lorrie Kirshenbaum
Cytokines such as TNFα have been implicated in cardiac dysfunction and toxicity associated with doxorubicin (DOX). While TNFα can elicit different cellular responses, including survival or death, the mechanisms underlying these divergent outcomes in the heart remain cryptic. The E3 ubiquitin ligase TRAF2 provides a critical signalling platform for K63-linked ubiquitination of RIPK1, crucial for NF-κB activation by TNFα. Whether alterations in TNFα-TRAF2 signalling underlie the cardiotoxic effects of DOX, remains poorly understood.
To investigate TRAF2 signalling in the pathogenesis of DOX cardiotoxicity.
Using a combination of
In contrast to vehicle-treated mice, severe ultrastructural defects, including cytoplasmic swelling, mitochondrial perturbations and elevated TNFα levels, were observed in the hearts of mice treated with DOX. While investigating the involvement of TNFα in DOX cardiotoxicity, we discovered that in the absence of DOX, NF-κB was readily activated by TNFα. However, TNFα-mediated NF-κB activation was impaired in cardiac myocytes treated with DOX. This coincided with the loss of K63-linked polyubiquitination of RIPK1, attributed to the proteasomal degradation of TRAF2. Further, TRAF2 protein expression was markedly reduced in the hearts of cancer patients treated with DOX. Impaired TRAF2 signalling resulted in the activation of Bnip3 and mitochondrial perturbations, including disrupted bioenergetics, loss of membrane potential and permeability transition pore opening. We further established that the reciprocal actions of the ubiquitinating and deubiquitinating enzymes c-IAP1 and USP19, respectively, regulated the proteasomal degradation of TRAF2 in DOX-treated cardiac myocytes. Importantly, an E3 ligase mutant of c-IAP1 (c-IAP1 H588A) or gain of function of USP19 prevented proteasomal degradation of TRAF2 and DOX-induced cell death. Further, wild-type TRAF2, but not a RING finger mutant defective for K63 ubiquitination of RIPK1, restored NF-κB signalling and suppressed DOX-induced cardiac cell death. Finally, cardiomyocyte-restricted expression of TRAF2 (AAV9-TRAF2)
Supported by Canadian Institute for Health Research.
Petra Kleinbongard
Cardioprotective strategies aim to preserve the myocardium from ischaemia/reperfusion injury and reduce infarct size and its consequences. Various stimuli delivered at sites remote from the heart (remote conditioning) activate molecular self-protection mechanisms in the target organ, the heart. Remote conditioning, that is ischaemic remote preconditioning (RIC) through short repeated cycles of ischaemia/reperfusion at a limb, as well as non-invasive electrical stimulation of the ear (ATS) through periodic very short electrical stimulation impulses of the heart, have been established in experimental studies and successfully transferred to patients. Remote conditioning is a systemic reaction. There are three levels of remote conditioning and the resulting cardioprotection: (I) the stimulus level; (II) the transmission level, neuronal and humoural mediators transmit the protective signal from the periphery to the heart and (III) the target level, receptor activation and intracellular signal transduction ultimately influence myocardial protection, as observed in various animal models and in humans/patients. Novel findings on signal transduction at different levels are highlighted: for example, the spleen as a relay organ for cardioprotective signal transduction and the non-neuronal cholinergic cardiac system at the target level.
Peter Krenek, Katarina Lelkova, Jana Veteskova, Eva Velasova, Gabriel Doka, Lenka Bies Pivackova, Diana Vavrincova, Jan Klimas
Endothelin-1 (ET-1) plays a critical role in pulmonary arterial hypertension (PAH) and is currently targeted by ET-1 receptor antagonists. However, strategies aimed at reducing cellular ET-1 expression have been less thoroughly explored. In preliminary experiments, we targeted multiple signalling pathways to identify those regulating endothelin-1 gene (
Supported by APVV-19-0458 and APVV-23-0502.
Branislav Kura1, Patricia Pavelkova1, Daria Kornieieva1, Lucia Kindernay1, Kristina Ferenczyova1, Jakub Strapec1, Barbora Kalocayova1,2, Margita Pobijakova3, Jan Slezak1
Radiation-induced heart disease (RIHD) is a known harmful side effect of mediastinal radiotherapy applied in oncological diseases. RIHD involves structural and functional abnormalities of the pericardium, coronary vessels, myocardium, valves and conduction system. The underlying pathological mechanisms are complex and mainly related to endothelial cell damage, oxidative stress and inflammation. These can cause cardiomyocyte disorders, tissue fibrosis and ultimately heart failure. To overcome these complications, it is necessary to look for specific therapeutic interventions, which are still lacking. Studies have shown that molecular hydrogen (H2) possesses radioprotective effects in different experimental models, including RIHD. H2 could be effective in alleviating RIHD through various mechanisms, for example selective neutralization of hydroxyl radicals, protection against inflammatory and apoptotic damage, antifibrotic and antihypertrophic effects, etc. Available information shows that microRNAs (miRNAs) might be involved in the radioprotective effect of H2. In this work, our research team investigated the effect of 6-weeks of H2 administration to 1-year-old Wistar rats irradiated in the mediastinal area with a single dose of 10 Gy (4–5 Gy/min). The results show that the inhalation of H2 effectively reduced heart damage (lactate dehydrogenase), improved oxidative stress (malondialdehyde, superoxide dismutase activity) and inflammatory (pro-inflammatory cytokines) parameters, as well as had a positive impact on heart function (infarct size, left ventricular end-diastolic pressure) parameters measured 6 weeks after irradiation. This study also reveals that H2 administration on the RIHD rat model also normalizes dysregulated expression of selected miRNAs (miRNA-21, -30e, -208 and miRNA-142). Based on these results, we conclude that H2 administration represents a promising treatment strategy for patients undergoing mediastinal irradiation with radiation-induced cardiac damage. However, to clarify other mechanisms of H2 action and to verify the effectiveness of H2 therapy in clinical practice, further studies are needed, especially focusing on the still unclear mechanism of its biological effect.
This work was funded by grants VEGA 2/0092/22 and APVV-19-0317.
Gary Lopaschuk
Alterations in cardiac energy metabolism are an important contributor to heart failure severity. Despite this, there is considerable confusion as to what changes in cardiac energy metabolism occur in heart failure with preserved ejection fraction (HFpEF). To assess this, we measured cardiac fatty acid oxidation and glucose oxidation in a mouse model of HFpEF [which involved administering 13-month-old C57BL/6N female mice a 60% high-fat diet and L-NAME (0.5 g/L/day) for 8 weeks to induce HFpEF]. We observed an increase in fatty acid oxidation in isolated working hearts from HFpEF mice, which was accompanied by a marked decrease in cardiac glucose oxidation. An increased reliance of the heart on fatty acid oxidation as a source of ATP production results in a decrease in cardiac efficiency (cardiac work/O2 consumed). Our goal was therefore to inhibit fatty acid oxidation in HFpEF mice as an approach to normalize cardiac energy metabolism. These studies were performed in a mouse model of diabetic cardiomyopathy (DbCM), which displays HFpEF symptoms and has high cardiac fatty acid oxidation rates and decreased glucose oxidation rates. A significant contributor to the increased fatty acid oxidation observed in DbCM hearts is the weakening of the regulatory control of mitochondrial fatty acid oxidation by malonyl-CoA. We therefore treated DbCM mice with an inhibitor of malonyl-CoA decarboxylase (MCD), which increases malonyl-CoA in the heart. A 4-week treatment with the MCD inhibitor, CBM-30011 (10 mg/kg/day), resulted in a significant decrease in cardiac fatty acid oxidation and a parallel increase in glucose oxidation in DbCM mice. An improvement in diastolic function was also observed in the DbCM mice (as assessed by echocardiographic E/e⊠). We conclude that in HFpEF mice the heart switches from glucose oxidation to fatty acid oxidation as the main source of ATP production. However, inhibiting fatty acid oxidation can increase the contribution of glucose oxidation to ATP production and lessen the severity of HFpEF.
Supported by CIHR Foundation Grant.
Lukáš Mach1,2
Heart failure is an important cause of mortality and morbidity globally and carries a significant economic burden. The advances in pharmacotherapy have led to improved outcomes for patients with heart failure. However, there remain patients who do not respond to the treatment; their condition deteriorates and ultimately results in untimely death or heart transplantation. Heart failure resulting from varied aetiologies is characterized by heterogeneous patterns of cardiac tissue remodelling that are not adequately reflected in the current therapies. Addressing this heterogeneity will be key for improving outcomes. Single-cell RNA sequencing allows addressing this unmet clinical need by studying transcriptomic phenotypes of individual cells contained in heart tissue in health and in disease at a large scale. Spatial transcriptomic technologies allow the localization of cells in niches, which enables inferences about cell-cell interactions. In patients with advanced heart failure, we have been able to interrogate the cell type abundance shifts and perturbations to cellular gene expression programmes that associate with heart failure in general but also with individual aetiologies and genotypes. With refined understanding of gene activation heterogeneity in heart failure, we will be able to validate putative disease mechanisms that may be amenable to therapeutic targeting.
This work was supported by The British Society for Heart Failure Research Fellowship, the British Heart Foundation Clinical Research Training Fellowship, the Rosetrees Trust and Alexander Jansons Myocarditis UK.
Marek Michalak
The endoplasmic reticulum (ER) plays a central role in cellular stress responses via mobilization of ER stress coping responses, such as the unfolded protein response (UPR). Activation of UPR is associated with multiple cardiac conditions and has been shown to be either protective or detrimental to the heart. The serine/threonine protein kinase/endoribonuclease (IRE1α) is the most evolutionarily conserved ER stress sensor and a key UPR stress signal transducer. Impaired calcium homeostasis activates ER stress responses, resulting in severe cardiac fibrosis that is prevented by the inhibition of IRE1α. There are two pools of IRE1α in cardiac muscle cells: one is localized to perinuclear ER responding to cellular stresses and the other is at the junctional sarcoplasmic reticulum (SR), a subspecialized membrane system involved in the regulation of excitation-contraction (E-C) coupling. The junctional SR containing ryanodine receptor/calcium channel forms membrane contacts with the T-tubules containing the L-type calcium channel. Deletion of the exons 20 and 21 of the
This work was funded by the Canadian Institutes for Health Research.
Ali. J. Marian
The nuclear genome is exposed to numerous internal and external stressors, which affect the structure and function of DNA. It is estimated that each mammalian cell develops 104–105 lesions in the nuclear genome every day. The stressors cause DNA oxidation, alkylation and cross-linking, among others, which disturb the delicate homeostatic balance that maintains genomic stability. The DNA lesions also affect the replicative function of the DNA and RNA transcription. A consequence of the replicative and transcriptional stress is the generation and accumulation of double-stranded breaks (DSBs) in the DNA, which are the most dangerous form of DNA lesions. The genomic stressors are increased in pathological conditions such as heart failure. Likewise, the DNA repair mechanisms are impaired. Collectively, these changes lead to the accumulation of DNA lesions, including DSBs. The increased DSBs in the nucleus activate the nuclear component of the DDR, including the tumour protein 53 (TP53) and its downstream target cyclin-dependent kinase inhibitor 1A (CDKN1A), leading to cell cycle arrest and cell death. Likewise, a subset of the DSBs are released into the cytosol, where they are sensed by the DNA-sensing proteins, leading to activation of the nuclear factor-kappa B1 (NFkB1) and interferon regulatory factor 3 (IRF3) and consequent expression of the senescence-associated secretory phenotype (SASP). Expression of SASP contributes to the pathogenesis of heart failure by inducing senescence, inflammation, cell cycle arrest and cell death. Genetic blockade of the cytosolic or nuclear DNA imparts salubrious effects in the experimental models of heart failure. The stage is set to test potential pharmacological inhibitions of these pathways, initially in large animals and subsequently in humans with heart failure.
This work was supported by the National Institutes of Health; National Heart, Lung and Blood Institute (Awards R01 HL151737 and R01 HL132401) and National Institute of Aging (R01AG082751).
Cristina Molina
Atrial fibrillation (AF) is the most common sustained arrhythmia in the clinical setting, which is associated with significant morbidity and mortality. AF is also associated with severe cellular calcium (Ca2+)-handling abnormalities in atrial myocytes that may contribute to the initiation and maintenance of the arrhythmia. This so-called remodelling is supposed to involve abnormal phosphorylation of key Ca2+-handling proteins. In addition, previous studies suggest that impaired phosphodiesterase (PDE)-dependent regulation of spatiotemporal distribution of cAMP may contribute to Ca2+-handling abnormalities associated with AF. We investigate how PDE8 controls the spatiotemporal response of intracellular cAMP in key relevant Ca2+-handling nanodomains by measuring real-time cAMP, ICa, L, spontaneous Ca2+ releases from the sarcoplasmic reticulum, protein expression and localization using highly sensitive and specifically targeted cAMP biosensors, patch-clamp, confocal Ca2+ imaging, contractility measurements and biochemical experiments.
Supported by DFG, Heisenberg programme, Gertraud und Heinz Rose Stiftung.
Martin Morad, Noemi Toth, Xiao-hua Zhang
While calcium signalling of SR by Ca2 transporters of the surface membrane (ICa and INCX) has been well studied, possible regulation of mitochondrial and nuclear Ca2 signalling by these proteins remains not fully explored. Here we investigate whether there are privileged interactions between Ca2 transporters of surface membrane and mitochondria, SR or nuclear envelope (NE), by comparing their Ca2 activity with cytosolic Ca2+, using genetically engineered fluorescent probes GCamP3/GCamP6 targeted to mitochondria, nuclear Lamin B, and SR lumen. In whole-cell clamped and TIRF- or confocal-imaged cardiomyocytes, caffeine-triggered SR Ca2 release caused rapid rise in cytosolic calcium (△F/F ~0.5: fura-2), followed by delayed and slower mitochondrial Ca2 uptake (△F/F =~1–2: mito-GCamp6), and much larger NE Ca2 uptake (△F/F ~10: LaminB-GCaMP6). Calcium-Induced Calcium Release (CICR)-triggered SR Ca2 release induces Ca2 uptake by NE that follows and outlasts cytosolic Ca2 signal significantly. Long-duration application of 5 mM caffeine to empty SR of its calcium content first transiently increases mitochondrial calcium, then triggers mitochondrial pacing despite the absence of SR calcium release. Lowering Na concentration in Ca2+-depleted SR increased the baseline calcium concentrations of SR, mitochondria and NE, enhancing the mitochondrial pacing rate. Spontaneous pacing was rapidly suppressed by blocking Ica but not by suppressing SR calcium uptake with CPA. We suggest that while activation of CICR is critical in triggering SR Ca2 release that replenishes NE Ca2 and initiates spontaneous pacing, calcium influx on NCX regulates mitochondrial pacing that sustains autorhythmicity in the absence of SR calcium release.
Danina M. Muntean1,2, Adrian Sturza1,2
Overweight and obesity represent serious health concerns whose prevalence and severity have been constantly rising worldwide. The global threat of the obesity pandemic is related to the high risk of cardiometabolic comorbidities/complications. Increased body fat leads to vascular and adipose tissue dysfunction mediated via increased oxidative stress and chronic systemic inflammation. Overwhelming experimental and clinical studies have reported the association of vitamin D deficiency with metabolic and cardiovascular pathologies, including chronic venous disease. Monoamine oxidases (MAOs) with two isoforms, A and B, are mitochondrial enzymes that catalyse the deamination of biogenic amines and neurotransmitters with the constant generation of hydrogen peroxide in both vessels and adipose tissue. We here present evidence that (i) suboptimal vitamin D status is associated with increased reactive oxygen species in visceral adipose tissue and vascular samples (mesenteric artery branches and varicose vein explants), (ii) serum levels of vitamin D are inversely correlated with the magnitude of local oxidative stress, (iii)
This research supported by the university internal funds awarded to the Centre for Translational Research and Systems Medicine.
Norbert Nagy1,2,3, Noémi Tóth1, Gergő Bitay1, Leila Topal1, Jenő Pintér4, Alexandra Polyák4, Zsófia Kohajda2, Zoltán Husti1, Naveed Muhammad1, Aiman Mohammed1, Norbert Jost1,2,3, László Virág1,3, István Baczkó1,3, Attila Farkas4, András Varró1,2,3
Regular physical exercise has well-known beneficial effects on health; however, increasing evidence indicates that extreme sports activities – although very rare – can induce various cardiac arrhythmias, such as atrial fibrillation or even sudden cardiac death. A significant number of sudden cardiac deaths among athletes have been attributed to ischaemic origins or other deviations revealed by autopsy. However, the underlying cause of the remaining cases remains unclear. Cardiac alternans indicate a periodic, regular oscillation of the action potential (short-long pattern) and parallel intracellular Ca2 release (large-small pattern). Alternans have been clearly associated with the development of ventricular fibrillation and sudden cardiac death. However, the potential role of cardiac alternans in the ‘athlete heart’ is unknown. In this study, the potential role of alternans was assessed in arrhythmia development in trained dogs. Beagle dogs underwent a vigorous 16-week long training program and were compared to a sedentary group. Cardiac action potentials were measured by the conventional microelectrode technique from right ventricular tissue samples. Ionic currents were recorded by the whole-cell configuration of the patch-clamp technique. Ca2 transients were monitored by employing fluo-4AM fluorescent dye. All experiments were performed at 37°C. Trained dogs exerted increased susceptibility to ventricular fibrillation as a response to burst pacing. At the cellular level, the Ito was downregulated in trained animals. Action potential alternans in endocardial and midmyocardial tissue layers were increased in trained animals. Purkinje-ventricle dispersion during alternans was found to be larger in trained dogs. Ca2 transient alternans were enhanced in trained animals. Ca2+ content of the sarcoplasmic reticulum was decreased in exercised animals, while cell shortening was identical between groups. Alternans may contribute to the development of ventricular fibrillation in intensive training-associated arrhythmias. The underlying mechanism of alternans could be SERCA downregulation in trained animals.
Supported by NKFIH-OTKA-142949.
Shigeo Ohta
Molecular hydrogen (H2) has emerged as a promising therapeutic and preventive medical gas. In 2007, we overturned the conventional wisdom that H2 has no effect on mammalian cells, leading to birth to the field of ‘hydrogen medical biology’. H2 has many advantages, including high efficacy, lack of toxic effects, and diverse functions such as antioxidant, anti-inflammatory, anti-cell death, and energy metabolism stimulation. In addition, H2 is beneficial not only for severe chronic and acute diseases but also for healthy people. The target molecule of H2 was identified as the oxidized form of porphyrin, which acts as a catalyst to stimulate the selective reaction of H2 with hydroxyl radicals. Various types of porphyrins are widely and abundantly distributed throughout the body. Therefore, the repair of oxidized porphyrins by H2 may bring multiple benefits to various cells. H2 suppresses free radical chain reactions and modifies signalling mediators involved in lipid peroxidation. We present the findings from animal experiments to clinical trials. The best example of H2's effectiveness is recovery after cardiac arrest. Inhalation of H2 gas has reduced the mortality rate by more than half and more than doubled the number of patients discharged without sequelae. The molecular mechanics will be discussed. The future mission of H2 medicine is to apply H2 for treatment, prevention, and quality of life across a wide range of fields. Furthermore, the use of H2 in agriculture has the potential to produce safe, productive and high-quality foods. H2 medicine could be practiced with simple and low-cost treatments without adverse effects and contribute to many unsolved serious problems facing today's society. H2 has the potential to address a wide range of issues, including cardiac arrest, cerebral infarction, metabolic syndrome, advanced cancer, inflammatory cytokine storm, Alzheimer's dementia, healthcare, beauty and agriculture.
Petr Ostadal
Cardiogenic shock is a critical circulatory condition in which a severely failing cardiac pump is unable to generate sufficient cardiac output, leading to tissue hypoperfusion in vital organs. Despite advances in intensive cardiology care, mortality from cardiogenic shock remains high. During the past years, mechanical circulatory support, including extracorporeal membrane oxygenation (ECMO), has become a standard therapy for cardiogenic shock with the aim of bridging the most critical conditions. Current evidence supporting the use of ECMO (and other mechanical circulatory support systems) in cardiogenic shock remains insufficient. The initiation of ECMO leads to rapid restoration of total circulatory output, blood pressure and tissue perfusion. On the other hand, it may be associated with a possible negative effect on the already severely damaged left ventricle as well as several specific complications. Furthermore, the haemodynamic effect of ECMO also depends on the cause of cardiogenic shock and modulating factors, such as the presence of valvular heart disease (e.g. aortic stenosis or mitral regurgitation) or mechanical complications (e.g. ventricular septal defect). The negative effect of ECMO could be at least partially prevented by synchronized pulsatile extracorporeal flow. Randomized clinical trials have reported a neutral effect of the use of ECMO on clinical outcomes in cardiogenic shock. However, these trials had significant limitations, particularly in the inclusion criteria. Several recently published analyses indicate that detailed haemodynamic examination allows for the identification of subjects who may benefit from mechanical support. Currently, numerous clinical questions remain unanswered, and further research in this field is fundamentally needed.
This work was supported by the Charles University Research Program ‘Cooperation Cardiovascular Sciences’ and Ministry of Health, Czech Republic – Conceptual Development of Research Organization – MH CZ DRO, Motol University Hospital, Prague, Czech Republic, 00064203.
