1
|
Unneland E, Norvik A, Bergum D, Buckler DG, Bhardwaj A, Eftestøl TC, Aramendi E, Nordseth T, Abella BS, Kvaløy JT, Skogvoll E. Re-arrest immediately after return of spontaneous circulation: A retrospective observational study of in-hospital cardiac arrest. Acta Anaesthesiol Scand 2025; 69:e14567. [PMID: 39692065 DOI: 10.1111/aas.14567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 12/02/2024] [Accepted: 12/06/2024] [Indexed: 12/19/2024]
Abstract
BACKGROUND Patients who achieve return of spontaneous circulation (ROSC) after in-hospital cardiac arrest (IHCA) may re-arrest. This phenomenon has not been sufficiently investigated. The aim of this study was to examine the immediate (1-min) and short-term (20-min) risks of re-arrest in IHCA. METHODS We retrospectively analyzed four datasets of IHCA episodes, comprising defibrillator recordings collected between 2002 and 2022. Re-arrest was defined as the resumption of chest compressions following a period of ROSC after cardiac arrest of any duration. Parametric models were applied to calculate the immediate risk of re-arrest. In addition, we estimated the short-term risk of re-arrest within 20 min. RESULTS In 763 episodes of IHCA, we observed 316 re-arrests: 68% to pulseless electrical activity (PEA), 25% to ventricular fibrillation/ventricular tachycardia (VF/VT), and 7% to asystole. Most re-arrests occurred with the same rhythm as in the initial arrest. When ROSC was achieved from a non-shockable rhythm, the risk of re-arrest to a non-shockable rhythm was initially 2% per minute and decreased to 1% per minute after 9 min. The corresponding risk of re-arrest to VF/VT was constant at 2% per minute. If ROSC was obtained from a shockable rhythm, the risk of re-arrest to a shockable rhythm was initially 5% per minute, decreasing to 4% per minute after 9 min. The corresponding risk to a non-shockable rhythm was constant at 1% per minute. The risk of re-arrest within 20 min was 27%, and the overall risk of at least one re-arrest per episode was 33%. CONCLUSIONS The immediate risk of re-arrest was approximately 2% per minute, with the highest risk occurring as a reversion to VF/VT if ROSC was obtained from VF/VT. The risk of re-arrest within 20 min of the initial arrest was 27%, and the overall risk of at least one re-arrest per episode was 33%.
Collapse
Affiliation(s)
- Eirik Unneland
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Anders Norvik
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Anesthesia and Intensive Care Medicine, St. Olav University Hospital, Trondheim, Norway
| | - Daniel Bergum
- Department of Anesthesia and Intensive Care Medicine, St. Olav University Hospital, Trondheim, Norway
| | - David G Buckler
- Department of Emergency Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Abhishek Bhardwaj
- Department of Medicine, University of California, Riverside, California, USA
| | - Trygve Christian Eftestøl
- Department of Electrical Engineering and Computer Science, University of Stavanger, Stavanger, Norway
| | - Elisabete Aramendi
- Engineering School of Bilbao, BioRes Group, University of the Basque Country, Bilbao, Spain
| | - Trond Nordseth
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Anesthesia and Intensive Care Medicine, St. Olav University Hospital, Trondheim, Norway
- Division of Emergencies and Critical Care, Department of Research and Development, Oslo University Hospital, Oslo, Norway
| | - Benjamin S Abella
- Center for Resuscitation Science, Department of Emergency Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jan Terje Kvaløy
- Department of Mathematics and Physics, University of Stavanger, Stavanger, Norway
| | - Eirik Skogvoll
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Anesthesia and Intensive Care Medicine, St. Olav University Hospital, Trondheim, Norway
| |
Collapse
|
2
|
Shi Q, Wang J, Malik H, Li X, Streeter J, Sharafuddin J, Weatherford E, Stein D, Itan Y, Chen B, Hall D, Song LS, Abel ED. IRS2 Signaling Protects Against Stress-Induced Arrhythmia by Maintaining Ca 2+ Homeostasis. Circulation 2024; 150:1966-1983. [PMID: 39253856 PMCID: PMC11631690 DOI: 10.1161/circulationaha.123.065048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 08/13/2024] [Indexed: 09/11/2024]
Abstract
BACKGROUND The docking protein IRS2 (insulin receptor substrate protein-2) is an important mediator of insulin signaling and may also regulate other signaling pathways. Murine hearts with cardiomyocyte-restricted deletion of Irs2 (cIRS2-KO) are more susceptible to pressure overload-induced cardiac dysfunction, implying a critical protective role of IRS2 in cardiac adaptation to stress through mechanisms that are not fully understood. There is limited evidence regarding the function of IRS2 beyond metabolic homeostasis regulation, particularly in the context of cardiac disease. METHODS A retrospective analysis of an electronic medical record database was conducted to identify patients with IRS2 variants and assess their risk of cardiac arrhythmias. Arrhythmia susceptibility was examined in cIRS2-KO mice. The underlying mechanisms were investigated using confocal calcium imaging of ex vivo whole hearts and isolated cardiomyocytes to assess calcium handling, Western blotting to analyze the involved signaling pathways, and pharmacological and genetic interventions to rescue arrhythmias in cIRS2-KO mice. RESULTS The retrospective analysis identified patients with IRS2 variants of uncertain significance with a potential association to an increased risk of cardiac arrhythmias compared with matched controls. cIRS2-KO hearts were found to be prone to catecholamine-sensitive ventricular tachycardia and reperfusion ventricular tachycardia. Confocal calcium imaging of ex vivo whole hearts and single isolated cardiomyocytes from cIRS2-KO hearts revealed decreased Ca²⁺ transient amplitudes, increased spontaneous Ca²⁺ sparks, and reduced sarcoplasmic reticulum Ca²⁺ content during sympathetic stress, indicating sarcoplasmic reticulum dysfunction. We identified that overactivation of the AKT1/NOS3 (nitric oxide synthase 3)/CaMKII (Ca²⁺/calmodulin-dependent protein kinase II)/RyR2 (type 2 ryanodine receptor) signaling pathway led to calcium mishandling and catecholamine-sensitive ventricular tachycardia in cIRS2-KO hearts. Pharmacological AKT inhibition or genetic stabilization of RyR2 rescued catecholamine-sensitive ventricular tachycardia in cIRS2-KO mice. CONCLUSIONS Cardiac IRS2 inhibits sympathetic stress-induced AKT/NOS3/CaMKII/RyR2 overactivation and calcium-dependent arrhythmogenesis. This novel IRS2 signaling axis, essential for maintaining cardiac calcium homeostasis under stress, presents a promising target for developing new antiarrhythmic therapies.
Collapse
Affiliation(s)
- Qian Shi
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Jinxi Wang
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Hamza Malik
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Xuguang Li
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Jennifer Streeter
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Jacob Sharafuddin
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Eric Weatherford
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA
- Fraternal Order of Eagles Diabetes Research Center, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - David Stein
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
- Department of Genetics and Genome Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Yuval Itan
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
- Department of Genetics and Genome Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Biyi Chen
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Duane Hall
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA
- Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Long-Sheng Song
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA
- Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, Iowa City, IA
- Fraternal Order of Eagles Diabetes Research Center, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - E. Dale Abel
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA
- Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, Iowa City, IA
- Fraternal Order of Eagles Diabetes Research Center, Carver College of Medicine, University of Iowa, Iowa City, IA
- Current address, Department of Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA
| |
Collapse
|
3
|
Alanova P, Alan L, Neckar J, Ostadal B, Kolar F. Cardioprotective Effect of Chronic Hypoxia Involves Inhibition of Mitochondrial Permeability Transition Pore Opening. Physiol Res 2024; 73:881-884. [PMID: 39560196 PMCID: PMC11629960 DOI: 10.33549/physiolres.935427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 06/25/2024] [Indexed: 12/13/2024] Open
Abstract
The aim of the study was to examine the potential role of mitochondrial permeability transition pore (mPTP) in the cardioprotective effect of chronic continuous hypoxia (CH) against acute myocardial ischemia/reperfusion (I/R) injury. Adult male Wistar rats were adapted to CH for 3 weeks, while their controls were kept under normoxic conditions. Subsequently, they were subjected to I/R insult while being administered with mPTP inhibitor, cyclosporin A (CsA). Infarct size and incidence of ischemic and reperfusion arrhythmias were determined. Our results showed that adaptation to CH as well as CsA administration reduced myocardial infarct size in comparison to the corresponding control groups. However, administration of CsA did not amplify the beneficial effect of CH, suggesting that inhibition of mPTP opening contributes to the protective character of CH.
Collapse
Affiliation(s)
- P Alanova
- Laboratory of Developmental Cardiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic.
| | | | | | | | | |
Collapse
|
4
|
Belosludtseva NV, Uryupina TA, Pavlik LL, Mikheeva IB, Talanov EY, Venediktova NI, Serov DA, Stepanov MR, Ananyan MA, Mironova GD. Pathological Alterations in Heart Mitochondria in a Rat Model of Isoprenaline-Induced Myocardial Injury and Their Correction with Water-Soluble Taxifolin. Int J Mol Sci 2024; 25:11596. [PMID: 39519147 PMCID: PMC11547074 DOI: 10.3390/ijms252111596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 10/25/2024] [Accepted: 10/26/2024] [Indexed: 11/16/2024] Open
Abstract
Mitochondrial damage and associated oxidative stress are considered to be major contributory factors in cardiac pathology. One of the most potent naturally occurring antioxidants is taxifolin, especially in its water-soluble form. Herein, the effect of a 14-day course of the peroral application of the water-soluble taxifolin (aqTAX, 15 mg/kg of body weight) on the progression of ultrastructural and functional disorders in mitochondria and the heart's electrical activity in a rat model of myocardial injury induced with isoprenaline (ISO, 150 mg/kg/day for two consecutive days, subcut) was studied. The delayed ISO-induced myocardial damage was accompanied by an increase in the duration of RR and QT intervals, and long-term application of aqTAX partially restored the disturbed intraventricular conduction. It was shown that the injections of ISO lead to profound ultrastructural alterations of myofibrils and mitochondria in cardiomyocytes in the left ventricle myocardium, including the impairment of the ordered arrangement of mitochondria between myofibrils as well as a decrease in the size and the number of these organelles per unit area. In addition, a reduction in the protein level of the subunits of the respiratory chain complexes I-V and the activity of the antioxidant enzymes catalase, glutathione peroxidase, and Mn-SOD in mitochondria was observed. The application of aqTAX caused an increase in the efficiency of oxidation phosphorylation and a partial restoration of the morphometric parameters of mitochondria in the heart tissue of animals with the experimental pathology. These beneficial effects of aqTAX are associated with the inhibition of lipid peroxidation and the normalization of the enzymatic activities of glutathione peroxidase and Mn-SOD in rat cardiac mitochondria, which may reduce the oxidative damage to the organelles. Taken together, these data allow one to consider this compound as a promising cardioprotector in the complex therapy of heart failure.
Collapse
Affiliation(s)
- Natalia V. Belosludtseva
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya 3, 142290 Pushchino, Russia
| | - Tatyana A. Uryupina
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya 3, 142290 Pushchino, Russia
| | - Lyubov L. Pavlik
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya 3, 142290 Pushchino, Russia
| | - Irina B. Mikheeva
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya 3, 142290 Pushchino, Russia
| | - Eugeny Yu. Talanov
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya 3, 142290 Pushchino, Russia
| | - Natalya I. Venediktova
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya 3, 142290 Pushchino, Russia
| | - Dmitriy A. Serov
- Prokhorov General Physics Institute of the Russian Academy of Sciences, Vavilov St. 38, 119991 Moscow, Russia
| | | | | | - Galina D. Mironova
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya 3, 142290 Pushchino, Russia
| |
Collapse
|
5
|
Mohammed ASA, Naveed M, Szabados T, Szatmári I, Lőrinczi B, Mátyus P, Czompa A, Orvos P, Husti Z, Hornyik T, Topal L, Déri S, Jost N, Virág L, Bencsik P, Baczkó I, Varró A. Effects of SZV-2649, a new multiple ion channel inhibitor mexiletine analogue. Sci Rep 2024; 14:23188. [PMID: 39369049 PMCID: PMC11455950 DOI: 10.1038/s41598-024-73576-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 09/18/2024] [Indexed: 10/07/2024] Open
Abstract
The antiarrhythmic and cardiac electrophysiological effects of SZV-2649 that contains a 2,6-diiodophenoxy moiety but lacks the benzofuran ring system present in amiodarone, were studied in mammalian cell line, 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 (IC50: 342 and 529 nM, respectively). In canine ventricular myocytes, SZV-2649 (10 µM) decreased the densities of IKr, and Ito outward and INaL and ICaL inward currents. The compound (2.5-10 µM) elicited Class IB type Vmax reducing and Class III type action potential duration prolonging effects in dog right ventricular muscle preparations. In canine atrial muscle, SZV-2629 (2.5-10 µM) moderately prolonged action potential duration and this effect was greatly augmented in preparations pretreated with 1 µM carbachol. In conclusion, SZV-2649, has antiarrhythmic effects based on its multiple ion channel blocking properties. Since its chemical structure substantially differs from that of amiodarone, it is expected that SZV-2649 would exhibit fewer adverse effects than the currently used most effective multichannel inhibitor drug amiodarone and may be a promising molecule for further development.
