693 results on '"Cerbai, Elisabetta"'
Search Results
302. Prenatal Exposure to Carbon Monoxide Delays Postnatal Cardiac Electrophysiological Maturation: Focus on Functional and Molecular Properties of f-Current.
- Author
-
Sartiani, Laura, Stillitano, Francesca, Brogioni, Simona, Suffredini, Silvia, Tattoli, Maria, Carratù, Maria R, Cuomo, Vincenzo, Mugelli, Alessandro, and Cerbai, Elisabetta
- Published
- 2005
303. Spatio-Temporal Relationship Between T-Tubular Electrical Activity and Ca2+Release in Heart Failure
- Author
-
Crocini, Claudia, Coppini, Raffaele, Ferrantini, Cecilia, Yan, Ping, Loew, Leslie M., Tesi, Chiara, Cerbai, Elisabetta, Poggesi, Corrado, Pavone, Francesco S., and Sacconi, Leonardo
- Published
- 2014
- Full Text
- View/download PDF
304. Acute appendicitis in a patient immunised with COVID‐19 vaccine: A case report with morphological analysis.
- Author
-
Marconi, Ettore, Crescioli, Giada, Bonaiuti, Roberto, Pugliese, Lavinia, Santi, Raffaella, Nesi, Gabriella, Cerbai, Elisabetta, Vannacci, Alfredo, and Lombardi, Niccolò
- Subjects
- *
COVID-19 , *COVID-19 vaccines , *APPENDICITIS , *COVID-19 pandemic , *COMPUTED tomography - Abstract
Although the benefit/risk profile for mRNA COVID‐19 vaccines is recognised as extremely favourable, appendicitis is currently considered an adverse event (AE) of special interest. We describe the case of a 58‐year‐old female who presented with signs and symptoms of appendicitis approximately 48 hours after her first injection of the Pfizer‐BioNTech vaccine. Abdominal ultrasound revealed fluid collection in the right iliac fossa and cecal wall thickening. Following the surgical visit, CT scan with contrast showed a distended appendix with thickened walls, suggestive of acute appendicitis. The patient tested negative to upper respiratory COVID‐19 reverse transcription‐polymerase chain reaction. Clinical trials and observational studies suggest a possible association between appendicitis and COVID‐19 vaccines. Th‐1 driven granulomatous inflammation reported in our case represents an infrequent nonspecific chronic inflammation of the appendix, especially in the setting of delayed or interval appendectomy. In view of the current paediatric vaccination campaign, we recommend monitoring the safety profile and potential gastrointestinal AEs associated with mRNA COVID‐19 vaccines to swiftly manage subjects with gastrointestinal symptoms and prevent potential complications. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
305. Spatiotemporal Relationship between Ca2+Release and Action Potential in Cardiomyocytes Probed by Random Access Microscopy
- Author
-
Sacconi, Leonardo, Crocini, Claudia, Coppini, Raffaele, Ferrantini, Cecilia, Yan, Ping, Loew, Leslie, Tesi, Chiara, Cerbai, Elisabetta, Poggesi, Corrado, and Pavone, Francesco
- Published
- 2013
- Full Text
- View/download PDF
306. Electrophysiological characterization of cardiac β2-adrenoceptors in sheep Purkinje fibers
- Author
-
Cerbai, Elisabetta, Masini, Ilaria, and Mugelli, Alessandro
- Published
- 1990
- Full Text
- View/download PDF
307. Photoresponsive Polymer‐Based Biomimetic Contractile Units as Building Block for Artificial Muscles.
- Author
-
Vitale, Giulia, Grandinetti, Bruno, Querceto, Silvia, Martella, Daniele, Tesi, Chiara, Poggesi, Corrado, Cerbai, Elisabetta, Wiersma, Diederik S., Parmeggiani, Camilla, Ferrantini, Cecilia, and Sacconi, Leonardo
- Subjects
- *
ARTIFICIAL muscles , *CARDIAC contraction , *SMART materials , *NEUROMUSCULAR diseases , *STRIATED muscle , *ARTIFICIAL implants , *BIOMIMETIC materials - Abstract
Loss of muscular mechanical function occurs in several diseases affecting millions of people worldwide, including heart failure, stroke, and neuromuscular disorders. To date, no medical or surgical treatments can restore muscular contractility, and the development of artificial muscles is of extreme interest. Mimicking biological muscles, which are optimized systems displaying quick reaction times, is not trivial; only few examples are reported, mainly focused on the use of biomimetic smart materials. Among them, liquid crystalline elastomers (LCEs) can be biocompatible, show contraction parameters comparable to those of native striated muscles, and are able to effectively potentiate cardiac contraction in vitro. To go further and develop in vivo implantable devices, the integration of the stimulation system with the LCE material represents an essential step. Here, a light‐stimulated biomimetic contractile unit (BCU), combining ultra‐thin photoresponsive LCE films and mini‐LED (mLED) matrixes is described. BCU performance (in terms of extent and kinetics of contractile force and shortening) can be fine‐tuned by modulating both mLED light power and spatial stimulation patterns, allowing to reproduce mechanical dynamics of native muscles. These results pave the way for the development of novel LCE‐based contraction assist devices for cardiac, skeletal, or smooth muscle support by assembling multiple BCUs. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
308. Modulation of the pacemaker current If by {beta}-adrenoceptor subtypes in ventricular myocytes isolated from hypertensive and normotensive rats
- Author
-
Cerbai, Elisabetta, Pino, Roberto, Rodriguez, Maria L, and Mugelli, Alessandro
- Abstract
Objective: Both β1- and β2-adrenoceptors (β1-AR and β2-AR) are functionally present in human and rat ventricular myocytes. The two receptor subtypes are differently regulated during the development of myocardial hypertrophy and failure. If is expressed in human and rat ventricular myocytes. In hypertrophied myocytes isolated from old spontaneously hypertensive rats (SHR) the density is much larger than in age-matched normotensive Wistar Kyoto (WKY). Due to the possible relevance of If as an arrhythmogenic mechanism in the rat and human ventricle, we studied and compared the effects of β1-AR and β2-AR stimulation on If in both hypertrophied and normal left ventricular myocytes of 18-month old SHR and WKY. Methods: The whole-cell configuration of the patch-clamp technique was employed. Noradrenaline (NA, 1 µM) was used to stimulate β1-AR and isoprenaline (ISO, 1 µM) in the presence of the β1-AR antagonist CGP 20712A (0.1 µM) to stimulate β2-AR. Results: In SHR, NA increased If by causing a 10.8±0.9 mV (n=10) positive shift in the voltage of maximal activation (V1/2); this effect was completely reversed by CGP 20712A. β2-AR stimulation was effective in seven out of 13 cells tested, where it caused a small positive shift in V1/2 (4.0±1.7 mV). Cyclopentyladenosine (CPA), a selective A1-receptor agonist, reversed the effect of NA; the antiadrenergic action of CPA was abolished in cells pre-incubated with pertussis toxin (PTX) to block inhibitory G proteins (Gi). In PTX-treated cells the shift in V1/2 caused by both β2-AR (9.6±1.7 mV, n=6, p<0.05) and β1-AR (17.6±1.9 mV, n=7, p<0.05) was significantly greater than in control cells. Both β-AR subtypes modulated If activation also in WKY: β1-AR shifted V1/2 by 16.0±1.4 mV (n=15) and β2-AR by 4.2±1.1 mV (n=7). However, in PTX-treated WKY cells only the β2-AR effect was potentiated (shift in V1/2: 11.4±1.4 mV, n=9, p<0.01), while the β1-AR response was unchanged (18.9±4.2 mV, n=5, n.s.). Conclusions: If expressed in SHR hypertrophied ventricular myocytes is modulated by catecholamines mainly through the stimulation of the β1-AR subtype. The β1-AR response is, however, significantly lower than that observed in myocytes from normotensive rats, probably as a consequence of the presence of an increased inhibitory activity of Gi proteins. This post-receptorial control may be seen as a mechanism to limit the arrhythmogenicity of β-AR stimulation in myocardial hypertrophy and failure.
- Published
- 1999
- Full Text
- View/download PDF
309. Electrophysiological alterations caused by free radicals in isolated myocytes
- Author
-
Cerbai, Elisabetta, Visentin, Sergio, Ambrosio, Giuseppe, and Mugelli, Alessandro
- Published
- 1990
- Full Text
- View/download PDF
310. Quantification of midkine gene expression in Patella caerulea (Mollusca, Gastropoda) exposed to cadmium
- Author
-
Stillitano, Francesca, Mugelli, Alessandro, Cerbai, Elisabetta, and Vanucci, Silvana
- Subjects
- *
GENE expression , *GENETIC regulation , *RISK management in business , *GASTROPODA - Abstract
Abstract: The release of cadmium into many coastal areas represents a threat to ecosystems and human health; cadmium is carcinogenic in mammals and in both marine invertebrates and vertebrates. The use of molluscs to assess the ecologic risk associated with contaminants is strongly recommended on account of their ecological role and on their highly conserved control and regulatory pathways that are often homologous to vertebrate systems. We previously identified a midkine family protein in the limpet Patella caerulea; the midkine is a recently discovered cytokines family with unequivocal informative value on repairing injury and neoplastic processes in mammals. Here we report on midkine (mdk) and α-tubulin (α-tub) gene expression patterns in P. caerulea exposed to cadmium. Limpets, collected on two occasions from a breakwater at a marina (Tyrrhenian Sea) were exposed to sublethal cadmium concentrations (0.5 and 1mgl−1 Cd) over a 10-day exposure period. RNA was extracted from the viscera of unexposed and exposed specimens. Real time TaqMan RT-PCR was performed to measure the relative mdk and α-tub gene expression levels. A remarkable mdk over-expression was observed in all exposed animals with respect to unexposed ones; mdk over-expression was significantly higher in both treatments when compared with un-treatment (mean expression levels: 23- and 38-fold, for 0.5 and 1mgl−1 Cd treatment, respectively; ANOVA, for both P <0.01). The study also indicates that the mdk up-regulation was significantly Cd-concentration dependent (P <0.05). A significant up-regulation of the constitutive α-tub gene was also observed in 1mgl−1 Cd-treated animals (mean expression level: 4-fold; ANOVA, P <0.05). In conclusion, these data provide the first evidence paving the way for the use of the midkine as a promising new biomarker of effect in the environment risk assessment policy. [Copyright &y& Elsevier]