Bohuslav Ostadal
Experimental and clinical studies have clearly demonstrated significant sex differences in myocardial structure and function, both under physiological and pathological conditions. The best example is significant sex differences in cardiac tolerance to ischaemia/reperfusion injury: pre-menopausal adult female hearts are more resistant than male myocardium. The importance of these findings is supported by the fact that the number of studies dealing with this issue has increased significantly in recent years. Detailed molecular and cellular mechanisms responsible for sex differences are yet to be elucidated; however, it has been stressed that the differences cannot be explained only by the effect of oestrogens. In recent years, a promising new hypothesis has been developed, suggesting that mitochondria may play a significant role in the sex differences in cardiac tolerance to oxygen deprivation. However, one thing is clear already today: sex differences are so important that they should be taken into consideration in clinical practice for the selection of the optimal diagnostic and therapeutic strategy in the treatment of ischaemic heart disease. The present review attempts to summarize the progress in cardiovascular research on sex-related differences in cardiac tolerance to oxygen deprivation during the last 40 years, that is from the first experimental observation. Particular attention was paid to the sex-related differences of the normal heart, sex-dependent tolerance to ischaemia-reperfusion injury, the role of hormones and, finally, to the possible role of cardiac mitochondria in the mechanism of sex-dependent differences in cardiac tolerance to ischaemia/reperfusion injury.
This study was supported by the Charles University Research Program ‘Cooperation Cardiovascular Sciences’; Ministry of Health, Czech Republic–Conceptual Development of Research Organization, Motol University Hospital, Prague, Czech Republic (No. 00064203); Czech Science Foundation (project No. 24-10497S); project National Institute for Research of Metabolic and Cardiovascular Diseases (Programme EXCELES, ID project No. LX22NPO5104) – Funded by the European Union – Next Generation EU.
Zoltán Papp1, Beáta Bódi1, Fruzsina Sárkány1, István Baczkó2, Róbert Sepp3, Dániel Priksz4, Miklós Fagyas1, Attila Tóth1
Mutations in genes encoding certain myocardial proteins result in hypercontractility of the left ventricle (LV) and are thought to drive pro-hypertrophic signalling in hypertrophic obstructive cardiomyopathy (HOCM). Accordingly, targeting the myosin heavy chain-β by small molecules appears to be an attractive approach to prevent the development of HOCM. Indeed, clinical evidence now supports that modulation of the myosin motor can be an effective treatment option for well-defined HOCM patient groups. Moreover, the potential benefit of myosin inhibitors has also been implicated for patients with heart failure with preserved ejection fraction (HFpEF) in the absence of known genetic alterations. However, it is unclear if cardiomyocytes on different genetic backgrounds respond identically to myosin inhibitors or not. Here, we tested the contractile effects of aficamten, a second-generation myosin inhibitor agent, in cardiac preparations of healthy rodents, dogs and humans. Moreover, these effects were also contrasted to those obtained in permeabilized cardiomyocytes isolated from HOCM patients following myectomy. Aficamten reduced maximal Ca2+- activated tensions (Fmax) and evoked robust rightward shifts in the Ca2+ sensitivities of isometric force production of permeabilized cardiomyocytes of all sources. Nevertheless, aficamten-induced Ca2+ desensitizations were higher in HOCM cardiomyocytes than those in cardiomyocytes of healthy animals or humans. Our results illustrate largely comparable contractile effects upon myosin inhibitor applications, irrespective of the myosin isoform composition or the presence or absence of mutated myocardial proteins in cardiomyocytes of different species. These data also implicate that pro-hypertrophic signalling can also be limited to similar degrees by aficamten in cardiomyocytes on different genetic backgrounds.
This work was supported by a grant from the National Research, Development and Innovation Office (K147173 to ZP). Project no. K147173 has been implemented with the support provided by the Ministry of Culture and Innovation of Hungary from the National Research, Development and Innovation Fund, financed under the K_23 ‘OTKA’ funding scheme.
Inna Rabinovich-Nikitin1,2, Huong Nguyen1,2, Erele Tzidon1,2
Pregnancy is a dynamic process associated with significant physiological changes in the cardiovascular system. During pregnancy, there is an increased metabolic demand on the mother and foetus to ensure adequate uteroplacental circulation for foetal growth and development. Insufficient haemodynamic changes can result in maternal and foetal changes to the cardiovascular system. Shift work during pregnancy results in significant changes to the circadian rhythm of both mother and foetus. Furthermore, shift work is linked to increased risk for developing cardiac disease. Therefore, engaging in shift work during pregnancy may adversely affect the cardiovascular health of the mother and have negative implications for the offspring, including increased risk for cardiovascular disease, obesity, and diabetes. While the link between shift work and negative pregnancy outcomes, such as miscarriage, preterm birth and reduced foetal growth, has been previously established, the outcomes of pregnancy on cardiovascular health in shift working mothers are not well understood. Herein, we utilize a night shift work model in mice and provide new evidence that shift work during pregnancy results in changes in genes controlling endothelial function and vascular regulation, coincident with increased cardiac haemodynamic demand. Furthermore, in silico analysis revealed that genes responsible for hemodynamic and endothelial functions are regulated by the circadian genes Clock and BMAL1. The downregulation of these genes following shift work leads to failed vascular response and insufficient metabolic supply to the maternal-foetal system, resulting in placental insufficiency and restricted foetal growth. Our findings provide the first evidence for a direct link between haemodynamic function and circadian regulation during pregnancy. Understanding the key genes and networks that are associated with circadian rhythms and cardiac haemodynamics during pregnancy may contribute to developing new therapeutic strategies to mitigate the adverse effects of shift work during pregnancy.
Arnela Saljic
Cardiac adipose tissue plays a crucial role in maintaining healthy cardiac function. It communicates with the myocardium through paracrine signalling and provides free fatty acids for beta-oxidation. However, an increase in cardiac adipose tissue has been associated with a higher risk of arrhythmias. This risk is linked to the release of adipokines, which signal fibroblasts to increase fibrosis and alter ion channel profiles in cardiomyocytes. These changes can lead to prolonged action potentials and re-entry circuits. Additionally, intramyocardial adipocytes can act as conduction blocks, slowing down conduction velocity and further increasing the risk of arrhythmias. In this study, we examine the adipose tissue of human atrial appendages in terms of fibrosis, adipose tissue volume and adipocyte size. The left and right atrial appendages were collected from 40 patients undergoing cardiac surgery at Rigshospitalet, Copenhagen (Denmark). Formalin-fixed and paraffin-embedded tissue was sectioned at 4 μm and stained with Picro Sirius Red. The sections were imaged using the AxioScanZ1 and subjected to automated intelligent image segmentation using the QuPath software in order to quantify fibrosis, adipose tissue and adipocyte size. We found that the content of fibrosis was significantly higher in the right atrial appendage (54.3%) compared to the left (35.9%) (
M. Saadeh Suleiman
Patients with occlusive coronary artery disease can be treated by coronary artery bypass surgery (CABG) using cardiopulmonary bypass (CPB) and cardioplegic arrest. However, cardioplegic arrest renders the heart ischaemic and susceptible to reperfusion injury. This injury is augmented by the effects of CPB, which can be largely avoided by performing surgery on a beating heart. However, despite extensive clinical research, adopting beating heart surgery remains controversial. Therefore, an approach to reduce ischaemia reperfusion (I/R) injury associated with cardioplegic arrest remains a major target. A significant number of additives to cardioplegia have thus far failed to translate to clinical practice. Several early studies reported cardioprotection by the general anaesthetic propofol against I/R. Our research aimed to investigate whether propofol cardioplegia is cardioprotective when used during CPB. Work in our unit started in 1996 and showed that the addition of propofol to cardioplegia is protective during cold or warm arrest and in hearts from different species and with different pathologies. Furthermore, CPB surgery on a pig model showed cardioprotection with propofol. Inhibition of the mitochondrial permeability transition pore seems to be the mechanism. Preparations for clinical research started in 2007/8. This involved approval by several regulatory and health agencies/committees. A detailed description of the trial has been published (1). Patient recruitment started in 2010 and finished in 2012. The primary analysis showed that cardiac injury using propofol (6 μg/mL) cardioplegia was not statistically different from that of the intralipid vehicle (2). This inconclusive outcome (
Supported by NIHR, BHF, NHRF, BRC and BRU.
Miloš P. Stojiljković, Ranko Škrbić, Sonja Marinković, Žana M. Maksimović, Tanja Šobot, Zorislava Bajić, Sanja Jovičić
Autonomic imbalance is one of the key components in the pathophysiology of heart failure (HF), with an increase in the sympathetic tone and a decrease in parasympathetic activity. Acetylcholinesterase (AChE) inhibitors constitute a large and chemically diverse group of compounds that inhibit AChE. Some of them are used to treat myasthenia gravis, such as pyridostigmine (PYR) or Alzheimer's disease (such as rivastigmine and donepezil), and they have been shown to exhibit some favourable cardiovascular effects. By using the isoprenaline (ISO) model of chronic HF (two doses of 85 mg/kg sc, 24 h apart), we aimed to investigate the potential protective effects of PYR, a peripheral AChE inhibitor. It was administered to Wistar rats in drinking water for 2 weeks at a daily dose of 20 mg/kg, while the control rats drank plain tap water instead. It was shown that PYR caused the attenuation of maladaptive cardiac remodelling, which was noted in histopathological preparations of the myocardium. PYR treatment also led to the preservation of ejection fraction (EF). In the untreated group, EF decreased significantly (from 68.71% to 53.20%), while in the PYR-treated group, EF remained stable (68.76%
Thus, by attenuating the decline in systolic function of the left ventricle and slowing the maladaptive cardiac tissue remodelling, PYR represents a promising agent in the treatment of HF.
Supported from intramural sources of the Faculty of Medicine, University of Banja Luka.
Zuzana Sumbalová1, Jarmila Kucharská2, Zuzana Rausová2, Anna Gvozdjáková3, Mária Szántová4, Branislav Kura5, Ján Slezák5
Metabolic dysfunction-associated steatotic liver disease (MASLD) is characterized by increased fat accumulation in liver tissue and is recognized as a hepatic manifestation of metabolic syndrome. Mitochondrial dysfunction and oxidative stress play significant roles in the progression of MASLD. Molecular hydrogen (H2) has been recognized as a medical gas with antioxidant, anti-inflammatory and antiapoptotic effects, and it can also stimulate energy metabolism. This study aimed to evaluate the effect of adjuvant therapy with hydrogen-rich water (HRW) on platelet mitochondrial bioenergetics and endogenous coenzyme Q10 (CoQ10) levels in patients with MASLD. The study included 30 patients with MASLD and 15 healthy volunteers. Over 8 weeks, 17 patients (H2 group) drank water (3 330 mL/day) enriched with HRW tablets (>4 mg/L H2), while 13 patients (P group) drank water with placebo tablets producing CO2. Mitochondrial bioenergetics in platelets isolated from whole blood was assessed using high-resolution respirometry. The concentration of CoQ10-TOTAL in platelets and plasma was determined by HPLC, and the oxidative stress parameter TBARS in plasma was measured spectrophotometrically. Patients with MASLD exhibited relatively higher CI-linked LEAK respiration, lower CI-linked OXPHOS, and reduced CII-linked ET capacity compared to the control group, along with lower CoQ10 concentrations. Plasma TBARS concentration was higher in the H2 group. After 8 weeks of HRW adjuvant therapy, CoQ10 concentration in platelets increased, plasma TBARS decreased, and the impaired parameters of platelet mitochondrial respiration improved. Our results support the evidence that mitochondria are the primary target of H2 therapy. The application of H2 appears to be a promising new strategy for the targeted treatment of mitochondrial disorders. Additional and longer-term studies are needed.
This work was supported by grants APVV-19-0317, VEGA 2/0063/18, 2/0092/22, 2/0148/22 and HRW Natural Products Inc.
Paramjit Tappia1, Mohammed Moghadasian2, Bram Ramjiawan3
It is now well established that obesity is a global epidemic and a major health hazard. Obesity is known to contribute to an increase in the risk of cardiovascular disease. Unhealthy dietary practices affect human health, and excessive fructose intake has been linked to metabolic syndrome and the occurrence of obesity and concomitant cardiac dysfunction. This study was undertaken to test the hypothesis that long-term consumption of a high fructose (HF) diet induces cardiac abnormalities during obesity in metabolic syndrome. Accordingly, our investigations, conducted in male SD rats fed an HF (60% w/w) diet plus a moderate level of saturated fat (10% w/w lard) for up to 20 weeks, revealed that systolic blood pressure and serum levels of insulin, triglycerides and total cholesterol were significantly increased in the HF fed group. Furthermore, an increase in visceral fat mass and reduced levels of the anti-inflammatory and anti-atherosclerotic cytokine, IL-10, were observed. Others have demonstrated an increase in cardiomyocytes size, left ventricular wall thickness and reduced ejection fraction and fractional shortening subsequent to high-fructose feeding of mice for 12 weeks. These data suggest that long-term consumption of HF can lead to cardiovascular dysfunction. Thus, an effective intervention to reduce HF intake may attenuate the risk for obesity-related morbidities.
The infrastructure support for this study was provided by the St. Boniface Hospital Albrechtsen Research Centre.
Belma Turan
Novel cardioprotective antidiabetic medications include treating patients with dual or multiple receptor agonists to have better cardiovascular benefits with consistently reduced major adverse cardiac events, improvement in heart failure, and diminished cardiovascular and all-cause mortality. Especially in recent years in this field, intensive studies have suggested that active incretin hormone glucagon-like peptide-1 amide (GLP-1) and its analogs have cardioprotective effects by modulating cardiomyocyte metabolism. In addition, a glucose-dependent insulinotropic polypeptide (GIP) is a hormone produced in the upper gut and supports the physiological and pharmacological relevance of GIP in metabolic disorders. Tirzepatide (TZPD), as a new generation dual agonist of GIP and GLP-1 receptors, has been shown to have metabolic effects in type 2 diabetic (T2DM) patients, as well as acting as an unbalanced and disproportionate agonist of GIP and GLP-1 receptors, and thereby showing a biased effect on GLP-1 receptors both in experimental animal studies and in cultured cells. In addition to the fact that TZPD administration affects the expression levels of these 2 receptors. It has been suggested that TZPD has a “biased pharmacologic” effect on GLP-1 receptors, and thus TZPD administration with a biased pharmacologic effect has the potential to affect other targets while activating some specific target proteins. Moreover, it has been proposed that GLP-1R activation in pancreatic beta-cells stimulates insulin secretion and activates adenylate cyclase (AC)-mediated cAMP production, cellular Ca2+-regulation, and modulates Gas protein activation. Although there are no clear studies to demonstrate a direct target effect of TZPD administration on heart function, the current data on pancreatic beta-cells strongly suggest a similar pathway is also possible in cardiomyocytes associated with its action. However, it is well accepted that off-target activity is the biological activity of a drug that is different from and not at that of its intended receptor. In some cases, off-target activity can be taken advantage of for therapeutic purposes. Therefore, we aimed to investigate both on-target effects and possible target effects of this drug in cardiomyocytes by considering its action in cAMP signaling. We first demonstrated its two receptors (GIP and GLP-1 receptors) localizations in cardiomyoblast (H9c2 cell line) and responses to TZPD application (40 nM). There was a significantly high response to the TZDP application in GIP-R under hyperglycemia and hyperinsulinemia mimicking cells compared to the GLP-1R. Furthermore, this application also induced a significant increase in the depressed cAMP level parallel to the increases in the expression levels of both depressed β1- and β2-adrenergic receptors. Moreover, the TZDP application significantly decreased the overexpressed β3-adrenergic receptor level in hyperglycemia and hyperinsulinemia mimicking cells parallel to a decrease in increased cGMP level. Our study overall demonstrated that a dual-action receptor agonist antidiabetic drug TZDP can provide both on-target receptor action and an off-target action via the cAMP signaling pathway in cardiomyoblasts under pathological stimuli such as in hyperglycemia and hyperinsulinemia, while both types of actions can include important therapeutic approaches fort he heart dysfunction under pathological conditions.
This study was supported by The Scientific and Technological Research Council of Turkiye (No. 124Z217).
Suresh Tyagi
Although the double and triple hits (i.e., hypertension, diabetes/diet/obesity, ageing) contribute to HFpEF, the contribution of a single-hit high-fat dysbiotic diet (HFD) to HFpEF is unclear. To test whether the HFD causes HFpEF, we administered HFD to normal wild-type (WT, C57) mice and measured HFpEF. Previous studies from our laboratory have observed gut dysbiosis instigating disruption in epigenetic memory (i.e. rhythmic methylation/de-methylation cycle on H3K4, K9, H4K20) via folate 1-carbon metabolism (FOCM) pathways by gene writer (DNMT), eraser (TET/FTO) and homocysteine (Hcy). We observed a link between mitochondrial TCA cycle and gene writer, eraser, and Hcy. A disruption in recycling Hcy back to methionine and impaired clearance by transsulfuration—specifically, mitochondrial sulfur metabolic pathways involving 3MST, CBS, and CSE—leads to a reduction in mitochondrial bioenergetics, ATP, OCR, H2S, PGC1α, and TFAM, along with an increase in the fission/fusion ratio, ultimately instigating cardiac dysfunction. The hypothesis is that the HFD causes epigenetic dysregulation and contributes to cardiac dysbiosis by HHcy and a decrease in H2S promoting HFpEF. To determine whether the lactobacillus lowers homocysteine and increases H2S, bidirectionally producing folate and lactone/ketone, we created a chronic dysbiotic environment by utilizing WT and DNMT1KO mice, treated w/o HFD and w/o lactobacillus. The
Zoltan Varga1,2,3
Immune checkpoint molecules are physiological regulators of the adaptive immune response. Immune checkpoint inhibitors (ICIs), such as anti-PD-1 or anti-CTLA-4 monoclonal antibodies, have revolutionized the treatment of cancer, and their clinical application is rapidly increasing, as reflected by thousands of ongoing clinical trials. However, ICIs cause various immune-related side effects, including acute and chronic cardiovascular toxicities. Among these, ICI-induced acute fulminant myocarditis is the most studied direct cardiotoxicity, although emerging clinical and preclinical data suggest that other ICI-related chronic cardiac toxicities are important, such as accelerated atherosclerosis, heart failure, arrhythmias and additional forms of cardiac dysfunction. These chronic cardiovascular toxicities are often hidden, as they may only occur in the presence of preexisting injuries (ischaemic necrosis, hypertrophy) and comorbidities (metabolic disease). It is therefore of high clinical importance to identify the risk factors and comorbidities that may precipitate ICI-induced cardiotoxicity. The mechanisms of ICI-induced myocarditis and cardiac dysfunction appear to be distinct: myocarditis develops as a specific autoimmune response against cardiac myocytes involving expansion of autoreactive CD8 T cells to cardiac self-antigens (e.g. against alpha myosin heavy chain), whereas cardiac dysfunction/heart failure following ICI therapy is associated with disturbances in cardiac transcriptomic and metabolic effects, likely due to increased expression of pro-inflammatory cytokines. The occurrence of these profound effects on the heart suggests a possible role for immune checkpoint molecules in the maintenance of cardiovascular homeostasis, and therefore, the disruption of physiological immune checkpoint signalling may lead to cardiac diseases, including heart failure.
Antonio Zaza
Ino-lusitropic therapy would be highly beneficial in heart failure (HF), even at its early stages, but it is underexploited because of the undesirable effects (mainly proarrhythmia) of currently available agents. In the early 2000, we came across a Na+/K+ pump blocker, named PST2744, in which we detected an ancillary SERCA2a stimulatory effect. After a thorough bench characterization, this compound, now known as istaroxime, has been tested in large animal models and patients with advanced heart failure, showing ino-lusitropic effects and an excellent safety profile (no proarrhythmia). Since then, we have synthetized and validated a series of derivatives that overcome istaroxime limitations: 1) lack of selectivity for SERCA and 2) pharmacokinetics suitable for acute use only. The outcome of this endeavour is a toolbox of ‘pure’ SERCA2a stimulators suitable for chronic HF treatment. My talk will go through the development process, what we have learned from it and where it is going.
This work was supported by the Windtree Therapeutics, INC. (
Michal Zeman1, Lubos Molcan1, Iveta Herichova1, Hana Mauer Sutovska1, Valentina Rumanova1,2, Monika Okuliarova1
Periods of biological rhythms in the cardiovascular system (CVS) range from milliseconds to days. Disruption of rhythmicity can have fatal consequences, and understanding physiological and molecular mechanisms is necessary to prevent and treat chronodisruption. Circadian rhythms (about 24 h) are important in CVS functioning. The disrupted daily blood pressure (BP) profile (non-dipping) can increase cardiovascular risk, but underlying mechanisms are not understood. We showed that hypertensive patients with the non-dipping BP profile had lower nighttime melatonin levels than dippers. However, the melatonin profile was not suppressed in transgenic rats with the upregulated renin-angiotensin-aldosterone system [TGR(mRen-2)27], which is accepted as the animal model of non-dipping. Aldosterone concentrations were upregulated, the rhythmicity of sympathetic activity was phase-shifted and expression of clock genes in the area postrema, a sensory transducer between blood and brain, was in antiphase in TGR compared to controls. Artificial light at night (ALAN) has increased dramatically, and epidemiological studies suggest its adverse effects on health, which can be mediated by disruption of circadian rhythms. We explored the consequences of ALAN on cardiovascular parameters in normotensive and spontaneously hypertensive rats. Control rats were kept in a light:dark cycle of 12:12, and ALAN groups were exposed to dim light (~2 lux) during the night. Heart rate, BP and locomotor activity were measured by telemetry, and gene expression in the heart was quantified by qPCR. The power of circadian oscillations in BP and heart rate decreased, and the rhythmic pattern of clock and clock-controlled genes and selected metabolic genes was disturbed by light at night. ALAN eliminated rhythmic expression of Nos3, Pkc1ɛ and glucocorticoid receptors and suppressed amplitude in Pdk4 and Pparγ daily rhythms. Results suggest that ALAN attenuates circadian rhythms in CVS, disrupts the expression of genes controlling heart metabolism and interferes with the ability of the heart to anticipate and compensate for altered workload.