Collapse
Grants
- EFOP-3.6.2-16-2017-00006, the UNKP for young researchers, UNKP-23-5-SZTE-704 Ministry of Human Capacities Hungary
- EFOP-3.6.2-16-2017-00006, the UNKP for young researchers, UNKP-23-5-SZTE-704 Ministry of Human Capacities Hungary
- EFOP-3.6.2-16-2017-00006, the UNKP for young researchers, UNKP-23-5-SZTE-704 Ministry of Human Capacities Hungary
- EFOP-3.6.2-16-2017-00006, the UNKP for young researchers, UNKP-23-5-SZTE-704 Ministry of Human Capacities Hungary
- EFOP-3.6.2-16-2017-00006, the UNKP for young researchers, UNKP-23-5-SZTE-704 Ministry of Human Capacities Hungary
- EFOP-3.6.2-16-2017-00006, the UNKP for young researchers, UNKP-23-5-SZTE-704 Ministry of Human Capacities Hungary
- EFOP-3.6.2-16-2017-00006, the UNKP for young researchers, UNKP-23-5-SZTE-704 Ministry of Human Capacities Hungary
- EFOP-3.6.2-16-2017-00006, the UNKP for young researchers, UNKP-23-5-SZTE-704 Ministry of Human Capacities Hungary
- EFOP-3.6.2-16-2017-00006, the UNKP for young researchers, UNKP-23-5-SZTE-704 Ministry of Human Capacities Hungary
- KDP-2020 Ministry for Innovation and Technology, Cooperative Doctoral Programme
- RRF-2.3.1-21-2022-00001 Recovery and Resilience Facility (RRF)
- NKFIH K 135464, K 142738, K 147212 TKP2021-EGA-32, FK 138223, GINOP-2.3.2.-15-2016-00006, GINOP-2.3.2.-15-2016-00040 National Research Development and Innovation Office
- NKFIH K 135464, K 142738, K 147212 TKP2021-EGA-32, FK 138223, GINOP-2.3.2.-15-2016-00006, GINOP-2.3.2.-15-2016-00040 National Research Development and Innovation Office
- NKFIH K 135464, K 142738, K 147212 TKP2021-EGA-32, FK 138223, GINOP-2.3.2.-15-2016-00006, GINOP-2.3.2.-15-2016-00040 National Research Development and Innovation Office
- NKFIH K 135464, K 142738, K 147212 TKP2021-EGA-32, FK 138223, GINOP-2.3.2.-15-2016-00006, GINOP-2.3.2.-15-2016-00040 National Research Development and Innovation Office
- NKFIH K 135464, K 142738, K 147212 TKP2021-EGA-32, FK 138223, GINOP-2.3.2.-15-2016-00006, GINOP-2.3.2.-15-2016-00040 National Research Development and Innovation Office
- NKFIH K 135464, K 142738, K 147212 TKP2021-EGA-32, FK 138223, GINOP-2.3.2.-15-2016-00006, GINOP-2.3.2.-15-2016-00040 National Research Development and Innovation Office
- NKFIH K 135464, K 142738, K 147212 TKP2021-EGA-32, FK 138223, GINOP-2.3.2.-15-2016-00006, GINOP-2.3.2.-15-2016-00040 National Research Development and Innovation Office
- SZTE AOK-KKA 2021, SZGYA 2021, SZTE AOK-KKA 2022 The Albert Szent-Györgyi Medical School institutional grant
- SZTE AOK-KKA 2021, SZGYA 2021, SZTE AOK-KKA 2022 The Albert Szent-Györgyi Medical School institutional grant
- SZTE AOK-KKA 2021, SZGYA 2021, SZTE AOK-KKA 2022 The Albert Szent-Györgyi Medical School institutional grant
- HUN-REN TKI project Hungarian Research Network
- HUN-REN TKI project Hungarian Research Network
- HUN-REN TKI project Hungarian Research Network
- bo_481_21 Hungarian Academy of Sciences, János Bolyai Research Scholarships
- RRF-2.3.1-21-2022-00003 National Heart Laboratory, Hungary
Collapse
Affiliation(s)
- Aiman Saleh A Mohammed
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Muhammad Naveed
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Tamara Szabados
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - István Szatmári
- Institute of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Szeged, Szeged, Hungary
- HUN-REN-SZTE Stereochemistry Research Group, Hungarian Research Network, Szeged, Hungary
| | - Bálint Lőrinczi
- Institute of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Szeged, Szeged, Hungary
| | - Péter Mátyus
- Department of Organic Chemistry, Semmelweis University, Budapest, Hungary
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine, Budapest, Hungary
| | - Andrea Czompa
- Department of Organic Chemistry, Semmelweis University, Budapest, Hungary
| | - Péter Orvos
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Zoltán Husti
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Tibor Hornyik
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Leila Topal
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Szilvia Déri
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
- HUN-REN-SZTE Research Group for Cardiovascular Pharmacology, Hungarian Research Network, Szeged, Hungary
| | - Norbert Jost
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
- HUN-REN-SZTE Research Group for Cardiovascular Pharmacology, Hungarian Research Network, Szeged, Hungary
- Interdisciplinary Research and Development and Innovation Centre of Excellence, University of Szeged, Szeged, Hungary
| | - László Virág
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
- Interdisciplinary Research and Development and Innovation Centre of Excellence, University of Szeged, Szeged, Hungary
| | - Péter Bencsik
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - István Baczkó
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary.
- Interdisciplinary Research and Development and Innovation Centre of Excellence, University of Szeged, Szeged, Hungary.
| | - András Varró
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary.
- HUN-REN-SZTE Research Group for Cardiovascular Pharmacology, Hungarian Research Network, Szeged, Hungary.
- Interdisciplinary Research and Development and Innovation Centre of Excellence, University of Szeged, Szeged, Hungary.
| |
Collapse
|
6
|
Mokhtari B, Jessri A, Ghaffari S, Badalzadeh R. Superior Anti-arrhythmogenic Effect of Combined Conditioning with Nicotinamide Mononucleotide and Ubiquinol in Myocardial Ischemia/Reperfusion Injury in Aged Rats. Adv Pharm Bull 2024; 14:686-695. [PMID: 39494250 PMCID: PMC11530879 DOI: 10.34172/apb.2024.044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 04/05/2024] [Accepted: 04/14/2024] [Indexed: 11/05/2024] Open
Abstract
Purpose Lethal ventricular arrhythmias are a significant clinical concern following reperfusion therapies in elderly patients with myocardial infarction. The combination of multi-target therapies to achieve optimal anti-arrhythmogenesis and improve the chances of successful translation for patient benefit has prompted considerable interest. This study examined the anti-arrhythmic effect of nicotinamide mononucleotide (NMN)/ubiquinol combination treatment following myocardial ischemia/reperfusion (IR) injury in aged rats, with an emphasis on the role of oxidative stress and nitric oxide (NO). Methods Male Wistar rats (n=30, 22-24 months old, 400-450 g) were randomized into five groups with or without IR and/or NMN and ubiquinol, either alone or in combination. NMN (100 mg/kg/48 hours) was administered intraperitoneally for 28 days before IR, and ubiquinol (30 mg/kg) was injected intravenously at early reperfusion. Electrocardiographic signals were recorded during the ischemia and the first 30 minutes of reperfusion. Two hours after reperfusion, myocardial hemodynamic and LDH release were measured, and the left ventricle samples were obtained to evaluate oxidative stress markers and NO levels. Results NMN/ubiquinol combination treatment significantly minimized the occurrence and severity of IR-induced arrhythmias, improved myocardial function, and reduced LDH release (P<0.05). It also decreased MDA content, increased superoxide dismutase (SOD), glutathione peroxidase (GPX), and catalase (CAT) activities, and enhanced NO formation (P<0.05). This combined treatment showed greater efficacy than the single treatments. Conclusion This study revealed the anti-arrhythmic effect of NMN/ubiquinol combination treatment in IR-treated aged rats, which may be associated with reduced oxidative stress and increased NO formation. This combinational approach deserves more investigation due to its potential to confer better anti-arrhythmic effect during aging.
Collapse
Affiliation(s)
- Behnaz Mokhtari
- Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amirhossein Jessri
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Samad Ghaffari
- Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Badalzadeh
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
7
|
Jiayu L, Xiaofeng L, Jinhong C, Fangjun D, Boya F, Xin Z, Zidong C, Rui T, Lu Y, Shule Q, Runying W, Wuxun D. Study on the mechanisms and Pharmacodynamic substances of Lian-Gui-Ning-Xin-Tang on Arrhythmia Therapy based on Pharmacodynamic-Pharmacokinetic associations. Heliyon 2024; 10:e36104. [PMID: 39253118 PMCID: PMC11381611 DOI: 10.1016/j.heliyon.2024.e36104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 08/08/2024] [Accepted: 08/09/2024] [Indexed: 09/11/2024] Open
Abstract
Background The Chinese herbal compound Lian-Gui-Ning-Xin-Tang (LGNXT), composed of 9 herbs, has a significant antiarrhythmic effect. Previous studies have confirmed that preventing intracellular Ca2+ overload and maintaining intracellular Ca2+ homeostasis may be the important antiarrhythmic mechanisms of LGNXT. Recent studies are focused on elucidating the mechanisms and pharmacodynamic substances of LGNXT. Purpose 1) To investigate the antiarrhythmic mechanisms of LGNXT; 2) to explore the association of pharmacodynamics (PD) and pharmacokinetics (PK) of the potential pharmacodynamic substances in LGNXT to further verify the mechanisms of action. Methods First, pharmacodynamic studies were conducted to determine the effect of LGNXT in arrhythmia at the electrophysiological, molecular, and tissue levels, and the "effect-time" relationship of LGNXT was further proposed. Next, an HPLC-MS/MS method was established to identify the "dose-time" relationship of the 9 potential compounds. Combining the "effect-time" and "dose-time" curves, the active ingredients closely related to the inhibition of inflammation, oxidative stress, and energy metabolism were identified to further verify the mechanisms and pharmacodynamic substances of LGNXT. Results Pretreatment with LGNXT could delay the occurrence of arrhythmias and reduce their duration and severity. LGNXT exerted antiarrhythmic effects by inhibiting MDA, LPO, IL-6, and cAMP; restoring Cx43 coupling function; and upregulating SOD, Ca2+-ATPase, and Na+-K+-ATPase levels. PK-PD association showed that nobiletin, methylophiopogonanone A, trigonelline, cinnamic acid, liquiritin, dehydropolisic acid, berberine, and puerarin were the main pharmacodynamic substances responsible for inhibiting the inflammatory response in arrhythmia. Methylophiopogonanone A, dehydropalingic acid, nobiletin, trigonelline, berberine, and puerarin in LGNXT exerted antiarrhythmic effects by inhibiting oxidative stress. Dehydropalingic acid, berberine, cinnamic acid, liquiritin, puerarin, trigonelline, methylophiopogonanone A, nobiletin, and tetrahydropalmatine exerted antiarrhythmic effects by inhibiting the energy-metabolism process. Conclusions LGNXT had a positive intervention effect on arrhythmias, especially ventricular tachyarrhythmias, which could inhibit inflammation, oxidative stress, and energy metabolism; positively stabilize the structure, and remodify the function of myocardial cell membranes. Additionally, the PD-PK association study revealed that methylophiopogonanone A, berberine, trigonelline, liquiritin, puerarin, tetrahydropalmatine, nobiletin, dehydropachymic acid, and cinnamic acid directly targeted inflammation, oxidative stress, and energy metabolism, which could be considered the pharmacodynamic substances of LGNXT. Thus, the antiarrhythmic mechanisms of LGNXT were further elucidated.
Collapse
Affiliation(s)
- Liang Jiayu
- Department of TCM, The First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang Hangzhou 310003, China
| | - Li Xiaofeng
- Department of Cardiology, The Second Affiliated Hospital of Tianjin University of TCM, Tianjin 300150, China
| | - Chen Jinhong
- School of Rehabilitation Medicine, Shandong Second Medical University, Shandong Weifang, 261053, China
| | - Deng Fangjun
- Department of Cardiology, Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin 300091, China
| | - Fan Boya
- Department of Medical qualification examination, National Administration of Traditional Chinese Medicine TCM Qualification Certification Center, Beijing 100120, China
| | - Zhen Xin
- Graduate School, Tianjin University of TCM, Tianjin 301617, China
| | - Cong Zidong
- Department of Cardiology, The Second Affiliated Hospital of Tianjin University of TCM, Tianjin 300150, China
| | - Tao Rui
- Department of TCM, Tianjin University of TCM, Tianjin, 301617, China
| | - Yu Lu
- Graduate School, Tianjin University of TCM, Tianjin 301617, China
| | - Qian Shule
- Graduate School, Tianjin University of TCM, Tianjin 301617, China
| | - Wang Runying
- Graduate School, Tianjin University of TCM, Tianjin 301617, China
| | - Du Wuxun
- Department of Cardiology, The Second Affiliated Hospital of Tianjin University of TCM, Tianjin 300150, China
| |
Collapse
|
8
|
Wulkan F, Romagnuolo R, Qiang B, Valdman Sadikov T, Kim KP, Quesnel E, Jiang W, Andharia N, Weyers JJ, Ghugre NR, Ozcan B, Alibhai FJ, Laflamme MA. Stem cell-derived cardiomyocytes expressing a dominant negative pacemaker HCN4 channel do not reduce the risk of graft-related arrhythmias. Front Cardiovasc Med 2024; 11:1374881. [PMID: 39045008 PMCID: PMC11263024 DOI: 10.3389/fcvm.2024.1374881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 06/11/2024] [Indexed: 07/25/2024] Open
Abstract
Background Human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) show tremendous promise for cardiac regeneration following myocardial infarction (MI), but their transplantation gives rise to transient ventricular tachycardia (VT) in large-animal MI models, representing a major hurdle to translation. Our group previously reported that these arrhythmias arise from a focal mechanism whereby graft tissue functions as an ectopic pacemaker; therefore, we hypothesized that hPSC-CMs engineered with a dominant negative form of the pacemaker ion channel HCN4 (dnHCN4) would exhibit reduced automaticity and arrhythmogenic risk following transplantation. Methods We used CRISPR/Cas9-mediated gene-editing to create transgenic dnHCN4 hPSC-CMs, and their electrophysiological behavior was evaluated in vitro by patch-clamp recordings and optical mapping. Next, we transplanted WT and homozygous dnHCN4 hPSC-CMs in a pig MI model and compared post-transplantation outcomes including the incidence of spontaneous arrhythmias and graft structure by immunohistochemistry. Results In vitro dnHCN4 hPSC-CMs exhibited significantly reduced automaticity and pacemaker funny current (I f ) density relative to wildtype (WT) cardiomyocytes. Following transplantation with either dnHCN4 or WT hPSC-CMs, all recipient hearts showed transmural infarct scar that was partially remuscularized by scattered islands of human myocardium. However, in contrast to our hypothesis, both dnHCN4 and WT hPSC-CM recipients exhibited frequent episodes of ventricular tachycardia (VT). Conclusions While genetic silencing of the pacemaker ion channel HCN4 suppresses the automaticity of hPSC-CMs in vitro, this intervention is insufficient to reduce VT risk post-transplantation in the pig MI model, implying more complex mechanism(s) are operational in vivo.