- Published
- 2007
- Full Text
- View/download PDF
311. Optical clearing in cardiac imaging: A comparative study.
- Author
-
Olianti, Camilla, Giardini, Francesco, Lazzeri, Erica, Costantini, Irene, Silvestri, Ludovico, Coppini, Raffaele, Cerbai, Elisabetta, Pavone, Francesco S., and Sacconi, Leonardo
- Subjects
- *
CARDIAC imaging , *DIAGNOSTIC imaging , *HYPERTROPHIC cardiomyopathy , *STRUCTURAL reliability , *FLUORESCENT dyes , *TRANSPARENCY (Optics) , *STAINS & staining (Microscopy) , *HEART - Abstract
The optical clearing of the cardiac tissue has always been a challenging goal to obtain successful three-dimensional reconstructions of entire hearts. Typically, the developed protocols are targeted at the clearing of the brain; cardiac tissue requires proper arrangements to the original protocols, which are usually tough and time-consuming to figure out. Here, we present the application of three different clearing methodologies on mouse hearts: uDISCO, CLARITY, and SHIELD. For each approach, we describe the required optimizations that we have developed to improve the outcome; in particular, we focus on comparing the features of the tissue after the application of each methodology, especially in terms of tissue preservation, transparency, and staining. We found that the uDISCO protocol induces strong fiber delamination of the cardiac tissue, thus reducing the reliability of structural analyses. The CLARITY protocol confers a high level of transparency to the heart and allows deep penetration of the fluorescent dyes; however, it requires long times for the clearing and the tissue loses its robustness. The SHIELD methodology, indeed, is very promising for tissue maintenance since it preserves its consistency and provides ideal transparency, but further approaches are needed to obtain homogeneous staining of the whole heart. Since the CLARITY procedure, despite the disadvantages in terms of tissue preservation and timings, is actually the most suitable approach to image labeled samples in depth, we optimized and performed the methodology also on human cardiac tissue from control hearts and hearts with hypertrophic cardiomyopathy. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
312. The HCN channel as a pharmacological target: Why, where, and how to block it.
- Author
-
Balducci, Valentina, Credi, Caterina, Sacconi, Leonardo, Romanelli, Maria Novella, Sartiani, Laura, and Cerbai, Elisabetta
- Subjects
- *
PHYSIOLOGY , *PACEMAKER cells , *CENTRAL nervous system , *CARDIAC pacemakers , *HEART cells , *HYPERPOLARIZATION (Cytology) - Abstract
Hyperpolarization-activated, cyclic nucleotide-gated (HCN) channels, expressed in a variety of cell types and in all tissues, control excitation and rhythm. Since their discovery in neurons and cardiac pacemaker cells, they attracted the attention of medicinal chemistry and pharmacology as novel targets to shape (patho)physiological mechanisms. To date, ivabradine represents the first-in-class drug as specific bradycardic agent in cardiac diseases; however, new applications are emerging in parallel with the demonstration of the involvement of different HCN isoforms in central and peripheral nervous system. Hence, the possibility to target specific isoforms represents an attractive development in this field; indeed, HCN1, HCN2 or HCN4 specific blockers have shown promising features in vitro and in vivo, with remarkable pharmacological differences likely depending on the diverse functional role and tissue distribution. Here, we show a recently developed compound with high potency as HCN2–HCN4 blocker; because of its unique profile, this compound may deserve further investigation. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
313. Edward Carmeliet (1930–2021)—channelling scientific curiosity: a tribute from the ESC Working Group on Cardiac Cellular Electrophysiology†.
- Author
-
Ravens, Ursula, Gomez, Ana M, Heijman, Jordi, Remme, Carol Ann, Dobrev, Dobromir, Smith, Godfrey, Volders, Paul G A, Cerbai, Elisabetta, Eisner, David A, Casadei, Barbara, Zaza, Antonio, Richard, Sylvain, Mugelli, Alessandro, Vassort, Guy, Brown, Hilary F, and Sipido, Karin R
- Subjects
- *
LIFE sciences , *CURIOSITY , *MEDICAL sciences , *POTASSIUM channels , *ION channels - Published
- 2021
- Full Text
- View/download PDF
314. Targeting Cyclic Guanosine Monophosphate to Treat Heart Failure: JACC Review Topic of the Week.
- Author
-
Emdin, Michele, Aimo, Alberto, Castiglione, Vincenzo, Vergaro, Giuseppe, Georgiopoulos, Georgios, Saccaro, Luigi Francesco, Lombardi, Carlo Mario, Passino, Claudio, Cerbai, Elisabetta, Metra, Marco, and Senni, Michele
- Subjects
- *
HEART failure , *NUCLEOTIDES , *TREATMENT effectiveness , *ANGIOTENSIN receptors , *STROKE volume (Cardiac output) - Abstract
The significant morbidity and mortality associated with heart failure with reduced ejection fraction (HFrEF) or heart failure with preserved ejection fraction (HFpEF) justify the search for novel therapeutic agents. Reduced cyclic guanosine monophosphate levels contribute to HF progression. Among molecules modulating the nitric oxide (NO)-GMP-phosphodiesterase (PDE) pathway, the evaluation of nitrates, synthetic natriuretic peptides (NP), and NP analogs has yielded mixed results. Conversely, sacubitril/valsartan, combining NP degradation inhibition through neprilysin and angiotensin receptor blockade, has led to groundbreaking findings in HFrEF. Other strategies to increase tissue cyclic guanosine monophosphate have been attempted, such as PDE-3 or PDE-5 inhibition (with negative or neutral results), NO-independent soluble guanylate cyclase (sGC) activation, or enhancement of sGC sensitivity to endogenous NO. Following the positive results of the phase 3 VICTORIA (A Study of Vericiguat in Participants With Heart Failure With Reduced Ejection Fraction) trial on the sGC stimulator vericiguat in HFrEF, the main open questions are the efficacy of the sacubitril/valsartan-vericiguat combination in HFrEF and of vericiguat in HFpEF. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
315. The hyperpolarization-activated cyclic nucleotide-gated 4 channel as a potential anti-seizure drug target.
- Author
-
Kharouf, Qays, Phillips, A. Marie, Bleakley, Lauren E., Morrisroe, Emma, Oyrer, Julia, Jia, Linghan, Ludwig, Andreas, Jin, Liang, Nicolazzo, Joseph A., Cerbai, Elisabetta, Romanelli, M. Novella, Petrou, Steven, and Reid, Christopher A.
- Subjects
- *
DRUG target , *THALAMIC nuclei , *SEIZURES (Medicine) , *KNOCKOUT mice , *RESEARCH , *ANIMAL experimentation , *RESEARCH methodology , *MEDICAL cooperation , *EVALUATION research , *NUCLEOTIDES , *COMPARATIVE studies , *DRUGS , *MEMBRANE transport proteins , *RESEARCH funding , *MEMBRANE proteins , *MICE - Abstract
Background and Purpose: Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels are encoded by four genes (HCN1-4) with distinct biophysical properties and functions within the brain. HCN4 channels activate slowly at robust hyperpolarizing potentials, making them more likely to be engaged during hyperexcitable neuronal network activity seen during seizures. HCN4 channels are also highly expressed in thalamic nuclei, a brain region implicated in seizure generalization. Here, we assessed the utility of targeting the HCN4 channel as an anti-seizure strategy using pharmacological and genetic approaches.Experimental Approach: The impact of reducing HCN4 channel function on seizure susceptibility and neuronal network excitability was studied using an HCN4 channel preferring blocker (EC18) and a conditional brain specific HCN4 knockout mouse model.Key Results: EC18 (10 mg·kg-1 ) and brain-specific HCN4 channel knockout reduced seizure susceptibility and proconvulsant-mediated cortical spiking recorded using electrocorticography, with minimal effects on other mouse behaviours. EC18 (10 μM) decreased neuronal network bursting in mouse cortical cultures. Importantly, EC18 was not protective against proconvulsant-mediated seizures in the conditional HCN4 channel knockout mouse and did not reduce bursting behaviour in AAV-HCN4 shRNA infected mouse cortical cultures.Conclusions and Implications: These data suggest the HCN4 channel as a potential pharmacologically relevant target for anti-seizure drugs that is likely to have a low side-effect liability in the CNS. [ABSTRACT FROM AUTHOR]- Published
- 2020
- Full Text
- View/download PDF
316. Abnormalities in sodium current and calcium homoeostasis as drivers of arrhythmogenesis in hypertrophic cardiomyopathy.