This work was supported by APVV-21-0223 and VEGA 1/0309/23.
Katarina Andelova1, Matus Sykora1, Veronika Farkasova1, Tatiana Stankovicova2, Tamara Egan Benova1, Barbara Szeiffova Bacova1, Vladimir Knezl1, Michal Pravenec3, Narcis Tribulova1
Poor adherence to lifestyle changes and antihypertensive drugs increases the risk of malignant arrhythmias and heart failure in chronic hypertension. Structural remodelling and altered Cx43 in the myocardium promote instability and dysfunction, but it is unclear if these changes are limited to the left ventricle (LV) or differ by sex. The aim of this study was to investigate key factors involved in the development of malignant arrhythmias and progression to heart failure in both the left and right ventricles of wild-type and hairless spontaneously hypertensive rats (SHR) males and females acclimated to ambient temperature. Notably, 6-month-old male and female wild SHR, hairless SHR, and normotensive Wistar strains were housed at 22°C. Biometric, electrocardiographic, and echocardiographic parameters were recorded. LV and right ventricle (RV) tissues were used for Cx43 immunolabeling, protein level analysis of Cx43, protein kinase C-ɛ (PKC-ɛ), protein kinase C-δ (PKC-δ), and extracellular markers (transforming growth factor-β (TGF-β), SMAD2/3, and matrix metalloproteinase-2 (MMP-2). Susceptibility to electrically inducible ventricular fibrillation (VF) was tested ex vivo in perfused hearts. Corrected QT interval was prolonged in male SHR strains compared to Wistar rats, with a lesser increase in hairless SHR. Ejection fraction was reduced in wild SHR males but not in hairless SHR compared to normotensive Wistar rats. Cx43 downregulation and abnormal topology were seen in the LV of wild SHR males and to a lesser extent in hairless SHR but not in females. Cx43 levels and topology in the RV were consistent across strains and sexes. Phosphorylated Cx43 at ser368 was reduced in the LV but not in the RV of SHR males. MMP2, PKC-ɛ, and PKC-δ levels were higher in the RV compared to the LV in both SHR strains. Hairless SHR males and females had reduced susceptibility to inducible VF compared to wild SHR males. Findings indicate that hypertension-induced pro-arrhythmic changes in Cx43 are primarily associated with the left, but not the right, ventricle in both hypertensive strains. Additionally, acclimation of hairless SHR appears to offer some protection against malignant cardiac arrhythmias and mechanical dysfunction.
This study was supported by grants VEGA 2/0133/24, 2/0006/23, 2/0002/20, and APVV 21-0410.
Marijana Andjic1,2, Nevena Lazarevic1,2,3, Aleksandar Kocovic1,2, Vladimir Jakovljevic2,3,4, Jovana Bradic1,2
Linda Bartošová1,2,3, Aleksandra Stamenkovic1,4,3, Craig Resch1,4,3, Tomáš Rajtík2, Harold Aukema3, Amir Ravandi1,4,5,3
Oxylipins are bioactive lipid molecules that are involved in multiple signalling pathways regulating inflammation, blood flow, or cardioprotection, and form an efficient signalling system across various cells and tissues. However, the understanding of oxylipins and their pathological site-of-action/molecular crosstalk is very limited. Given the activation of inflammatory pathways during ischaemia -reperfusion (I/R) injury, we aimed to investigate the oxylipidome across the systemic circulation to determine the impact of I/R injury on oxylipins in various blood pools. Arterial (Art), venous (Ven), and coronary sinus (CS) blood samples were obtained at baseline, 60 min of ischaemia and 120 min of reperfusion. Oxylipins were measured by LC-MS/MS. We detected 45 oxylipins in plasma. There were neither significant differences in total nor individual oxylipins among Art, Ven, and CS. At 60 min of ischaemia, we detected a significant decrease in total oxylipin levels in Ven and CS blood, as well as altered levels of individual oxylipins (9-HETE, 12-HETE, 12,13-diHOME, and TXB2). Following the reperfusion, TXB2 was significantly decreased. There were no differences between Art and CS blood. In conclusion, there are no significant differences in pig plasma oxylipidome between three blood groups during stable non-pathological conditions. Oxylipin levels were altered during cardiac I/R in Ven and CS blood. Further depiction of more comprehensive oxylipin mechanisms in I/R injury is required.
This study was supported by the Heart and Stroke Foundation of Canada.
Linda Bartošová1, Andrej Kováč2, Gabriel Dóka1, Peter Balis3, Vladimír Garaj4, Kristína Ferenczyová3, Ulrika Duľová3, Juraj Piešťanský5, Tomáš Rajtík1,3
Inhibition of a transient receptor potential vanilloid 2 channels (TRPV2) has been described as a promising tool to affect the course and outcome of several diseases, from biomarker role in various types of cancer to modulation of immunological, neurological, and cardiovascular conditions such as heart failure and adverse remodelling. However, the lack of selective inhibitors has been a significant limitation. Therefore, we aimed to characterise the pharmacokinetics of drug probe SET2, the first selective TRPV2 inhibitor. Healthy Wistar rats underwent left carotid artery cannulation for arterial blood access. SET2 pharmacokinetics were assessed following a single intraperitoneal dose (25 mg/kg), with urine excretion monitored for 48 h in metabolic cages. Plasma and urine concentrations were measured using an UHPLC-MS method. Pharmacokinetic parameters such as maximum plasma concentration (Cmax), time to reach maximum plasma concentration (Tmax), biological half-life (T1/2), mean residence time (MRT), and clearance (CL) were analysed by non-compartmental analysis. Acute cardiovascular effects, including electrocardiogram (ECG) and blood pressure (BP), along with plasma markers of cardiac and renal damage, were also evaluated. SET2 reached Tmax at 2 min with a Cmax of 1,428 ng/mL. The estimated T1/2 was 65.1 min, and MRT was 70.5 min. The clearance rate was 0.448 mL/min/kg. Importantly, the compound maintained its inhibitory concentration for nearly 3 h without causing any acute changes in ECG or BP. This study is the first to characterise the pharmacokinetics and acute cardiovascular effects of SET2, demonstrating its safety and favourable pharmacokinetic profile following intraperitoneal administration.
This study was supported by grant VEGA 1/0775/21.
Gergő Bitay1, Kálmán Benke2, Ali Alex Sayour2, Tamás Radovits2, Béla Merkely2, Miklós Bitay3, András Varró1,4, István Baczkó1, Norbert Nagy1,4
Heart failure is a progressive, multifactorial clinical syndrome, leading to development of several life threating arrhythmias such as ventricular fibrillation. A possible mechanism contributed to arrhythmia development in heart failure is the so-called alternans, indicating periodic short-long oscillation of the action potential that leads to repolarisation inhomogeneity. Alternans are considered suitable predictors of sudden cardiac death; however, there is no specific pharmacological intervention to avoid or reduce alternans. In this study, we tested a novel, selective Na+/Ca2+ exchanger inhibitor ORM-10962 on alternans development in human heart failure and in undiseased human hearts. Left and right ventricular papillary muscles and trabeculae were obtained from explanted hearts of heart failure patients undergoing cardiac transplantation. Undiseased human hearts were obtained from an organ donor after removal of pulmonary and aortic valves for transplant surgery. Action potentials were measured by conventional microelectrode technique. Alternans were evoked by rapid pacing from 500 ms to 190 ms. All experiments were made at 37°C. Results show that ventricular action potential duration (APD) alternans in heart failure were larger than alternans recorded in undiseased ventricle. In failed hearts, left ventricle exerted larger alternans compared to right ventricle. Importantly, failing hearts – especially the left ventricle – often exerted considerable action potential amplitude alternans, typically in higher pacing rates that amplify APD alternans and ultimately lead to 2:1 conduction block. Data indicate that 1 μmol/L ORM-10962 shortens APD in the failed hearts although does not influence APD alternans magnitude, however, delays the development of action potential amplitude alternans and conduction block. These data support animal models claiming increased APD alternans in heart failure. Additionally, human failing hearts often exert large action potential amplitude alternans leading to conduction block that could be an important link between sudden cardiac death and alternans. Selective NCX inhibition may shift the development of amplitude alternans presumably due to APD shortening; however, other Ca2+-dependent mechanism could also contribute but these require further experiments.
This study was supported by grants TKP2021-EGA-23, RRF-2.3.1-21-2022-00003, K134939, and Nemzeti Tudós Akadémia.
Andjela Bojanic1, Aneta Stojmenovski1, Milica Gajic Bojic1,2, Djordje Djukanovic1,3, Zorislava Bajic1,4, Ranko Skrbic1,2
Diazepam, primarily known for its central anxiolytic and muscle relaxant effects, also exhibits vasodilating and hypotensive properties. However, its potential effect on vascular contractility, particularly in the human umbilical artery model, remains relatively unexplored. The aim of this study was to examine the effect of diazepam on human umbilical artery contractility and to identify the mechanisms behind these effects, such as the role of potassium channels, calcium channels and endothelial function. To examine the mechanisms of diazepam-induced relaxation (10−8–10−4 M), a series of functional tests were applied to the organ bath system. Human umbilical artery rings were incubated with different selective blockers: 4-Aminopyridine (10−5 M), TEA (10−5 M), and glibenclamide (10−5 M) to examine the role of potassium channels, amlodipine (10−5 M) to examine the role of voltage-gated calcium channels, and L-NAME (10−4 M) and indomethacin (10−4 M) to examine the role of endothelium. Diazepam caused a significant percentage of relaxation of human umbilical arteries (about 90%, expressed in relation to serotonin-induced precontraction). No significance was found in the relaxation responses of diazepam in the presence of potassium channel blockers, as well as endothelium-related relaxation mediators. However, the percentage of contraction of rings treated with BaCl2 was significantly lower in the presence of diazepam (Emax 9.95%) compared to control rings (Emax 87.358%). The relaxation effects of diazepam on human umbilical arteries indicate its vasodilation potential, which can be significant in conditions of pronounced use of diazepam during pregnancy. The presented functional tests suggest that the relaxation effects of diazepam are primarily mediated through calcium channels, but additional research is needed to explain the exact mechanisms.
Almos Boroš1,2, Miloslava Chalupova1, Jaroslav Hrdlička1, Michal Behuliak2, Jan Neckar1, Josef Zicha2
It is well known that cholinergic potentiation by acetylcholinesterase inhibitors (AChEi) has reduced proinflammatory cytokine production and slowed the progression of heart failure. In this study, chronic cholinergic potentiation with the peripherally acting AChEi pyridostigmine (PYR) was studied in male: (a) Normotensive Wistar-Kyoto (WKY) rat, (b) spontaneously hypertensive rat (SHR), a well-studied model of hypertension and cardiac hypertrophy with autonomic nervous system (ANS) imbalance, and (c) SHR-CRP rats, a transgenic SHR strain with systemic inflammation due to expression of human C-reactive protein (CRP). Mean arterial pressure (MAP), cardiac ischaemic tolerance, ANS balance, and splenic cytokine gene expressions were determined in rats chronically treated with PYR at 25 mg/kg/day for 8 weeks in drinking water and untreated controls. In both hypertensive strains, ANS balance was reduced, and PYR treatment markedly improved it by augmenting parasympathetic tone. SHR-CRP rats had significantly higher MAP than SHR (by 15 ± 1 mmHg) regardless of daytime. PYR significantly reduced MAP in SHR and SHR-CRP strains, and this effect was greater in SHR-CRP. Infarct sizes were similar among all groups. PYR treatment slightly (but not significantly) reduced the infarct size normalised to the area at risk in SHR-CRP rats (by 9%). Gene expression of tumour necrosis factor-1α (TNF-1α) was significantly higher in the spleens of control SHR-CRP rats compared to WKY and SHR groups, which were similar levels. Interleukine-10 (Il-10) gene expression was comparable between SHR and SHR-CRP groups, and WKY rats showed higher expression regardless of treatment. Therefore, chronic PYR treatment reduced the TNF-α/Il-10 gene expression ratio in SHR-CRP rat spleens by reducing TNF-α gene expression. In conclusion, our data suggest that chronic peripheral cholinergic potentiation has the potential to significantly slow the progression of cardiovascular diseases associated with systemic inflammation state through immunomodulation.
This study was supported by the National Institute for Research of Metabolic and Cardiovascular Diseases (Programme EXCELES, ID Project No. LX22NPO5104) – Funded by the European Union – Next Generation EU.
Jovana Bradic1,2, Marijana Andjic1,2, Vladimir Jakovljevic2,3,4, Jovana Novakovic1,2
Despite its extensive traditional use in treating various disorders, the potential of
Andrea Brunetti1, Nefeli Grammatica Pavlidou2, David Revuelta1, Franziska Reinhardt3, Sven Margenburg1, Till Joscha Demal1, Cristina Molina1
3′,5′-cyclic adenosine monophosphate (cAMP) is a ubiquitous second messenger, which acts in nanodomains to regulate cardiac excitation-contraction coupling by activating phosphorylation of Ca2+-handling proteins. The sarcoplasmic reticulum (SR) Ca2+ATPase2a (SERCA2a) is a key regulator of Ca2+ reuptake into the SR during diastole, and thus, alterations of protein phosphorylation in this specific nanodomain could trigger atrial fibrillation (AF)-associated mechanisms of the Ca2+-handling leading to arrhythmias. Our aim was to investigate the cAMP-dependent regulation of SERCA2a in the human atrium. A total of 119 atrial myocytes were isolated from tissues of 80 patients in sinus rhythm. Föster-resonance energy transfer (FRET) was used to measure cAMP levels. Cells were transfected with an adenovirus (E1-PLN) to express the phospholamban FRET-based cAMP sensor and cultured during 48 h. Inhibition of phosphodiesterases (PDEs) 1 to 4 promoted a huge increase in cAMP levels in SERCA2a nanodomain. Surprisingly, this increase was reduced to half when a PDE8 inhibitor (PF-04957325, 30 nmol/L) was applied on top. Same tendency was observed when 1 mmol/L Ca2+or Bay-K 8644 (an L-type Ca2+ channel agonist, 1 mmol/L) were applied on top of 3-isobutyl-1-methylxanthine, while ryanodine receptor inhibition after PF-04957325 increased cAMP levels even further. Notably, 10 μmol/L forskolin was used at the end of the protocol as positive control. Stimulation with the beta-adrenergic agonist isoprenaline (ISO, 100 nmol/L) was also performed in a number of cells before applying the aforementioned PDE inhibitors. This regulation of cAMP levels in SERCA2a nanodomain indicates a possible Ca2+-dependent phosphorylation of SERCA2a modulated by L-type Ca2+ channel activity.
Katarína Bujnová, Andrej Barta, Martina Cebová
Myocardial infarction (MI) is the most serious manifestation of coronary artery disease defined as the death of cardiomyocytes due to prolonged ischaemia. Cardiovascular disease is often associated with obesity, which increases inflammation and oxidative stress, negatively affecting the bioavailability of nitric oxide (NO), a key vasodilator synthesized by nitric oxide synthase (NOS). The aim of this study was to examine the effect of a high-fat diet on NO bioavailability in the context of oxidative stress and inflammation after MI. Nine weeks old Wistar Kyoto rats were divided into two groups, one fed high-fat diet while the other continued with standard lab chow for 4 weeks. In week 13, the induction of MI was performed by the reversible ligation of the left descending coronary artery. The occlusion lasted 20 min. Seven days after surgery, animals were sacrificed. Plasma levels of proinflammatory cytokines and superoxide dismutase (SOD) activity were measured using a Bioplex kit and a colorimetric assay using a commercially available kit, respectively. The total NOS activity was assessed by converting [3H]-L-arginine to [3H]-L-citrulline. Protein expression levels of endothelial nitric oxide synthase (eNOS), inducible nitric oxide synthase (iNOS), and nuclear factor kappa B (NFκB) were analysed by Western blot. Concentration of conjugated dienes in the heart was measured spectrophotometrically. The results showed that a high-fat diet significantly decreased total NOS activity and eNOS expression, while conversely increasing the expression of iNOS and inflammatory markers such as tumor necrosis factor-α, interleukin-6, and NFκB. Likewise, the increased concentration of conjugated dienes in the heart and the trend of decreased activity of the antioxidant system SOD demonstrates oxidative stress. This suggests that a high-fat diet negatively affects physiological NO bioavailability after MI and contributes to further myocardial damage via inflammation and oxidative stress.
This study was supported by grants VEGA 2/0131/24, APVV-22-0271, and VV Bujnová.
Johannes Castelein1,2, Amanda Duus2, Pernille Baekke3, Ingolf Sack4, Noah Jaitner4, Adam Hansen5,6, Ole de Backer3, Rudi Dierckx7, Niels Vejlstrup3, Morten Lund2,3, Thomas Jespersen2, Ronald Borra1,7
Ventricular stiffness is a crucial diagnostic and prognostic indicator of diastolic function, influenced by both physiological and pathological factors. However, assessing ventricular stiffness and diastolic function in a clinical setting remains challenging. Cardiac Multifrequency Magnetic Resonance Elastography (MMRE) has emerged as a promising tool for evaluating stiffness in both left ventricle (LV) and right ventricle (RV). This study aimed to explore the capability of cardiac MMRE to non-invasively quantify the diastolic ventricular stiffness in adult healthy controls (HCs) and its physiological variation with age and sex. We prospectively included 40 HCs who underwent cardiac MMRE. Shear wave speed (SWS) maps were reconstructed for the LV and RV as surrogate for tissue stiffness. No significant correlations were found between age and SWS in either the LV (
Pinar Cevikbas Ilik1, Merve Sayin2, Yasemin Atici3, Belma Turan1,3
Magnolol (MAGNO) is a hydroxylated biphenyl compound, isolated from the bark of Magnolia officinalis. This product, as a traditional Chinese medicine, demonstrated potential resources for natural antioxidants to treat a variety of disorders via scavenging of free radicals, protecting DNA, providing growth performance, and disease resistance at the cellular and system levels in mammalians. Since previous studies have shown that magnolia bark extract administration provided a significant antioxidant effect in isolated rats, protected the myocardium against ischaemic injury, and suppressed ventricular arrhythmia, in this study, we aimed to evaluate the cardioprotective effects of administration of MAGNO (100 mg/kg; a cumulative dose of 15 mg/kg in 2-week) in Doxorubicin (DOX)-treated rat hearts. One group of adult male Wistar rats was injected with DOX (DOX-group) or saline (CON-group), while other group of DOX-treated rats was administered with MAGNO before DOX (Pre-MAGNO group) or after DOX (Post-MAGNO group). The MAGNO administration, before or after DOXO, provided full survival of animals during 12–14 weeks and significant recoveries in the systemic parameters of animals such as high systolic and diastolic blood pressures. This treatment also significantly improved heart function including recoveries in end-diastolic volume, left ventricular end-systolic volume, heart rate, and cardiac output. Furthermore, the MAGNO administrations improved the structure of left ventricles such as recoveries in loss of myofibrils, degenerative nuclear changes, fragmentation of cardiomyocytes, and interstitial oedema, while all of these benefits were observed in the pre-MAGNO treatment group, more prominently. Overall, one can point out the beneficial antioxidant effects of MAGNO in chronic heart diseases as a supporting and complementing agent to conventional therapies.
This study was supported by the Lokman Hekim University Scientific Research Projects Coordination Unit. Project No. 202AP404, 2022.
Francisco Cruz1, Ana Isabel Moreno1, Patricia Sánchez1, Lilian Gutierrez1, Juan Manuel Ruiz1, Paula García1, Gema Mondejar1, Alvaro Macías1, María Vera1, Amaia Talavera1, Isabel Martínez1, José Jalife1,2,3
Flecainide and other class Ic antiarrhythmic drugs (AADs) are widely used in Andersen-Tawil syndrome type 1 (ATS1) patients with life-threatening arrhythmias but structurally normal hearts. However, class Ic AADs may fail and exhibit proarrhythmic effect in some ATS1 patients through mechanisms that remain poorly understood. We aimed to provide novel insight into the molecular mechanisms of AAD-induced arrhythmias and whether they might increase the risk of life-threatening ventricular arrhythmias and sudden cardiac death (SCD). A review of data from 48 ATS1 patients reported in the literature showed that 50% of patients responded partially to flecainide, with ventricular arrhythmias (VA) reduction in only 18.2% and of those patients, VA persisted in 20%–50%. Flecainide was ineffective in 29.5% of patients, and of those, 13.5% suffered a non-fatal cardiac arrest. In AAV-mediated cardiac-specific mouse models expressing five different mutations (Kir2.1Δ314-315, Kir2.1C122Y, Kir2.1G215D, Kir2.1R67W, and Kir2.1S136F), treatment with flecainide or propafenone (40 mg/kg) differentially altered the cardiac conduction velocity by prolonging the duration of the P wave, PR, QRS, and QTc intervals compared to Kir2.1WT, with Kir2.1S136F showing milder effects. Flecainide also increased VA inducibility in all mutant mice except Kir2.1S136F, which exhibited a significant reduction in VA. Voltage-clamp studies revealed a mutation-dependent effect on inward rectifier K+ (IK1) and Na+(INa) current densities. Kir2.1G215D cardiomyocytes showed a higher impact on IK1, followed by Kir2.1C122Y, Kir2.1R67W, and Kir2.1S136F. Additionally, Kir2.1C122Y cardiomyocytes had a significant decrease in INa. Superfusion of flecainide (10 μmol/L) slightly increased IK1 density in Kir2.1WT and Kir2.1S136F, while Kir2.1C122Y and Kir2.1R67W showed a decrease in both IK1 and INa densities, despite normal trafficking of mutant channels. Optical mapping experiments in ATS1 patient-specific iPSC-CM monolayers expressing Kir2.1C122Y, Kir2.1G215D, and Kir2.1R67W mutations showed an increase in rotor incidence at baseline and under flecainide treatment, confirming the drug's proarrhythmic effect. Lastly, in silico molecular docking studies predict that the Kir2.1-Cys311 pharmacophore binding site is altered in Kir2.1C122Y heterotetramers, reducing flecainide accessibility and leading to channel closure and arrhythmias. Class Ic AADs are only partially effective and might be proarrhythmic in some ATS1 patients. Our findings indicate that the efficacy of AADs may vary depending on the specific ATS1 mutation. We submit that mutations impacting both the resting membrane potential and cellular excitability potentially create a substrate for life-threatening arrhythmias. These data raise concern about the widespread use of class Ic AADs in ATS1 patients, urgently calling for further mechanistic studies to advance personalised therapy.