Collapse
Affiliation(s)
- Fanny Wulkan
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
| | - Rocco Romagnuolo
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
| | - Beiping Qiang
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
| | | | | | - Elya Quesnel
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
| | - Wenlei Jiang
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
| | - Naaz Andharia
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
| | - Jill J. Weyers
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Nilesh R. Ghugre
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, ON, Canada
- Schulich Heart Program, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Bilgehan Ozcan
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
| | - Faisal J. Alibhai
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
| | - Michael A. Laflamme
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
- Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
9
|
Yao ZJ, Jiang YP, Yuan D, Hong P, He MJ, Li FX, Xu SY, Lin HB, Zhang HF. Decreased connexin 40 expression of the sinoatrial node mediates ischemic stroke-induced arrhythmia in mice. Exp Neurol 2024; 376:114773. [PMID: 38599368 DOI: 10.1016/j.expneurol.2024.114773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 03/23/2024] [Accepted: 04/06/2024] [Indexed: 04/12/2024]
Abstract
BACKGROUND Arrhythmia is the most common cardiac complication after ischemic stroke. Connexin 40 is the staple component of gap junctions, which influences the propagation of cardiac electrical signals in the sinoatrial node. However, the role of connexin 40 in post-stroke arrhythmia remains unclear. METHODS In this study, a permanent middle cerebral artery occlusion model was used to simulate the occurrence of an ischemic stroke. Subsequently, an electrocardiogram was utilized to record and assess variations in electrocardiogram measures. In addition, optical tissue clearing and whole-mount immunofluorescence staining were used to confirm the anatomical localization of the sinoatrial node, and the sinoatrial node tissue was collected for RNA sequencing to screen for potential pathological mechanisms. Lastly, the rAAV9-Gja5 virus was injected with ultrasound guidance into the heart to increase Cx40 expression in the sinoatrial node. RESULTS We demonstrated that the mice suffering from a permanent middle cerebral artery occlusion displayed significant arrhythmia, including atrial fibrillation, premature ventricular contractions, atrioventricular block, and abnormal electrocardiogram parameters. Of note, we observed a decrease in connexin 40 expression within the sinoatrial node after the ischemic stroke via RNA sequencing and western blot. Furthermore, rAAV9-Gja5 treatment ameliorated the occurrence of arrhythmia following stroke. CONCLUSIONS In conclusion, decreased connexin 40 expression in the sinoatrial node contributed to the ischemic stroke-induced cardiac arrhythmia. Therefore, enhancing connexin 40 expression holds promise as a potential therapeutic approach for ischemic stroke-induced arrhythmia.
Collapse
Affiliation(s)
- Zhi-Jun Yao
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Yan-Pin Jiang
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China; Department of Anesthesiology, The First Hospital Affiliated to the Army Medical University, Chongqing 400038, China
| | - Dan Yuan
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Pu Hong
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Meng-Jiao He
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Feng-Xian Li
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Shi-Yuan Xu
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Hong-Bin Lin
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China.
| | - Hong-Fei Zhang
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China.
| |
Collapse
|
10
|
Shi Q, Malik H, Crawford RM, Streeter J, Wang J, Huo R, Shih JC, Chen B, Hall D, Abel ED, Song LS, Anderson EJ. Cardiac monoamine oxidase-A inhibition protects against catecholamine-induced ventricular arrhythmias via enhanced diastolic calcium control. Cardiovasc Res 2024; 120:596-611. [PMID: 38198753 PMCID: PMC11074799 DOI: 10.1093/cvr/cvae012] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 11/01/2023] [Accepted: 11/22/2023] [Indexed: 01/12/2024] Open
Abstract
AIMS A mechanistic link between depression and risk of arrhythmias could be attributed to altered catecholamine metabolism in the heart. Monoamine oxidase-A (MAO-A), a key enzyme involved in catecholamine metabolism and longstanding antidepressant target, is highly expressed in the myocardium. The present study aimed to elucidate the functional significance and underlying mechanisms of cardiac MAO-A in arrhythmogenesis. METHODS AND RESULTS Analysis of the TriNetX database revealed that depressed patients treated with MAO inhibitors had a lower risk of arrhythmias compared with those treated with selective serotonin reuptake inhibitors. This effect was phenocopied in mice with cardiomyocyte-specific MAO-A deficiency (cMAO-Adef), which showed a significant reduction in both incidence and duration of catecholamine stress-induced ventricular tachycardia compared with wild-type mice. Additionally, cMAO-Adef cardiomyocytes exhibited altered Ca2+ handling under catecholamine stimulation, with increased diastolic Ca2+ reuptake, reduced diastolic Ca2+ leak, and diminished systolic Ca2+ release. Mechanistically, cMAO-Adef hearts had reduced catecholamine levels under sympathetic stress, along with reduced levels of reactive oxygen species and protein carbonylation, leading to decreased oxidation of Type II PKA and CaMKII. These changes potentiated phospholamban (PLB) phosphorylation, thereby enhancing diastolic Ca2+ reuptake, while reducing ryanodine receptor 2 (RyR2) phosphorylation to decrease diastolic Ca2+ leak. Consequently, cMAO-Adef hearts exhibited lower diastolic Ca2+ levels and fewer arrhythmogenic Ca2+ waves during sympathetic overstimulation. CONCLUSION Cardiac MAO-A inhibition exerts an anti-arrhythmic effect by enhancing diastolic Ca2+ handling under catecholamine stress.
Collapse
MESH Headings
- Animals
- Female
- Humans
- Male
- Mice
- Calcium/metabolism
- Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism
- Catecholamines/metabolism
- Cells, Cultured
- Cyclic AMP-Dependent Protein Kinases/metabolism
- Diastole/drug effects
- Disease Models, Animal
- Heart Rate/drug effects
- Mice, Inbred C57BL
- Mice, Knockout
- Monoamine Oxidase/metabolism
- Monoamine Oxidase Inhibitors/pharmacology
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/enzymology
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Phosphorylation
- Reactive Oxygen Species/metabolism
- Ryanodine Receptor Calcium Release Channel/metabolism
- Tachycardia, Ventricular/enzymology
- Tachycardia, Ventricular/physiopathology
Collapse
Affiliation(s)
- Qian Shi
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, 285 Newton Rd, Iowa City, IA 52242, USA
| | - Hamza Malik
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, 285 Newton Rd, Iowa City, IA 52242, USA
| | - Rachel M Crawford
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, 180 S Grand Ave., Iowa City, IA 52242, USA
| | - Jennifer Streeter
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, 285 Newton Rd, Iowa City, IA 52242, USA
| | - Jinxi Wang
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, 285 Newton Rd, Iowa City, IA 52242, USA
| | - Ran Huo
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, 180 S Grand Ave., Iowa City, IA 52242, USA
| | - Jean C Shih
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, 1985 Zonal Avenue, Los Angeles, CA 90089, USA
| | - Biyi Chen
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, 285 Newton Rd, Iowa City, IA 52242, USA
| | - Duane Hall
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, 285 Newton Rd, Iowa City, IA 52242, USA
- Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, CBRB 2267285, Newton Rd, Iowa City, IA 52242, USA
| | - E Dale Abel
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, 285 Newton Rd, Iowa City, IA 52242, USA
- Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, CBRB 2267285, Newton Rd, Iowa City, IA 52242, USA
- Fraternal Order of Eagles Diabetes Research Center, Carver College of Medicine, University of Iowa, 169 Newton Rd, Iowa City, IA 52242, USA
| | - Long-Sheng Song
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, 285 Newton Rd, Iowa City, IA 52242, USA
- Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, CBRB 2267285, Newton Rd, Iowa City, IA 52242, USA
- Fraternal Order of Eagles Diabetes Research Center, Carver College of Medicine, University of Iowa, 169 Newton Rd, Iowa City, IA 52242, USA
| | - Ethan J Anderson
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, 180 S Grand Ave., Iowa City, IA 52242, USA
- Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, CBRB 2267285, Newton Rd, Iowa City, IA 52242, USA
- Fraternal Order of Eagles Diabetes Research Center, Carver College of Medicine, University of Iowa, 169 Newton Rd, Iowa City, IA 52242, USA
| |
Collapse
|
11
|
Kucera C, Ramalingam A, Srivastava S, Bhatnagar A, Carll AP. Nicotine Formulation Influences the Autonomic and Arrhythmogenic Effects of Electronic Cigarettes. Nicotine Tob Res 2024; 26:536-544. [PMID: 38011908 PMCID: PMC11033561 DOI: 10.1093/ntr/ntad237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 10/09/2023] [Accepted: 11/20/2023] [Indexed: 11/29/2023]
Abstract
INTRODUCTION Evidence is mounting that electronic cigarette (e-cig) use induces cardiac sympathetic dominance and electrical dysfunction conducive to arrhythmias and dependent upon nicotine. A variety of nicotine types and concentrations are available in e-cigs, but their relative cardiovascular effects remain unclear. Here we examine how different nicotine forms (racemic, free base, and salt) and concentrations influence e-cig-evoked cardiac dysfunction and arrhythmogenesis and provide a mechanism for nicotine-salt-induced autonomic imbalance. METHODS ECG-telemetered C57BL/6J mice were exposed to filtered air (FA) or e-cig aerosols from propylene glycol and vegetable glycerin solvents either without nicotine (vehicle) or with increasing nicotine concentrations (1%, 2.5%, and 5%) for three 9-minute puff sessions per concentration. Spontaneous ventricular premature beat (VPB) incidence rates, heart rate, and heart rate variability (HRV) were compared between treatments. Subsequently, to test the role of β1-adrenergic activation in e-cig-induced cardiac effects, mice were pretreated with atenolol and exposed to either FA or 2.5% nicotine salt. RESULTS During puffing and washout phases, ≥2.5% racemic nicotine reduced heart rate and increased HRV relative to FA and vehicle controls, indicating parasympathetic dominance. Relative to both controls, 5% nicotine salt elevated heart rate and decreased HRV during washout, suggesting sympathetic dominance, and also increased VPB frequency. Atenolol abolished e-cig-induced elevations in heart rate and declines in HRV during washout, indicating e-cig-evoked sympathetic dominance is mediated by β1-adrenergic stimulation. CONCLUSIONS Our findings suggest that inhalation of e-cig aerosols from nicotine-salt-containing e-liquids could increase the cardiovascular risks of vaping by inducing sympathetic dominance and cardiac arrhythmias. IMPLICATIONS Exposure to e-cig aerosols containing commercially relevant concentrations of nicotine salts may increase nicotine delivery and impair cardiac function by eliciting β1-adrenoceptor-mediated sympathoexcitation and provoking ventricular arrhythmias. If confirmed in humans, our work suggests that regulatory targeting of nicotine salts through minimum pH standards or limits on acid additives in e-liquids may mitigate the public health risks of vaping.
Collapse
Affiliation(s)
- Cory Kucera
- Department of Physiology, University of Louisville School of Medicine (ULSOM), Louisville, KY, USA
- Christina Lee Brown Envirome Institute, ULSOM, Louisville, KY, USA
- American Heart Association Tobacco Regulation and Addiction Center 2.0 (A-TRAC 2.0), ULSOM, Louisville, KY, USA
- Center for Cardiometabolic Science, ULSOM, Louisville, KY, USA
| | - Anand Ramalingam
- Christina Lee Brown Envirome Institute, ULSOM, Louisville, KY, USA
- American Heart Association Tobacco Regulation and Addiction Center 2.0 (A-TRAC 2.0), ULSOM, Louisville, KY, USA
- Center for Cardiometabolic Science, ULSOM, Louisville, KY, USA
| | - Shweta Srivastava
- Christina Lee Brown Envirome Institute, ULSOM, Louisville, KY, USA
- American Heart Association Tobacco Regulation and Addiction Center 2.0 (A-TRAC 2.0), ULSOM, Louisville, KY, USA
- Center for Cardiometabolic Science, ULSOM, Louisville, KY, USA
| | - Aruni Bhatnagar
- Christina Lee Brown Envirome Institute, ULSOM, Louisville, KY, USA
- American Heart Association Tobacco Regulation and Addiction Center 2.0 (A-TRAC 2.0), ULSOM, Louisville, KY, USA
- Center for Cardiometabolic Science, ULSOM, Louisville, KY, USA
- Division of Environmental Medicine, ULSOM, Louisville, KY, USA
- Center for Integrative Environmental Health Sciences, ULSOM, Louisville, KY, USA
| | - Alex P Carll
- Department of Physiology, University of Louisville School of Medicine (ULSOM), Louisville, KY, USA
- Christina Lee Brown Envirome Institute, ULSOM, Louisville, KY, USA
- American Heart Association Tobacco Regulation and Addiction Center 2.0 (A-TRAC 2.0), ULSOM, Louisville, KY, USA
- Center for Cardiometabolic Science, ULSOM, Louisville, KY, USA
- Division of Environmental Medicine, ULSOM, Louisville, KY, USA
- Center for Integrative Environmental Health Sciences, ULSOM, Louisville, KY, USA
| |
Collapse
|
12
|
Yu Y, Liu JY, Yang HJ, Luo XQ, Gao XP, Huang XX, Tang AX, Mary Cheng HY, Liu WC, Zhang P. Circadian disruption during fetal development promotes pathological cardiac remodeling in male mice. iScience 2024; 27:109008. [PMID: 38352228 PMCID: PMC10863319 DOI: 10.1016/j.isci.2024.109008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 12/19/2023] [Accepted: 01/22/2024] [Indexed: 02/16/2024] Open
Abstract
Disruption of circadian rhythms during fetal development may predispose mice to developing heart disease later in life. Here, we report that male, but not female, mice that had experienced chronic circadian disturbance (CCD) in utero were more susceptible to pathological cardiac remodeling compared with mice that had developed under normal intrauterine conditions. CCD-treated males showed ventricular chamber dilatation, enhanced myocardial fibrosis, decreased contractility, higher rates of induced tachyarrhythmia, and elevated expression of biomarkers for heart failure and myocardial remodeling. In utero CCD exposure also triggered sex-dependent changes in cardiac gene expression, including upregulation of the secretoglobin gene, Scgb1a1, in males. Importantly, cardiac overexpression of Scgb1a1 was sufficient to induce myocardial hypertrophy in otherwise naive male mice. Our findings reveal that in utero CCD exposure predisposes male mice to pathological remodeling of the heart later in life, likely as a consequence of SCGB1A1 upregulation.