- Author
-
Coppini, Raffaele, Santini, Lorenzo, Olivotto, Iacopo, Ackerman, Michael J, and Cerbai, Elisabetta
- Subjects
- *
HYPERTROPHIC cardiomyopathy , *BRUGADA syndrome , *VENTRICULAR tachycardia , *CARDIAC arrest , *TACHYARRHYTHMIAS , *VENTRICULAR fibrillation , *GENETIC disorders - Abstract
Hypertrophic cardiomyopathy (HCM) is a common inherited monogenic disease with a prevalence of 1/500 in the general population, representing an important cause of arrhythmic sudden cardiac death (SCD), heart failure, and atrial fibrillation in the young. HCM is a global condition, diagnosed in >50 countries and in all continents. HCM affects people of both sexes and various ethnic and racial origins, with similar clinical course and phenotypic expression. The most unpredictable and devastating consequence of HCM is represented by arrhythmic SCD, most commonly caused by sustained ventricular tachycardia or ventricular fibrillation. Indeed, HCM represents one of the main causes of arrhythmic SCD in the young, with a marked preference for children and adults <30 years. SCD is most prevalent in patients with paediatric onset of HCM but may occur at any age. However, risk is substantially lower after 60 years, suggesting that the potential for ventricular tachyarrhythmias is mitigated by ageing. SCD had been linked originally to sports and vigorous activity in HCM patients. However, it is increasingly clear that the majority of events occurs at rest or during routine daily occupations, suggesting that triggers are far from consistent. In general, the pathophysiology of SCD in HCM remains unresolved. While the pathologic and physiologic substrates abound and have been described in detail, specific factors precipitating ventricular tachyarrhythmias are still unknown. SCD is a rare phenomenon in HCM cohorts (<1%/year) and attempts to identify patients at risk, while generating clinically useful algorithms for primary prevention, remain very inaccurate on an individual basis. One of the reasons for our limited understanding of these phenomena is that limited translational research exists in the field, while most efforts have focused on clinical markers of risk derived from pathology, instrumental patient evaluation, and imaging. Specifically, few studies conducted in animal models and human samples have focused on targeting the cellular mechanisms of arrhythmogenesis in HCM, despite potential implications for therapeutic innovation and SCD prevention. These studies found that altered intracellular Ca2+ homoeostasis and increased late Na+ current, leading to an increased likelihood of early and delayed after-depolarizations, contribute to generate arrhythmic events in diseased cardiomyocytes. As an array of novel experimental opportunities have emerged to investigate these mechanisms, including novel 'disease-in-the-dish' cellular models with patient-specific induced pluripotent stem cell-derived cardiomyocytes, important gaps in knowledge remain. Accordingly, the aim of the present review is to provide a contemporary reappraisal of the cellular basis of SCD-predisposing arrhythmias in patients with HCM and discuss the implications for risk stratification and management. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
317. Hyperpolarization-activated cyclic-nucleotide-gated channels: pathophysiological, developmental, and pharmacological insights into their function in cellular excitability1.
- Author
-
Spinelli, Valentina, Sartiani, Laura, Mugelli, Alessandro, Romanelli, Maria Novella, and Cerbai, Elisabetta
- Subjects
- *
HYPERPOLARIZATION (Cytology) , *BLADDER pacemakers , *ATRIAL arrhythmias , *CARDIAC hypertrophy , *CELL survival - Abstract
The hyperpolarization-activated cyclic-nucleotide-gated (HCN) proteins are voltage-dependent ion channels, conducting both Na+ and K+, blocked by millimolar concentrations of extracellular Cs+ and modulated by cyclic nucleotides (mainly cAMP) that contribute crucially to the pacemaker activity in cardiac nodal cells and subsidiary pacemakers. Over the last decades, much attention has focused on HCN current, If, in non-pacemaker cardiac cells and its potential role in triggering arrhythmias. In fact, in addition to pacemakers, HCN current is constitutively present in the human atria and has long been proposed to sustain atrial arrhythmias associated to different cardiac pathologies or triggered by various modulatory signals (catecholamines, serotonin, natriuretic peptides). An atypical If occurs in diseased ventricular cardiomyocytes, its amplitude being linearly related to the severity of cardiac hypertrophy. The properties of atrial and ventricular If and its modulation by pharmacological interventions has been object of intense study, including the synthesis and characterization of new compounds able to block preferentially HCN1, HCN2, or HCN4 isoforms. Altogether, clues emerge for opportunities of future pharmacological strategies exploiting the unique properties of this channel family: the prevalence of different HCN subtypes in organs and tissues, the possibility to target HCN gain- or loss-of-function associated with disease, the feasibility of novel isoform-selective drugs, as well as the discovery of HCN-mediated effects for old medicines. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
318. Late sodium current inhibitors to treat exercise-induced obstruction in hypertrophic cardiomyopathy: an in vitro study in human myocardium.
- Author
-
Ferrantini, Cecilia, Pioner, Josè Manuel, Mazzoni, Luca, Gentile, Francesca, Tosi, Benedetta, Rossi, Alessandra, Belardinelli, Luiz, Tesi, Chiara, Palandri, Chiara, Matucci, Rosanna, Cerbai, Elisabetta, Olivotto, Iacopo, Poggesi, Corrado, Mugelli, Alessandro, and Coppini, Raffaele
- Subjects
- *
HYPERTROPHIC cardiomyopathy , *LEFT ventricular hypertrophy , *CATECHOLAMINES , *DISOPYRAMIDE , *ADRENERGIC receptors - Abstract
Background and Purpose: In 30-40% of hypertrophic cardiomyopathy (HCM) patients, symptomatic left ventricular (LV) outflow gradients develop only during exercise due to catecholamine-induced LV hypercontractility (inducible obstruction). Negative inotropic pharmacological options are limited to β-blockers or disopyramide, with low efficacy and tolerability. We assessed the potential of late sodium current (INaL )-inhibitors to treat inducible obstruction in HCM.Experimental Approach: The electrophysiological and mechanical responses to β-adrenoceptor stimulation were studied in human myocardium from HCM and control patients. Effects of INaL -inhibitors (ranolazine and GS-967) in HCM samples were investigated under conditions simulating rest and exercise.Key Results: In cardiomyocytes and trabeculae from 18 surgical septal samples of patients with obstruction, the selective INaL -inhibitor GS-967 (0.5 μM) hastened twitch kinetics, decreased diastolic [Ca2+ ] and shortened action potentials, matching the effects of ranolazine (10μM). Mechanical responses to isoprenaline (inotropic and lusitropic) were comparable in HCM and control myocardium. However, isoprenaline prolonged action potentials in HCM myocardium, while it shortened them in controls. Unlike disopyramide, neither GS-967 nor ranolazine reduced force at rest. However, in the presence of isoprenaline, they reduced Ca2+ -transient amplitude and twitch tension, while the acceleration of relaxation was maintained. INaL -inhibitors were more effective than disopyramide in reducing contractility during exercise. Finally, INaL -inhibitors abolished arrhythmias induced by isoprenaline.Conclusions and Implications: Ranolazine and GS-967 reduced septal myocardium tension during simulated exercise in vitro and therefore have the potential to ameliorate symptoms caused by inducible obstruction in HCM patients, with some advantages over disopyramide and β-blockers. [ABSTRACT FROM AUTHOR]- Published
- 2018
- Full Text
- View/download PDF
319. Content of mitochondrial calcium uniporter (MCU) in cardiomyocytes is regulated by microRNA-1 in physiologic and pathologic hypertrophy.
- Author
-
Zaglia, Tania, Ceriotti, Paola, Campo, Antonio, Borile, Giulia, Armani, Andrea, Carullo, Pierluigi, Prando, Valentina, Coppini, Raffaele, Vida, Vladimiro, Stølen, Tomas O., Ulrik, Wisløff, Cerbai, Elisabetta, Stellin, Giovanni, Faggian, Giuseppe, De Stefani, Diego, Sandri, Marco, Rizzuto, Rosario, Lisa, Fabio Di, Pozzan, Tullio, and Catalucci, Daniele
- Subjects
- *
CARDIAC hypertrophy , *CALCIUM channels , *MICRORNA , *BETA adrenoceptors , *MITOCHONDRIAL physiology , *HEART cells , *PHYSIOLOGY - Abstract
The mitochondrial Ca2+ uniporter complex (MCUC) is a multimeric ion channel which, by tuning Ca2+ influx into the mitochondrial matrix, finely regulates metabolic energy production. In the heart, this dynamic control of mitochondrial Ca2+ uptake is fundamental for cardiomyocytes to adapt to either physiologic or pathologic stresses. Mitochondrial calcium uniporter (MCU), which is the core channel subunit of MCUC, has been shown to play a critical role in the response to β-adrenoreceptor stimulation occurring during acute exercise. The molecular mechanisms underlying the regulation of MCU, in conditions requiring chronic increase in energy production, such as physiologic or pathologic cardiac growth, remain elusive. Here, we show that microRNA-1 (miR-1), a member of the muscle-specific microRNA (myomiR) family, is responsible for direct and selective targeting of MCU and inhibition of its translation, thereby affecting the capacity of the mitochondrial Ca2+ uptake machinery. Consistent with the role of miR-1 in heart development and cardiomyocyte hypertrophic remodeling, we additionally found that MCU levels are inversely related with the myomiR content, in murine and, remarkably, human hearts from both physiologic (i.e., postnatal development and exercise) and pathologic (i.e., pressure overload) myocardial hypertrophy. Interestingly, the persistent activation of β-adrenoreceptors is likely one of the upstream repressors of miR-1 as treatment with β-blockers in pressure-overloaded mouse hearts prevented its down-regulation and the consequent increase in MCU content. Altogether, these findings identify the miR-1/MCU axis as a factor in the dynamic adaptation of cardiac cells to hypertrophy. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
320. Mechanisms of pro-arrhythmic abnormalities in ventricular repolarisation and anti-arrhythmic therapies in human hypertrophic cardiomyopathy.