This study was supported by the National heart, Lung and Blood Institute, NIH grant number R01HL163943; La Caixa Banking Foundation project code HR18-00304 (LCF/PR/HR19/52160013); grants PI-FIS-2020 # PI20/01220 and PI-FIS-2023 # PI23/01039 from Instituto de Salud Carlos III (ISCIII) and co-funded by Fondo Europeo de Desarrollo Regional (FEDER); and by the European Union, respectively; grant PID2020-116935RB-I00 and BFU2016-75144-R funded by MCIN/AEI/10.13039/501100011033; Fundación La Marató de TV3 (736/C/2020) ‘amb el suport de la Fundació La Marató de TV3’; CIBERCV (CB16/11/00458; CB/11/00222 to CV); European Union's Horizon 2020 grant agreement GA-965286; and Program S2022/BMD7229 -CM ARCADIA-CM funded by Comunidad de Madrid to JJ.
Vivien Demeter-Haludka1, Szilvia Déri1, Alexandra Polyák1, Leila Topal1, Noémi Zombori-Tóth1, Jenő Antal Pintér2, Aiman Saleh A. Mohammed1, István Baczkó1, András Varró1,
Growing evidence suggests that rigorous exercise regimens in elite athletes increase the risk of arrhythmias, including ventricular fibrillation, which can lead to sudden cardiac death. Our results have demonstrated that 4 months of sustained, high-intensity endurance training in dogs prolonged the corrected QT interval and lengthened APD90, accompanied by a decrease in transient outward potassium current [I(to)] density. This training also increased repolarisation variability and decreased the resting heart rate. Left ventricular hypertrophy developed in trained dogs. Compared to the sedentary dogs, trained dogs exhibited significantly higher level of fibrosis in the left ventricle, which may contribute to an increased ectopic activity and heightened arrhythmia susceptibility. In this study, we have examined the expression of ion channel subunits potentially involved in cardiac arrhythmic remodelling induced by sustained, high-intensity endurance training. Western blot and immunocytochemistry were used to analyse the expression and density of I(to) subunit Kv4.3 and KChiP2 in left ventricular tissue and isolated cardiomyocytes from both sedentary and trained dogs. Hyperpolarization-activated cyclic nucleotide-gated channels (HCN)-1, HCN2, and HCN4 were immunolabeled in left ventricular cardiomyocytes, and the ultrarapid delayed-rectifying potassium current [I(Kur)] subunit Kv1.5 expression was measured in atrial cells. High-intensity training did not alter Kv4.3 or KChiP2 expression. Interestingly, HCN4 density significantly increased in trained dogs (10391 ± 616 a.u.;
This work was supported by the National Research Development and Innovation Office (NKFIH K 135464, K 142738, K-147212, GINOP-2.3.2.-15-2016-0006, GINOP-2.3.2.-15-2016-00047 GINOP-2.3.2.-15-2016-00040, and TKP2021-EGA-32), the Ministry of Human Capacities Hungary (EFOP-3.6.2-16-2017-00006), the Albert Szent-Györgyi Medical School institutional grant (SZTE AOK-KKA 2021 and SZTE AOK-KKA 2022), and by Hungarian Research Network (HUN-REN TKI project). This study was also supported by Project RRF-2.3.1-21-2022-00003 ‘National Heart Laboratory, Hungary’ implemented with the support provided by the European Union and by the Pharmaceutical and Medical Device Developments Competence Centre of the Life Sciences Cluster of the Centre of Excellence for Interdisciplinary Research, Development and Innovation of the University of Szeged, Hungary.
Mikkel Eiksen Dupont1, Stine Bøttcher Jacobsen1, Stefan Noe Niikanof Christiansen1,2, Jacob Tfelt-Hansen1,3, Morten Holdgaard Smerup4, Jeppe Dyrberg Andersen1, Niels Morling1
Untreated fresh cardiac tissue is the optimal tissue material for investigating DNA methylation patterns of cardiac biology and diseases. However, fresh tissue is difficult to obtain. Therefore, tissue stored as frozen or formalin-fixed, paraffin-embedded (FFPE) is widely used for DNA methylation studies. It is unknown whether storage conditions alter the DNA methylation in cardiac tissue. In this study, we compared the DNA methylation patterns of fresh, frozen, and FFPE cardiac tissue to investigate if the storage method affected the DNA methylation results. We used the Infinium MethylationEPIC assay to obtain genome-wide methylation levels in fresh, frozen, and FFPE tissues from nine individuals. We found that the DNA methylation levels of 21.4% of the examined CpG sites were overestimated in the FFPE samples compared to that of fresh and frozen tissues, whereas 5.7% were underestimated. Duplicate analyses of the DNA methylation patterns showed high reproducibility (precision) for frozen and FFPE tissues. In conclusion, we found that frozen and FFPE tissues gave reproducible DNA methylation results and that frozen and fresh tissues gave similar results.
This project was internally funded by the Department of Forensic Medicine.
Anar Dushpanova1, Valentina Casieri1, Eugenio Ridolfi Riva2, Ciro Zinno2, Filippo Agnesi2, Letizia Brogi3, Valentina Sassu1, Lisa Alibrandi1, Silvestro Micera2, Vincenzo Lionetti1
The precise effect of acute unilateral damage of cardiac vagus nerve (CVN) on the spontaneous electrical activity of contralateral vagus nerve at the cardiac level is not well comprehended. We examined the transient neuroelectrical response to a transection of the right or left cardiac vagal nerve (VNTx) by measuring the spiking activity of each CVN. Fourteen male Gottingen minipigs were subjected to sternotomy. Multiple electrode cuffs were placed below the cut level to capture vagal electroneurographic signals while monitoring electrocardiogram and hemodynamics, both before and immediately after cardiac vagotomy (left: L-cut,
This study was supported by H2020-FETPROACT-2018-2020 NEUHEART project #824071.
Chantal Eickelmann1, Nilgün Gedik1, Helmut Raphael Lieder1, Svenja Idel2, Laxmikanth Kollipara2, Albert Sickmann2,3,4, Michael Sturek5, Gerd Heusch1, Petra Kleinbongard1
Ossabaw minipigs differ from Göttingen minipigs in their unique genetic predisposition to develop a full metabolic syndrome and their non-responsiveness to cardioprotective interventions, even before the development of their diseased phenotype. Previous DNA sequencing data revealed differences in a cluster of mitochondrial genes between the two pig strains. Obviously, alterations in their protein composition affect the function of mitochondria, i.e., Krebs cycle, oxidative phosphorylation, mitochondrial biogenesis, and mitochondrial dysfunction play a crucial role in the development of metabolic syndrome as well as in myocardial ischaemia-reperfusion injury and cardioprotection.
Mitochondria were isolated from cardiac left ventricular tissue samples of both pig strains (
A total of 810 proteins were identified in at least three of four samples, and 654 were classified as mitochondrial proteins. Among them, 22 proteins were differentially expressed between both pig strains. Most of them were classified as related to mitochondrial metabolism (9 higher/7 lower expressed in Ossabaw minipigs), four (two higher/two lower expressed) as related to mitochondrial transcription and translation, and three as related to small molecule transport (one higher/two lower expressed), two as related to oxidative phosphorylation (one higher/one lower expressed), and one as related to dynamic and surveillance (lower expressed).
The mitochondrial proteome is largely comparable between Ossabaw and Göttingen minipigs. However, the proteins that were differentially expressed are related to mitochondrial metabolism, which play a crucial role in the development of metabolic syndrome and ischaemic heart disease. In the next step, the results of the proteomic approach must be validated via protein biochemistry.
This work was supported by the German Research Foundation [SFB 1116 B08 to G.H. and P.K.] and the European COST ACTION in CARDIOPROTECTION [CA16225 to G.H. and IG16225 to G.H. and P.K.]. S.I., L.K. and A.S. acknowledge the support by the‘Ministerium für Kultur und Wissenschaft des Landes Nordrhein-Westfalen’ and ‘Der Regierende Bürgermeister von Berlin, Senatskanzlei Wissenschaft und Forschung’.
Enikő Edit Enyedi1, Attila Ádám Szabó1, Tamás Bence Pintér1, Csongor Váradi2, Petra Erzsébet Bíró1, Zoltán Balázs Tóth1, Zoltán Papp1, Attila Tóth1, Miklós Fagyas1
In severe cases of COVID-19, autoantibodies appear in the circulation, targeting lung and heart tissues. The pathophysiological role and long-term effects of these antibodies remain unknown, and there is little information on the transient or permanent persistence or detectability of these antibodies. Lung-specific autoantibodies developed during COVID-19 may reappear or increase in titre due to tissue damage accompanying lung surgery, which may affect the wound healing and recovery from surgery. The objective of this study was to investigate the long-term behaviour of anti-cardiac and anti-lung autoantibodies in patients who had recovered from COVID-19. Autoantibodies were detected by a Western blot-based method in patients who had undergone lung surgery for various reasons with partial lung resection; all relevant clinical data were also collected. Patients who underwent lung surgery (
This study was supported by the EKÖP-24-3-II-DE-302 (E.E.E.), EKÖP-24-3-II-DE-277 (A.Á.Sz.) University Research Scholarship Program (EKÖP) of the Ministry of Culture and Innovation from the source of the National Research, Development and Innovation Fund.
Veronika Farkašová, Miroslav Ferko, Natalia Andelova, Tanya Ravingerova
Ischaemic heart disease represents a major ailment leading to heart remodelling, cardiac hypertrophy, and subsequent heart failure. Cardioprotective adaptive interventions can mitigate the deleterious consequences of ischaemia/reperfusion (I/R) injury to the heart. Intermittent hypoxic preconditioning (IHPC) is a relatively well-established cardioprotective non-pharmacological intervention in various models, such as cardiac injury and I/R injury in particular. However, its mode of action is currently not completely understood. The study aimed to investigate the mechanisms and cellular cardioprotective signalling of specific IHPC protocol in rat hearts. For this purpose, we subjected the male Wistar rats of age 3 months to IHPC (10% O2, normal pressure) for 8 h once per week for four weeks. Subsequently, the rat hearts were isolated for Langendorff perfusion and subjected to 20-min stabilization, 30-min ischaemia, and 40-min of reperfusion to evaluate post-ischaemic contractile dysfunction and reperfusion arrhythmias and to harvest the tissue samples for analysis by WB and mass spectrometry. The recovery of contractile function after I/R injury was significantly increased in IHPC animals compared to non-IHPC controls. Overall activity of reperfusion injury salvage kinase (RISK) pathway components was increased in IHPC hearts, suggesting the role in cellular cardioprotective signalling mediated by intermittent IHPC. On the contrary, the activity of survivor activating factor enhancement (SEFE) pathway components were non-affected in IHPC hearts compared to controls. The cardioprotective effect of intermittent IHPC in rat hearts can be mediated by the upregulation of RISK but not SAFE pathway signalization.
This study was supported by grants APVV-19-0540; APVV-22-0264; VEGA 2/104/22; and VEGA 1/0775/21.
Kristina Ferenczyova1,2, Barbora Kalocayova1,2, Lucia Kindernay1, Matus Sykora1, Miroslav Barancik1, Lubomira Tothova3, Linda Bartosova2, Tomas Rajtik1,2, Monika Bartekova1,3
Cardiovascular system diseases belong to the most prevalent civilization diseases. To date, several cardioprotective effects of quercetin (QCT) have been confirmed in young healthy individuals. The aim of our work was to reveal the effects of QCT on cardiovascular system in elderly animals with type 2 diabetes. In this study, QCT was orally administered for 6 weeks at a dose of 20 mg/kg/day to 6-month-old/1-year-old lean (fa/+) and obese (fa/fa) Zucker diabetic fatty (ZDF) rats. Blood pressure measurement and echocardiography were performed. Oxidative stress markers were analysed from blood plasma, and the expression of the selected proteins were monitored in myocardium. Isolated hearts of ZDF rats were subjected to I/R (30 min/120 min). The total collagen content was determined in the left ventricle. Our data showed that administration of QCT significantly reduced blood pressure in 6-month-old rats, while it had no effect in 1-year-old rats. QCT significantly increased the antioxidant activity parameter in blood plasma of obese 1-year-old rats. Administration of QCT did not have cardioprotective effects against I/R damage in any group. Complex activation of the RISK pathway was not induced by QCT. In 1-year-old obese ZDF rats, administration of QCT normalized the E/A ratio (marker of diastolic dysfunction), reduced the thickness of the left ventricular wall, and significantly decreased the collagen content. QCT has been shown to have beneficial effects on blood pressure in a model of type 2 diabetes, but increasing age and/or development of diabetes impair its antihypertensive effects. In the prevention of I/R damage, QCT was not effective in both ages. Potential explanation of failing of QCT cardioprotection in increasing age and diabetes may be the failure to activate the RISK pathway. On the contrary, QCT suppressed diabetes-induced diastolic dysfunction and reduced the collagen content in the left ventricle in elderly individuals.
This study was supported by grants APVV-21-0194, VEGA 1/0775/21, VEGA 1/0775/21, APVV-20-0242, and VEGA 2/0148/22.
Marta Gaburjáková, Jana Gaburjáková
Catecholaminergic polymorphic ventricular tachycardia (CPVT) is a highly arrhythmogenic syndrome triggered by stress, primarily linked to gain-of-function point mutations in the cardiac ryanodine receptor (RyR2). This Ca2+ release channel in the sarcoplasmic reticulum (SR) plays a fundamental role in the Ca2+ cycling essential for heart contraction. CPVT is effectively suppressed by flecainide. This antiarrhythmic agent is a known inhibitor of the surface-membrane Na+channel while also inhibiting the intracellular RyR2 channel. Initially, the flecainide-RyR2 interaction was considered therapeutically irrelevant since RyR2 activity was affected only when the channel conducted ions in the cytosol-to-lumen direction, opposite to physiological SR Ca2+ release. This statement has been revisited as the RyR2 channel is able to carry charge-compensating countercurrent in the cytosol-to-lumen direction, and reducing this current may in fact suppress pathologically increased SR Ca2+ release. Monitoring single-channel properties, we examined whether flecainide can target the RyR2-mediated countercurrent. Additionally, we assessed the impact of flecainide on the SR Cl-channel as it may also play a role in conducting a charge-compensating countercurrent. RyR2 and Cl- channels isolated from ventricles of the rat heart were incorporated into planar lipid membranes (BLMs) and examined under voltage-clamp conditions. Flecainide was added to the RyR2 cytosolic face in a dose-dependent manner, while Ca2+ current flowed via the channel pore from lumen to cytosol, and the countercurrent carried by Tris + cations was in the opposite direction. These conditions more closely mimic the cell situation. The activity and conductive properties of the Cl- channel were assessed with a Cl- gradient applied across the BLM. We demonstrate that flecainide significantly reduced the RyR2-mediated countercurrent without affecting channel activity. In contrast, the SR Cl- channel was completely resistant to flecainide. Based on these findings, it is reasonable to propose that the primary intracellular target of flecainide in vivo is the RyR2-mediated countercurrent.
This study was supported by grants VEGA 2/0018/21 and APVV-22-0061.
Žarko Gagić1, Đorđe Đukanović1,2
Thromboembolic disorders represent a significant cause of mortality leading to life threatening conditions such as myocardial infarction, pulmonary embolism, and deep vein thrombosis. Factor Xa (fXa) plays a key role in blood coagulation cascade and thrombus formation. It is a serine protease that catalyses hydrolysis of prothrombin into thrombin, which further promote conversion of fibrinogen into fibrin, a final stage of blood clotting. In recent years, fXa has been identified as a promising target for the design of efficient and safer antithrombotics, compared to conventional warfarin therapy. Today, several (fXa) inhibitors has been approved, such as rivaroxaban, apixaban, betrixaban, edoxaban, and fondaparinux, but research in this area is still in the focus of pharmaceutical industry. To identify the most important structural determinants that influence fXa inhibitory activity, 3D-quantitative structure–activity relationship (QSAR) studies were performed on dataset consisting of 54 structural analogues of betrixaban. Experimentally determined IC50 activities (expressed as pIC50) were collected from literature, and all structures were previously optimized using the semiempirical PM3 method. The 3D-QSAR model was created using partial least squares regression analysis. Internal (R2 = 0.93; Q2 = 0.75) and external (R2pred = 0.66) validation parameters confirmed that the model can be used for prediction of fXa inhibitory activity. The 3D-QSAR model revealed most important variables that influence activity (distances between H-bond donors, acceptors, hydrophobic, and steric regions), and this information will be further used for the design of new selective fXa inhibitors.
The authors did not receive any financial support from any organization for the submitted work.
Ferenc Gallyas1, Robert Halmosi2, Kalman Toth2
Cardiovascular diseases are the leading cause of morbidity and mortality worldwide, imposing significant health and economic burdens on the individual, families, and societies. Accordingly, a substantial effort is exerted for finding novel effective therapeutics for these diseases. Recently, inhibitors of the poly(ADP-ribose) polymerase (PARPi), originally developed for cancer therapy, show promise for repurposing in cardiovascular diseases. These drugs' cytoprotective, antioxidant, and anti-inflammatory properties can be pivotal in treating heart conditions characterised by tissue damage and inflammation. Studies suggest that PARPi could protect cardiac cells from ischaemia-reperfusion injury and reduce adverse remodelling post-myocardial infarction. Additionally, their anti-inflammatory properties might mitigate atherosclerosis progression. Although prolonged administration of PARPi could lead to malignant transformation due to DNA damage accumulation, short-term application of these drugs may be beneficial in post-infarct cardiac remodelling. We are presenting experimental evidence for the use of PARPi in the aforementioned conditions, and we provide mechanistic background for the protective effects of PARPi including the effects of these drugs on protein kinase B/Akt and mitogen-activated protein kinase signalling.
Tibor Hornyik1, Zoltan Husti1, Luca Soattin2, Leila Topál1, Jenő Pintér1, Aiman Mohammed1, Eleonora Torre3, Gabriella Forte4, Mark R. Boyett5, László Virág1, Norbert Jost1, Matteo E. Mangoni3, Bo H. Bentzen2, Gwilym M. Morris6, István Baczkó1, András Varró1, Alicia D'Souza7
Clinical studies suggest an increased incidence of atrial fibrillation (AF) in endurance athletes. However, the underlying mechanisms of AF associated with endurance training are not understood. In this study, we examined the electrophysiological role of pulmonary veins in a canine model of high-intensity endurance exercise. Beagle dogs were randomized into sedentary group (SG,
Csaba Horvath1, Izabela Jarabicova1, Andrea Marcinikova1, Jaroslav Hrdlicka2, Almos Boros2, Veronika Olejnickova2,3, Jan Neckar2, Frantisek Kolar2, M.Saadeh Suleiman4, Adriana Duris Adameova1,5
Recent studies have highlighted the receptor-interacting protein kinase 3 (RIP3) as a critical regulator of necroptosis, oxidative stress, and inflammation in various tissues, including the heart. This study investigates the pleiotropic effects of RIP3 inhibition under ischaemic conditions, focusing on its role in myocardial protection. Utilising various models of ischaemia-reperfusion injury (IRI) ex vivo (30-min I, 10-min R) and in vivo (1-h I, 7 days-R), we administered a specific RIP3 inhibitor – GSK'872 during the whole respective R period. Under ex vivo conditions, RIP3 inhibition significantly reduced myocardial necrosis-like cell death evidenced by decreased LDH activity after IRI; however, without altering the expression of the main pro-necroptotic proteins – pRIP3 and pMLKL. Interestingly, although this intervention reversed the IRI-induced increase in the extent and rate of mitochondrial swelling, it did not affect the level of RIP3-associated mitochondrial proteins – PGAM5 and pDrp1. In an in vivo IRI model, a 7-day administration of GSK'872 prevented the likely formation of the necroptosis, but not pyroptosis-associated membrane pores, proven by decreased pRIP3 and pMLKL and unchanged NT-GSDMD expression, respectively. Furthermore, RIP3 inhibition was associated with a likely reduction of mitochondrial damage based on the PGAM5 and pDrp1 expression limitation. Interestingly, consistently with human patients with an ischaemic insult to the heart, such rats also exhibited elevated plasma RIP3 levels, while GSK'872 mitigated such an increase. However, such intervention was unable to preserve cardiac function since it did not affect any echocardiographic parameters after IRI. This research indicates RIP3 as a promising target for cardiovascular protection and recovery following ischaemic events and underscores the potential of RIP3 inhibition as a therapeutic strategy for mitigating post-ischaemic cardiac damage.
This study was supported by grants APVV-20-0242; APPV-15-0607, and VEGA 1/0016/20.