Collapse
Affiliation(s)
- Yang Yu
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan 646000, China
- Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Jing-Yu Liu
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Hui-Jiao Yang
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xiao-Qin Luo
- Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Xiao-Ping Gao
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xiao-Xin Huang
- School of Basic Medical Science, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Ao-Xue Tang
- School of Basic Medical Science, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Hai-Ying Mary Cheng
- Department of Biology, University of Toronto Mississauga, Mississauga, Ontario L5L 1C6, Canada
| | - Wei-Chao Liu
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Peng Zhang
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan 646000, China
| |
Collapse
|
13
|
Oknińska M, Duda MK, Czarnowska E, Bierła J, Paterek A, Mączewski M, Mackiewicz U. Sex- and age-dependent susceptibility to ventricular arrhythmias in the rat heart ex vivo. Sci Rep 2024; 14:3460. [PMID: 38342936 PMCID: PMC10859380 DOI: 10.1038/s41598-024-53803-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 02/05/2024] [Indexed: 02/13/2024] Open
Abstract
The incidence of life-threatening ventricular arrhythmias, the most common cause of sudden cardiac death (SCD), depends largely on the arrhythmic substrate that develops in the myocardium during the aging process. There is a large deficit of comparative studies on the development of this substrate in both sexes, with a particular paucity of studies in females. To identify the substrates of arrhythmia, fibrosis, cardiomyocyte hypertrophy, mitochondrial density, oxidative stress, antioxidant defense and intracellular Ca2+ signaling in isolated cardiomyocytes were measured in the hearts of 3- and 24-month-old female and male rats. Arrhythmia susceptibility was assessed in ex vivo perfused hearts after exposure to isoproterenol (ISO) and hydrogen peroxide (H2O2). The number of ventricular premature beats (PVBs), ventricular tachycardia (VT) and ventricular fibrillation (VF) episodes, as well as intrinsic heart rate, QRS and QT duration, were measured in ECG signals recorded from the surfaces of the beating hearts. After ISO administration, VT/VFs were formed only in the hearts of males, mainly older ones. In contrast, H2O2 led to VT/VF formation in the hearts of rats of both sexes but much more frequently in older males. We identified several components of the arrhythmia substrate that develop in the myocardium during the aging process, including high spontaneous ryanodine receptor activity in cardiomyocytes, fibrosis of varying severity in different layers of the myocardium (nonheterogenic fibrosis), and high levels of oxidative stress as measured by nitrated tyrosine levels. All of these elements appeared at a much greater intensity in male individuals during the aging process. On the other hand, in aging females, antioxidant defense at the level of H2O2 detoxification, measured as glutathione peroxidase expression, was weaker than that in males of the same age. We showed that sex has a significant effect on the development of an arrhythmic substrate during aging. This substrate determines the incidence of life-threatening ventricular arrhythmias in the presence of additional stimuli with proarrhythmic potential, such as catecholamine stimulation or oxidative stress, which are constant elements in the pathomechanism of most cardiovascular diseases.
Collapse
Affiliation(s)
- Marta Oknińska
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, Marymoncka 99/103, 01-813, Warsaw, Poland
| | - Monika Katarzyna Duda
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, Marymoncka 99/103, 01-813, Warsaw, Poland
| | - Elżbieta Czarnowska
- Department of Pathology, The Children's Memorial Health Institute, Aleja Dzieci Polskich 20, 04-736, Warsaw, Poland
- Department of Pathology, Medical University of Warsaw, Żwirki i Wigury 61, 02-091, Warsaw, Poland
| | - Joanna Bierła
- Department of Pathology, The Children's Memorial Health Institute, Aleja Dzieci Polskich 20, 04-736, Warsaw, Poland
| | - Aleksandra Paterek
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, Marymoncka 99/103, 01-813, Warsaw, Poland
| | - Michał Mączewski
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, Marymoncka 99/103, 01-813, Warsaw, Poland
| | - Urszula Mackiewicz
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, Marymoncka 99/103, 01-813, Warsaw, Poland.
| |
Collapse
|
14
|
Ferrand MC, Giordano G, Mougenot N, Laporte PL, Vignier N, Leclerc A, Algalarrondo V, Extramiana F, Charpentier F, Neyroud N. Intracardiac electrophysiology to characterize susceptibility to ventricular arrhythmias in murine models. Front Physiol 2024; 15:1326663. [PMID: 38322613 PMCID: PMC10846502 DOI: 10.3389/fphys.2024.1326663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 01/08/2024] [Indexed: 02/08/2024] Open
Abstract
Introduction: Sudden cardiac death (SCD) and ventricular fibrillation are rare but severe complications of many cardiovascular diseases and represent a major health issue worldwide. Although the primary causes are often acute or chronic coronary diseases, genetic conditions, such as inherited channelopathies or non-ischemic cardiomyopathies are leading causes of SCD among the young. However, relevant experimental models to study the underlying mechanisms of arrhythmias and develop new therapies are still needed. The number of genetically engineered mouse models with cardiac phenotype is growing, making electrophysiological studies in mice essential tools to study arrhythmogenicity and arrhythmia mechanisms and to test novel treatments. Recently, intracardiac catheterization via the jugular vein was described to induce and record ventricular arrhythmias in living anesthetized mice. Several strategies have been reported, developed in healthy wild-type animals and based on aggressive right ventricular stimulation. Methods: Here, we report a protocol based on programmed electrical stimulation (PES) performed in clinical practice in patients with cardiac rhythm disorders, adapted to two transgenic mice models of arrhythmia - Brugada syndrome and cardiolaminopathy. Results: We show that this progressive protocol, based on a limited number of right ventricular extrastimuli, enables to reveal different rhythmic phenotypes between control and diseased mice. In this study, we provide detailed information on PES in mice, including catheter positioning, stimulation protocols, intracardiac and surface ECG interpretation and we reveal a higher susceptibility of two mouse lines to experience triggered ventricular arrhythmias, when compared to control mice. Discussion: Overall, this technique allows to characterize arrhythmias and provides results in phenotyping 2 arrhythmogenic-disease murine models.
Collapse
Affiliation(s)
- Marine C. Ferrand
- Sorbonne Université, Inserm, Research Unit on Cardiovascular and Metabolic Diseases, UMRS-1166, Paris, France
| | - Gauthier Giordano
- Sorbonne Université, Inserm, Research Unit on Cardiovascular and Metabolic Diseases, UMRS-1166, Paris, France
- Competence Center for Hereditary or Rare Heart Diseases, Centre Hospitalier Régional Universitaire de Nancy, Vandœuvre-lès-Nancy, France
| | | | - Pierre-Léo Laporte
- Sorbonne Université, Inserm, Research Unit on Cardiovascular and Metabolic Diseases, UMRS-1166, Paris, France
- Reference Center for Inherited Arrhythmic Syndromes, Hôpital Bichat, APHP, Université de Paris Cité, Paris, France
| | - Nicolas Vignier
- Sorbonne Université, Inserm, UMRS-974, Center of Research in Myology, Institute of Myology, Paris, France
| | - Arnaud Leclerc
- Sorbonne Université, Inserm, Research Unit on Cardiovascular and Metabolic Diseases, UMRS-1166, Paris, France
| | - Vincent Algalarrondo
- Reference Center for Inherited Arrhythmic Syndromes, Hôpital Bichat, APHP, Université de Paris Cité, Paris, France
| | - Fabrice Extramiana
- Sorbonne Université, Inserm, Research Unit on Cardiovascular and Metabolic Diseases, UMRS-1166, Paris, France
- Reference Center for Inherited Arrhythmic Syndromes, Hôpital Bichat, APHP, Université de Paris Cité, Paris, France
| | | | - Nathalie Neyroud
- Sorbonne Université, Inserm, Research Unit on Cardiovascular and Metabolic Diseases, UMRS-1166, Paris, France
| |
Collapse
|
15
|
Mouchtouri ET, Konstantinou T, Lekkas P, Lianopoulou A, Kotsaridou Z, Mourouzis I, Pantos C, Kolettis TM. Endothelin Modulates Rhythm Disturbances and Autonomic Responses to Acute Emotional Stress in Rats. BIOLOGY 2023; 12:1401. [PMID: 37998000 PMCID: PMC10669295 DOI: 10.3390/biology12111401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 10/11/2023] [Accepted: 11/03/2023] [Indexed: 11/25/2023]
Abstract
The ubiquitous peptide endothelin is currently under investigation as a modulatory factor of autonomic responses to acute emotional stress. Baseline plasma levels of endothelin alter blood pressure responses, but it remains unclear whether autonomic activity and arrhythmogenesis (i.e., brady- or tachyarrhythmias) are affected. We recorded sympathetic and vagal indices (derived from heart rate variability analysis), rhythm disturbances, voluntary motion, and systolic blood pressure after acute emotional stress in conscious rats with implanted telemetry devices. Two strains were compared, namely wild-type and ETB-deficient rats, the latter displaying elevated plasma endothelin. No differences in heart rate or blood pressure were evident, but sympathetic responses were blunted in ETB-deficient rats, contrasting prompt activation in wild-type rats. Vagal withdrawal was observed in both strains at the onset of stress, but vagal activity was subsequently restored in ETB-deficient rats, accompanied by low voluntary motion during recovery. Reflecting such distinct autonomic patterns, frequent premature ventricular contractions were recorded in wild-type rats, as opposed to sinus pauses in ETB-deficient rats. Thus, chronically elevated plasma endothelin levels blunt autonomic responses to acute emotional stress, resulting in vagal dominance and bradyarrhythmias. Our study provides further insights into the pathophysiology of stress-induced tachyarrhythmias and syncope.
Collapse
Affiliation(s)
- Eleni-Taxiarchia Mouchtouri
- Department of Cardiology, Medical School, University of Ioannina, 45500 Ioannina, Greece; (E.-T.M.); (T.K.)
- Cardiovascular Research Institute, 45500 Ioannina, Greece;
| | - Thomas Konstantinou
- Department of Cardiology, Medical School, University of Ioannina, 45500 Ioannina, Greece; (E.-T.M.); (T.K.)
- Cardiovascular Research Institute, 45500 Ioannina, Greece;
| | | | - Alexandra Lianopoulou
- School of Applied Biology and Biotechnology, Agricultural University of Athens, 10447 Athens, Greece; (A.L.); (Z.K.)
| | - Zoi Kotsaridou
- School of Applied Biology and Biotechnology, Agricultural University of Athens, 10447 Athens, Greece; (A.L.); (Z.K.)
| | - Iordanis Mourouzis
- Department of Pharmacology, National and Kapodistrian University of Athens, 11527 Athens, Greece; (I.M.); (C.P.)
| | - Constantinos Pantos
- Department of Pharmacology, National and Kapodistrian University of Athens, 11527 Athens, Greece; (I.M.); (C.P.)
| | - Theofilos M. Kolettis
- Department of Cardiology, Medical School, University of Ioannina, 45500 Ioannina, Greece; (E.-T.M.); (T.K.)
- Cardiovascular Research Institute, 45500 Ioannina, Greece;
| |
Collapse
|
16
|
Yaar S, Filatova TS, England E, Kompella SN, Hancox JC, Bechtold DA, Venetucci L, Abramochkin DV, Shiels HA. Global Air Pollutant Phenanthrene and Arrhythmic Outcomes in a Mouse Model. ENVIRONMENTAL HEALTH PERSPECTIVES 2023; 131:117002. [PMID: 37909723 PMCID: PMC10619431 DOI: 10.1289/ehp12775] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 09/19/2023] [Accepted: 09/25/2023] [Indexed: 11/03/2023]
Abstract
BACKGROUND The three-ringed polycyclic aromatic hydrocarbon (PAH) phenanthrene (Phe) has been implicated in the cardiotoxicity of petroleum-based pollution in aquatic systems, where it disrupts the contractile and electrical function of the fish heart. Phe is also found adsorbed to particulate matter and in the gas phase of air pollution, but to date, no studies have investigated the impact of Phe on mammalian cardiac function. OBJECTIVES Our objectives were to determine the arrhythmogenic potential of acute Phe exposure on mammalian cardiac function and define the underlying mechanisms to provide insight into the toxicity risk to humans. METHODS Ex vivo Langendorff-perfused mouse hearts were used to test the arrhythmogenic potential of Phe on myocardial function, and voltage- and current-clamp recordings were used to define underlying cellular mechanisms in isolated cardiomyocytes. RESULTS Mouse hearts exposed to ∼ 8 μ M Phe for 15-min exhibited a significantly slower heart rate (p = 0.0006 , N = 10 hearts), a prolonged PR interval (p = 0.036 , N = 8 hearts), and a slower conduction velocity (p = 0.0143 , N = 7 hearts). Whole-cell recordings from isolated cardiomyocytes revealed action potential (AP) duration prolongation (at 80% repolarization; p = 0.0408 , n = 9 cells) and inhibition of key murine repolarizing currents-transient outward potassium current (I to ) and ultrarapid potassium current (I Kur )-following Phe exposure. A significant reduction in AP upstroke velocity (p = 0.0445 , n = 9 cells) and inhibition of the fast sodium current (I Na ; p = 0.001 , n = 8 cells) and calcium current (I Ca ; p = 0.0001 ) were also observed, explaining the slowed conduction velocity in intact hearts. Finally, acute exposure to ∼ 8 μ M Phe significantly increased susceptibility to arrhythmias (p = 0.0455 , N = 9 hearts). DISCUSSION To the best of our knowledge, this is the first evidence of direct inhibitory effects of Phe on mammalian cardiac electrical activity at both the whole-heart and cell levels. This electrical dysfunction manifested as an increase in arrhythmia susceptibility due to impairment of both conduction and repolarization. Similar effects in humans could have serious health consequences, warranting greater regulatory attention and toxicological investigation into this ubiquitous PAH pollutant generated from fossil-fuel combustion. https://doi.org/10.1289/EHP12775.