- Author
-
Passini, Elisa, Mincholé, Ana, Coppini, Raffaele, Cerbai, Elisabetta, Rodriguez, Blanca, Severi, Stefano, and Bueno-Orovio, Alfonso
- Subjects
- *
HEART ventricle abnormalities , *PROARRHYTHMIA , *ARRHYTHMIA treatment , *MYOCARDIAL depressants , *HYPERTROPHIC cardiomyopathy , *DEPOLARIZATION (Cytology) - Abstract
Introduction Hypertrophic cardiomyopathy (HCM) is a cause of sudden arrhythmic death, but the understanding of its pro-arrhythmic mechanisms and an effective pharmacological treatment are lacking. HCM electrophysiological remodelling includes both increased inward and reduced outward currents, but their role in promoting repolarisation abnormalities remains unknown. The goal of this study is to identify key ionic mechanisms driving repolarisation abnormalities in human HCM, and to evaluate anti-arrhythmic effects of single and multichannel inward current blocks. Methods Experimental ionic current, action potential (AP) and Ca 2 + -transient (CaT) recordings were used to construct populations of human non-diseased and HCM AP models (n = 9118), accounting for inter-subject variability. Simulations were conducted for several degrees of selective and combined inward current block. Results Simulated HCM cardiomyocytes exhibited prolonged AP and CaT, diastolic Ca 2 + overload and decreased CaT amplitude, in agreement with experiments. Repolarisation abnormalities in HCM models were consistently driven by L-type Ca 2 + current (I CaL ) re-activation, and I CaL block was the most effective intervention to normalise repolarisation and diastolic Ca 2 + , but compromised CaT amplitude. Late Na + current (I NaL ) block partially abolished repolarisation abnormalities, with small impact on CaT. Na + /Ca 2 + exchanger (I NCX ) block effectively restored repolarisation and CaT amplitude, but increased Ca 2 + overload. Multichannel block increased efficacy in normalising repolarisation, AP biomarkers and CaT amplitude compared to selective block. Conclusions Experimentally-calibrated populations of human AP models identify I CaL re-activation as the key mechanism for repolarisation abnormalities in HCM, and combined I NCX , I NaL and I CaL block as effective anti-arrhythmic therapies also able to partially reverse the HCM electrophysiological phenotype. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
321. R.O.C. RyR2 mutation impairs atrial and ventricular contractility.
- Author
-
Ferrantini, Cecilia, Coppini, Raffaele, Scellini, Beatrice, Ferrara, Claudia, Pioner, Jose Manuel, Mazzoni, Luca, Priori, Silvia, Cerbai, Elisabetta, Tesi, Chiara, and Poggesi, Corrado
- Subjects
- *
RYANODINE receptors , *GENETIC mutation , *CALCIUM channels , *SARCOPLASMIC reticulum , *CARDIAC contraction - Abstract
Ryanodine receptor (RyR2) is the major Ca2+ channel of the cardiac sarcoplasmic reticulum (SR) and plays a crucial role in the generation of myocardial force. Changes in RyR2 gating properties and resulting increases in its open probability (Pu) are associated with Ca2+ leakage from the SR and arrhythmias; however, the effects of RyR2 dysfunction on myocardial contractility are unknown. Here, we investigated the possibility that a RyR2 mutation associated with catecholaminergic polymorphic ventricular tachycardia, R4496C, affects the contractile function of atrial and ventricular myocardium. We measured isometric twitch tension in left ventricular and atrial trabeculae from wild-type mice and heterozygous transgenic mice carrying the R4496C RyR2 mutation and found that twitch force was comparable under baseline conditions (30°C, 2 mM [Ca2+]0, 1 Hz). However, the positive inotropic responses to high stimulation frequency, 0.1 µM isoproterenol, and 5 mM [Ca2+]0 were decreased in R4496C trabeculae, as was post-rest potentiation. We investigated the mechanisms underlying inotropic insufficiency in R4496C muscles in single ventricular myocytes. Under baseline conditions, the amplitude of the Ca2+ transient was normal, despite the reduced SR Ca2+ content. Under inotropic challenge, however, R4496C myocytes were unable to boost the amplitude of Ca2+ transients because they are incapable of properly increasing the amount of Ca2+ stored in the SR because of a larger SR Ca2+ leakage. Recovery of force in response to premature stimuli was faster in R4496C myocardium, despite the unchanged rates of recovery of L-type Ca2+ channel current (ICa-L) and SR Ca2+ content in single myocytes. A faster recovery from inactivation of the mutant R4496C channels could explain this behavior. In conclusion, changes in RyR2 channel gating associated with the R4496C mutation could be directly responsible for the alterations in both ventricular and atrial contractility. The increased RyR2 P0 and fractional Ca2+ release from the SR induced by the R4496C mutation preserves baseline contractility despite a slight decrease in SR Ca2+ content, but cannot compensate for the inability to increase SR Ca2+ content during inotropic challenge. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
322. Cellular determinants of arrhythmic risk in hypertrophic cardiomyopathy.
- Author
-
Santini, Lorenzo, Bacchi, Beatrice, Ferrantini, Cecilia, Olivotto, Iacopo, Coppini, Raffaele, and Cerbai, Elisabetta
- Subjects
- *
HYPERTROPHIC cardiomyopathy , *VENTRICULAR fibrillation , *ACTION potentials , *CARDIAC arrest , *VENTRICULAR arrhythmia , *VENTRICULAR tachycardia , *BRUGADA syndrome - Abstract
The most devastating consequence of HCM is sudden cardiac death (SCD) due to ventricular fibrillation. The positive correlation between the extent of late gadolinium enhancement and the arrhythmic risk in HCM suggests that ventricular arrhythmias originate from the fibrotic regions. However, recent data suggest that enhanced cellular automaticity (early- or delayed-afterdepolarizations, EADs or DADs-) may be clinically more relevant in promoting ventricular arrhythmias in patients. Aiming to better understand the cellular and molecular mechanisms of arrhythmogenesis in HCM and to establish a reliable arrhythmic risk stratification in patients, we performed a translational study in HCM patients who underwent surgical myectomy, by combining a clinical follow-up study with in vitro assessments of cellular arrhythmogenicity. We retrospectively studied 61 HCM patients who underwent surgical myectomy. At the time of surgery, fresh ventricular tissue was collected and used to isolate single ventricular cardiomyocytes (CMs), which were used for patch-clamp and Ca2+ imaging experiments to assess the occurrence of EADs and DADs. Patients were followed up for a median time of 8 years and the occurrence of non-sustained ventricular tachycardia (NSVT) or life-threateningarrhythmic events (LAE) was monitored. EADs occurred in CMs from 36% of patients and were associated with prolonged action potential duration. DADs occurred in 24% of patients and were associated with abnormalities of intracellular Ca2+ handling. The occurrence of NSVT/ LAE in patients strongly correlated with the presence of DADs in cardiomyocytes. The presence of pro-arrhythmic changes appears to be necessary for arrhythmia generation in HCM and seems to be related with specific alterations at ECG level, that might be used as clinical arrhythmia predictors in HCM patients. Fibrosis per se is not a major predictor of arrhythmias in HCM but may contribute to generate sustained arrhythmias in the presence of substantial cellular triggers (DADs). [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
323. Effects of mavacamten in human myocardium from hypertrophic obstructive cardiomyopathy (HOCM) patients: A comparison with disopyramide.
- Author
-
Musumeci, Monica, Palandri, Chiara, Santini, Lorenzo, Giammarino, Lucrezia, Vitale, Giulia, Langione, Marianna, Ferrantini, Cecilia, Pioner, Manuel Jose, Coppini, Raffaele, and Cerbai, Elisabetta
- Subjects
- *
HYPERTROPHIC cardiomyopathy , *MYOCARDIUM , *HUMAN beings - Published
- 2022
- Full Text
- View/download PDF
324. Muscle RING finger-1 is required to prevent age-related cardiac hypertrophy and interstitial remodelling.
- Author
-
Prando, Valentina, Parry, Traci L., Suryadevara, Vidyani, Scalco, Arianna, Pianca, Nicola, Moro, Nicola, Pesce, Paola, Tang, Wei, Ma, Hong, Qian, Li, Liu, Jiandong, Tessari, Maddalena, Coppini, Raffaele, Cerbai, Elisabetta, Patterson, Cam, Basso, Cristina, Faggian, Giuseppe, Bonaldo, Paolo, Sandri, Marco, and Mongillo, Marco
- Subjects
- *
CARDIAC hypertrophy - Published
- 2022
- Full Text
- View/download PDF
325. Mesoscopic optical imaging of whole mouse heart.
- Author
-
Giardini, Francesco, Lazzeri, Erica, Olianti, Camilla, Beconi, Giada, Costantini, Irene, Silvestri, Ludovico, Cerbai, Elisabetta, Pavone, Francesco S., and Sacconi, Leonardo
- Subjects
- *
OPTICAL images , *MICE , *HEART - Published
- 2022
- Full Text
- View/download PDF
326. Interleukin 6 rapidly modifies hyperpolarization-activated Cyclic nucleotide–gated (HCN) channel expression in vitro and its level is linearly related to HCN isoforms in human atria.
- Author
-
Laurino, Annunziatina, Gencarelli, Manuela, Balducci, Valentina, Cameli, Matteo, Sartiani, Laura, and Cerbai, Elisabetta
- Subjects
- *
ATRIUMS (Architecture) , *HUMAN beings - Published
- 2022
- Full Text
- View/download PDF
327. Extracellular stiffness as a determinant of cardiac dysfunction in Duchenne's muscular dystrophy: A study on human iPSC-derived cardiomyocytes.
- Author
-
Giammarino, Lucrezia, Santini, Lorenzo, Palandri, Chiara, Musumeci, Monica, Pioner, Manuel Jose, Ferrantini, Cecilia, Langione, Marianna, Coppini, Raffaele, and Cerbai, Elisabetta
- Subjects
- *
DUCHENNE muscular dystrophy , *HEART diseases , *HUMAN experimentation , *DETERMINANTS (Mathematics) - Published
- 2022
- Full Text
- View/download PDF
328. Dual effect of hydrogen-sulfide on the hyperpolarization-activated inward current (If) in human atrial cardiomyocytes.
- Author
-
Balducci, Valentina, Gencarelli, Manuela, Laurino, Annunziatina, Spinelli, Valentina, Stefano, Pierluigi, Cerbai, Elisabetta, and Sartiani, Laura
- Subjects
- *
HUMAN beings , *HYDROGEN sulfide - Published
- 2022
- Full Text
- View/download PDF
329. Optogenetic manipulation of cardiac electrical dynamics using sub-threshold illumination: Dissecting the role of cardiac alternans in terminating rapid rhythms.