Izabela Jarabicová1, Csaba Horváth1, Jaroslav Hrdlička2, Almos Boroš2, Veronika Olejníčková2,3, Eva Zábrodská2,3, Hana Mauer Šutovská4, Ľuboš Molčan4, Libor Kopkan5, Martin Chudý6, Eva Goncalvesová6, Jan Neckář2, Michal Zeman4, František Kolář2, Adriana Adameová1,7
Cell loss due to necrosis-like cell death is a crucial mechanism underlying the pathogenesis of heart failure (HF). However, it is unknown whether a role of various types of programmed necrosis differs depending on the HF aetiology. In this study, we aimed to examine necroptosis and pyroptosis in failing hearts due to myocardial infarction (MI) or pressure overload induced genetically or surgically. The effects of a pharmacological inhibitor of receptor-interacting protein kinase 3 (RIP3), a molecule connecting both these pro-death pathways, were also studied. As a clinical translational approach targeting necrosis-like events, a plasma screening of certain circulating molecules in the human HF subjects was performed. All three HF models showed active necroptosis as documented by the increased activation of RIP3 and mixed lineage kinase domain-like pseudokinase (MLKL). On the contrary, only hearts failing due to MI were positive for the markers of pyroptosis (NLR family pyrin domain containing 3, caspase-1, gasdermin D, and interleukin-1β). In post-MI HF, the RIP3 inhibitor was able to reduce the activation of both RIP3 and MLKL, advocating for its protective effect against necroptosis, but it had no effect on pyroptosis and did not modulate the compromised cardiac function. A two-fold increase in the serum levels of high mobility group box 1 (HMGB1) was found in HF patients regardless of the HF aetiology or severity, while they positively correlated with the serum C-reactive protein. This is the first study showing that different necrosis-like cell death pathways likely underlie the development of HF of various origins. The RIP3 inhibition mitigates the necroptotic cell loss in post-MI HF, highlighting its potential clinical perspective in HF management. The serum HMGB1 was suggested as a novel promising marker in the HF diagnosis.
This study was supported by grants VEGA 1/0016/20, APVV-20-0242, APPV-15-0607, NU21J-02-00039, and LX22NPO5104.
Jazzar Ashley, Jacques Danielle, Bkaily Ghassan
A lot of work was done dealing with insulin regulation of cardiomyocyte function. However, little is known about the remodelling of morphology and ionic homeostasis of ventricular heart cells. The non-essential amino acid taurine, which is present in seafood and produced by the heart and the brain, contributes to the excitation-contraction coupling of cardiomyocytes. However, whether taurine prevents the remodelling of heart cells is to be explored. In the first series of experiments, we studied the effect of chronic high concentrations of insulin in hypertrophy and intracellular concentrations of sodium and calcium in cultured adult rat heart cells using quantitative 3D confocal microscopy. In the second series of experiments, we verified whether taurine prevents insulin-induced remodelling of cardiomyocytes. In this study, we show that treatment with a high concentration of insulin induces an increase in the volume of ventricular cardiomyocytes that is associated with an increase of cytosolic and nuclear sodium and calcium as well as a decrease in the ratio of pCREB/tCREB. Long-term treatment with taurine prevented insulin-induced remodelling of ventricular cardiomyocytes. This effect of taurine was prevented by the taurine-sodium symporter blocker. In conclusion, our results show that insulin induces ventricular cardiomyocyte hypertrophy via downregulation of the pCREB/tCREB level and that chronic taurine treatment prevents this effect.
This work was funded by NSERC and CHIR.
Jovana Joksimovic Jovic1,2, Jovan Rudic3, Nikola Jovic4,5, Maja Nikolic1,2, Jasmina Sretenovic1,2, Milica Milinkovic2,6, Slobodanka Mitrovic5,7, Kristina Andric5,8, Vladimir Jakovljevic1,2,9
Polycystic ovary syndrome (PCOS) is a prevalent endocrine disorder affecting women of reproductive age, often linked to a range of cardiometabolic complications, including obesity, insulin resistance, and dyslipidemia. Despite the high incidence of these metabolic disturbances, the underlying mechanisms remain poorly understood. Dietary approaches that enhance antioxidant levels by incorporating polyphenol-rich foods may offer promising therapeutic options for the prevention of chronic metabolic diseases. This study aimed to evaluate the effects of standardised Aronia extract (SEA) and metformin (MET) on metabolic regulation in a rat model of PCOS. This research involved female Wistar albino rats, aged 6 weeks, with body weights ranging from 150 g to 170 g. PCOS was induced through subcutaneous administration of dehydroepiandrosterone during 5 weeks. Following successful confirmation of PCOS, the rats were categorized into the following four treatment groups: PCOS + distilled water, PCOS + MET, PCOS + SEA, and PCOS + MET + SEA. Results demonstrated that treatment with MET and SEA, both independently and in combination, significantly decreased final body weight, ovarian weight, total cholesterol, triglycerides, low-density lipoprotein (LDL), glycaemia, and insulin levels, while increasing high-density lipoprotein (HDL) levels compared to the PCOS control group. Additionally, the PCOS group showed a prolonged diestrus phase in the estrous cycle; whereas, the other treatment groups exhibited regular estrous cycles lasting 4–5 days. Cystic follicles significantly decreased, while corpora lutea significantly increased in all treated groups
Barbora Kalocayova1,2, Daria Kornieieva1, Matus Sykora1, Denisa Snurikova1, Kristina Ferenczyova1,2, Monika Bartekova1,3, Katarina Krskova4, Stefan Zorad4, Jana Vlkovicova1
Diabetic nephropathy is a complication of diabetes mellitus type 2 (DM2) that affects transmembrane enzymes such as Na,K-ATPase (NKA) and angiotensin-converting enzyme (ACE)-2. This study aimed to assess the effect of hyperglycaemia and quercetin (QCT) on these enzymes and the degree of renal tissue fibrosis. We used 6-month-old obese Zucker diabetic fatty (ZDF) rats as the model of DM2 and included lean ZDF rats as the control group. QCT (20 mg/kg/day) was orally administered to the rats for 6 weeks. The ZDF model exhibited varying glucose concentrations, resulting in the categorisation into groups with lower (9–11 mmol/L, D-↓, DQ-↓) and higher (> 11 mmol/L, D-↑, DQ-↑) hyperglycaemic levels. Glucose levels were positively correlated with both the Vmax value of NKA and hydroxyproline (a marker of fibrosis). We observed a significant upregulation in the expression levels of NKA (α1, β1) in the D-↓ group relative to the control group. However, this alteration did not significantly impact the expression of ACE2. QCT reduced the NKA activity in both control and DQ-↓ rats. The DQ-↑ group showed an increase in NKA and ACE2 expression compared to DQ-↓, while D-↑ had significantly lower NKA and ACE2 expression than D-↓. Administration of QCT reduced hydroxyproline in the DQ-group more significantly than in the DQ-↓ group. Administration of QCT to lean control ZDF rats and obese diabetic rats with a milder form of hyperglycaemia partially reduced NKA activity but mainly ACE2 and NKA expression. On the contrary, administration of QCT to obese diabetic rats with such severe hyperglycaemia significantly increased the expression of both enzymes, ACE2 and NKA. We found that QCT administration to obese ZDF rats had an anti-fibrotic effect regardless of the degree of hyperglycaemia, which we consider positive.
This project was supported by grants VEGA: 2/0148/22, 2/0104/20, 2/0159/24, 2/0123/24, APVV-20-0421, and APVV-21-0194.
Aleksandar Kocovic1,2, Nedeljko Manojlovic1, Jovana Novakovic1,2, Jelena Terzic1,2, Jovana Bradic1,2, Marijana Andjic1,2, Nevena Lazarevic1,2,3, Nevena Jeremic1,2,4, Vladimir Zivkovic2,5,6, Vladimir Jakovljevic2,3,5
Lichens represent a distinctive symbiotic association between fungi (mycobiont) and algae (photobiont). While lichens have been employed in traditional medicine and specific secondary metabolites have been explored for their effects, research on their cardioprotective properties remains limited. This study investigated the potential cardioprotective effects of the acetonic extract of Xanthoparmelia stenophylla (XSA) lichen in a rat model of doxorubicin-induced cardiotoxicity. The lichen sample was collected from the Stara Planina region. The acetone extract of XSA was prepared using cold maceration. This study involved 40 male Wistar albino rats, with the extract administered orally at a dose of 125 mg/kg for 28 days. Following the treatment, doxorubicin was administered intraperitoneally at a cumulative dose of 15 mg/kg. Three days after the final dose of doxorubicin, the hearts were isolated and examined ex vivo using the Langendorff apparatus. Coronary venous effluent samples were collected to assess oxidative stress markers via spectrophotometric analysis. Additionally, myocardial tissue samples were analysed using histological staining with haematoxylin and eosin. The administration of XSA at a dose of 125 mg/kg for 28 days significantly reduced oxidative stress parameters in the coronary venous effluent in a model of doxorubicin-induced cardiotoxicity. Furthermore, the architecture of the myocardial tissue was preserved, and degenerative changes were reduced in the group treated with XSA, indicating its potential as a cardioprotective agent. Our findings demonstrate that XSA possesses significant cardioprotective and antioxidant properties, suggesting its potential utility as a cardioprotective agent. Further investigation is required to elucidate the underlying mechanisms through which XSA or some of its ingredients exert its effects'.
This work was supported by the Ministry of Science, Technological Development and Innovation of the Republic of Serbia (Agreement No: 451-03-65/2024-03/200111 and 451-03-65/2024-03/200124)
Daria Kornieieva1,2, Matus Sykora1, Barbora Kalocayova1,3, Adriana Martišková1, Barbara Szeiffova Bacova1, Branislav Kura1
Despite many improving in medical procedures and techniques, heart failure (HF) still remains a global health issue with high rates of mortality, morbidity, and reduced quality of life. Among the key mechanisms implicated in HF development are oxidative stress and inflammation, resulting in myocardial damage, blood congestion, and poor tissue perfusion. Molecular hydrogen (H2) has emerged as a potential therapeutic agent mainly due to its strong antioxidant and anti-inflammatory effects. Intraperitoneal administration of isoproterenol is considered a reliable and proven model of HF induction in experimental animals. In this study, six-month-old male Wistar rats were divided into the following four groups: Control (C), isoproterenol-induced HF (ISO, 50 mg/kg for 5 consecutive days), HF with H2 treatment (ISO + H2, inhalation of 4% H2-enriched air for 30 min daily for 6 weeks), and HF with vitamin C treatment (ISO + C, 75 mg/kg in drinking water ad libitum). Biochemical markers such as lipid profile, glucose, aspartate aminotransferase (AST), alanine transaminase (ALT), and uric acid were colorimetrically analysed in blood plasma, while the expression of pro-inflammatory proteins, such as interleukins (IL-6), tumour necrosis factor alpha (TNF-α), and nuclear factor kappa B (NF-κB) in the heart tissue was assessed using the Western blot method. Isoproterenol administration led to a non-significant rise in uric acid, ALT, and AST levels, alongside a significant increase in blood glucose. In contrast, mostly H2 treatment slightly reduced all these parameters, approaching values seen in the control group. Expression of inflammatory proteins was notably elevated in the ISO group, while both treatment groups exhibited reduced protein levels, especially H2 treatment showing a more substantial decrease. Conclusion: H2 treatment attenuated inflammatory protein expression and improved biochemical parameters in ISO-induced HF, suggesting its therapeutic potential in cardiovascular diseases. These findings indicate that H2 inhalation offers a promising approach for novel HF treatment strategies.
This study was supported by grants VEGA 2/0092/22 and APVV 19-0317
Aleksandra Krivokuća1, Zorislava Bajić1, Tanja Šobot1, Anđela Bojanić1, Aneta Stojmenovski1, Snežana Uletilović1, Tanja Cvjetković1, Nebojša Mandić-Kovačević1, Maja Barudžija1, Sanja Jovičić1, Miloš Stojiljković1, Ranko Škrbić1
Cardiovascular disease (CVD) remains a leading cause of mortality and disability worldwide. Myocardial infarction (MI), a critical manifestation of CVD, arises from an imbalance between myocardial oxygen supply and demand. Isoprenaline (ISO), a synthetic non-selective β-adrenergic receptor agonist, has been utilized in experimental models to induce myocardial infarction in rats at high doses by disrupting the balance between free radical production and antioxidant defences. Urolithins, derived from the intestinal enzymatic breakdown of ellagic acid (EA), are notable gut metabolites. Among these, urolithin A is the most extensively studied and is recognized for its significant health benefits, including anti-inflammatory, anti-cancer, and antioxidant effects. Despite these promising findings, the specific effects of urolithin A on oxidative stress and myocardial injury in the isoprenaline rat model have not been thoroughly investigated. The aim of this study was to explore the cardioprotective effects of urolithin A against isoprenaline-induced myocardial injury in rats. All the animals were divided into the following four groups: control group (Con) (
Tatyana Kuropatkina1, Dmitrii Atiakshin2,3, Fedor Sychev4, Marina Artemieva1,4, Tatyana Samoilenko2, Olga Gerasimova2, Viktoriya Shishkina2, Khaydar Gufranov1, Natalia Medvedeva4, Tyler LeBaron5,6, Oleg Medvedev1,7
Hydrogen gas has demonstrated selective antioxidant properties, specifically against hydroxyl radicals, and possesses both antioxidant and anti-inflammatory effects. This study explored the therapeutic potential of hydrogen in a monocrotaline-induced model of pulmonary hypertension, which is characterized by oxidative stress—a common feature in cardiovascular diseases. Male Wistar rats were administered a subcutaneous injection of a water-alcohol solution containing monocrotaline to induce pulmonary hypertension or a control solution. One group of monocrotaline-treated rats was exposed to 4% molecular hydrogen in atmospheric air, while other groups were ventilated with standard atmospheric air. After 21 days, hemodynamic parameters, including systemic and pulmonary pressures, were measured under urethane narcosis. The results indicated that hydrogen inhalation did not significantly alter the primary markers of pulmonary hypertension. However, there was a significant reduction in systemic blood pressure, particularly in the systolic component. Furthermore, hydrogen exposure led to a decrease in TGF-β expression and a reduction in tryptase-positive mast cells, suggesting anti-inflammatory and vascular protective effects. These findings highlight hydrogen's potential as a therapeutic agent in mitigating cardiovascular complications associated with oxidative stress and inflammation.
This study was supported by the Russian Science Foundation, grant number 22-13-00111 (accessed on 11 October 2023) (Oleg Medvedev).
Simon Libak Haugaard1, Mélodie Schneider1, Sofie Troest Kjeldsen1, Stefan Michael Sattler2, Joakim Armstrong Bastrup3, Helena Carstensen1, Charlotte Hopster Iversen1, Ali Altintas4, Thomas Andrew Jepps3, Kate M. Herum5, Arnela Saljic2, Thomas Jespersen2, Sarah Dalgas Nissen1,2, Rikke Buhl1
Atrial fibrillation (AF) is a common arrhythmia in both horses and humans, sustained by electrical, structural, and metabolic changes that compromise treatment efficacy. This study investigates the molecular and electrophysiological remodelling associated with chronic AF and evaluates the potential of metformin, an indirect activator of AMP-activated protein kinase (AMPK), in mitigating these pathological changes. Twenty retired racehorses underwent AF induction via two weeks of right atrial tachypacing and were randomized to receive either metformin (
The project was funded by the Danish Independent Research Foundation (grant number 1032-00053B). SLH was funded by a research grant from the Danish Cardiovascular Academy, which is funded by the Novo Nordisk Foundation (grant number NNF20SA0067242) and the Danish Heart Foundation. MJS was funded by a research grant from the Danish Cardiovascular Academy, which is funded by the Novo Nordisk Foundation (grant number PhD 2022002-HF) and the Danish Heart Foundation.
Patricia Lima1, Dalton Vassallo1, Tatiani Nascimento1, Héber Silva2, César Filho2, Vinícius Marques1, Michael Dashwood3, Melchior Lima2
This study aimed to analyse the vascular reactivity of great saphenous vein (GSV) segments harvested using conventional, endoscopic, and No-Touch (NT) techniques in coronary artery bypass grafting (CABG). A total of 416 vein rings from 41 patients were studied, including 136 rings from the conventional technique (without perivascular fat), 104 from the NT technique (with perivascular fat), and 176 from the endoscopic technique (without perivascular fat). Vascular reactivity was evaluated based on the contractile response to phenylephrine, and nitric oxide (NO) bioavailability was assessed using the NO synthase inhibitor L-NAME. Results showed that vascular reactivity was significantly lower in the NT technique compared to the conventional (CV) and endoscopic (EVH) methods. The application of L-NAME revealed that the NT technique resulted in lower vascular contraction due to the influence of perivascular fat, which promoted vascular relaxation. NO detection was significantly higher in the NT technique, while the CV and EVH techniques showed limited NO presence and no significant change after L-NAME application. Morphological analysis confirmed the presence of perivascular fat in the NT technique and vessel stretching in the CV and EVH techniques due to hydraulic distention. In conclusion, the NT technique demonstrated reduced vasoconstrictive response and greater NO bioavailability, suggesting better vascular function and potentially lower risks of acute graft thrombosis compared to the CV and EVH techniques. The findings support the benefits of preserving perivascular fat in improving graft outcomes in CABG.
Viktória Líšková, Barbora Svetláková, Miroslav Barančík
Doxorubicin (DOX) is an anticancer drug with toxic effects on normal cells. Important role in DOX-induced cytotoxicity plays in increased production of reactive oxygen species (ROS) and oxidative stress. Increased ROS production can also induce autophagy, process playing an important role in maintaining normal cellular homeostasis through degradation of macromolecules. Autophagy can have a dual role in the cellular responses to pathological conditions. It may represent a beneficial adaptive response to stress but can also lead to maladaptive responses, which are linked to disease pathogenesis and cell-death induction. It has been found that excessive autophagy may lead to cell death. Several proteins, such as Beclin-1, p62, LC3A/B, and AuTophaGy-related (Atg), play an essential role in the regulation of autophagosome formation and the proper functioning of autophagy. To investigate the influence of short- and long-term exposure of HEK293 cells to effects of DOX on proteins involved in autophagy regulation. Human embryonic kidney cells HEK293 were used in studies. The cells were exposed to the effects of DOX for 3 h or 21 h. Protein samples obtained after cell lysis were used for the determination of levels of specific proteins using Western blot analysis. We found that DOX induced time-dependent changes in protein levels of studied autophagy markers. Prolonged exposure of HEK293 cells to 2.5 μmol/L DOX was associated with significant up-regulation of all studied autophagy markers (Atg5, Atg12, LC3A/B, and Beclin-1). Similar to the effects observed with long-term cell exposure to doxorubicin, Atg5 increases after 3-h of exposure to 2.5 μmol/L to DOX. Doxorubicin induced also an increase in LC3A/B, Atg5, and Atg3 protein levels after 3 h, with the most pronounced effects observed at a DOX concentration of 1 μmol/L. The results point to a significant role of proteins involved in autophagy processes in the mechanisms of DOX action on HEK293 cells. The effects of DOX were for some autophagy markers depended on the concentration of used DOX and the duration of cell exposure to this cytostatic agent.
This study was supported by grants VEGA 2/0169/24 and APVV-18-0548.
Natasa Macak1, Tamara Djuric1, Milica Dekleva2, Jovana Kuveljic1, Ivana Kolic1, Maja Zivkovic1, Ana Djordjevic1
Left ventricular (LV) dilatation following myocardial infarction (MI) is an important predictor of prognosis and indicates which patients may eventually develop heart failure (HF). The hallmark of systolic HF is a reduced LV ejection fraction (LVEF) due to impaired LV contractility, which can occur after MI as a result of cardiac remodelling and LV dilatation. Up-regulation of lncRNA Plasmacytoma Variant Translocation 1 (PVT1) has been observed in hypertrophy and myocardial fibrosis, and circulating PVT1 is a potential diagnostic biomarker for chronic HF. The aim of this preliminary study was to investigate the potential effects of the PVT1 gene variant rs4410871 on the parameters of LV structure and function, i.e., LV end-systolic volume (LVESV), LV end-diastolic volume (LVEDV), and LVEF, in patients after first acute MI. This study included 126 patients. The rs4410871 C > T variant was genotyped using Applied Biosystems TaqMan® technology. Statistical analyses were performed using Statistica 8 software. LVESV and LVEDV values > 62 mL/m2 and > 156 mL/m2 in men and > 47 mL/m2 and > 121 mL/m2 in women, respectively, were defined as dilatation. Patients with LVEF < 50% were defined as having impaired LVEF, i.e., systolic dysfunction. We found that genotypes carrying the T allele according to the dominant model (CC
This study was funded by the Ministry of Science, Technological Development and Innovation of the Republic of Serbia (Agreement No. 451-03-66/2024-03/200017).