Collapse
Affiliation(s)
- Sana Yaar
- Faculty of Biology, Medicine, and Health, Division of Cardiovascular Sciences, University of Manchester, Manchester, UK
| | - Tatiana S. Filatova
- Department of Human and Animal Physiology, Lomonosov Moscow State University, Moscow, Russia
| | - Ellie England
- Faculty of Biology, Medicine, and Health, Division of Cardiovascular Sciences, University of Manchester, Manchester, UK
| | - Shiva N. Kompella
- Faculty of Biology, Medicine, and Health, Division of Cardiovascular Sciences, University of Manchester, Manchester, UK
| | - Jules C. Hancox
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | - David A. Bechtold
- Faculty of Biology, Medicine, and Health, Division of Cardiovascular Sciences, University of Manchester, Manchester, UK
| | - Luigi Venetucci
- Faculty of Biology, Medicine, and Health, Division of Cardiovascular Sciences, University of Manchester, Manchester, UK
| | - Denis V. Abramochkin
- Department of Human and Animal Physiology, Lomonosov Moscow State University, Moscow, Russia
| | - Holly A. Shiels
- Faculty of Biology, Medicine, and Health, Division of Cardiovascular Sciences, University of Manchester, Manchester, UK
| |
Collapse
|
17
|
Hadova K, Kmecova J, Ochodnicka‐Mackovicova K, Kralova E, Doka G, Bies Pivackova L, Vavrinec P, Stankovicova T, Krenek P, Klimas J. Rapid changes of mRNA expressions of cardiac ion channels affected by Torsadogenic drugs influence susceptibility of rat hearts to arrhythmias induced by Beta-Adrenergic stimulation. Pharmacol Res Perspect 2023; 11:e01134. [PMID: 37715323 PMCID: PMC10504435 DOI: 10.1002/prp2.1134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 08/16/2023] [Accepted: 08/17/2023] [Indexed: 09/17/2023] Open
Abstract
Drug-induced long QT syndrome (LQTS) and Torsades de Pointes (TdP) are serious concerns in drug development. Although rats are a useful scientific tool, their hearts, unlike larger species, usually do not respond to torsadogenic drugs. Consequently, their resistance to drug-induced arrhythmias is poorly understood. Here, we challenged rats with rapid delayed rectifier current (Ikr)-inhibiting antibiotic clarithromycin (CLA), loop diuretic furosemide (FUR) or their combination (CLA + FUR), and examined functional and molecular abnormalities after stimulation with isoproterenol. Clarithromycin and furosemide were administered orally at 12-h intervals for 7 days. To evaluate electrical instability, electrocardiography (ECG) was recorded either in vivo or ex vivo using the Langendorff-perfused heart method under basal conditions and subsequently under beta-adrenergic stimulation. Gene expression was measured using real-time quantitative PCR in left ventricular tissue. Indeed, FUR and CLA + FUR rats exhibited hypokalemia. CLA and CLA + FUR treatment resulted in drug-induced LQTS and even an episode of TdP in one CLA + FUR rat. The combined treatment dysregulated gene expression of several ion channels subunits, including KCNQ1, calcium channels and Na+/K + -ATPase subunits, while both monotherapies had no impact. The rat with recorded TdP exhibited differences in the expression of ion channel genes compared to the rest of rats within the CLA + FUR group. The ECG changes were not detected in isolated perfused hearts. Hence, we report rapid orchestration of ion channel reprogramming of hearts with QT prolongation induced by simultaneous administration of clarithromycin and furosemide in rats, which may account for their ability to avoid arrhythmias triggered by beta-adrenergic stimulation.
Collapse
Affiliation(s)
- Katarina Hadova
- Department of Pharmacology and Toxicology, Faculty of PharmacyComenius University BratislavaBratislavaSlovakia
| | - Jana Kmecova
- Department of Pharmacology and Toxicology, Faculty of PharmacyComenius University BratislavaBratislavaSlovakia
- State Institute for Drug ControlBratislavaSlovakia
| | | | - Eva Kralova
- Department of Pharmacology and Toxicology, Faculty of PharmacyComenius University BratislavaBratislavaSlovakia
| | - Gabriel Doka
- Department of Pharmacology and Toxicology, Faculty of PharmacyComenius University BratislavaBratislavaSlovakia
| | - Lenka Bies Pivackova
- Department of Pharmacology and Toxicology, Faculty of PharmacyComenius University BratislavaBratislavaSlovakia
| | - Peter Vavrinec
- Department of Pharmacology and Toxicology, Faculty of PharmacyComenius University BratislavaBratislavaSlovakia
| | - Tatiana Stankovicova
- Department of Pharmacology and Toxicology, Faculty of PharmacyComenius University BratislavaBratislavaSlovakia
| | - Peter Krenek
- Department of Pharmacology and Toxicology, Faculty of PharmacyComenius University BratislavaBratislavaSlovakia
| | - Jan Klimas
- Department of Pharmacology and Toxicology, Faculty of PharmacyComenius University BratislavaBratislavaSlovakia
| |
Collapse
|
18
|
Unneland E, Norvik A, Bergum D, Buckler DG, Bhardwaj A, Christian Eftestøl T, Aramendi E, Nordseth T, Abella BS, Terje Kvaløy J, Skogvoll E. Non-shockable rhythms: A parametric model for the immediate probability of return of spontaneous circulation. Resuscitation 2023; 191:109895. [PMID: 37406761 DOI: 10.1016/j.resuscitation.2023.109895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 06/21/2023] [Accepted: 06/23/2023] [Indexed: 07/07/2023]
Abstract
BACKGROUND Cardiac arrest can present with asystole, Pulseless Electrical Activity (PEA), or Ventricular Fibrillation/Tachycardia (VF/VT). We investigated the transition intensity of Return of spontaneous circulation (ROSC) from PEA and asystole during in-hospital resuscitation. MATERIALS AND METHODS We included 770 episodes of cardiac arrest. PEA was defined as ECG with >12 QRS complexes per min, asystole by an isoelectric signal >5 seconds. The observed times of PEA to ROSC transitions were fitted to five different parametric time-to-event models. At values ≤0.1, transition intensities roughly represent next-minute probabilities allowing for direct interpretation. Different entities of PEA and asystole, dependent on whether it was the primary or a secondary rhythm, were included as covariates. RESULTS The transition intensities to ROSC from primary PEA and PEA after asystole were unimodal with peaks of 0.12 at 3 min and 0.09 at 6 min, respectively. Transition intensities to ROSC from PEA after VF/VT, or following transient ROSC, exhibited high initial values of 0.32 and 0.26 at 3 minutes, respectively, but decreased. The transition intensity to ROSC from initial asystole and asystole after PEA were both about 0.01 and 0.02; while asystole after VF/VT had an intensity to ROSC of 0.15 initially which decreased. The transition intensity from asystole after temporary ROSC was constant at 0.08. CONCLUSION The immediate probability of ROSC develops differently in PEA and asystole depending on the preceding rhythm and the duration of the resuscitation attempt. This knowledge may aid simple bedside prognostication and electronic resuscitation algorithms for monitors/defibrillators.
Collapse
Affiliation(s)
- Eirik Unneland
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway.
| | - Anders Norvik
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway; Clinic of Anesthesia and Intensive Care Medicine, St Olav University Hospital, Trondheim, Norway
| | - Daniel Bergum
- Clinic of Anesthesia and Intensive Care Medicine, St Olav University Hospital, Trondheim, Norway
| | - David G Buckler
- Department of Emergency Medicine, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | | | - Trygve Christian Eftestøl
- Department of Electrical Engineering and Computer Science, University of Stavanger, Stavanger, Norway
| | - Elisabete Aramendi
- University of the Basque Country, Engineering School of Bilbao, BioRes Group, Bilbao, Spain
| | - Trond Nordseth
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway; Clinic of Anesthesia and Intensive Care Medicine, St Olav University Hospital, Trondheim, Norway; Department of Research and Development, Division of Emergencies and Critical Care, Oslo University Hospital. Oslo, Norway
| | - Benjamin S Abella
- Center for Resuscitation Science, Department of Emergency Medicine, University of Pennsylvania, Philadelphia, USA
| | - Jan Terje Kvaløy
- Department of Mathematics and Physics, University of Stavanger, Stavanger, Norway
| | - Eirik Skogvoll
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway; Clinic of Anesthesia and Intensive Care Medicine, St Olav University Hospital, Trondheim, Norway
| |
Collapse
|
19
|
Fu Y, Snelder N, Guo T, van der Graaf PH, van Hasselt JGC. Evaluation of a Cardiovascular Systems Model for Design and Analysis of Hemodynamic Safety Studies. Pharmaceutics 2023; 15:pharmaceutics15041175. [PMID: 37111660 PMCID: PMC10143046 DOI: 10.3390/pharmaceutics15041175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 03/20/2023] [Accepted: 04/05/2023] [Indexed: 04/29/2023] Open
Abstract
Early prediction, quantification and translation of cardiovascular hemodynamic drug effects is essential in pre-clinical drug development. In this study, a novel hemodynamic cardiovascular systems (CVS) model was developed to support these goals. The model consisted of distinct system- and drug-specific parameter, and uses data for heart rate (HR), cardiac output (CO), and mean atrial pressure (MAP) to infer drug mode-of-action (MoA). To support further application of this model in drug development, we conducted a systematic analysis of the estimation performance of the CVS model to infer drug- and system-specific parameters. Specifically, we focused on the impact on model estimation performance when considering differences in available readouts and the impact of study design choices. To this end, a practical identifiability analysis was performed, evaluating model estimation performance for different combinations of hemodynamic endpoints, drug effect sizes, and study design characteristics. The practical identifiability analysis showed that MoA of drug effect could be identified for different drug effect magnitudes and both system- and drug-specific parameters can be estimated precisely with minimal bias. Study designs which exclude measurement of CO or use a reduced measurement duration still allow the identification and quantification of MoA with acceptable performance. In conclusion, the CVS model can be used to support the design and inference of MoA in pre-clinical CVS experiments, with a future potential for applying the uniquely identifiable systems parameters to support inter-species scaling.
Collapse
Affiliation(s)
- Yu Fu
- Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Nelleke Snelder
- LAP&P Consultants BV, Archimedesweg 31, 2333 CM Leiden, The Netherlands
| | - Tingjie Guo
- Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Piet H van der Graaf
- Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
- Certara QSP, Canterbury CT2 7FG, UK
| | - Johan G C van Hasselt
- Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| |
Collapse
|
20
|
Heger J, Szabados T, Brosinsky P, Bencsik P, Ferdinandy P, Schulz R. Sex Difference in Cardioprotection against Acute Myocardial Infarction in MAO-B Knockout Mice In Vivo. Int J Mol Sci 2023; 24:ijms24076443. [PMID: 37047416 PMCID: PMC10094730 DOI: 10.3390/ijms24076443] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 03/22/2023] [Accepted: 03/25/2023] [Indexed: 03/31/2023] Open
Abstract
The cardiomyocyte-specific knockout (KO) of monoamine oxidase (MAO)-B, an enzyme involved in the formation of reactive oxygen species (ROS), reduced myocardial ischemia/reperfusion (I/R) injury in vitro. Because sex hormones have a strong impact on MAO metabolic pathways, we analyzed the myocardial infarct size (IS) following I/R in female and male MAO-B KO mice in vivo. Method and Results: To induce the deletion of MAO-B, MAO-B KO mice (Myh6 Cre+/MAO-Bfl/fl) and wild-type (WT, Cre-negative MAO-Bfl/fl littermates) were fed with tamoxifen for 2 weeks followed by 10 weeks of normal mice chow. Myocardial infarction (assessed by TTC staining and expressed as a percentage of the area at risk as determined by Evans blue staining)) was induced by 45 min coronary occlusion followed by 120 min of reperfusion. Results: The mortality following I/R was higher in male compared to female mice, with the lowest mortality found in MAO-B KO female mice. IS was significantly higher in male WT mice compared to female WT mice. MAO-B KO reduced IS in male mice but had no further impact on IS in female MAO-B KO mice. Interestingly, there was no difference in the plasma estradiol levels among the groups. Conclusion: The cardiomyocyte-specific knockout of MAO-B protects male mice against acute myocardial infarction but had no effect on the infarct size in female mice.
Collapse
|
21
|
Boulay E, Miraucourt LS, Pugsley MK, Abernathy MM, Chui R, Dalton J, Demers M, Dybdal N, Gazaille E, Greiter-Wilke A, Hoffmann P, Huang H, LaDuke C, Norton K, Pierson JB, Reeves I, Roche B, Rossman EI, Schultze AE, Tang HM, Wisialowski T, Authier S. The incidence of spontaneous arrhythmias in telemetered beagle dogs, Göttingen Minipigs and Cynomolgus non-human primates: A HESI consortium retrospective analysis. J Pharmacol Toxicol Methods 2023; 121:107266. [PMID: 36963703 DOI: 10.1016/j.vascn.2023.107266] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 03/17/2023] [Accepted: 03/20/2023] [Indexed: 03/26/2023]
Abstract
INTRODUCTION Characterization of the incidence of spontaneous arrhythmias to identify possible drug-related effects is often an important part of the analysis in safety pharmacology studies using telemetry. METHODS A retrospective analysis in non-clinical species with and without telemetry transmitters was conducted. Electrocardiograms (24 h) from male and female beagle dogs (n = 131), Göttingen minipigs (n = 108) and cynomolgus non-human primates (NHP; n = 78) were analyzed. RESULTS Ventricular tachycardia (VT) was observed in 3% of the dogs but was absent in minipigs and NHPs. Ventricular fibrillation (VF) was not observed in the 3 species. Ventricular premature beats (VPBs) were more frequent during daytime and atrioventricular blocks (AVBs) were more frequent at night in all species. A limited number of animals exhibited a high arrhythmia frequency and there was no correlation between animals with higher frequency of an arrhythmia type and the frequency of other arrythmias in the same animals. Clinical chemistry or hematology parameters were not different with or without telemetry devices. NHP with a transmural left ventricular pressure (LVP) catheter exhibited a greater incidence of VPBs and PJCs compared to telemetry animals without LVP. DISCUSSION All species were similar with regards to the frequency of ventricular ectopic beats (26-46%) while the dog seemed to have more frequent junctional complexes and AVB compared to NHP and minipigs. Arrhythmia screening may be considered during pre-study evaluations, to exclude animals with abnormally high arrhythmia incidence.