- Author
-
Biasci, Valentina, Santini, Lorenzo, Hussaini, Sayedeh, Ferrantini, Cecilia, Coppini, Raffaele, Loew, Leslie, Luther, Stefan, Campione, Marina, Poggesi, Corrado, Pavone, Francesco Saverio, Cerbai, Elisabetta, Bub, Gil, and Sacconi, Leonardo
- Subjects
- *
RHYTHM , *LIGHTING , *CARDIAC contraction - Published
- 2022
- Full Text
- View/download PDF
330. Electrophysiological effects of ivabradine in dog and human cardiac preparations: Potential antiarrhythmic actions
- Author
-
Koncz, István, Szél, Tamás, Bitay, Miklós, Cerbai, Elisabetta, Jaeger, Kristian, Fülöp, Ferenc, Jost, Norbert, Virág, László, Orvos, Péter, Tálosi, László, Kristóf, Attila, Baczkó, István, Papp, Julius Gy., and Varró, András
- Subjects
- *
ELECTROPHYSIOLOGY , *IVABRADINE , *LABORATORY dogs , *MYOCARDIAL depressants , *CARDIAC pacemakers , *PURKINJE cells , *HEART function tests - Abstract
Abstract: Ivabradine is a novel antianginal agent which inhibits the pacemaker current. The effects of ivabradine on maximum rate of depolarization (Vmax), repolarization and spontaneous depolarization have not yet been reported in human isolated cardiac preparations. The same applies to large animals close to human in heart size and spontaneous frequency. Using microelectrode technique action potential characteristics and by applying patch-clamp technique ionic currents were studied. Ivabradine exerted concentration-dependent (0.1–10μM) decrease in the amplitude of spontaneous diastolic depolarization and reduction in spontaneous rate of firing of action potentials and produced a concentration- and frequency-dependent Vmax block in dog Purkinje fibers while action potential duration measured at 50% of repolarization was shortened. In the presence of ivabradine, at 400ms cycle length, Vmax block developed with an onset kinetic rate constant of 13.9±3.2beat−1 in dog ventricular muscle. In addition to a fast recovery of Vmax from inactivation (τ=41–46ms) observed in control, a second slow component for recovery of Vmax was expressed (offset kinetics of Vmax block) having a time constant of 8.76±1.34s. In dog after attenuation of the repolarization reserve ivabradine moderately but significantly lengthened the repolarization. In human, significant prolongation of repolarization was only observed at 10μM ivabradine. Ivabradine in addition to the Class V antiarrhythmic effect also has Class I/C and Class III antiarrhythmic properties, which can be advantageous in the treatment of patients with ischemic heart disease liable to disturbances of cardiac rhythm. [Copyright &y& Elsevier]
- Published
- 2011
- Full Text
- View/download PDF
331. The effect of losartan treatment on the response of diabetic cardiomyocytes to ATP depletion
- Author
-
Alfarano, Chiara, Suffredini, Silvia, Fantappiè, Ornella, Mugelli, Alessandro, Cerbai, Elisabetta, Manni, Maria Elena, and Raimondi, Laura
- Subjects
- *
LOSARTAN , *HEART cells , *TREATMENT of diabetes , *ADENOSINE triphosphate , *LABORATORY rats , *ISCHEMIA , *ION channels , *CARDIAC contraction - Abstract
Abstract: The present work aimed to investigate the effect of losartan treatment of healthy and diabetic rats on cardiomyocyte response to ATP depletion. Cells were isolated from normoglycemic (N) and streptozotocin-injected (55mg/kg) rats (D) treated or not treated with losartan (20mg/kg/day in the drinking water; NL and DL, respectively) for 3 weeks. In each group of cells, enzyme activities such as glucose-6-phosphate (G6PDH) and glycerol-3-phosphate dehydrogenases (G3PDH), lactate/pyruvate, glycogen levels and citrate synthase were measured as an index of glycolytic dysregulation and mitochondrial mass, respectively. Cells were then challenged with NaCN (2mM) in glucose-free Tyrode solution (metabolic intoxication, MI), a protocol to study ischemia at cell level. Under these conditions, the time to contractile failure up to rigor-type hyper-contracture in field-stimulated cells and KATP current activation by patch-clamp recordings were measured. In comparison with N and NL, D cells presented higher G6PDH and cytoplasmic G3PDH activities, lactate/pyruvate, glycogen content but similar levels of citrate synthase, and decay time of contraction. When subjected to MI, D cells showed delayed activation of the KATP current (25.7±7.1min; p <0.001 vs. N and NL), increased time to contractile failure and rigor-type hyper-contracture (p <0.001 vs. N and NL). In cells from DL rats both functional (time to rigor and to KATP current activation) and metabolic parameters, approached values similar to those measured in N and NL cells. These results demonstrate that diabetic cardiomyocytes from rats treated with losartan, maintain the capacity to respond promptly to ATP depletion reaching contractile failure, rigor-type hypercontracture and KATP opening with a similar timing of N cells. [Copyright &y& Elsevier]
- Published
- 2011
- Full Text
- View/download PDF
332. Oleuropein aglycon prevents cytotoxic amyloid aggregation of human amylin
- Author
-
Rigacci, Stefania, Guidotti, Valentina, Bucciantini, Monica, Parri, Matteo, Nediani, Chiara, Cerbai, Elisabetta, Stefani, Massimo, and Berti, Andrea
- Subjects
- *
PHYTOCHEMICALS , *AMYLIN , *ANTINEOPLASTIC agents , *CELL aggregation , *FIBERS , *TYPE 2 diabetes treatment , *OLIGOMERS , *CELL-mediated cytotoxicity , *THERAPEUTICS - Abstract
Abstract: Pancreatic amyloid deposits of amylin are a hallmark of Type II diabetes and considerable evidence indicates that amylin oligomers are cytotoxic to β-cells. Many efforts are presently spent to find out naturally occurring molecules, or to design synthetic ones, able to hinder amylin aggregation or to protect cells against aggregate cytotoxicity. In this context, a protective effect of some polyphenols against amyloid cytotoxicity was reported. Actually dietary polyphenols are endowed with multiple health benefits, and extra virgin olive oil is attracting increasing interest as a source of these substances. Here, we investigated the effects on amylin aggregation and cytotoxicity of the secoiridoid oleuropein aglycon, the main phenolic component of extra virgin olive oil. We found that oleuropein, when present during the aggregation of amylin, consistently prevented its cytotoxicity to RIN-5F pancreatic β-cells, as determined by the 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyl tetrazolium bromide test and caspase-3 activity assay. A lack of interaction with the cell membrane of amylin aggregates grown in the presence of oleuropein was shown by fluorescence microscopy and synthetic lipid vesicle permeabilization. Moreover, our ThT assay, circular dichroism analysis and electron microscopy images suggested that oleuropein interferes with amylin aggregation, resulting in a different path skipping the formation of toxic pre-fibrillar aggregates. These results provide a molecular basis for some of the benefits potentially coming from extra virgin olive oil consumption and pave the way to further studies on the possible pharmacological use of oleuropein to prevent or to slow down the progression of type II diabetes. [Copyright &y& Elsevier]
- Published
- 2010
- Full Text
- View/download PDF
333. Molecular basis of funny current (If) in normal and failing human heart
- Author
-
Stillitano, Francesca, Lonardo, Giuseppe, Zicha, Stephen, Varro, Andras, Cerbai, Elisabetta, Mugelli, Alessandro, and Nattel, Stanley
- Subjects
- *
HEART diseases , *HEART failure , *GENE expression , *ARRHYTHMIA , *MUSCLE cells , *MESSENGER RNA , *ION channels , *ELECTROPHYSIOLOGY ,MOLECULAR aspects - Abstract
Abstract: If overexpression has been functionally demonstrated in ventricular myocytes from failing human hearts. Altered expression of If-channels as a consequence of electrophysiological remodeling may represent an arrhythmogenic mechanism in heart failure; however, the molecular basis of If overexpression in human cardiac disease is unknown. HCN1, 2 and 4 subtypes, which encode If-channels, have been identified in the heart. The present study was designed to characterize HCN isoform expression in failing and non-failing hearts. Ventricular and atrial samples were obtained from normal or failing hearts explanted from patients with end-stage ischemic cardiomyopathy. If was recorded in patch-clamped left ventricular myocytes. mRNA and protein expression of HCN subunits were measured in both atria and ventricles of control and diseased hearts. HCN2 and HCN4 were detected in human myocardium. Both mRNA and protein levels of HCN2/4 were significantly augmented in failing ventricles (p <0.01 for mRNA, p <0.05 for protein). These results are consistent with the electrophysiological data showing that, in failing ventricular myocytes, If is of larger amplitude and activates at less negative potential. Changes in mRNA and protein expression of both HCN2/4 isoforms in atrial specimens from patients with heart failure mirrored those observed in ventricles (p <0.001 for mRNA, p <0.05 for protein). No disease-dependent alteration was detected for MiRP1, the putative β-subunit of the If-channel. In conclusion, HCN4 is the predominant channel subtype in normal human heart, and its expression is further amplified by disease. HCN upregulation likely contributes to increased If and may play a role in ventricular and atrial arrhythmogenesis in heart failure. [Copyright &y& Elsevier]
- Published
- 2008
- Full Text
- View/download PDF
334. Expression of the hyperpolarization-activated current, I f, in cultured adult rat ventricular cardiomyocytes and its modulation by hypertrophic factors
- Author
-
Stillitano, Francesca, Sartiani, Laura, DePaoli, Petra, Mugelli, Alessandro, and Cerbai, Elisabetta
- Subjects
- *
CYCLIC nucleotides , *MUSCLE cells , *HYPERTROPHY , *ANGIOTENSINS - Abstract