Ugljesa Malicevic, Vikrant Rai, Devendra K. Agrawal
An arteriovenous fistula (AVF) is a surgically created connection between an artery and a vein that is used to provide vascular access for haemodialysis in patients with end-stage renal disease. Compared to a central venous catheter (CVC), AVF offers numerous benefits, including lower infection rates, reduced thrombosis, and improved patient survival. However, AVF is also associated with issues such as maturation failure, posing a significant medical challenge. AVF surgery triggers a complex interplay of cellular processes, including inflammation, proliferation, and differentiation. Muscle dissection and adjacent structural changes during AVF creation may have paracrine effects via vascular cuffing and inflammation, potentially influencing AVF maturation. Myogenic Differentiation 1 (MYOD1) is a key regulator of muscle differentiation, and its expression increases after muscle injury. This study aimed to examine the expression of MYOD1 and pro-inflammatory genes and proteins in an AVF swine model and assess the effect of a TREM-1 antagonist on their expression and AVF maturation. AVF was surgically created between the femoral artery (FA) and femoral vein (FV) using a side-to-side anastomosis in Yucatan miniswine. Swine were randomly divided into the following three groups: (I) Control with no treatment, (II) treatment with the TREM-1 antagonist, LR12 peptide, and (III) treatment with a scrambled peptide. LR12 or the scrambled peptide was administered at the AVF site. Three months after AVF creation and the application of LR12 or scrambled peptide, FA, FV, and surrounding tissues were harvested for histology, immunostaining, and qPCR to analyse the expression of various molecules, including MYOD1, MMP9, NF-κB, and TGFβ3. AVF maturation was assessed by ultrasound and angiography. Increased expression of mRNA transcripts and proteins for MYOD1, MMP9, NF-κB, and TGFβ3 was observed in the control and scrambled peptide groups. LR12 treatment significantly reduced the expression of all these molecules and was associated with less fibrosis and improved AVF patency, as revealed by ultrasound and angiography. The findings of this study suggest that targeting perivascular cuffing and muscle injury during AVF creation may offer potential therapeutic targets or biomarkers for early fibrosis in AVF maturation failure.
I am recipient of the Martin Morad Young Investigator Exchange Award.
Andrea Marciníková1, Csaba Horváth1, Izabela Jarabicová1, Petra Majerová2, Dominika Olešová2,3, Miroslav Ferko4, Natália Andelová4, M. Saadeh Suleiman5, Adriana Adameová1,4
Regulated forms of necrosis-like cellular demise have been shown to contribute to cardiac ischaemia/reperfusion (I/R) injury. Nonetheless, the underlying mechanisms occurring in the very first minutes of cardiac I/R remain partially elucidated. Thus, this study was conducted to provide a comprehensive protein screening focusing on pro-pyroptotic and mitochondrial damage-associated events. In Langendorff-perfused rat hearts subjected to 30 min I followed by ultrashort 10 min R. The proteomics was examined utilising liquid chromatography with mass spectrometry (LC-MS/MS) and immunoblotting techniques. Furthermore, calcium-induced mitochondrial swelling and lactate dehydrogenase (LDH) release were assessed to prove mitochondrial stress and necrosis-like phenotype, respectively. An assay determining adenosine 5′-triphosphatase (ATPase) activity was used to assess energy metabolism and overall mitochondrial function. About 160 proteins related to cell-death pathways, cellular metabolism, and post-translational modifications exhibited significant differential expression in hearts affected by such a short R. In spite of the signs of the plasma membrane structure disruption, conventional proteins involved in pyroptotic signalling, both canonical and non-canonical, were unchanged by this R interval. Importantly, this brief I/R was linked to the upregulation of p25 fragment of poly (ADP-ribose) polymerase 1 (PARP1 p25) and mature apoptosis-inducing factor (AIF) proteins, alongside nitrosative stress and mitochondrial swelling, while ATPase activity was lower. Conversely, a receptor-interacting protein kinase 3 (RIP3) inhibitor (GSK'872, 250 nmol/L) mitigated the elevation in PARP1 p25 and AIF expression, as well as prevented such a reduction of ATPase activity due to short R in previously ischaemic hearts. This study showed for the first time that during early I/R, necrosis-like injury is likely associated with RIP3-mediated mitochondrial, non-pyroptotic events, while the inhibition of RIP3 confers cardioprotective effects.
This study was supported by grants VEGA1/0016/20, APVV-20-0242, APVV-22-0264, and VEGA 2/0016/23
Sonja T. Marinković1,2, Žana M. Maksimović1, Tanja Šobot1,3, Đorđe Đukanović1,4, Sanja Jovičić1,5, Ranko Škrbić1,6, Miloš P. Stojiljković1,6
The isoprenaline (ISO) model of heart failure is a long-standing model. However, since its introduction in 1965, a wide range of approaches has been implemented. With the array of used doses and different application durations, various heart failure or myocardial damage models have been derived. This raises a challenge of how to choose the appropriate dose and duration, as well as the question of comparability and reproducibility of the results. This study aimed to compare time-related functional and structural changes of the ISO-induced model of chronic heart failure. Male Wistar rats, weighing 300–350 g, were used. The baseline echocardiogram was performed, and afterwards, an 85 mg/kg/day dose of ISO was applied subcutaneously for two consecutive days. Repeated echocardiograms and histopathology samples were obtained. At the 2-week time point, significant changes in left ventricular internal diameter end diastole and end systole (LVIDd and LVIDs) and end-diastolic and end-systolic volume (EDV and ESV) were noted. At the same time point, histological examination showed inflammatory infiltrate with initial fibrotic changes. However, no significant change in ejection fraction (EF) was found. After an additional week, further progression of heart failure was noted; however, there was no significance when compared to the two-week time point, and therefore, no samples for histopathology were obtained. Five weeks after the ISO application, the echocardiographic assessment showed a further increase of LVIDd, LVIDs, EDV, and ESV, as well as a significant reduction of the ejection fraction (< 55%). Furthermore, histological examination showed the progression of the fibrotic changes in the myocardium. Based on the results, a five-week period was found to be sufficient for the development of chronic heart failure using the isoprenaline model of 85 mg/kg/day subcutaneously for two consecutive days.
Rikke Marion-Knudsen1, Helena Dominguez1,2, Thomas Jespersen1, Arnela Saljic1
Atrial fibrillation (AF) is the most common arrhythmia in humans, affecting 59 million people worldwide. It is difficult to predict AF since initial symptoms are mild and passing. Once the disease progresses, symptoms become more prominent, and treatment is initiated. These challenges highlight the importance of researching the underlying mechanisms leading to AF, to start treatment earlier and improve quality of life. Studies show that the hearts of AF patients are characterised by remodelling of the tissue, and with the use of biomarkers, this remodelling can be examined. Based on preliminary results, the biomarker periostin is suggested as a new existing marker for structural remodelling in the heart. The project aimed to determine if periostin levels in heart biopsies and blood samples from patients undergoing open heart surgery can help predict AF. Heart biopsies and blood samples from 17 patients were included and grouped according to whether the patients developed post-operative AF (POAF) or stayed in sinus rhythm (SR) (SR:
Zuzana Matiko, Roman Moravčík, Michal Zeman
Elevated plasma levels of homocysteine (HCY) represent a risk factor for the development of several cardiovascular diseases. Better understanding of mechanisms how HCY can induce endothelial dysfunction are needed for prevention and treatment of these diseases. The aim of study was to investigate negative effects of HCY in the following two models: (1) Human endothelial cells isolated from the vein of human umbilical cord (HUVEC) and (2) hybrid immortalised endothelial cells (EA.hy926). We treated these endothelial cells with the indoleamine melatonin to attenuate HCY-induced endothelial damage. The cell viability was significantly decreased after 48 h HCY treatment in dose-dependent manner (1–6 mmol/L) in HUVEC. In contrast, only higher doses of HCY (4–6 mmol/L) decreased the viability of EA.hy926 cells. HCY significantly inhibited cell proliferation in HUVEC, and this negative effect was reversed by cotreatment with melatonin. Surprisingly, proliferation of EA.hy926 cells increased after 1–3 mmol/L HCY, and coincubation with melatonin did not influence cell proliferation. Morphology and cytoskeletal remodelling were analysed after 48 h exposure to 5 mmol/L HCY by immunocytochemistry. HUVEC and EA.hy926 responded differently to the treatment, and their morphology was dramatically changed compared to control cells. We observed an altered actin cytoskeleton and reduced cytoplasm in both types of endothelial cells. However, actin remodelling and structure change were not prevented by melatonin treatment. Based on results, we can conclude that both models of endothelial cells respond to hyperhomocysteinemia in partially different ways, and EA.hy926 cells are less sensitive. Findings highlight the importance of considering the specific characteristics of both models. The role of melatonin as a protective agent against hyperhomocysteinemia requires further investigation, especially in relation to its differential effects in various endothelial cell types.
This study was supported by grant APVV-21-0223.
Hana Mauer Sutovska1, Katarina Babarikova1, Pavel Svitok2, Libor Kopkan3, Michal Zeman1, Lubos Molcan1
Maternal-offspring interaction, behaviour, and the quantity and quality of maternal milk during early postnatal period can significantly impact the risk of cardiovascular disease later in life. In our study, hypertensive homozygous transgenic (mRen2) 27 rats (TGR) and normotensive Hannover Sprague-Dawley (HanSD) rats were used, with litters culled to eight pups per dam and cross-fostered, resulting in the following four groups: HanSD male pups fostered with HanSD mothers (HanSD), HanSD male pups fostered with TGR mothers (HanSDx), TGR male pups fostered with TGR mothers (TGR), and TGR male pups fostered with HanSD mothers (TGRx). Post-weaning, we implanted telemetry to measure blood pressure (BP) and heart rate in 5 to 12-week-old offspring. BP and heart rate were consistently higher in TGR than in HanSD offspring at all ages, regardless of fostering. On the base of heart rate beat-to-beat variability (marker of the autonomic nervous system), we found that TGR had a higher LF/HF ratio, indicating increased sympathetic activity, while HanSD showed a developmental decrease in LF/HF ratios into adulthood. Cross-fostering partially restored the normal decreasing LF/HF ratios in TGRx. At week 12, a salt load (2%) was introduced to assess its effects on BP and renal function. TGRx exhibited exaggerated BP responses to salt load, and cross-fostering in this strain accelerated renal injury. Cross-fostering did not prevent genetically determined hypertension in TGRx, although it reduced sympathetic activity. TGRx showed increased BP responses to salt, more severe renal damage, and altered sodium handling, possibly influenced by differences in the ionic composition of maternal milk between hypertensive and normotensive mothers. These findings highlight the importance of early maternal factors in hypertension development.
This study was supported by APVV-17-0178, APVV-21-0223, and ACCORD (the Operation Program of Integrated Infrastructure for the project, Advancing University Capacity and Competence in Research, Development and Innovation, ITMS2014 + : 313021 × 329, co-financed by the European Regional Development Fund) projects.
Zoran Miloradović1, Jelica Grujić Milanović1, Vesna Jaćević2, Slađan Milanović1, Milan Ivanov1, Una Jovana Vajić1, Danijela Karanović1, Nevena Mihailović Stanojević1, Đurđica Jovović1
Influence of natural polyphenol resveratrol on cardiorenal injuries induced by malignant hypertension (MH) has not been completely elucidated. The aim of this study was to examine long-term resveratrol treatment on cardiac and kidney structure, glomerular filtration, oxidative stress, and antioxidant defence in MH rats.
This study was performed on 6-month-old female spontaneously hypertensive rats (SHRs) with induced MH. MH was induced by 4-week L-NAME (10 mg/kg/day) treatment. Resveratrol groups (R) received resveratrol (10 mg/kg/day) during the same period. Animals were randomly divided into the following four groups: SHR (
Resveratrol attenuates heart and kidney damages in MH rats at the same way but less intensive, than in SHR, mainly due to its antioxidant activity. This promotes it as therapeutic agent against cardiorenal damages induced by malignant hypertension.
This study was supported by the Ministry of Education, Science and Technological Development of the Republic of Serbia, Grant No. 451-03-68/2022-14/200015.
Isidora Milosavljevic1, Jelena Terzic1, Aleksandar Kocovic1, Maja Muric2, Jasmina Sretenovic2, Vladimir Jakovljevic2,3
Lemon balm (
Gábor Mohácsi1, Leila Topal1, Aiman Saleh A Mohammed1, Muhammad Naveed1, Szilvia Déri1, Norbert Nagy1, László Virág1, Prof. István Baczkó1,2, Prof. András Varró1,2
Xention/Servier pharmaceutical companies developed XEN-D0103 as a selective inhibitor of IKur/Kv1.5 current to treat atrial fibrillation. The design of XEN-D0103 was motivated by the general consensus that IKur is expressed and functional primarily in the atrial tissue, with a minimal presence in the ventricular tissue. Consequently, it was anticipated that XEN-D0103 would circumvent the proarrhythmic side effects commonly linked to the currently available therapies. However, a previous study casted doubt on whether IKur is actually absent from the ventricles and started a contentious debate that remains unresolved within the scientific community. To provide further clarity on the expression of Kv1.5 channels within cardiac ventricular muscle and to elucidate the impact of Kv1.5 channel inhibition on the dynamics of repolarisation both on cardiac ventricular muscle and Purkinje fibres. Immunocytochemistry was utilised to detect the potential expression of Kv1.5 channels in isolated canine ventricular myocytes. Action potentials were recorded using standard conventional microelectrode techniques from both canine right ventricular papillary muscle and Purkinje fibres. Patch-clamp measurements were conducted to explore the impact of XEN-D0103 on isolated canine ventricular myocytes. Immunocytochemical analysis revealed abundant expression of Kv1.5 channels in canine ventricular myocytes. Action potential recordings demonstrated a significant prolongation of APD90 in both canine ventricular (244 ± 26 ms
Ana I Moreno-Manuel1, Álvaro Macías1, Francisco M Cruz1, Lilian K Gutiérrez1, Fernando Martínez-de Benito1,2, Andrés González-Guerra1, Patricia Sánchez-Pérez1, Isabel Martínez Carrascoso1, Francisco José Bermúdez-Jiménez3, María Linarejos Vera-Pedrosa1, Juan Manuel Ruiz-Robles1, Juan A Bernal1, José Jalife1,2,4
Short QT Syndrome Type 3 (SQTS3) is a rare arrhythmogenic channelopathy caused by gain-of-function mutations in KCNJ2, the gene coding the inward rectifier potassium channel Kir2.1 responsible for IK1. We aimed to enhance the understanding of SQTS3 by investigating its electrophysiologic and arrhythmogenic mechanisms. It is characterized by variable expressivity, which led me to hypothesised that the phenotype depends on the specific gain-of-function mechanisms triggered by each mutation. We used
Ministerio de Universidades (Spanish Government) awarded me the ‘Formación del Personal Universitario’ or FPU Fellowship [FPU20/01569] to conduct my PhD.
Lucia Mosná1, Miroslava Hardoňová2, Miroslav Vlček1, Pavol Šiarnik2, Andrea Havranová1, Adela Penesová1, Richard Imrich1, Ingrid Žitňanová3, Peter Turčáni2, Branislav Kollár2, Žofia Rádiková1
Multiple sclerosis (MS) is a chronic immune-mediated disease characterized by inflammation, demyelination, and axonal degeneration in the CNS, leading to motor impairment, visual disturbances, sensory problems, fatigue, and cognitive impairment. Patients with MS are at an increased risk of metabolic and cardiovascular comorbidities as well. Glucagon-like peptide-1 (GLP-1), an incretin secreted from the GIT after food intake, is not only involved in maintaining body weight, glucose, and lipid metabolism but also affects cognitive functions and acts as a neurotransmitter in the CNS. GLP-1 receptor agonists (GLP-1RA) are used in Type 2 DM treatment and have cardioprotective, neuroprotective, neurotrophic, anti-inflammatory, and antioxidant properties. The aim of our study was to assess effects of concomitant therapy with GLP-1RA on atherosclerosis risk and oxidative damage in treated people with MS. A total of 28 people (age: 39.4 ± 8.7 years, BMI: 25.0 ± 3.9 kg/m2) with relapse-remitting MS treated with natalizumab participated in this study. The subjects were randomly divided into the following two groups: – GLP1 group (
This study was supported by grant APVV-21-0261.
Maja Murić1,2, Jovana Novaković2,3, Vladimir Živković1,2,4, Jovana Joksimović Jović1,2, Jasmina Sretenović1,2, Nevena Lazarević2,3,5, Marijana Anđić2,3, Nemanja Murić6,7, Danijela Đoković6,7, Ksenija Vučićević2,3, Vladimir Jakovljević1,2,5
Heart failure (HF) is a complex syndrome often associated with oxidative stress. Empagliflozin and sacubitril/valsartan have shown individual benefits in HF management. In this study, we investigated whether the combination of empagliflozin and sacubitril/valsartan provides enhanced effects on oxidative stress in HF. Forty Wistar Albino rats were divided into control group (CTRL,
This work was supported by the Faculty of Medical Sciences, University of Kragujevac (JP 01/24) and the Ministry of Science, Technical Development, and Innovation of the Republic of Serbia (contract No. 451-03-65/2024-03/200111).
Reyhaneh Nejati Bervanlou1, Jan Graban1, Tatiana Ravingerova2, Daniela Jezova1, Natasa Hlavacova1
Exercise is a widely recognised intervention for preventing and treating cardiovascular diseases. One potential mediator of these benefits could be brain-derived neurotrophic factor (BDNF), which is released during exercise and may contribute to cardioprotection. While the role of BDNF is well known in neurocognition, its involvement in cardioprotection is underexplored. This study aimed to test the hypothesis that exercise-induced BDNF signalling via the tropomyosin receptor kinase B (TrkB) receptor in the heart is one of the mechanisms underlying improved cardiac function, particularly in hypertensive conditions. This study was performed in adult male Wistar (
This study was supported by grants Slovak Grant Agency VEGA 2/0158/22, VEGA SR 2/0104/22, APVV-19-0540, and APVV-20-0242.
Huynh Thanh Trai Nguyen
Despite the time passed since the SARS-CoV-2 pandemic, its negative effects persist, with patients experiencing complications linked to the infection and other health disorders. In terms of long-term outcomes, it became crucial to define a condition encompassing all such complications, now termed post-COVID syndrome. The wide range of these complications has led to the adoption of a multidisciplinary approach that identified organ-specific effects, though the underlying mechanisms of post-COVID remain unclear. This study aimed to explore potential immunological mechanisms in cardiac tissue that may contribute to the development of post-COVID syndrome.
The significant presence of autoantibodies in the samples suggests that they may play a role in the pathophysiology of post-COVID syndrome, potentially involving autoimmune responses. Additionally, the large number of detected autoantibodies points to potential issues with isotype-switching or other immune dysfunctions, raising hypotheses about immune system disruption or the alteration of proteins into autoantigens.
This study was supported by the University of Debrecen Scientific Research Bridging Fund (DETKA).
Marina Nikolic1,2, Aleksandra Stojanovic2,3, Andjela Milojevic Samanovic2,4, Milos Nikolic3, Ksenija Vucicevic2,3, Nikola Andric5,6, Slobodanka Mitrovic6,7, Nevena Jeremic2,3,8, Vladimir Jakovljevic1,2,9
This research aimed to evaluate the potential of using a combination of neprilysin inhibitor (sacubitril) and an AT1 receptor antagonist (valsartan) to induce white adipocyte browning and to evaluate whether this combined approach is more effective than cold exposure in promoting the formation of beige adipocytes. Forty male Wistar albino rats (8 weeks old; body weight: 200 ± 20g) were included in this study and further divided according to diet regime and applied treatment into the following four equal groups: CTRL + 4°C – healthy untreated rats; CTRL + E + 4°C – healthy rats treated with S/V; MetS + 4°C – rats with metabolic syndrome (MetS); and MetS + E + 4°C – rats with MetS treated with S/V. All rats were exposed to a low temperature (+4°C) for 4 h daily over a period of 4 weeks. Experimental groups of rats were administered per os a drug combination at a dosage of 68 mg/kg daily for 4 weeks. On completion of the experimental protocol, white adipose tissue was isolated for further analyses. The results of our study singled out combined cold exposure with S/V treatment as a more effective approach in inducing multilocular morphology of white adipocytes compared to untreated rats exposed to low temperatures alone. A significantly higher gene expression of thermogenic molecules was observed in white adipocytes of rats with MetS treated with S/V compared to expression found in animals from the MetS + 4°C group. In line with these results, the percentage of UCP1 stained adipocytes in MetS + E + 4°C was significantly higher in comparison to MetS + 4°C rats. In summary, due to more noticeable promotion of browning achieved by sacubitril/valsartan treatment in cold environment compared to untreated rats, the findings of this study indicate strong potential of this fixed drug combination to induce browning, making it an agent of quite similar potency in the formation of beige adipocytes as exposure to cold temperatures, which is defined as the gold standard.
This study was supported by the Faculty of Medical Science, University of Kragujevac, Grant/Award Number: 04/22; Ministry of Science, Technological Development and Innovation, Republic of Serbia, Grant/Award Number: 451-03-47/2023-01/200111.