Collapse
Affiliation(s)
- Emmanuel Boulay
- Charles River, Laval, Canada; Faculty of Veterinary Medicine, University of Montreal, QC, Canada
| | | | | | | | - Ray Chui
- Cardiovascular Analytics, Newbury Park, CA, USA
| | | | - Marjorie Demers
- Faculty of Veterinary Medicine, University of Montreal, QC, Canada
| | | | - Elissa Gazaille
- Faculty of Veterinary Medicine, University of Montreal, QC, Canada
| | - Andrea Greiter-Wilke
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center, Basel, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | | - Simon Authier
- Charles River, Laval, Canada; Faculty of Veterinary Medicine, University of Montreal, QC, Canada.
| |
Collapse
|
22
|
Mohammed Abdulsalam T, Hasanin AH, Hussein Mohamed R, Khairy E, Mahmoud D, Habib EK, Badawy AES. A comparative study between angiotensin receptor neprilysin inhibitor (thiorphan/irbesartan) with each of nitrate and carvedilol in a rat model of myocardial ischemic reperfusion injury. Fundam Clin Pharmacol 2023. [PMID: 36868872 DOI: 10.1111/fcp.12886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 02/02/2023] [Accepted: 02/22/2023] [Indexed: 03/05/2023]
Abstract
The combined angiotensin receptor neprilysin inhibitor is a promising cardioprotective pharmacological agent. This study investigated the beneficial effects of thiorphan (TH)/irbesartan (IRB), in myocardial ischemia-reperfusion (IR) injury, compared to each of nitroglycerin and carvedilol. Male Wistar rats were divided into five groups (10 rats/group): Sham, untreated I/R, TH/IRB + IR (0.1/10 mg/kg), nitroglycerin + IR (0.2 mg/kg), and carvedilol + IR (10 mg/kg). Mean arterial blood pressure, cardiac functions and arrhythmia incidence, duration and score were assessed. Cardiac levels of creatine kinase-MB (CK-MB), oxidative stress, endothelin-1, ATP, Na+ /K+ ATPase pump activity and mitochondria complexes activities were measured. Histopathological examination, Bcl/Bax immunohistochemistry studies and electron microscopy examination of left ventricle were performed. TH/IRB preserved the cardiac functions and mitochondrial complexes activities, mitigated cardiac damage, reduced oxidative stress and arrhythmia severity, improved the histopathological changes and decreased cardiac apoptosis. TH/IRB showed a comparable effect to each of nitroglycerin and carvedilol in alleviating the IR injury consequences. TH/IRB showed significant preservation of mitochondrial complexes activity I and II compared to nitroglycerin. TH/IRB significantly increased LVdP/dtmax and decreased oxidative stress, cardiac damage and endothelin-1 along with increasing the ATP content, Na+ /K+ ATPase pump activity and mitochondrial complexes activity when compared to carvedilol. TH/IRB showed a cardioprotective effect in reducing IR injury that is comparable to each of nitroglycerin and carvedilol that could be explained in part by its ability to preserve mitochondrial function, increase ATP, decrease oxidative stress as well as endothelin 1.
Collapse
Affiliation(s)
| | - Amany H Hasanin
- Department of Clinical Pharmacology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Reham Hussein Mohamed
- Department of Clinical Pharmacology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Eman Khairy
- Department of Medical Biochemistry & Molecular Biology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Dalia Mahmoud
- Department of Medical Biochemistry & Molecular Biology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Eman K Habib
- Department of Anatomy and Embryology, Faculty of Medicine, Galala University, Al Galala, Egypt
| | - Ahmed El Sayed Badawy
- Department of Clinical Pharmacology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| |
Collapse
|
23
|
Polyák A, Topal L, Zombori-Tóth N, Tóth N, Prorok J, Kohajda Z, Déri S, Demeter-Haludka V, Hegyi P, Venglovecz V, Ágoston G, Husti Z, Gazdag P, Szlovák J, Árpádffy-Lovas T, Naveed M, Sarusi A, Jost N, Virág L, Nagy N, Baczkó I, Farkas AS, Varró A. Cardiac electrophysiological remodeling associated with enhanced arrhythmia susceptibility in a canine model of elite exercise. eLife 2023; 12:80710. [PMID: 36815557 PMCID: PMC10014074 DOI: 10.7554/elife.80710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 02/16/2023] [Indexed: 02/24/2023] Open
Abstract
The health benefits of regular physical exercise are well known. Even so, there is increasing evidence that the exercise regimes of elite athletes can evoke cardiac arrhythmias including ventricular fibrillation and even sudden cardiac death (SCD). The mechanism of exercise-induced arrhythmia and SCD is poorly understood. Here, we show that chronic training in a canine model (12 sedentary and 12 trained dogs) that mimics the regime of elite athletes induces electrophysiological remodeling (measured by ECG, patch-clamp, and immunocytochemical techniques) resulting in increases of both the trigger and the substrate for ventricular arrhythmias. Thus, 4 months sustained training lengthened ventricular repolarization (QTc: 237.1±3.4 ms vs. 213.6±2.8 ms, n=12; APD90: 472.8±29.6 ms vs. 370.1±32.7 ms, n=29 vs. 25), decreased transient outward potassium current (6.4±0.5 pA/pF vs. 8.8±0.9 pA/pF at 50 mV, n=54 vs. 42), and increased the short-term variability of repolarization (29.5±3.8 ms vs. 17.5±4.0 ms, n=27 vs. 18). Left ventricular fibrosis and HCN4 protein expression were also enhanced. These changes were associated with enhanced ectopic activity (number of escape beats from 0/hr to 29.7±20.3/hr) in vivo and arrhythmia susceptibility (elicited ventricular fibrillation: 3 of 10 sedentary dogs vs. 6 of 10 trained dogs). Our findings provide in vivo, cellular electrophysiological and molecular biological evidence for the enhanced susceptibility to ventricular arrhythmia in an experimental large animal model of endurance training.
Collapse
Affiliation(s)
- Alexandra Polyák
- Department of Pharmacology and Pharmacotherapy, University of SzegedSzegedHungary
| | - Leila Topal
- Department of Pharmacology and Pharmacotherapy, University of SzegedSzegedHungary
| | - Noémi Zombori-Tóth
- Department of Pharmacology and Pharmacotherapy, University of SzegedSzegedHungary
| | - Noémi Tóth
- Department of Pharmacology and Pharmacotherapy, University of SzegedSzegedHungary
| | - János Prorok
- Department of Pharmacology and Pharmacotherapy, University of SzegedSzegedHungary
- ELKH-SZTE Research Group for Cardiovascular Pharmacology, Eötvös Loránd Research NetworkSzegedHungary
| | - Zsófia Kohajda
- ELKH-SZTE Research Group for Cardiovascular Pharmacology, Eötvös Loránd Research NetworkSzegedHungary
| | - Szilvia Déri
- Department of Pharmacology and Pharmacotherapy, University of SzegedSzegedHungary
| | | | - Péter Hegyi
- Centre for Translational Medicine and Institute of Pancreatic Diseases, Semmelweis UniversityBudapestHungary
- Institute for Translational Medicine, Medical School, University of PécsPécsHungary
- Translational Pancreatology Research Group, Interdisciplinary Centre of Excellence for Research Development and Innovation, University of SzegedSzegedHungary
| | - Viktória Venglovecz
- Department of Pharmacology and Pharmacotherapy, University of SzegedSzegedHungary
| | - Gergely Ágoston
- Institute of Family Medicine, University of SzegedSzegedHungary
| | - Zoltán Husti
- Department of Pharmacology and Pharmacotherapy, University of SzegedSzegedHungary
| | - Péter Gazdag
- Department of Pharmacology and Pharmacotherapy, University of SzegedSzegedHungary
| | - Jozefina Szlovák
- Department of Pharmacology and Pharmacotherapy, University of SzegedSzegedHungary
| | - Tamás Árpádffy-Lovas
- Department of Pharmacology and Pharmacotherapy, University of SzegedSzegedHungary
| | - Muhammad Naveed
- Department of Pharmacology and Pharmacotherapy, University of SzegedSzegedHungary
| | - Annamária Sarusi
- Department of Pharmacology and Pharmacotherapy, University of SzegedSzegedHungary
| | - Norbert Jost
- Department of Pharmacology and Pharmacotherapy, University of SzegedSzegedHungary
- ELKH-SZTE Research Group for Cardiovascular Pharmacology, Eötvös Loránd Research NetworkSzegedHungary
- Department of Pharmacology and Pharmacotherapy, Interdisciplinary Excellence Centre, University of SzegedSzegedHungary
| | - László Virág
- Department of Pharmacology and Pharmacotherapy, University of SzegedSzegedHungary
- Department of Pharmacology and Pharmacotherapy, Interdisciplinary Excellence Centre, University of SzegedSzegedHungary
| | - Norbert Nagy
- Department of Pharmacology and Pharmacotherapy, University of SzegedSzegedHungary
- ELKH-SZTE Research Group for Cardiovascular Pharmacology, Eötvös Loránd Research NetworkSzegedHungary
| | - István Baczkó
- Department of Pharmacology and Pharmacotherapy, University of SzegedSzegedHungary
- Department of Pharmacology and Pharmacotherapy, Interdisciplinary Excellence Centre, University of SzegedSzegedHungary
| | - Attila S Farkas
- Department of Internal Medicine, Cardiology ward, University of SzegedSzegedHungary
| | - András Varró
- Department of Pharmacology and Pharmacotherapy, University of SzegedSzegedHungary
- ELKH-SZTE Research Group for Cardiovascular Pharmacology, Eötvös Loránd Research NetworkSzegedHungary
- Department of Pharmacology and Pharmacotherapy, Interdisciplinary Excellence Centre, University of SzegedSzegedHungary
| |
Collapse
|
24
|
Cachorro E, Günscht M, Schubert M, Sadek MS, Siegert J, Dutt F, Bauermeister C, Quickert S, Berning H, Nowakowski F, Lämmle S, Firneburg R, Luo X, Künzel SR, Klapproth E, Mirtschink P, Mayr M, Dewenter M, Vettel C, Heijman J, Lorenz K, Guan K, El-Armouche A, Wagner M, Kämmerer S. CNP Promotes Antiarrhythmic Effects via Phosphodiesterase 2. Circ Res 2023; 132:400-414. [PMID: 36715019 PMCID: PMC9930893 DOI: 10.1161/circresaha.122.322031] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Ventricular arrhythmia and sudden cardiac death are the most common lethal complications after myocardial infarction. Antiarrhythmic pharmacotherapy remains a clinical challenge and novel concepts are highly desired. Here, we focus on the cardioprotective CNP (C-type natriuretic peptide) as a novel antiarrhythmic principle. We hypothesize that antiarrhythmic effects of CNP are mediated by PDE2 (phosphodiesterase 2), which has the unique property to be stimulated by cGMP to primarily hydrolyze cAMP. Thus, CNP might promote beneficial effects of PDE2-mediated negative crosstalk between cAMP and cGMP signaling pathways. METHODS To determine antiarrhythmic effects of cGMP-mediated PDE2 stimulation by CNP, we analyzed arrhythmic events and intracellular trigger mechanisms in mice in vivo, at organ level and in isolated cardiomyocytes as well as in human-induced pluripotent stem cell-derived cardiomyocytes. RESULTS In ex vivo perfused mouse hearts, CNP abrogated arrhythmia after ischemia/reperfusion injury. Upon high-dose catecholamine injections in mice, PDE2 inhibition prevented the antiarrhythmic effect of CNP. In mouse ventricular cardiomyocytes, CNP blunted the catecholamine-mediated increase in arrhythmogenic events as well as in ICaL, INaL, and Ca2+ spark frequency. Mechanistically, this was driven by reduced cellular cAMP levels and decreased phosphorylation of Ca2+ handling proteins. Key experiments were confirmed in human iPSC-derived cardiomyocytes. Accordingly, the protective CNP effects were reversed by either specific pharmacological PDE2 inhibition or cardiomyocyte-specific PDE2 deletion. CONCLUSIONS CNP shows strong PDE2-dependent antiarrhythmic effects. Consequently, the CNP-PDE2 axis represents a novel and attractive target for future antiarrhythmic strategies.
Collapse
Affiliation(s)
- Eleder Cachorro
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
| | - Mario Günscht
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
| | - Mario Schubert
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
| | - Mirna S. Sadek
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
| | - Johanna Siegert
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
| | - Fabian Dutt
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
| | - Carla Bauermeister
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
| | - Susann Quickert
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
| | - Henrik Berning
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
| | - Felix Nowakowski
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
| | - Simon Lämmle
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
| | - Rebecca Firneburg
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
| | - Xiaojing Luo
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
| | - Stephan R. Künzel
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
| | - Erik Klapproth
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
| | - Peter Mirtschink
- Institute of Clinical Chemistry and Laboratory Medicine, Department of Clinical Pathobiochemistry, University Hospital Dresden, Germany (P.M.)
| | - Manuel Mayr
- The James Black Centre, King’s College, University of London, United Kingdom (M.M.)
- Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany (M.M.)
| | - Matthias Dewenter
- Department of Molecular Cardiology and Epigenetics, Heidelberg University, Germany (M.D.)
- DZHK (German Center for Cardiovascular Research), Partner Site, Heidelberg-Mannheim, Germany (M.D., C.V.)
| | - Christiane Vettel
- DZHK (German Center for Cardiovascular Research), Partner Site, Heidelberg-Mannheim, Germany (M.D., C.V.)
- Institute of Experimental and Clinical Pharmacology and Toxicology, University Medical Center Mannheim, Germany (C.V.)
| | - Jordi Heijman
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine, and Life Sciences, Maastricht University, The Netherlands (J.H.)
| | - Kristina Lorenz
- Institut für Pharmakologie und Toxikologie, Julius-Maximilians-Universität Würzburg, Germany (K.L.)