Abstract: The hyperpolarization-activated, cyclic nucleotide-gated (HCN) current, I f, is typically overexpressed in myocytes from hypertrophied and failing hearts, where it may play an arrhythmogenic role. Signaling pathways activated by agonists such as angiotensin-II, endothelin-1 and phenylephrine, via G protein-coupled receptors (GPCR), promote myocardial hypertrophy, but their effect on cellular electrophysiological remodeling, particularly I f expression is largely unknown. Thus, I f expression was measured by patch-clamp and quantitative RT-PCR measurement in cultured adult rat ventricular cardiomyocytes (VCM) exposed to different culture conditions, that is, in the absence or presence of: fetal bovine serum (FBS, 5%), 0.1μM angiotensin-II, 0.1μM endothelin-1 or 20μM phenylephrine. Membrane capacitance (C m) was used to estimate cell size and current density in patch-clamped VCM. At 8 days of culture, about 60% of VCM showed I f. In serum-free medium, I f density was increased by phenylephrine (2.28±0.51 vs. 0.84±0.30pA/pF in CTR, p <0.05) and endothelin-1 (2.20±0.38 vs. 1.03±0.34pA/pF in control, p <0.05), not by angiotensin-II (1.60±0.50pA/pF, not significant vs. control). Similarly, in cells cultured with 5% FBS, phenylephrine and endothelin-1 significantly increased I f density by 159.3% and 59.5% (p <0.05 vs. untreated cells), while angiotensin-II did not modify it. The effect of endothelin-1 was abolished by using the selective endothelin receptor type A (ET1A) antagonist BQ-123 (1μM). mRNA levels for HCN2 and HCN4 were significantly increased during in vitro culture; exposure to endothelin-1 increased HCN2 mRNA. A similar pattern of I f overexpression was detected in hypertrophied left ventricular cardiomyocytes from old hypertensive rats. Thus, adult VCM in primary culture undergo changes in I f expression reminiscent of in vivo hypertrophy; endothelin-1 (but no angiotensin-II) seems to play a role in ionic remodeling. [Copyright &y& Elsevier]
- Published
- 2008
- Full Text
- View/download PDF
335. NADPH oxidase-dependent redox signaling in human heart failure: Relationship between the left and right ventricle
- Author
-
Nediani, Chiara, Borchi, Elisabetta, Giordano, Carla, Baruzzo, Serena, Ponziani, Vanessa, Sebastiani, Mariangela, Nassi, Paolo, Mugelli, Alessandro, d'Amati, Giulia, and Cerbai, Elisabetta
- Subjects
- *
HEART ventricles , *HEART diseases , *CARDIAC arrest , *PEROXIDATION - Abstract
Abstract: Oxidative stress resulting from increased superoxide generation by NADPH oxidase is implicated in the pathophysiology of human heart failure. Downstream targets of NADPH oxidase remain undefined and available information is restricted to the left ventricle (LV). Thus, we aimed to assess the cascade of events triggered by increased NADPH oxidase activity (lipid peroxidation and activation of mitogen-activated protein kinases ERK1/2, JNK and p38) and their mutual relationship in right (RV) and (LV) of end-stage failing human hearts. When compared to control ventricles, diseased RV and LV showed a significant increase in NADPH oxidase superoxide production that positively correlated with p47phox membrane translocation (RV: r =0.76, P <0.001; LV: r =0.79, P <0.001). MDA content, a marker of lipid peroxidation, was also enhanced and ERK and p38, but not JNK, were activated. For all these relevant steps of the oxidative stress pathway, a significant correlation was observed between LV and RV from the same heart (NADPH-dependent superoxide production: r =0.678, P <0.0055; MDA: r =0.95, P <0.0001; p-p38/p38 ratio: r =0.926, P <0.0001; p-ERK/ERK ratio: r =0.913, P <0.0001). We concluded that in human heart failure, both ventricles are targets of NADPH oxidase superoxide generation which in turn may trigger the coordinated activation of downstream signaling components. This pathway may contribute to adverse remodeling of the LV and RV and subsequent progression toward end-stage heart failure, suggestive of new therapeutic targeting strategy. [Copyright &y& Elsevier]
- Published
- 2007
- Full Text
- View/download PDF
336. Functional remodeling in post-myocardial infarcted rats: focus on beta-adrenoceptor subtypes
- Author
-
Sartiani, Laura, De Paoli, Petra, Stillitano, Francesca, Aimond, Frank, Vassort, Guy, Mugelli, Alessandro, and Cerbai, Elisabetta
- Subjects
- *
VENTRICULAR remodeling , *MYOCARDIAL infarction , *RATS , *ARRHYTHMIA , *ANIMAL experimentation - Abstract
Abstract: Cellular electrophysiological remodeling of the infarcted heart may lead to the deterioration of cardiac function and/or to arrhythmias. The present study was designed to characterize the functional expression of the hyperpolarization-activated current (I f) and its modulation by β1-, β2- and β3-adrenoceptor (AR) subtypes, in patch-clamped ventricular myocytes isolated from the heart of post-myocardial infarcted (PMI) rats and sham-operated control (SHAM) rats. Maximum specific conductance of I f was significantly higher in left ventricular myocytes (LVM) from PMI rats compared to right ventricular myocytes from PMI rats as well as LVM and RVM from SHAM rats. All other basic properties of I f were similar. β1AR stimulation with noradrenaline caused a rightward shift of V H in LVM from PMI rats which was significantly smaller (52.2%) than in LVM from SHAM rats. Incubation with pertussis toxin (PTX) largely restored the effect of β1AR in PMI cells (86.6% vs. SHAM cells), but did not affect β1AR response in SHAM cells. β2AR response was significantly and equally increased by PTX-pretreatment (by 94% in SHAM and 87% in PMI cells). Conversely, β3AR stimulation by the selective agonist SR 58611A caused a leftward shift of the activation curve which was significantly larger in PMI cells than in SHAM cells (P <0.01). β3AR response was blunted by PTX-pretreatment, by incubation with NG-monomethyl-l-arginine acetate or by the selective β3AR antagonist SR 59230A 1 μM. In conclusion, I f is significantly overexpressed in LVM from PMI rat hearts. In these cells, I f modulation by β1AR is significantly depressed while β3AR modulation is markedly enhanced, probably reflecting the increased activity of PTX-sensitive Gi proteins in PMI cells. [Copyright &y& Elsevier]
- Published
- 2006
- Full Text
- View/download PDF
337. The effects of gender on electrical therapies for the heart: physiology, epidemiology, and access to therapies
- Author
-
Stefano Lorenzetti, Giuseppe Boriani, Gianluca Botto, Alessandro Biffi, Elisabetta Cerbai, Gabriele Bronzetti, Giuseppe Oreto, Igor Diemberger, Luigi Padeletti, Vincenzo Livio Malavasi, Boriani, Giuseppe, Lorenzetti, Stefano, Cerbai, Elisabetta, Oreto, Giuseppe, Bronzetti, Gabriele, Malavasi, Vincenzo Livio, Biffi, Alessandro, Padeletti, Luigi, Botto, Gianluca, and Diemberger, Igor
- Subjects
Male ,medicine.medical_specialty ,Action Potentials ,Review ,Ablation ,030204 cardiovascular system & hematology ,QT interval ,Health Services Accessibility ,Sudden cardiac death ,Defibrillator ,Electrocardiography ,03 medical and health sciences ,QRS complex ,Sex Factors ,0302 clinical medicine ,Heart Conduction System ,Heart Rate ,Pregnancy ,Physiology (medical) ,Internal medicine ,Epidemiology ,medicine ,Animals ,Humans ,cardiovascular diseases ,030212 general & internal medicine ,Healthcare Disparities ,Exercise ,business.industry ,Gender ,Arrhythmias, Cardiac ,Health Status Disparities ,medicine.disease ,Cardiovascular physiology ,Clinical trial ,Treatment Outcome ,Heart failure ,cardiovascular system ,Cardiology ,Physical therapy ,Sex ,Female ,Electrophysiologic Techniques, Cardiac ,Cardiology and Cardiovascular Medicine ,business ,Arrhythmia - Abstract
The difference between men and women is clear even just by looking at an electrocardiogram: females present higher resting heart rate, a shorter QRS complex length and greater corrected QT interval. The development of these differences from pubertal age onward suggests that sexual hormones play a key role, although their effect is far from being completely understood. Different incidences between sexes have been reported for many arrhythmias, both ventricular and supraventricular, and also for sudden cardiac death. Moreover, arrhythmias are an important issue during pregnancy, both for diagnosis and treatment. Interestingly, cardiovascular structural and electrophysiological remodelling promoted by exercise training enhances this 'gender effect'. Despite all these relevant issues, we lack gender specific recommendations in the current guidelines for electrical therapies for heart rhythm disorders and heart failure. Even more, we continue to see that fewer women are included in clinical trials and are less referred than men for these treatments.
- Published
- 2017
338. Cellular mechanisms of arrhythmias
- Author
-
Cerbai, Elisabetta and Mugelli, Alessandro
- Published
- 1992
- Full Text
- View/download PDF
339. The effect of temperature on intracellular Ca 2+ homeostais and ventricular arrhythmias
- Author
-
Gambassi, Giovanni, Cerbai, Elisabetta, Pahor, Marco, Carbonin, Pierugo, and Mugelli, Alessandro
- Published
- 1994
- Full Text
- View/download PDF
340. Modelling genetic diseases for drug development: Hypertrophic cardiomyopathy.
- Author
-
Santini, Lorenzo, Palandri, Chiara, Nediani, Chiara, Cerbai, Elisabetta, and Coppini, Raffaele
- Subjects
- *
HYPERTROPHIC cardiomyopathy , *GENETIC disorders , *GENETIC models , *DRUGS , *BRUGADA syndrome , *DRUG development , *GENOME editing - Abstract
Different tools for HCM disease modeling and pharmaceutical drug screening. This figure summarizes the different research platforms to model HCM and to screen novel drugs for this disease. Hypertrophic cardiomyopathy (HCM) is the commonest genetic cardiac disease, with a prevalence of 1/500. It is caused by over 1400 different mutations, mainly involving the genes coding for sarcomere proteins. The main pathological features of HCM are left ventricular hypertrophy, diastolic dysfunction and the increased ventricular arrhythmogenesis. Predicting the risk of heart failure and lethal arrhythmias is the most challenging clinical task for HCM patient management. Moreover, there are no disease-modifying therapies that can prevent disease progression or sudden arrhythmic death in HCM patients. In this review, we will illustrate the most advanced research models and methods that have been employed for HCM studies, including preclinical tests of novel or existing drugs, along with visionary future development based on gene editing approaches. Acknowledging the advantages and limitations of the different models, and a critical consideration of the different, often conflicting result obtained using different approaches is essential for a deep understanding of HCM pathophysiology and for obtaining meaningful information on novel treatments, in order to improve patient risk stratification and therapeutic management. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
341. Update on antithrombotic therapy and body mass: a clinical consensus statement of the European Society of Cardiology Working Group on Cardiovascular Pharmacotherapy and the European Society of Cardiology Working Group on Thrombosis.