Alisha Niskala1, Arnela Saljic1, Thomas Jespersen1, Dobromir Dobrev2,3,4
An enhanced inflammatory response is commonly observed in patients with atrial fibrillation (AF) contributing to increased cardiac fibrosis. Recent studies have identified that the expression of S100A8 and S100A9, calcium binding proteins, is significantly enhanced in the atrial tissue of patients with AF, with a 2.8-fold increase for S100A8 and 5.3-fold increase for S100A9. These proteins act as endogenous ligands for toll-like receptor 4 (TLR4) and, on binding, may downstream activate the NLRP3 inflammasome, a known key player in the pathogenesis of AF. However, the specific mechanisms by which S100A8/A9 proteins activate the inflammasome and their involvement in AF-related structural remodelling remains unclear. The aim of this project was to develop a macrophage-fibroblast co-culture system to explore the potential role of S100A8/A9 in fibroblast activation and their downstream pro-inflammatory effects. Given that S100A8/A9 proteins are released in response to enhanced inflammatory signals, selectively inhibiting atrial inflammation through these proteins could constitute novel therapeutic strategies to prevent or slow the progression of fibrosis driven AF. This study will examine the role S100A8/A9 in NLRP3 inflammasome activation and fibroblast activation using commercial mouse macrophage and fibroblast such as cell lines, human macrophages differentiated from isolated peripheral blood mononuclear cells (PBMCs), and human atrial fibroblasts. To activate the NLRP3 inflammasome and initiate macrophage-fibroblast interactions, cells will be treated with lipopolysaccharide from
Jovana Novakovic1,2, Tanja Jesic Petrovic3, Jovana Bradic1,2, Isidora Milosavljevic1,2, Nevena Jeremic1,2,4, Ivan Srejovic2,4,5, Aleksandar Kocovic1,2, Vladimir Zivkovic2,4,5, Vladimir Jakovljevic2,5,6
This study aimed to assess the impact of a 2-week administration of wild garlic (Allium ursinum) essential oil (WG) and allitridin (a biologically active compound found in wild garlic) on doxorubicin (DOX)-induced cardiotoxicity in a rat model. The experiment involved the following four groups: Healthy untreated rats (CTRL,
Gregoris Orphanides, Lujain Alsadder
Electrocardiography (ECG) is an invaluable tool in diagnosing cardiovascular diseases. This study introduced a novel method to reconstruct the heart's Three-Dimensional Electrical Current Vector (ECV3) by combining a subset of the standard 12-lead ECG leads. ECV3 was then used to estimate the perfusion angle and any other leads. This study aimed to investigate how ECV3 and perfusion angle could be used to assess electrophysiological changes in myocardial infarction. For this purpose, an open-source online ECG dataset from 45152 subjects was utilised. ECV3 was estimated using three independent components (x, y, and z) from the available ECG recordings. Lead I was marked as the x component, lead aVF as the z counterpart, and lead V2 as the y component. Based on the specifications of each lead measurement, trigonometric equations were applied to reconstruct ECV3 and use it to estimate the electrical activity in the other leads. The perfusion angle was measured as the mean angle between the S and T waves of the ECV3. ECV3 estimated leads yielded a significant cross-correlation for 94% of all patients in the database with a median correlation of 0.999 for limb leads and 0.829 for chest leads. The mean perfusion angle significantly decreased from an average of 140.7° for subjects with normal sinus rhythm (
Benjámin Paskuj, Muhammad Naveed, Gábor Mohácsi, Leila Topal, Norbert Nagy, Tibor Hornyik, András Varró, István Baczkó
Sodium-glucose cotransporter 2 (SGLT2) inhibitors have been shown to improve the clinical outcome of chronic heart failure (CHF) and atrial fibrillation (AF) and reduce the risk of arrhythmias and sudden cardiac death (SCD). The current ESC guidelines recommend the use of SGLT2 inhibitors as first-line agents in the treatment of CHF. Despite the outstanding clinical efficacy of SGLT2 inhibitors, the underlying mechanisms for their antiarrhythmic effects are poorly understood. Therefore, we aimed to investigate the cardiac electrophysiological effect of the following two SGLT2 inhibitors: Empagliflozin (EMPA) and Dapagliflozin (DAPA). Action potential (AP) measurements were carried out by conventional microelectrode technique using healthy canine and failing human (explanted) cardiac preparations. The effect of EMPA and DAPA on AP parameters [AP duration at 90% of repolarisation (APD 90), maximal rate of depolarisation (V max), AP amplitude (AMP), and conduction time (CT)] and their potential frequency dependency were measured. All data shown as mean ± SEM. Compared to control, 1 μmol/L and 3 μmol/L EMPA increased the maximal rate of depolarisation (166.5 ± 14.5 V/s
Bozidar Pindovic1, Milos Glisic2, Tamara Nikolic Turnic1,3,4, Vladimir Zivkovic2,4,5, Natalia Vasilievna Chichkova6, Vladimir Petrovich Fisenko5, Marina Nikolic2, Lazar Stijak7, Lemina Elena Yurievna8, Mirjana Veselinovic9,10, Milena Jovicic11, Katarina Mihajlovic1, Sergey Bolevich12, Vladimir Jakovljevic2,4,12
Data on the effects of different types of training activities on the functional recovery of the heart after ischaemia/reperfusion damage are still very inconsistent. An additional problem is the fact that it is still not known which type of training and its intensity (aerobic or anaerobic) achieves the best results in myocardial preconditioning. The aim of this study was to investigate the effects of exercise preconditioning on an animal model of ischaemia/reperfusion (I/R) injury in rat myocardium, specifically to determine the impact of different types of training processes on I/R, as well as the role of oxidative stress in the observed effects. This study was conducted on 40 rats divided into one control group (sedentary rats) and four experimental groups. Rats in the experimental groups underwent a training protocol for preconditioning through running and swimming (in both aerobic and anaerobic forms). After the training protocols, the animals were sacrificed, and the isolated hearts were perfused using the Langendorff retrograde perfusion method at a constant coronary perfusion pressure. Following stabilization, the hearts were subjected to global ischaemia for 20 min and then reperfusion for 30 min. Oxidative stress parameters were assessed in samples of coronary venous effluent as well as in the blood of rats using spectrophotometric methods. Both types of training were associated with increased production of cardiac pro-oxidants, with anaerobic intensity leading to the greatest increase in their production.
Tamás Bence Pintér, Petra Erzsébet Bíró, Attila Ádám Szabó, Enikő Edit Enyedi, Miklós Fagyas
Nowadays, the neprilysin inhibitor sacubitril in combination with an angiotensin II receptor antagonist is successfully used in the treatment of heart failure patients, which illustrates the prominent role of neprilysin (NEP) in the pathogenesis of cardiovascular diseases. By inhibiting the NEP enzyme, sacubitril significantly reduces the number of hospitalisations and mortality from cardiovascular causes. There are significant individual differences in the efficacy of the medication and the response to therapy, and we wanted to study the mechanisms underlying this. Serum NEP concentration was determined by commercially available ELISA kits and serum NEP activity by end-point fluorescence assay. The NEP gene was analysed by Sanger sequencing and the detected genetic variations by restriction fragment length polymorphism analysis. Serum NEP concentrations in cardiovascular patients showed a significant interindividual variation (mean: 35.41 ng/L [min-max: 0.38-10765 ng/L;
This study was supported by the EKÖP-24-3-II-DE-277 (A.Á.Sz.), EKÖP-24-3-II-DE-302 (E.E.E), and University Research Scholarship Programme (EKÖP) of the Ministry for Culture and Innovation from the source of the National Research, Development and Innovation Fund.
Žana Radić Savić, Vanja Vidović, Stojko Vidović, Jelena Bećarević, Mirna Popović Šarić, Ranko Škrbić
Oxidative stress is known to lead to cardiovascular diseases, including myocardial infarction (MI). The major antioxidant enzyme that maintains redox homeostasis and cellular response to oxidative stress is glutathione S-transferase P (GSTP), a member of the GST enzyme superfamily. Besides, new evidence suggests that this gene's polymorphisms can enable oxidative stress-related lipid abnormalities, influence lipid levels, and contribute to cardiovascular disease risk. The two most commonly occurring single nucleotide polymorphisms (SNPs), i.e., GSTPAB rs1695 and GSTPCD rs1138272, result in a deficiency of the GSTP antioxidant capacity. Therefore, we speculated that polymorphic variants of GSTP1 are related to MI risk and lipid traits in patients with MI. The case-control study comprised 75 patients with MI not receiving antilipemic therapy and 79 age and sex-matched controls. The GSTP1 genotypes were determined by qPCR using Applied Biosystems Taqman Drug Metabolism Genotyping assays. We found that SNP rs1138272 (*CT + TT) in the GSTP1 gene, associated with the lack of GSTP activity, modifies the risk for MI. Our results showed that carriers of variant GSTP1CD*CT + TT genotype are at almost five-fold increased risk of MI (OR = 4.68, 95% CI = 1.47–14.78,
Ivana Radosová1, Izabela Jarabicová1, Jaroslav Hrdlička2, Veronika Olejníčková2,3, Ján Neckář2, František Kolář2, Adriana Adameová1,4, Csaba Horváth1
Necroptosis, a regulated form of necrotic cell death, is associated with the pathomechanisms of various cardiovascular diseases. However, the relevance of the necroptotic process in cardiomegaly, especially considering potential sex differences, remains elusive. In this study, we used male and female Wistar rats in which cardiomegaly was induced by constriction of the abdominal aorta 2 days after birth. Animals were sacrificed by thiopental overdose after 90 days, and the left ventricular tissue was used for molecular analysis by SDS-PAGE/Western blotting. The left ventricular weight to the body weight ratio was significantly increased in both males and females after constriction of the abdominal aorta, while the right ventricular and lung weight to the body weight ratio was higher only in males. Echocardiographic analysis revealed decreased fractional shortening in both sexes, but ejection fraction was decreased only in males. Both sexes exhibited higher levels of the main necroptotic markers in the left ventricles – phosphorylated receptor-interacting protein kinase 3 (RIP3) and mixed lineage kinase domain-like protein (MLKL), indicating necroptosis activation. However, regarding the molecular processes associated with necroptosis, we found increased protein expression of tumour necrosis factor (TNF), interleukin 6 (IL-6), and cyclooxygenase 2 (COX-2) only in females, indicating a pro-inflammatory environment. Contrary, males were rather positive for upregulated expression of NADPH oxidase 2 and 4 (NOX2 and NOX4) and inducible NO synthase (iNOS), indicating the presence of pro-oxidative damage. Concerning the upstream receptors, females had higher expression of TNFR1 while had increased protein expression of TLR4 and MyD88. Conclusively, we indicated for the first time that although necroptosis is potentially involved in the pathomechanisms in both sexes, males exhibited more pronounced cardiac enlargement and pro-oxidative stress, while females were positive for a pro-inflammatory environment. Such data might advocate for a need for an individual pharmacological approach between the two sexes.
This study was supported by grants APVV-20-0242; APVV-15-0607, and VEGA 1/0016/20.
Táňa Ravingerova1, Veronika Farkašová1, Ľubomír Lonek1, Ján Graban2, Silvia Mužiková1,3, Vladislava Zohdi3,4
Cardiovascular diseases, especially ischaemic heart disease (IHD) as the leading cause of heart failure and mortality, will not reduce over the coming decades despite the progress in pharmacotherapy, interventional cardiology, and surgery. This represents a danger, especially in the elderly with hypertension and metabolic disorders. The infarct-sparing effect of ischaemic ‘preconditioning’ (IPC) is the most robust form of innate cardioprotection based on heart adaptation to moderate stress. However, its translation to clinical practice is limited. There are novel forms of adaptive interventions, such as ‘remote’IPC (RPC) and other non-invasive approaches, such as exercise-induced preconditioning (EPC) or adaptation to hypoxia (HPC). We explored that these non-pharmacological interventions in the adult male Wistar rats in vivo aimed to increase cardiac resistance to I/R ex vivo. RPC was induced by three cycles of 5-min inflation (200 mmHg)/5-min deflation of pressure cuff on the hind limb. For EPC, rats were placed in cages equipped with wheels for 2 weeks of free running. In a hypoxic chamber, HPC was induced by staying animals (10% O2, 4 weeks). The efficacy of these interventions was tested in the Langendorff-perfused hearts exposed to 30 min global ischaemia/2 h reperfusion, focused on the post-I/R functional recovery, arrhythmogenesis, and lethal injury (infarct size, IS). In parallel groups, heart tissue samples were processed (WB) to investigate the levels and activity of proteins involved in the ‘pro-survival’ RISK cascade. All interventions reduced contractile dysfunction, IS, and incidence and severity of reperfusion-induced arrhythmias. Cardioprotection was associated with significant up-regulation of selected pro-survival RISK proteins, such as PKB, PKCɛ, eNOS, and anti-apoptotic and anti-oxidative effects. Protective effects were maintained under conditions of hyperglycaemia and in the hearts of SHR rats. The beneficial effects of these ‘conditioning’ interventions suggest their potential that could be used in the management of ischaemic heart disease in patients with comorbidities.
This study was supported by grants Slovak Grant Agency VEGA SR 2/0104/22, APVV-19-0540, and APVV-20-0242.
David Revuelta1,2, Andrea Brunetti1,2, Franziska Reinhardt3, Sven Margenburg3, Simon Pecha3, Cristina E. Molina1,2
Atrial fibrillation (AF), the most common arrhythmia, is associated with altered Ca2+-handling in atrial myocytes. Up-regulation of adenosine A2A receptors in AF has been linked to increased protein kinase A (PKA)-mediated phosphorylation of RyR2, leading to spontaneous sarcoplasmic reticulum Ca2+ release. It is well accepted that 3′5′-cyclic adenosine monophosphate (cAMP)/PKA signalling is compartmentalised into specific nanodomains due to the action of phosphodiesterases (PDEs), the enzymes that degrade cAMP. The aim of this study was to characterise cAMP dynamics in key Ca2+-handling nanodomains on A2A receptor activation and to study the roles of PDE3, PDE4, and PDE8 in human atrial myocytes (HAMs) from patients in sinus rhythm (SR) and with AF. A total of 198 HAMs were isolated from atrial samples of 45 patients undergoing cardiac surgery. Cells were transfected with adenoviral vectors encoding FRET biosensors targeted to measure cAMP specifically in main Ca2+-handling nanodomains: The sarcolemma, cytosol, RyR2, or phospholamban (PLN)/SERCA2a. After 48 h of culture to allow for biosensor expression, FRET live imaging was used to measure cAMP levels. Our results revealed larger cytosolic cAMP increases in AF on A2A receptors activation with CGS. However, cAMP increase in the RyR2 nanodomain was lower in AF, indicating that the reported increase in PKA-mediated phosphorylation of RyR2 in AF might be surprisingly mediated by cytosolic cAMP. PDE3 and PDE4 were key regulators of cytosolic and PLN/SERCA2a compartments, though PDE4 regulation was reduced in AF. The overall response in the sarcolemma was low, and only PDE4 seemed to regulate this compartment. Finally, PDE8 emerged as a novel regulator of the A2A response in RyR2, aligning with recent electrophysiological studies in murine models. These findings highlight the complex PDE-mediated regulation of cAMP in the adenosine A2A response in HAMs, offering new insights into how cAMP signalling is altered in AF. Understanding these nanodomain-specific disruptions could provide potential therapeutic targets for restoring normal cAMP signalling in patients with AF.
Patricia Sánchez-Pérez1, Álvaro Macías1, Ana I Moreno-Manuel1, Lilian K Gutiérrez1, María Linarejos Vera-Pedrosa1, Francisco M Cruz1, Isabel Martínez-Carrascoso1, Juan M Ruiz-Robles1, Juan Antonio López1, Manuel J Gómez-Rodríguez1, Francisco M Espinosa2, Ricardo García2, José Jalife1,3,4
The advent of human-induced pluripotent stem cell-derived cardiomyocyte (hiPSC-CM) technology may revolutionise cardiac medicine. However, the use of hiPSC-CMs has been restricted due to their immature phenotype, more similar to foetal than adult cardiomyocytes. Multiple strategies have been proposed for improving hiPSC-CM maturation, but the most promising are those that mimic the cardiac microenvironment, provided in part by the extracellular matrix (ECM). In this study, we proposed a human cardiac decellularized ECM to enhance the structural and functional maturation of hiPSC-CMs compared to other commercial matrices. Our matrix derives from the deposition of ECM components of human cardiac fibroblasts aligned in micropattern on polydimethylsiloxane (PDMS) for 7 days, providing heart stiffness (≈ 10 kPa), ECM components, and scaffold to mature hiPSC-CMs. We also tested on our matrix that an excess of ECM deposition, becoming a more fibrotic ECM, is counterproductive to hiPSC-CM functional maturation. The human cell matrix significantly increases sarcomere length and alignment, cell circularity index, anisotropy, and the expression of structural genes such as CTnT and actinin in hiPSC-CMs. Also, the provided scaffold and stiffness of our matrix improve Conexin 43 and N-Cadherin location. Most importantly, this decellularized human cardiac matrix improves hiPSC-CM function in terms of metabolism and electrophysiology, which is a principal limitation of these cells. Patch-clamp and optical mapping experiments revealed an adult-like cardiomyocyte phenotype: maximum diastolic potential (MDP) (≈-78 mV), amplitude (≈115 mV), upstroke velocity (≈130 V/s), and AP duration at 90% repolarization (APD90) (≈300 ms). NaV1.5 expression and sodium current (INa) are similar to the adult cardiomyocyte. In addition, action potential conduction velocity (≈40 cm/s) in hiPSC-CM monolayers cultured in our matrix is close to the adult heart. In conclusion, we present a cost-effective and easy-to-make platform that mimics the ECM cardiac microenvironment. This platform represents an optimal strategy for hiPSC-CM maturation, which will help investigate molecular mechanisms of inheritable arrhythmogenic diseases.
Merve Sayin1, Pinar Cevikbas Ilik1, Yasemin Atici2, Belma Turan3
A magnolia bark extract with the active component mainly magnolol (MAGNO) is in a group of bioactive phytochemicals. Studies mentioned that this compound has antioxidant-like actions, known as an uncommon antioxidant. The action of MAGNO includes a marked protection against increased oxidative stress in cells through various signalling pathways. In addition, it has been demonstrated that the magnolia bark extract radicals do not react with molecular oxygen and produce no superoxide radical under the typical settings of inhibited autoxidation. Therefore, in this study, we treated either insulin-resistant male elderly (24-month-old) rats/mice or doxorubicin-applied male adult rats with MAGNO (100 mg/kg) for 12–14 weeks besides their standard chow. When compared with untreated groups (elderly or adult animals), MAGNO treatment significantly provided antioxidant-like effects on biochemical parameters of heart tissues such as increases in the activity of arylesterase and total antioxidant levels as well as significant decreases in the activity of the mitochondrial enzyme succinate dehydrogenase, one of the sources of reactive oxygen species and the total oxidant level with a significant increase in the ratio of ATP to ADP. MAGNO treatment of doxorubicin-applied rats (before or after) provided significant recoveries in the systemic parameters of animals such as plasma levels of manganese and zinc, total oxidant, and antioxidant statuses. Furthermore, we determined important cardioprotective effects of this treatment such as improvements in the redox regulation of the hearts, such as marked improvements in activities of glutathione peroxidase and glutathione reductase and oxygen radical-absorbing capacities in these doxorubicin-treated animals. Overall, our data presented significant myocardial ameliorations in physiological ageing-associated biochemical alterations in the metabolic heart with MAGNO supplementation as well as significant cardioprotection against doxorubicin toxicity. Finally, one can propose the beneficial antioxidant effects of MAGNO in chronic heart diseases as a supporting and complementing agent to conventional therapies.
This study was supported by the Lokman Hekim University Scientific Research Projects Coordination Unit. Project No. 202AP404, 2022.
Mélodie Schneider1, Simon Libak Haugaard1, Sarah Dalgas Nissen1, Sheyla Barrado Ballestero2, Rikke Buhl1, Urmas Roostalu2
Activation of the autonomic nervous system (ANS) and local sympathetic hyperinnervation plays a pivotal, yet poorly understood role in the initiation and maintenance of atrial fibrillation (AF). Changes in the intricate three-dimensional (3D) network of nerves are difficult to characterise using traditional histological methods. To gain insights into the role of innervation in cardiac arrhythmia, non-destructive imaging techniques capable of visualising large tissue samples from large animal models of AF are crucial. In this study, we aimed to investigate the feasibility of 3D light sheet fluorescence microscopy (LSFM) in equine atrial tissue and provide a detailed 3D characterisation of the sympathetic nervous system in the epicardium, myocardium, and endocardium, while exploring innervation changes in a horse model of experimentally induced chronic AF. Biopsies from the left posterior atrial wall were harvested from horses after 4 months of induced AF (
This work was supported by a research grant from the Danish Cardiovascular Academy and the Danish Heart Foundation (PhD 2022002-HF).
Jan Slezak
Heart transplantation, although routine, faces significant challenges due to ischaemia reperfusion injury (I/RI) after cold ischaemic storage. Reperfusion with warm, oxygenated blood generates reactive oxygen species (ROS) such as nitrosyl and hydroxyl radicals, exacerbating cell damage. In addition, extracorporeal circulation (ECC) induces systemic oxidative stress, further complicating transplant outcomes. This study investigated the therapeutic potential of hydrogen gas in reducing I/RI by targeting hydroxyl radicals, reducing lipid peroxidation, inflammation, and increasing antioxidant enzyme activity. Porcine hearts stored in cold ischaemia for 3 h were perfused with blood after 2% inhalation and blood saturation in an ECC oxygenator with 2% hydrogen gas in air. Pigs treated with hydrogen showed significantly less ventricular fibrillation and improved histopathological results compared to controls. Hydrogen administration significantly reduced systemic oxidative stress, markers of ischaemia, inflammation, and lipid peroxidation. Hydrogen gas delivered via conventional ventilators and ECC oxygenators shows promise as an adjunctive therapy in heart transplantation, improving post-transplant cardiac function and ameliorating systemic oxidative stress.
This research was supported by grants from the Agency for Research and Development Support (APVV-0241-11, APVV-15-0376, APVV-19-0317), the Slovak Academy of Sciences (VEGA 2/0092/22 and 2/0063/18), the Structural Funds of the European Union (ITMS 26230120009), and the Ministry of Health of the Slovak Republic (2019/4-CEMSAV-1).