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., Dortmund, Germany (K.L.)
| | - Kaomei Guan
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
| | - Ali El-Armouche
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
| | - Michael Wagner
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
- Bereich Rhythmologie, Klinik für Innere Medizin und Kardiologie, Herzzentrum Dresden, Dresden University of Technology, Germany (M.W.)
| | - Susanne Kämmerer
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
| |
Collapse
|
25
|
Effects of Bempedoic Acid in Acute Myocardial Infarction in Rats: No Cardioprotection and No Hidden Cardiotoxicity. Int J Mol Sci 2023; 24:ijms24021585. [PMID: 36675100 PMCID: PMC9860765 DOI: 10.3390/ijms24021585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 12/22/2022] [Accepted: 12/30/2022] [Indexed: 01/15/2023] Open
Abstract
Lipid-lowering drugs have been shown to have cardioprotective effects but may have hidden cardiotoxic properties. Therefore, here we aimed to investigate if chronic treatment with the novel lipid-lowering drug bempedoic acid (BA) exerts hidden cardiotoxic and/or cardioprotective effects in a rat model of acute myocardial infarction (AMI). Wistar rats were orally treated with BA or its vehicle for 28 days, anesthetized and randomized to three different groups (vehicle + ischemia/reperfusion (I/R), BA + I/R, and positive control vehicle + ischemic preconditioning (IPC)) and subjected to cardiac 30 min ischemia and 120 min reperfusion. IPC was performed by 3 × 5 min I/R cycles before ischemia. Myocardial function, area at risk, infarct size and arrhythmias were analyzed. Chronic BA pretreatment did not influence cardiac function or infarct size as compared to the vehicle group, while the positive control IPC significantly reduced the infarct size. The incidence of reperfusion-induced arrhythmias was significantly reduced by BA and IPC. This is the first demonstration that BA treatment does not show cardioprotective effect although moderately reduces the incidence of reperfusion-induced arrhythmias. Furthermore, BA does not show hidden cardiotoxic effect in rats with AMI, showing its safety in the ischemic/reperfused heart.
Collapse
|
26
|
Minnebaeva EV, Durkina AV, Azarov JE, Bernikova OG. Myocardial Electrophysiological Response to Ischemia and Reperfusion Depends on the Age of Rats. J EVOL BIOCHEM PHYS+ 2022. [DOI: 10.1134/s0022093022070079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
|
27
|
Oknińska M, Mączewski M, Mackiewicz U. Ventricular arrhythmias in acute myocardial ischaemia-Focus on the ageing and sex. Ageing Res Rev 2022; 81:101722. [PMID: 36038114 DOI: 10.1016/j.arr.2022.101722] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 08/17/2022] [Accepted: 08/20/2022] [Indexed: 01/31/2023]
Abstract
Annually, approximately 17 million people die from cardiovascular diseases worldwide, half of them suddenly. The most common direct cause of sudden cardiac death is ventricular arrhythmia triggered by an acute coronary syndrome (ACS). The study summarizes the knowledge of the mechanisms of arrhythmia onset during ACS in humans and in animal models and factors that may influence the susceptibility to life-threatening arrhythmias during ACS with particular focus on the age and sex. The real impact of age and sex on the arrhythmic susceptibility within the setting of acute ischaemia is masked by the fact that ACSs result from coronary artery disease appearing with age much earlier among men than among women. However, results of researches show that in ageing process changes with potential pro-arrhythmic significance, such as increased fibrosis, cardiomyocyte hypertrophy, decrease number of gap junction channels, disturbances of the intracellular Ca2+ signalling or changes in electrophysiological parameters, occur independently of the development of cardiovascular diseases and are more severe in male individuals. A review of the literature also indicates a marked paucity of research in this area in female and elderly individuals. Greater awareness of sex differences in the aging process could help in the development of personalized prevention methods targeting potential pro-arrhythmic factors in patients of both sexes to reduce mortality during the acute phase of myocardial infarction. This is especially important in an era of aging populations in which women will predominate due to their longer lifespan.
Collapse
Affiliation(s)
- Marta Oknińska
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, Marymoncka 99/103, 01-813 Warsaw, Poland
| | - Michał Mączewski
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, Marymoncka 99/103, 01-813 Warsaw, Poland
| | - Urszula Mackiewicz
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, Marymoncka 99/103, 01-813 Warsaw, Poland.
| |
Collapse
|
28
|
Lyhne MK, Debes KP, Helgogaard T, Vegge A, Kildegaard J, Pedersen-Bjergaard U, Olsen LH. Electrocardiography and heart rate variability in Göttingen Minipigs: Impact of diurnal variation, lead placement, repeatability and streptozotocin-induced diabetes. J Pharmacol Toxicol Methods 2022; 118:107221. [PMID: 36100059 DOI: 10.1016/j.vascn.2022.107221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 08/26/2022] [Accepted: 09/07/2022] [Indexed: 12/01/2022]
Abstract
BACKGROUND The Göttingen Minipig is widely used in preclinical research and safety pharmacology, but standardisation of porcine electrocardiography (ECG) is lacking. The aim of this study was to investigate diurnal effects, change over time and choice of lead on ECG morphology and heart rate variability (HRV) in healthy and streptozotocin (STZ) induced diabetic Göttingen Minipigs. METHODS Diabetes was experimentally induced using STZ in 11 Göttingen Minipigs (DIA). Seven controls (CON) were included. 24-h ECG was recorded at baseline and four months. Morphological parameters (QRS and T wave duration, P- and T-wave amplitude, PR and QT (Bazett's (QTcb) or Fridericia (QTcf) correction) intervals and ST segment), presence of cardiac arrhythmias, heart rate (HR) and HRV (time and frequency domain) were analysed. RESULTS Four months after induction, DIA had decreased P-wave amplitude (P < 0.0001) and T-wave duration (P = 0.017), compared to CON. QTcb was lower in DIA, but not in CON. Both groups had decreased HR (P < 0.0001) and QRS duration (lead II, P = 0.04) and length of PR-segment increased (lead I and II, P < 0.01) while selected HRV parameters also increased (all P < 0.01). Time of day influenced HR, QRS duration, PR segment, ST segment, T- and P-wave amplitude and some parameters of HRV. Inter- and intra-observer variability of morphological measurements was low (<6%). CONCLUSION ECG parameters were influenced by time setting, diurnal variation and lead. Some ECG and HRV changes were found in diabetic minipigs four months after STZ induction. The findings underline the need for standardisation of ECG and HRV in Göttingen Minipigs.
Collapse
Affiliation(s)
- Mille Kronborg Lyhne
- Department of Veterinary and Animal Sciences, University of Copenhagen, Ridebanevej 9, 1870 Frederiksberg, Denmark.
| | - Karina Poulsdóttir Debes
- Department of Veterinary and Animal Sciences, University of Copenhagen, Ridebanevej 9, 1870 Frederiksberg, Denmark.
| | - Terese Helgogaard
- Department of Veterinary and Animal Sciences, University of Copenhagen, Ridebanevej 9, 1870 Frederiksberg, Denmark
| | - Andreas Vegge
- Global Drug Discovery, Novo Nordisk A/S, Novo Nordisk Park 1, 2760 Måløv, Denmark.
| | - Jonas Kildegaard
- Global Drug Discovery, Novo Nordisk A/S, Novo Nordisk Park 1, 2760 Måløv, Denmark.
| | - Ulrik Pedersen-Bjergaard
- Department of Endocrinology and Nephrology, Nordsjællands Hospital Hillerød, Dyrehavevej 29, 3400 Hillerød, Denmark.
| | - Lisbeth Høier Olsen
- Department of Veterinary and Animal Sciences, University of Copenhagen, Ridebanevej 9, 1870 Frederiksberg, Denmark.
| |
Collapse
|
29
|
Li S, Wang H, Zhou Y. JAK2/STAT3 pathway mediates beneficial effects of pterostilbene on cardiac contractile and electrical function in the setting of myocardial reperfusion injury. Physiol Res 2022. [DOI: 10.33549/physiolres.934919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Contractile dysfunction and fatal arrhythmias are the hallmarks of myocardial ischemia/reperfusion (I/R) injury. Pterostilbene has notable cardioprotective effects, but its main mechanisms are not fully understood. Here, we investigated the effect of PTE on myocardial hemodynamics, arrhythmias, inflammatory/oxidative responses, and the causal role of the JAK2/STAT3 pathway in rats with acute myocardial I/R injury. Sixty male 7-8 months Sprague-Dawley rats (n=10/each group) experienced in vivo model of myocardial I/R injury through 40-min LAD coronary artery occlusion and subsequent 24-h reperfusion. PTE at concentrations of 5 and 25 mg/kg was intraperitoneally administered to rats five min before reperfusion. Cardiac hemodynamics, reperfusion-induced ventricular arrhythmias, infarct size, inflammatory cytokines, oxidative stress markers, the activity of the JAK2/STAT3 pathway were measured as the endpoints. Administration of PTE to I/R-injured rats recovered myocardial contractile function and reduced infarct size and ventricular arrhythmias counts and incidence in a dose-dependent manner. PTE at 25 mg/kg significantly and more potently reduced the levels of inflammatory mediators NF-κB, TNF-α, and IL-1β, suppressed intracellular ROS production, augmented the activity of glutathione, and manganese-superoxide dismutase, and upregulated the JAK2 and STAT3 phosphorylation. Importantly, pretreatment of rats with Ag490 as a JAK2 inhibitor significantly abolished the cardioprotective and signaling effects of PTE in I/R rats. PTE exerts significant protective effects on reducing arrhythmias and myocardial infarction and enhancing cardiac function by stimulating JAK2/STAT3-related suppression of inflammatory and oxidative reactions in the I/R injury setting.
Collapse
Affiliation(s)
| | | | - Y Zhou
- Department of Cardiology, Jiangxi Provincial People's Hospital, Nanchang, China
| |
Collapse
|
30
|
Favere K, Van Fraeyenhove J, Jacobs G, Bosman M, Eens S, De Sutter J, Miljoen H, Guns PJ, De Keulenaer GW, Segers VFM, Heidbuchel H. Cardiac electrophysiology studies in mice via the transjugular route: a comprehensive practical guide. Am J Physiol Heart Circ Physiol 2022; 323:H763-H773. [PMID: 36018757 DOI: 10.1152/ajpheart.00337.2022] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cardiac arrhythmias are associated with cardiovascular morbidity and mortality. Cardiac electrophysiology studies (EPS) use intracardiac catheter recording and stimulation for profound evaluation of the heart's electrical properties. The main clinical application is investigation and treatment of rhythm disorders. These techniques have been translated to the murine setting to open opportunities for detailed evaluation of the impact of different characteristics (including genetics) and interventions on cardiac electrophysiology and -pathology. Currently, a detailed description of the technique of murine transjugular EPS (which is the standard route of catheter introduction) is lacking. This article provides detailed information on EPS in mice via the transjugular route. This includes catheter placement, stimulation protocols, intracardiac tracing interpretation, artefact reduction and surface ECG recording. In addition, reference values as obtained in C57BL/6N mice are presented for common electrophysiological parameters. This detailed methodological description aims to increase accessibility and standardisation of EPS in mice. Ultimately, also human research and patient care may benefit from translation of the knowledge obtained in preclinical models using this technique.
Collapse
Affiliation(s)
- Kasper Favere
- Laboratory of Physiopharmacology, GENCOR, University of Antwerp, Antwerp, Belgium.,Research Group Cardiovascular Diseases, GENCOR, University of Antwerp, Antwerp, Belgium.,Department of Cardiology, Antwerp University Hospital, Antwerp, Belgium.,Department of Internal Medicine, Ghent University, Ghent, Belgium
| | - Jens Van Fraeyenhove
- Laboratory of Physiopharmacology, GENCOR, University of Antwerp, Antwerp, Belgium
| | - Griet Jacobs
- Laboratory of Physiopharmacology, GENCOR, University of Antwerp, Antwerp, Belgium
| | - Matthias Bosman
- Laboratory of Physiopharmacology, GENCOR, University of Antwerp, Antwerp, Belgium
| | - Sander Eens
- Laboratory of Physiopharmacology, GENCOR, University of Antwerp, Antwerp, Belgium.,Research Group Cardiovascular Diseases, GENCOR, University of Antwerp, Antwerp, Belgium
| | - Johan De Sutter
- Department of Internal Medicine, Ghent University, Ghent, Belgium
| | - Hielko Miljoen
- Department of Cardiology, Antwerp University Hospital, Antwerp, Belgium
| | - Pieter-Jan Guns
- Laboratory of Physiopharmacology, GENCOR, University of Antwerp, Antwerp, Belgium
| | - Gilles W De Keulenaer
- Laboratory of Physiopharmacology, GENCOR, University of Antwerp, Antwerp, Belgium.,Department of Cardiology, ZNA Middelheim Hospital, Antwerp, Belgium
| | - Vincent F M Segers
- Laboratory of Physiopharmacology, GENCOR, University of Antwerp, Antwerp, Belgium.,Research Group Cardiovascular Diseases, GENCOR, University of Antwerp, Antwerp, Belgium.,Department of Cardiology, Antwerp University Hospital, Antwerp, Belgium
| | - Hein Heidbuchel
- Research Group Cardiovascular Diseases, GENCOR, University of Antwerp, Antwerp, Belgium.,Department of Cardiology, Antwerp University Hospital, Antwerp, Belgium
| |
Collapse
|
31
|
Weber BY, Brenner GB, Kiss B, Gergely TG, Sayour NV, Tian H, Makkos A, Görbe A, Ferdinandy P, Giricz Z. Rosiglitazone Does Not Show Major Hidden Cardiotoxicity in Models of Ischemia/Reperfusion but Abolishes Ischemic Preconditioning-Induced Antiarrhythmic Effects in Rats In Vivo. Pharmaceuticals (Basel) 2022; 15:ph15091055. [PMID: 36145276 PMCID: PMC9503202 DOI: 10.3390/ph15091055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 08/19/2022] [Accepted: 08/22/2022] [Indexed: 11/16/2022] Open
Abstract
Clinical observations are highly inconsistent with the use of the antidiabetic rosiglitazone regarding its associated increased risk of myocardial infarction. This may be due to its hidden cardiotoxic properties that have only become evident during post-marketing studies. Therefore, we aimed to investigate the hidden cardiotoxicity of rosiglitazone in ischemia/reperfusion (I/R) injury models. Rats were treated orally with either 0.8 mg/kg/day rosiglitazone or vehicle for 28 days and subjected to I/R with or without cardioprotective ischemic preconditioning (IPC). Rosiglitazone did not affect mortality, arrhythmia score, or infarct size during I/R. However, rosiglitazone abolished the antiarrhythmic effects of IPC. To investigate the direct effect of rosiglitazone on cardiomyocytes, we utilized adult rat cardiomyocytes (ARCMs), AC16, and differentiated AC16 (diffAC16) human cardiac cell lines. These were subjected to simulated I/R in the presence of rosiglitazone. Rosiglitazone improved cell survival of ARCMs at 0.3 μM. At 0.1 and 0.3 μM, rosiglitazone improved cell survival of AC16s but not that of diffAC16s. This is the first demonstration that chronic administration of rosiglitazone does not result in major hidden cardiotoxic effects in myocardial I/R injury models. However, the inhibition of the antiarrhythmic effects of IPC may have some clinical relevance that needs to be further explored.