- Author
-
Gigante B, Tamargo J, Agewall S, Atar D, Ten Berg J, Campo G, Cerbai E, Christersson C, Dobrev D, Ferdinandy P, Geisler T, Gorog DA, Grove EL, Kaski JC, Rubboli A, Wassmann S, Wallen H, and Rocca B
- Subjects
- Humans, Risk Factors, Thrombosis prevention & control, Body Mass Index, Treatment Outcome, Hemorrhage chemically induced, Bariatric Surgery adverse effects, Cardiology standards, Obesity diagnosis, Obesity complications, Risk Assessment, Thinness diagnosis, Fibrinolytic Agents pharmacokinetics, Fibrinolytic Agents therapeutic use, Fibrinolytic Agents adverse effects, Fibrinolytic Agents administration & dosage, Consensus
- Abstract
Obesity and underweight are a growing health problem worldwide and a challenge for clinicians concerning antithrombotic therapy, due to the associated risks of thrombosis and/or bleeding. This clinical consensus statement updates a previous one published in 2018, by reviewing the most recent evidence on antithrombotic drugs based on body size categories according to the World Health Organization classification. The document focuses mostly on individuals at the extremes of body weight, i.e. underweight and moderate-to-morbid obesity, who require antithrombotic drugs, according to current guidelines, for the treatment or prevention of cardiovascular diseases or venous thromboembolism. Managing antithrombotic therapy or thromboprophylaxis in these individuals is challenging, due to profound changes in body composition, metabolism and organ function, and altered drug pharmacokinetics and pharmacodynamics, as well as weak or no evidence from clinical trials. The document also includes artificial intelligence simulations derived from in silico pharmacokinetic/pharmacodynamic models, which can mimic the pharmacokinetic changes and help identify optimal regimens of antithrombotic drugs for severely underweight or severely obese individuals. Further, bariatric surgery in morbidly obese subjects is frequently performed worldwide. Bariatric surgery causes specific and additional changes in metabolism and gastrointestinal anatomy, depending on the type of the procedure, which can also impact the pharmacokinetics of antithrombotic drugs and their management. Based on existing literature, the document provides consensus statements on optimizing antithrombotic drug management for underweight and all classes of obese patients, while highlighting the current gaps in knowledge in these complex clinical settings, which require personalized medicine and precision pharmacology., (© 2024 the European Society of Cardiology.)
- Published
- 2024
- Full Text
- View/download PDF
342. Physiological and pathological ventricular hypertrophy: a role for cavin-2 mediated Akt regulation.
- Author
-
Coppini R, Giammarino L, and Cerbai E
- Abstract
Competing Interests: Conflict of interest: none declared.
- Published
- 2024
- Full Text
- View/download PDF
343. Ticagrelor and Statins: Dangerous Liaisons?
- Author
-
Rocca B, Bigagli E, and Cerbai E
- Abstract
Polypharmacy is often necessary in complex, chronic, comorbid and cardiovascular patients and is a known risk factor for potential drug-drug interaction (DDI) that can cause adverse reactions (toxicity or therapeutic failure). Anti-thrombotic drugs (largely low-dose aspirin and a platelet P2Y12 receptor inhibitor) and statins are among the most co-administered drugs in cardiovascular patients. Ticagrelor is a selective antagonist of the platelet P2Y12-receptor, highly effective in inhibiting platelet aggregation and bio-transformed by the CYP3A4 and substrate of transporters, such as the breast cancer resistance protein (BCRP). Statins have different pharmacokinetic profiles; some undergo CYP3A4-mediated metabolism; rosuvastatin is primarily metabolized by the CYP2C9; and they have different affinities for drug transporters. Rhabdomyolysis is a very rare but severe adverse event, which is specific for statins which can be triggered by DDIs that increase statin's concentrations through blockade of their biotransformation and/or elimination. Large pharmacovigilance and small observational studies reported increased rhabdomyolysis in patients treated with some statins and ticagrelor but not aspirin, clopidogrel or prasugrel. Recent studies in vitro, pharmacokinetic trials and in silico drug modelling identified and validated the BCRP inhibition by ticagrelor, as a mechanism contributing to the DDI with statins, as 'victim' drugs, leading to increased rhabdomyolysis. While the clinical impact of this DDI deserves further investigation, a careful evaluation should be advised when ticagrelor is co-prescribed with some statins., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
344. Oral anticoagulation in patients with left ventricular thrombus: a systematic review and meta-analysis.
- Author
-
Haller PM, Kazem N, Agewall S, Borghi C, Ceconi C, Dobrev D, Cerbai E, Grove EL, Kaski JC, Lewis BS, Niessner A, Rocca B, Rosano G, Savarese G, Schnabel RB, Semb AG, Sossalla S, Wassmann S, and Sulzgruber P
- Subjects
- Humans, Administration, Oral, Treatment Outcome, Risk Factors, Anticoagulants adverse effects, Anticoagulants administration & dosage, Heart Ventricles drug effects, Female, Risk Assessment, Male, Stroke mortality, Stroke diagnosis, Stroke prevention & control, Aged, Factor Xa Inhibitors adverse effects, Factor Xa Inhibitors administration & dosage, Vitamin K antagonists & inhibitors, Middle Aged, Thrombosis mortality, Thrombosis drug therapy, Thrombosis prevention & control, Thrombosis diagnosis, Hemorrhage chemically induced, Heart Diseases mortality, Heart Diseases diagnosis, Heart Diseases drug therapy, Heart Diseases complications
- Abstract
Aims: Direct oral anticoagulants (DOACs) are increasingly used off-label to treat patients with left ventricular thrombus (LVT). We analysed available meta-data comparing DOACs and vitamin K antagonists (VKAs) for efficacy and safety., Methods: We conducted a systematic search and meta-analysis of observational and randomized data comparing DOACs vs. VKAs in patients with LVT. Endpoints of interest were stroke or systemic embolism, thrombus resolution, all-cause death, and a composite bleeding endpoint. Estimates were pooled using a random-effects model meta-analysis, and their robustness was investigated using sensitivity and influential analyses., Results: We identified 22 articles (18 observational studies, 4 small randomized clinical trials) reporting on a total of 3587 patients (2489 VKA vs. 1098 DOAC therapy). The pooled estimates for stroke or systemic embolism [odds ratio (OR): 0.81; 95% confidence interval (CI): 0.57, 1.15] and thrombus resolution (OR: 1.12; 95% CI: 0.86, 1.46) were comparable, and there was low heterogeneity overall across the included studies. The use of DOACs was associated with lower odds of all-cause death (OR: 0.65; 95% CI: 0.46, 0.92) and a composite bleeding endpoint (OR: 0.67; 95% CI: 0.47, 0.97). A risk of bias was evident particularly for observational reports, with some publication bias suggested in funnel plots., Conclusion: In this comprehensive analysis of mainly observational data, the use of DOACs was not associated with a significant difference in stroke or systemic embolism, or thrombus resolution, compared with VKA therapy. The use of DOACs was associated with a lower rate of all-cause death and fewer bleeding events. Adequately sized randomized clinical trials are needed to confirm these findings, which could allow a wider adoption of DOACs in patients with LVT., (© The Author(s) 2024. Published by Oxford University Press on behalf of the European Society of Cardiology.)
- Published
- 2024
- Full Text
- View/download PDF
345. New pharmacological agents and novel cardiovascular pharmacotherapy strategies in 2023.
- Author
-
Tamargo J, Agewall S, Borghi C, Ceconi C, Cerbai E, Dan GA, Ferdinandy P, Grove EL, Rocca B, Magavern E, Sulzgruber P, Semb AG, Sossalla S, Niessner A, Kaski JC, and Dobrev D
- Subjects
- Humans, Treatment Outcome, Animals, Drug Repositioning, Drug Development, Cardiovascular Diseases drug therapy, Cardiovascular Diseases prevention & control, Cardiovascular Agents therapeutic use, Cardiovascular Agents adverse effects
- Abstract
Although cardiovascular diseases (CVDs) are the leading cause of death worldwide, their pharmacotherapy remains suboptimal. Thus, there is a clear unmet need to develop more effective and safer pharmacological strategies. In this review, we summarize the most relevant advances in cardiovascular pharmacology in 2023, including the approval of first-in-class drugs that open new avenues for the treatment of atherosclerotic CVD and heart failure (HF). The new indications of drugs already marketed (repurposing) for the treatment of obstructive hypertrophic cardiomyopathy, hypercholesterolaemia, type 2 diabetes, obesity, and HF; the impact of polypharmacy on guideline-directed drug use is highlighted as well as results from negative clinical trials. Finally, we end with a summary of the most important phase 2 and 3 clinical trials assessing the efficacy and safety of cardiovascular drugs under development for the prevention and treatment of CVDs., (© The Author(s) 2024. Published by Oxford University Press on behalf of the European Society of Cardiology.)
- Published
- 2024
- Full Text
- View/download PDF
346. Cardiac safety assessment of a novel recombinant bispecific antibody targeting the ether-à-go-go related gene 1 (hERG1)-β1 integrin macromolecular complex.
- Author
-
Santini L, Duranti C, Palandri C, Giammarino L, Musumeci M, Carlucci L, Capitani C, Colasurdo R, Recchia F, Cerbai E, Coppini R, and Arcangeli A
- Abstract
Introduction: In the last decades, mounting evidence has pointed out the human ether-á-go-go-related gene (hERG1) potassium channel as a novel biomarker in human cancers. However, hERG1 sustains the cardiac repolarizing current IKr and its blockade can induce a prolonged QT interval at the ECG, which increases the risk of life-threatening arrhythmias. This represents a major hindrance for targeting hERG1 for antineoplastic therapeutic purposes. Based on our discovery that hERG1 resides in a macromolecular complex with the β1 subunit of integrin adhesion receptors only in tumors, and not in the heart, we generated (and patented WO2019/015936) a novel engineered, single chain, bispecific antibody in the format of a diabody (scDb-hERG1-β1). This antibody has been proven to target with high affinity the hERG1/β1 integrin complex and to exert a good antineoplastic activity in preclinical mouse models. Methods: In the present study, we evaluated the cardiac safety of the scDb-hERG1-β1, determining the action potential duration (APD) of human cardiomyocytes, either atrial (from valve-disease patients) or ventricular (from aortic stenosis patients). Cardiac cells were incubated in vitro with i) the scDb-hERG1-β1, ii) the full length anti-hERG1 monoclonal antibody (mAb-hERG1) and iii) its single chain Fragment variable derivative (scFv-hERG1), from which the scDb-hERG1-β1 was assembled. All the tests were performed before and after treatment with the specific hERG1 blocker E4031. In addition, we have performed preliminary experiments, analyzing the effects of the scDb-hERG1/β1 in vivo measuring the QT interval length of the surface ECG after its injection intravenously in farm-pigs. Results: The scDb-hERG1-β1 did not produce any lengthening of APD compared to control (vehicle) conditions, either in atrial or ventricular cardiomyocytes, whereas both the hERG1-mAb and the scFv-hERG1 produced a significant APD prolongation. The addition of E4031 further prolonged APD. The scDb-hERG1-β1 did not produce any alterations of the QT (and QTc) interval values, once injected intravenously in farm pigs. Discussion: Overall, the above evidences plead for the cardiac safety of the scDb-hERG1-β1, suggesting that an application of this antibody for anti-cancer therapy will be untainted by cardiotoxicity., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 Santini, Duranti, Palandri, Giammarino, Musumeci, Carlucci, Capitani, Colasurdo, Recchia, Cerbai, Coppini and Arcangeli.)