Paula G Socuéllamos1,2, Fran M Cruz1, Álvaro Macías1, Alba Vera-Zambrano2, Ana I Moreno-Manuel1, Carmen Prior3, Teresa González2, Antonio J Cartón3, Carmen Valenzuela2,4, José Jalife1,4,5
Nav1.5 channels, encoded by SCNA5, are essential for the genesis of the cardiac action potential (AP) and contribute to its duration. Gain-of-function mutations in SCNA5 are associated with long QT syndrome (LQTS); whereas, loss-of-function mutations are linked with Brugada syndrome. MOG1 was described as a partner of the Nav1.5 channelosome, increasing Nav1.5 current amplitude and membrane expression. Two loss-of-function variants in the MOG1 gene (E61X and E83D) have been found in patients with Brugada Syndrome, but no relationship with LQTS has been reported. We have identified the first gain-of-function variant in MOG1 (L18F) in a patient with LQTS. The patient presented complete atrial-ventricular (AV) block, prolonged QT intervals and ventricular tachycardia. We generated AAV9-mediated cardiac-specific mouse models expressing MOG1WT and MOG1L18F. MOG1L18F mice recapitulated the patient's phenotype, with prolonged QT interval corrected for heart rate on surface ECG, and an increased incidence of arrhythmia on programmed electrical stimulation. In current-clamp experiments, the incidence of delayed afterdepolarizations (DADs) and spontaneous APs was higher in MOG1L18F compared to MOG1WT cardiomyocytes (CMs). AP duration at 90% of repolarisation (APD90) was dramatically increased at low stimulation frequencies (0.1–2 Hz) in MOG1L18F but very similar to MOG1WT CMs at higher frequencies (from 4 Hz to 10 Hz), resulting in a very steep APD90 restitution curve in MOG1L18F, which was proarrhythmic. On voltage clamping, late sodium current (INaL) was extremely enhanced in MOG1L18F CMs compared to MOG1WT, especially at depolarised membrane potentials. This effect was mediated by Nav1.8, at least in part. The increase in INaL opposed repolarising currents, lengthening the AP and the QT, ultimately leading to ventricular tachyarrhythmias. In conclusion, we demonstrated for the first time that cardiac-specific expression of the L18F gain-of-function variant of MOG1 in live mice impairs AV conduction and increases INaL, which prolongs the APD and the QT interval and provides a new genetic basis for LQTS.
Ivan Srejovic1,2,3, Maja Muric1,2, Jovana Novakovic2,4, Vladimir Zivkovic1,2,3, Jasmina Sretenovic1,2, Marina Nikolic1,2, Nevena Lazarevic2,4,5, Marijana Andjic2,4, Aleksandar Kocovic2,4, Vladimir Jakovljevic1,2,5
Heart failure (HF) continues to be a major global health challenge, characterised by impaired cardiac function and remodelling. While both empagliflozin and sacubitril/valsartan have demonstrated individual efficacy in HF, their combined effects have not been thoroughly investigated. Understanding the potential synergistic benefits of these therapies could lead to more effective management of HF. To evaluate the combined effects of empagliflozin and sacubitril/valsartan on cardiac function and myocardial remodelling in a HF rat model. Forty male Wistar Albino rats were initially allocated into the following two groups: A healthy control group (
This study was supported by the Faculty of Medical Sciences, University of Kragujevac (JP 01/24) and the Ministry of Science, Technical Development, and Innovation of the Republic of Serbia (contract No. 451-03-65/2024-03/200111).
Aleksandra Stojanovic1,2, Marina Nikolic2,3, Jasmina Sretenovic2,3, Goran Marjanovic4, Tatjana Vulovic5,6, Vladimir Jakovljevic2,3,7
Type 1 diabetes mellitus (T1DM) is regarded as autoimmune disorder, primarily manifested during early childhood and adolescence rather than adulthood. Individuals with T1DM have greater cardiovascular and total morbidity and mortality than general population. The return of consciousness and alertness after anaesthesia may be accompanied by heart failure, high blood pressure, increased abdominal and intracranial pressure, and myocardial ischaemia. This study aimed to examine the effect of thiopental (TIO) monotherapy as well as its combination with different agents used in anaesthesia induction, on cardiac function and redox state of rats with T1DM. A total of 40 Wistar albino male rats were used in this study and randomly divided into the following five groups: TIO, fentanyl + thiopental (FEN + TIO), remifentanil + hiopental (REM + TIO), midazolam + thiopental (MID + TIO), and dexmedetomidine + thiopental (DEX + TIO). After T1DM induction was confirmed, all animals were subjected to a short narcosis of tested anaesthetic or combination of anaesthetics by intraperitoneal injection: TIO 85 mg/kg, FEN 0.005 mg/kg, REM 0.04 mg/kg, MID 2.5 mg/kg, and DEX 0.05 mg/kg of body weight. The hearts were retrogradely perfused according to Langendorff technique. The parameters of the redox status were evaluated from coronary venous effluent and blood samples. Our results demonstrated that REM in combination with TIO had the most prominent positive effect on cardiac function due to significantly improved parameters of cardiac contractility and enhanced antioxidative capacity. Moreover, combination of TIO with other drugs, especially with DEX and MID, had depressive effects on cardiac contractility, HR, and parameters of oxidative stress as well as histological changes. Given that diabetes is an increasingly common entity in the population, this research supplemented knowledge about the effects of anaesthetic agents (belonging to different pharmacological groups, with different pharmacokinetic properties) on myocardial function, oxidative stress, and histological changes in conditions of diabetes.
Aneta Stojmenovski1, Andjela Bojanic1, Milica Gajic Bojic1,2, Nebojsa Mandic Kovacevic1,3, Snezana Uletilovic1,4, Sanja Jovicic1,5, Milka Maticic1, Aleksandra Smitran1,6, Ranko Skrbic1,2
Hyperhomocysteinemia, characterised by elevated levels of homocysteine, is a known risk factor for vascular dysfunction, potentially leading to structural changes in blood vessels and can serve as a predictor for pre-eclampsia. While magnesium sulphate is the primary treatment for the prevention and management of seizures in severe pre-eclampsia and eclampsia, diazepam may also be used in certain situations. The aim of this study was to investigate the effects of diazepam on oxidative status and histological alterations of human umbilical arteries in a hyperhomocysteinemia model. Umbilical arteries, isolated from the umbilical cord of healthy women, were cleaned and cut into strips 2–3 mm long. These arterial rings were incubated in Krebs-bicarbonate solution for 3 h at 37°C, aerated with a mixture of 95% oxygen and 5% carbon dioxide. This study included the following four treatment groups: Control (only buffer solution), diazepam (100 μmol/L), homocysteine (1 mmol/L), and combination diazepam (100 μmol/L) + homocysteine (1 mmol/L). The impact of diazepam was assessed by measuring oxidative stress markers, including thiobarbituric acid reactive substances (TBARS), NO2, superoxide dismutase (SOD), glutathione (GSH), and catalase (CAT). Additionally, histological examination was performed to evaluate structural changes in the arterial wall, using classic haematoxylin-eosin, as well as pentachrome, Movat staining. Findings indicate that diazepam significantly (
Jakub Strapec1, Kristína Ferenczyová1,2, Lucia Kindernay1, Ulrika Duľová1, Ľubomíra Tóthová3, Monika Barteková1,4
Quercetin (QCT), a plant-derived flavonoid, provides cardiovascular benefits. Previous studies showed its protective effect on ischaemia-reperfusion (I/R) injury in young, healthy animals. This study investigated its cardioprotective potential in ageing rats. We administered QCT (20 mg/kg/day orally) to 2-year-old aged rats for 6 weeks. Afterwards, the animals were anesthetised, and their hearts were excised, placed on a Langendorff apparatus and subjected to 30 min of ischaemia followed by 120 min of reperfusion. The infarct size was assessed via TTC staining, and cardiac function recovery was monitored during the first 40 min of reperfusion. Western blotting was used to evaluate the expression of reperfusion injury salvage kinase (RISK) pathway proteins (Akt, PKC-ɛ, eNOS, GSK-3β), antioxidant enzymes (SOD1, SOD2), and apoptosis markers (Bax/Bcl-2). The oxidative status was assessed using markers, thiobarbituric acid reactive substances (TBARS), advanced oxidation protein products (AOPP), ferric reducing ability of plasma (FRAP), fructosamine, and advanced glycation end-products (AGEs), in blood plasma. According to our research, there was no cardioprotective advantage from extended QCT treatment in the heart's recovery after ischaemia. In fact, the impact of QCT was associated with a tendency towards decreasing cardiac functional recovery. In line with physiological data, QCT was unable to activate the RISK signalling pathway in the aged hearts. On the contrary, QCT decreased the cardiac Bax/Bcl-2 ratio suggesting its anti-apoptotic effect in aged rat hearts. Finally, elevated FRAP and AOPP levels suggest that QCT may have a pro-oxidant impact on ageing hearts. According to our results, QCT had less cardioprotective effectiveness in older individuals, most likely as a result of insufficient RISK pathway activation and decreased antioxidant efficiency. Although QCT exerts anti-apoptotic effect, this does not confer protection against I/R injury, making its use for cardioprotection in ageing populations unpromising.
This study was supported by grants APVV-21-0194, VEGA 2/0104/20, and VEGA 2/0159/24.
Adrian Sturza1, Valentina Buda2, Daliana Minda3, Cristina A. Dehelean4, Zorita Diaconeasa5, Cristian Iuhas6, Rodica Lighezan7, Corina Danciu3, Danina M. Muntean1,
Black chokeberry (Aronia melanocarpa) is a rich source of dietary phytochemicals, mainly the bioactive polyphenols. This study performed in mice aortic rings was aimed to complement previous research on the vascular protective effects of phytochemicals by assessing the
This study was supported by the University internal grant 5DOC/1425/03.02.2020.
Tamara Szabados1, András Makkos2, Márta Szabó2, Imre Vörös2, Anikó Görbe2, Péter Ferdinandy2,3, Péter Bencsik1,3
MicroRNA (miRNA)-based RNA therapeutics are advancing, offering novel approaches to treat various diseases by restoring miRNA expression patterns. Although there is a growing body of literature investigating the function and biology of miRNAs, data on miRNA pharmacokinetics are severely limited, leading to unanticipated pitfalls. We have previously identified several microRNAs as ProtectomiRs against myocardial ischaemia/reperfusion injury, such as miR-125b* and miR-450a. To investigate their potential use as RNA therapeutics, we performed pharmacokinetic studies in mice. A single injection of 10 μg miR-125b* mimic or its scramble miRNA control or vehicle alone (neutral lipid emulsion, NLE) was administered to C57BL/6 mice intravenously. MiR-125b* expression was measured from plasma and left ventricle (LV) at 1, 2, 4, 8, and 24-h post-injection. MiR-125b* level in blood plasma and in LV showed a marked increase at 1-h post-injection, which was decreased at later time points. Time-matched vehicle and scramble miRNA did not alter miR-125b* expression at any time point in plasma or in LV. For the next step of investigation, the vehicle compound NLE was not available on the market anymore; therefore, we have chosen a widely used cation-based new vehicle, jetPEI®. The pharmacokinetic studies of miR-450a with jetPEI and saline have been performed. As a pilot experiment, separate groups of C57BL/6 mice were administered 0.25 mg/kg or 2.5 mg/kg miR-450a intravenously dissolved either in jetPEI or saline. MiR-450a expression was measured from blood plasma and LV at 1- and 60-min post-injection and was increased at both time points only when dissolved in jetPEI. We repeated miR-450a pharmacokinetics with jetPEI using several sampling points (at 1 min and 1, 2, 4, 8, and 24-h post-injection) only at 0.25 mg/kg dose and found that miR-450a expression was significantly increased in LV only at 1 min. Pharmacokinetic properties of miRNAs are not well-understood yet; therefore, such pioneering experiments are of utmost need.
This study was supported by the National Heart Program (NVKP 16-1-2016-0017), Cooperative Doctoral Program (KDP-2020), János Bolyai Research Scholarships of the Hungarian Academy of Sciences (bo_481_21), EKÖP-24-4, and EKÖP-398.
Jelena Terzic1,2, Aleksandar Kocovic1,2, Nedeljko Manojlovic1, Jovana Novakovic1,2, Jovana Bradic1,2, Marijana Andjic1,2, Nevena Lazarevic1,2,3, Nevena Jeremic1,2,4, Vladimir Zivkovic2,5,6, Vladimir Jakovljevic2,3,5
Usnic acid (UA), a natural dibenzofuran secondary metabolite present in various lichens, has garnered extensive research interest due to its diverse biological activities. Its potential therapeutic applications are significant, spanning antimicrobial, antitumour, and antioxidant effects. Despite the historical use of lichen-derived UA in traditional medicine and the established benefits of UA, there is a notable gap in research concerning its cardioprotective properties. This study aimed to investigate the impact of UA on doxorubicin-induced cardiotoxicity in a rat model. UA was extracted from the acetonic extract of the lichen Xanthoparmelia stenophylla and identified through comparison with a standard reference. This study involved 40 male Wistar albino rats. UA was administered orally at a dose of 25 mg/kg daily for 28 days. Following this treatment period, doxorubicin was administered intraperitoneally at a cumulative dose of 15 mg/kg. Three days after the doxorubicin administration, the rats' hearts were excised and examined ex vivo using a Langendorff apparatus. Coronary venous effluent samples were collected to assess oxidative stress markers via spectrophotometry. Additionally, myocardial tissue samples were analysed using histological staining with haematoxylin and eosin. Administration of UA at a dose of 25 mg/kg for 28 days leads to the alleviation of oxidative stress parameters in coronary venous effluent in a model of doxorubicin-induced cardiotoxicity. Also, in the group treated with UA, the architecture of the myocardial tissue was preserved, and degenerative changes are reduced. Our findings demonstrate that UA possesses significant cardioprotective and antioxidant properties, suggesting its potential utility as a cardioprotective agent. Further investigation is required to elucidate the underlying mechanisms through which UA exerts its effects.
This study was supported by the Ministry of Education, Science, and Technological Development of the Republic of Serbia (Agreement Nos. 451-03-9/2021-14/200111 and 451-03-68/2022-14/200378).
Leila Topal1, Eleonora Torre2, Jenő Pintér1, Aimen Mohammed1, Naveed Muhammad1, Alaa Almagid1, Luca Soattin3, Tibor Hornyik1, Zoltán Husti1, Vivien Demeter-Haludka1, Tamás Zombori4, Alexandra Polyák5, László Virág1, Norbert Jost1, István Baczkó1, András Varró1
Paradoxically, endurance athletes are prone to several cardiac arrhythmias, including atrial fibrillation (AF). Current guidelines suggest ‘an approximately five-fold increased lifetime risk of atrial fibrillation compared to sedentary individuals, despite a lower prevalence of traditional atrial fibrillation risk factors’. Since no comprehensive study has been conducted in a large animal model relevant to humans; therefore, the purpose of our study was to determine the underlying cardiac changes caused by the long-term intense endurance exercise that led to increased susceptibility to arrhythmias. Male beagle dogs were randomised into a ‘Sedentary’ (SED) and an ‘Exercised’ (EXE) group (
This study was supported by EKÖP-24-4 - Tudománnyal fel! Felsőoktatási Doktorvárományosi és Posztdoktori Kutatói Ösztöndíj; EKÖP-403.
Vanja Vidović1,2, Jelena Bećarević1,2, Nikola Malešević2, Nela Maksimović3, Žana Radić Savić2,4, Irina Milovac1,2, Tamara Kovačević Preradović5, Mirna Popović Šarić3, Stojko Vidović1,2, Ranko Škrbić2,6
The β2 adrenergic receptors (ADRB2) are expressed on the various types of cells, including cardiomyocytes and white adipocytes, playing an important role in cardiovascular system control, glucose, and insulin maintenance as well as energy expenditure and lipolysis. Among the ADRB2 gene, Arg16Gly and Gln27Glu polymorphisms have been linked to the risk of myocardial infarction (MI) and altered lipid metabolism; therefore, the aim of this study was to assess the risk of the selected polymorphisms with MI and with mean values of lipid parameters in patients from the Republic of Srpska. The case-control study comprised 75 patients with MI who were not taking lowering lipid drugs and 79 age and sex-matched healthy controls. The ADRB2 genotypes were determined by qPCR. The results have shown that in the group of patients, carriers of GA or AA genotypes among Arg16Gly polymorphism have statistically lower mean values of high-density lipoprotein (HDL) cholesterol in comparison to the carriers of GG genotype (
Jana Vlkovicova1, Barbora Kalocayova1,2, Denisa Snurikova1, Katarina Krskova3, Stefan Zorad3, Tijana Culafic4, Snezana Tepavcevic4, Norbert Vrbjar1
Excessive fructose consumption is associated with metabolic changes and serious health problems. Regular drinking of fructose-sweetened beverages results in obesity, inflammation, and cardiometabolic disorders. In addition, high fructose intake increases oxidative stress. Therefore, administration of antioxidant quercetin, which naturally occurs in fruits and vegetables, could protect against fructose-induced renal impairment. This study aimed to investigate the renal consequences of a 15-week intake of 10% fructose, focusing on the properties of the enzyme Na,K-ATPase and the effect of supplementation with the antioxidant quercetin. We confirmed experimentally that when drinking a 10% fructose solution for 9 weeks, young Wistar rats developed several signs of insulin resistance, as indicated by glucose, insulin, and homeostasis model assessment of insulin resistance (HOMA-IR) values. At the same time, we also focused on monitoring the effect of quercetin administered by gavage for 6 weeks (20 mg/kg/day in 1% methylcellulose solution). Rats from control groups received methylcellulose vehicles. Our study demonstrated that elevated fructose was associated with increased body weight and plasma glucose levels in the rats. Oral administration of quercetin normalised the intake of the fructose solution to levels comparable to water intake. Furthermore, quercetin administration significantly reduced glycaemia. Through the action of polyphenols, we expected protection from renal damage during the development of metabolic disease and preservation of tubular Na,K-ATPase functionality, but this was not confirmed. Also, the increase in plasma creatinine and the decrease in the glutathione GSH/GSSG ratio in the renal tissue homogenate suggest that quercetin administration may not be beneficial in a state of pre-existing renal pathology.
This study was supported by grants APVV-20-0421, VEGA 2/0123/24, and VEGA 2/0148/22.
Barbara Wysokińska1, Jerzy Bełtowski2, Aleksandra Skora3, Wiktor Gaik3
Hypertension in children is an emerging clinical issue increasingly recognised for its potential to cause early vascular damage and increase cardiovascular risk in adulthood. The pathogenesis of primary hypertension (PHT) in the paediatric population has been linked to oxidative stress, with paraoxonase 1 (PON1) playing a crucial role. PON1, primarily associated with high-density lipoproteins (HDL), hydrolyses lipid peroxides, mitigating oxidative stress and protecting against low-density lipoprotein (LDL) oxidation, which is a significant factor in endothelial dysfunction and atherosclerosis. One of the natural PON1 substrates, homocysteine thiolactone, is involved in the detrimental vascular effect of homocysteine by spontaneously reacting with proteins (protein N-homocysteinylation). Several clinical studies demonstrated the relationship among PON1 deficiency, protein N-homocysteinylation, and cardiovascular disease in adults; however, this relationship in children has not been studied so far. The aim of this study was to analyse PON1 status and N-homocysteinylation of plasma proteins in children with primary hypertension. We analysed 97 children/adolescents aged 11–17 years: 46 with primary hypertension and 51 without cardiovascular abnormalities. Surprisingly, PON1 activity and concentration were significantly higher in the hypertensive group compared to controls. HDL-cholesterol as well as apolipoproteins A-I and A-II did not differ between groups. Despite higher PON1 activity against homocysteine thiolactone in the hypertensive group, the degree of N-homocysteinylation of proteins did not differ significantly from the control group. Additionally, lipolactonase activity of PON1 (activity towards thiobutylbutyrolactone) was lower in hypertensive children with obesity than in those with normal body weight. Hypertension had no effect on PON3 concentration and activity. In conclusion, PON1 concentration and activity are higher in children and adolescents with arterial hypertension, suggesting detrimental role of the enzyme in its pathogenesis. Obesity impairs lipolactonase activity of PON1, which is crucial for its antioxidant activity.
Hannah Zhang1,2, Aleksandra Stamenkovic2, Craig Resch2, Ashish Shah1,2, Amir Ravandi1,2
Cardiogenic shock (CS) is a life-threatening state characterised by acutely failing heart function, and despite significant advances in therapy options, CS continues to have a high in-hospital mortality rate of nearly 50%. Lack of standardised defining features of CS leads to delay in treatment, resulting in significant morbidity and mortality. Given the rapid metabolic shifts that occur during CS, we aimed to define the plasma lipidomic shifts during acute myocardial infarction (AMI)-related CS in hemodynamically well-defined patient population. Plasma samples and detailed hemodynamic assessment by non-invasive cardiac system (NICaS) were obtained for 259 patients presenting with ST-elevation myocardial infarction (STEMI) prior to their intervention. Plasma lipidomics was performed using liquid chromatography/tandem mass spectrometry (LC/MS/MS). The lipidomic shifts seen in patients presenting with did not correlate with systolic blood pressure or cardiac index. High lactate levels were negatively correlated with several lysophosphatidylcholine (LPC) species and positively correlated with acylcarnitines. There was a significant reduction in 18 lipid species in patients with high lactate levels, including several LPCs. Hemodynamic assessment of these patients showed that heart rate, stroke index, and Granov Goor Index (GGI, a proxy for left ventricular ejection fraction) had significant correlations with LPC levels, while cardiac output and cardiac index had few or no correlation to lipid profile, respectively. Furthermore, low LPC and high carnitine levels had a significant correlation with in-hospital mortality in patients with cardiogenic shock. In our study, lack of LPC production in setting of acute MI is a predicator of poor cardiovascular hemodynamics and, as a result, in-hospital mortality. This indicates that low plasma phospholipase activity could be used as a marker of poor clinical outcomes and can be a novel therapeutic avenue in cardiogenic shock patients.
This study was supported by the Heart and Stroke Foundation.