- Published
- 2023
- Full Text
- View/download PDF
347. Corrigendum: Calcium handling maturation and adaptation to increased substrate stiffness in human iPSC-derived cardiomyocytes: the impact of full-length dystrophin deficiency.
- Author
-
Pioner JM, Santini L, Palandri C, Langione M, Grandinetti B, Querceto S, Martella D, Mazzantini C, Scellini B, Giammarino L, Lupi F, Mazzarotto F, Gowran A, Rovina D, Santoro R, Pompilio G, Tesi C, Parmeggiani C, Regnier M, Cerbai E, Mack DL, Poggesi C, Ferrantini C, and Coppini R
- Abstract
[This corrects the article DOI: 10.3389/fphys.2022.1030920.]., (Copyright © 2023 Pioner, Santini, Palandri, Langione, Grandinetti, Querceto, Martella, Mazzantini, Scellini, Giammarino, Lupi, Mazzarotto, Gowran, Rovina, Santoro, Pompilio, Tesi, Parmeggiani, Regnier, Cerbai, Mack, Poggesi, Ferrantini and Coppini.)
- Published
- 2023
- Full Text
- View/download PDF
348. Beneficial effects of chronic mexiletine treatment in a human model of SCN5A overlap syndrome.
- Author
-
Nasilli G, Yiangou L, Palandri C, Cerbai E, Davis RP, Verkerk AO, Casini S, and Remme CA
- Subjects
- Humans, Mexiletine pharmacology, Cardiac Conduction System Disease, NAV1.5 Voltage-Gated Sodium Channel genetics, NAV1.5 Voltage-Gated Sodium Channel metabolism, Action Potentials, Myocytes, Cardiac, Long QT Syndrome, Brugada Syndrome genetics
- Abstract
Aims: SCN5A mutations are associated with various cardiac phenotypes, including long QT syndrome type 3 (LQT3), Brugada syndrome (BrS), and cardiac conduction disease (CCD). Certain mutations, such as SCN5A-1795insD, lead to an overlap syndrome, with patients exhibiting both features of BrS/CCD [decreased sodium current (INa)] and LQT3 (increased late INa). The sodium channel blocker mexiletine may acutely decrease LQT3-associated late INa and chronically increase peak INa associated with SCN5A loss-of-function mutations. However, most studies have so far employed heterologous expression systems and high mexiletine concentrations. We here investigated the effects of a therapeutic dose of mexiletine on the mixed phenotype associated with the SCN5A-1795insD mutation in HEK293A cells and human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs)., Methods and Results: To assess only the chronic effects on trafficking, HEK293A cells transfected with wild-type (WT) SCN5A or SCN5A-1795insD were incubated for 48 h with 10 µm mexiletine followed by wash-out, which resulted in an increased peak INa for both SCN5A-WT and SCN5A-1795insD and an increased late INa for SCN5A-1795insD. Acute re-exposure of HEK293A cells to 10 µm mexiletine did not impact on peak INa but significantly decreased SCN5A-1795insD late INa. Chronic incubation of SCN5A-1795insD hiPSC-CMs with mexiletine followed by wash-out increased peak INa, action potential (AP) upstroke velocity, and AP duration. Acute re-exposure did not impact on peak INa or AP upstroke velocity, but significantly decreased AP duration., Conclusion: These findings demonstrate for the first time the therapeutic benefit of mexiletine in a human cardiomyocyte model of SCN5A overlap syndrome., Competing Interests: Conflict of interest: None declared, (© The Author(s) 2023. Published by Oxford University Press on behalf of the European Society of Cardiology.)
- Published
- 2023
- Full Text
- View/download PDF
349. New pharmacological agents and novel cardiovascular pharmacotherapy strategies in 2022.
- Author
-
Tamargo J, Agewall S, Borghi C, Ceconi C, Cerbai E, Dan GA, Ferdinandy P, Grove EL, Rocca B, Sulzgruber P, Semb AG, Sossalla S, Niessner A, Kaski JC, and Dobrev D
- Abstract
Cardiovascular diseases (CVD) remain the leading cause of death worldwide and pharmacotherapy of most of them is suboptimal. Thus, there is a clear unmet clinical need to develop new pharmacological strategies with greater efficacy and better safety profiles. In this review, we summarize the most relevant advances in cardiovascular pharmacology in 2022 including the approval of first-in-class drugs that open new avenues for the treatment of obstructive hypertrophic cardiomyopathy (mavacamten), type 2 diabetes mellitus (tirzepatide), and heart failure (HF) independent of left ventricular ejection fraction (sodium-glucose cotransporter 2 inhibitors). We also dealt with fixed dose combination therapies repurposing different formulations of "old" drugs with well-known efficacy and safety for the treatment of patients with acute decompensated HF (acetazolamide plus loop diuretics), atherosclerotic cardiovascular disease (moderate-dose statin plus ezetimibe), Marfan syndrome (angiotensin receptor blockers plus β-blockers), and secondary cardiovascular prevention (i.e. low-dose aspirin, ramipril and atorvastatin), thereby filling existing gaps in knowledge, and opening new avenues for the treatment of CVD. Clinical trials confirming the role of dapagliflozin in patients with HF and mildly reduced or preserved ejection fraction, long-term evolocumab to reduce the risk of cardiovascular events, vitamin K antagonists for stroke prevention in patients with rheumatic heart disease-associated atrial fibrillation, antibiotic prophylaxis in patients at high risk for infective endocarditis before invasive dental procedures, and vutrisiran for the treatment of hereditary transthyretin-related amyloidosis with polyneuropathy were also reviewed. Finally, we briefly discuss recent clinical trials suggesting that FXIa inhibitors may have the potential to uncouple thrombosis from hemostasis and attenuate/prevent thromboembolic events with minimal disruption of hemostasis., (© The Author(s) 2023. Published by Oxford University Press on behalf of the European Society of Cardiology.)
- Published
- 2023
- Full Text
- View/download PDF
350. Slower Calcium Handling Balances Faster Cross-Bridge Cycling in Human MYBPC3 HCM.
- Author
-
Pioner JM, Vitale G, Steczina S, Langione M, Margara F, Santini L, Giardini F, Lazzeri E, Piroddi N, Scellini B, Palandri C, Schuldt M, Spinelli V, Girolami F, Mazzarotto F, van der Velden J, Cerbai E, Tesi C, Olivotto I, Bueno-Orovio A, Sacconi L, Coppini R, Ferrantini C, Regnier M, and Poggesi C
- Subjects
- Humans, Calcium metabolism, Carrier Proteins genetics, Carrier Proteins metabolism, Myocardium metabolism, Myocytes, Cardiac metabolism, Mutation, Calcium, Dietary metabolism, Cytoskeletal Proteins genetics, Induced Pluripotent Stem Cells metabolism, Cardiomyopathy, Hypertrophic pathology
- Abstract
Background: The pathogenesis of MYBPC3 -associated hypertrophic cardiomyopathy (HCM) is still unresolved. In our HCM patient cohort, a large and well-characterized population carrying the MYBPC3 :c772G>A variant (p.Glu258Lys, E258K) provides the unique opportunity to study the basic mechanisms of MYBPC3 -HCM with a comprehensive translational approach., Methods: We collected clinical and genetic data from 93 HCM patients carrying the MYBPC3 :c772G>A variant. Functional perturbations were investigated using different biophysical techniques in left ventricular samples from 4 patients who underwent myectomy for refractory outflow obstruction, compared with samples from non-failing non-hypertrophic surgical patients and healthy donors. Human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes and engineered heart tissues (EHTs) were also investigated., Results: Haplotype analysis revealed MYBPC3 :c772G>A as a founder mutation in Tuscany. In ventricular myocardium, the mutation leads to reduced cMyBP-C (cardiac myosin binding protein-C) expression, supporting haploinsufficiency as the main primary disease mechanism. Mechanical studies in single myofibrils and permeabilized muscle strips highlighted faster cross-bridge cycling, and higher energy cost of tension generation. A novel approach based on tissue clearing and advanced optical microscopy supported the idea that the sarcomere energetics dysfunction is intrinsically related with the reduction in cMyBP-C. Studies in single cardiomyocytes (native and hiPSC-derived), intact trabeculae and hiPSC-EHTs revealed prolonged action potentials, slower Ca
2+ transients and preserved twitch duration, suggesting that the slower excitation-contraction coupling counterbalanced the faster sarcomere kinetics. This conclusion was strengthened by in silico simulations., Conclusions: HCM-related MYBPC3 :c772G>A mutation invariably impairs sarcomere energetics and cross-bridge cycling. Compensatory electrophysiological changes (eg, reduced potassium channel expression) appear to preserve twitch contraction parameters, but may expose patients to greater arrhythmic propensity and disease progression. Therapeutic approaches correcting the primary sarcomeric defects may prevent secondary cardiomyocyte remodeling.- Published
- 2023
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.