Collapse
Affiliation(s)
- Bennet Y. Weber
- MTA-SE System Pharmacology Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1089 Budapest, Hungary
| | - Gábor B. Brenner
- MTA-SE System Pharmacology Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1089 Budapest, Hungary
| | - Bernadett Kiss
- MTA-SE System Pharmacology Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1089 Budapest, Hungary
| | - Tamás G. Gergely
- MTA-SE System Pharmacology Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1089 Budapest, Hungary
| | - Nabil V. Sayour
- MTA-SE System Pharmacology Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1089 Budapest, Hungary
| | - Huimin Tian
- MTA-SE System Pharmacology Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1089 Budapest, Hungary
| | - András Makkos
- MTA-SE System Pharmacology Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1089 Budapest, Hungary
| | - Anikó Görbe
- MTA-SE System Pharmacology Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1089 Budapest, Hungary
- Pharmahungary Group, H-6722 Szeged, Hungary
| | - Péter Ferdinandy
- MTA-SE System Pharmacology Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1089 Budapest, Hungary
- Pharmahungary Group, H-6722 Szeged, Hungary
| | - Zoltán Giricz
- MTA-SE System Pharmacology Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1089 Budapest, Hungary
- Pharmahungary Group, H-6722 Szeged, Hungary
- Correspondence:
| |
Collapse
|
32
|
Shen R, Zuo D, Chen K, Yin Y, Tang K, Hou S, Han B, Xu Y, Liu Z, Chen H. K2P1 leak cation channels contribute to ventricular ectopic beats and sudden death under hypokalemia. FASEB J 2022; 36:e22455. [PMID: 35899468 DOI: 10.1096/fj.202200707r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/28/2022] [Accepted: 07/06/2022] [Indexed: 11/11/2022]
Abstract
Hypokalemia causes ectopic heartbeats, but the mechanisms underlying such cardiac arrhythmias are not understood. In reduced serum K+ concentrations that occur under hypokalemia, K2P1 two-pore domain K+ channels change ion selectivity and switch to conduct inward leak cation currents, which cause aberrant depolarization of resting potential and induce spontaneous action potential of human cardiomyocytes. K2P1 is expressed in the human heart but not in mouse hearts. We test the hypothesis that K2P1 leak cation channels contribute to ectopic heartbeats under hypokalemia, by analysis of transgenic mice, which conditionally express induced K2P1 specifically in hearts, mimicking K2P1 channels in the human heart. Conditional expression of induced K2P1 specifically in the heart of hypokalemic mice results in multiple types of ventricular ectopic beats including single and multiple ventricular premature beats as well as ventricular tachycardia and causes sudden death. In isolated mouse hearts that express induced K2P1, sustained ventricular fibrillation occurs rapidly after perfusion with low K+ concentration solutions that mimic hypokalemic conditions. These observed phenotypes occur rarely in control mice or in the hearts that lack K2P1 expression. K2P1-expressing mouse cardiomyocytes of transgenic mice much more frequently fire abnormal single and/or rhythmic spontaneous action potential in hypokalemic conditions, compared to wild type mouse cardiomyocytes without K2P1 expression. These findings confirm that K2P1 leak cation channels induce ventricular ectopic beats and sudden death of transgenic mice with hypokalemia and imply that K2P1 leak cation channels may play a critical role in human ectopic heartbeats under hypokalemia.
Collapse
Affiliation(s)
- Rongrong Shen
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Pan-Vascular Research Institute, Heart, Lung, and Blood Center, Tongji University School of Medicine, Shanghai, China
| | - Dongchuan Zuo
- Key Laboratory of Medical Electrophysiology, Institute of Cardiovascular Research, Ministry of Education, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Southwest Medical University, Luzhou, China.,Department of Biological Sciences, University at Albany, State University of New York, Albany, New York, USA
| | - Kuihao Chen
- Department of Biological Sciences, University at Albany, State University of New York, Albany, New York, USA.,Department of Pharmacology, Ningbo University School of Medicine, Ningbo, China
| | - Yiheng Yin
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Pan-Vascular Research Institute, Heart, Lung, and Blood Center, Tongji University School of Medicine, Shanghai, China
| | - Kai Tang
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Pan-Vascular Research Institute, Heart, Lung, and Blood Center, Tongji University School of Medicine, Shanghai, China
| | - Shangwei Hou
- Key Laboratory for Translational Research and Innovative Therapeutics of Gastrointestinal Oncology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bo Han
- Key Laboratory for Translational Research and Innovative Therapeutics of Gastrointestinal Oncology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yawei Xu
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Pan-Vascular Research Institute, Heart, Lung, and Blood Center, Tongji University School of Medicine, Shanghai, China
| | - Zheng Liu
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Pan-Vascular Research Institute, Heart, Lung, and Blood Center, Tongji University School of Medicine, Shanghai, China.,Cryo-Electron Microscopy Center, Southern University of Science and Technology, Shenzhen, China
| | - Haijun Chen
- Department of Biological Sciences, University at Albany, State University of New York, Albany, New York, USA
| |
Collapse
|
33
|
Liu Q, Sun J, Dong Y, Li P, Wang J, Wang Y, Xu Y, Tian X, Wu B, He P, Yu Q, Lu X, Cao J. Tetramisole is a new I K1 channel agonist and exerts I K1 -dependent cardioprotective effects in rats. Pharmacol Res Perspect 2022; 10:e00992. [PMID: 35880674 PMCID: PMC9316008 DOI: 10.1002/prp2.992] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 06/22/2022] [Accepted: 07/04/2022] [Indexed: 11/30/2022] Open
Abstract
Cardiac ischemia, hypoxia, arrhythmias, and heart failure share the common electrophysiological changes featured by the elevation of intracellular Ca2+ (Ca2+ overload) and inhibition of the inward rectifier potassium (IK1 ) channel. IK1 channel agonists have been considered a new type of anti-arrhythmia and cardioprotective agents. We predicted using a drug repurposing strategy that tetramisole (Tet), a known anthelminthic agent, was a new IK1 channel agonist. The present study aimed to experimentally identify the above prediction and further demonstrate that Tet has cardioprotective effects. Results of the whole-cell patch clamp technique showed that Tet at 1-100 μmol/L enhanced IK1 current, hyperpolarized resting potential (RP), and shortened action potential duration (APD) in isolated rat cardiomyocytes, while without effects on other ion channels or transporters. In adult Sprague-Dawley (SD) rats in vivo, Tet showed anti-arrhythmia and anticardiac remodeling effects, respectively, in the coronary ligation-induced myocardial infarction model and isoproterenol (Iso, i.p., 3 mg/kg/day, 10 days) infusion-induced cardiac remodeling model. Tet also showed anticardiomyocyte remodeling effect in Iso (1 μmol/L) infused adult rat ventricular myocytes or cultured H9c2 (2-1) cardiomyocytes. Tet at 0.54 mg/kg in vivo or 30 μmol/L in vitro showed promising protections on acute ischemic arrhythmias, myocardial hypertrophy, and fibrosis. Molecular docking was performed and identified the selective binding of Tet with Kir2.1. The cardioprotection of Tet was associated with the facilitation of IK1 channel forward trafficking, deactivation of PKA signaling, and inhibition of intracellular calcium overload. Enhancing IK1 may play dual roles in anti-arrhythmia and antiventricular remodeling mediated by restoration of Ca2+ homeostasis.
Collapse
Affiliation(s)
- Qinghua Liu
- Department of PathophysiologyShanxi Medical UniversityTaiyuanChina
| | - Jiaxing Sun
- Department of PathophysiologyShanxi Medical UniversityTaiyuanChina
| | - Yangdou Dong
- Department of PathophysiologyShanxi Medical UniversityTaiyuanChina
| | - Pan Li
- Department of PathophysiologyShanxi Medical UniversityTaiyuanChina
| | - Jin Wang
- Key Laboratory of Cellular Physiology, Ministry of EducationShanxi Medical UniversityTaiyuanChina
- Department of PhysiologyShanxi Medical UniversityTaiyuanChina
| | - Yulan Wang
- Key Laboratory of Cellular Physiology, Ministry of EducationShanxi Medical UniversityTaiyuanChina
- Department of PhysiologyShanxi Medical UniversityTaiyuanChina
| | - Yanwu Xu
- Department of BiochemistryShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Xinrui Tian
- Department of Respiratory and Critical Care MedicineSecond Hospital of Shanxi Medical UniversityTaiyuanChina
| | - Bowei Wu
- Key Laboratory of Cellular Physiology, Ministry of EducationShanxi Medical UniversityTaiyuanChina
- Department of PhysiologyShanxi Medical UniversityTaiyuanChina
| | - Peifeng He
- Shanxi Key Laboratory of Big Data for Clinical Decision Research, School of ManagementShanxi Medical UniversityTaiyuanChina
| | - Qi Yu
- Shanxi Key Laboratory of Big Data for Clinical Decision Research, School of ManagementShanxi Medical UniversityTaiyuanChina
| | - Xuechun Lu
- Department of Hematology, The Second Medical Center, Chinese PLA General HospitalNational clinical research center for geriatric diseaseBeijingChina
| | - Jimin Cao
- Key Laboratory of Cellular Physiology, Ministry of EducationShanxi Medical UniversityTaiyuanChina
- Department of PhysiologyShanxi Medical UniversityTaiyuanChina
| |
Collapse
|
34
|
Grune J, Lewis AJM, Yamazoe M, Hulsmans M, Rohde D, Xiao L, Zhang S, Ott C, Calcagno DM, Zhou Y, Timm K, Shanmuganathan M, Pulous FE, Schloss MJ, Foy BH, Capen D, Vinegoni C, Wojtkiewicz GR, Iwamoto Y, Grune T, Brown D, Higgins J, Ferreira VM, Herring N, Channon KM, Neubauer S, Sosnovik DE, Milan DJ, Swirski FK, King KR, Aguirre AD, Ellinor PT, Nahrendorf M. Neutrophils incite and macrophages avert electrical storm after myocardial infarction. NATURE CARDIOVASCULAR RESEARCH 2022; 1:649-664. [PMID: 36034743 PMCID: PMC9410341 DOI: 10.1038/s44161-022-00094-w] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 06/06/2022] [Indexed: 12/24/2022]
Abstract
Sudden cardiac death, arising from abnormal electrical conduction, occurs frequently in patients with coronary heart disease. Myocardial ischemia simultaneously induces arrhythmia and massive myocardial leukocyte changes. In this study, we optimized a mouse model in which hypokalemia combined with myocardial infarction triggered spontaneous ventricular tachycardia in ambulatory mice, and we showed that major leukocyte subsets have opposing effects on cardiac conduction. Neutrophils increased ventricular tachycardia via lipocalin-2 in mice, whereas neutrophilia associated with ventricular tachycardia in patients. In contrast, macrophages protected against arrhythmia. Depleting recruited macrophages in Ccr2 -/- mice or all macrophage subsets with Csf1 receptor inhibition increased both ventricular tachycardia and fibrillation. Higher arrhythmia burden and mortality in Cd36 -/- and Mertk -/- mice, viewed together with reduced mitochondrial integrity and accelerated cardiomyocyte death in the absence of macrophages, indicated that receptor-mediated phagocytosis protects against lethal electrical storm. Thus, modulation of leukocyte function provides a potential therapeutic pathway for reducing the risk of sudden cardiac death.
Collapse
Affiliation(s)
- Jana Grune
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Andrew J. M. Lewis
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- These authors contributed equally and are listed in alphabetical order: Andrew J. M. Lewis, Masahiro Yamazoe
| | - Masahiro Yamazoe
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- These authors contributed equally and are listed in alphabetical order: Andrew J. M. Lewis, Masahiro Yamazoe
| | - Maarten Hulsmans
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - David Rohde
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Ling Xiao
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Shuang Zhang
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Christiane Ott
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), Nuthetal, Germany
| | - David M. Calcagno
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
| | - Yirong Zhou
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Kerstin Timm
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Mayooran Shanmuganathan
- Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- National Institute for Health (NIHR) Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, UK
| | - Fadi E. Pulous
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Maximilian J. Schloss
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Brody H. Foy
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Diane Capen
- Program in Membrane Biology, Nephrology Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Claudio Vinegoni
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Gregory R. Wojtkiewicz
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Yoshiko Iwamoto
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Tilman Grune
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), Nuthetal, Germany
| | - Dennis Brown
- Program in Membrane Biology, Nephrology Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - John Higgins
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | | | - Neil Herring
- National Institute for Health (NIHR) Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, UK
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Keith M. Channon
- Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- National Institute for Health (NIHR) Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, UK
| | - Stefan Neubauer
- Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- National Institute for Health (NIHR) Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, UK
| | | | - David E. Sosnovik
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Filip K. Swirski
- Cardiovascular Research Institute and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kevin R. King
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
- Department of Medicine, Division of Cardiovascular Medicine, University of California, San Diego La Jolla, CA, USA
| | - Aaron D. Aguirre
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Patrick T. Ellinor
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Matthias Nahrendorf
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Internal Medicine, University Hospital Wuerzburg, Wuerzburg, Germany
| |
Collapse
|
35
|
Lin Q, Lam HK, Curtis MJ, Cvetkovic Z. Similarity Maps for Ventricular Arrhythmia Classification. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2022; 2022:1927-1930. [PMID: 36086299 DOI: 10.1109/embc48229.2022.9870989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Ventricular arrhythmias are the primary arrhythmias that cause sudden cardiac death. In current clinical and preclinical research, the discovery of new therapies and their translation is hampered by the lack of consistency in diagnostic criteria for distinguishing between ventricular tachycardia (VT) and ventricular fibrillation (VF). This study develops a new set of features, similarity maps, for discrimination between VT and VF using deep neural network architectures. The similarity maps are designed to capture the similarity and the regularity within an ECG trace. Our experiments show that the similarity maps lead to a substantial improvement in distinguishing VT and VF.
Collapse
|
36
|
Champéroux P, Fares R, Bastogne T, Richard S, Le Guennec JY, Thireau J. Contribution of hemodynamic side effects and associated autonomic reflexes to ventricular arrhythmias triggering by torsadogenic hERG blocking drugs. Br J Pharmacol 2022; 179:4549-4562. [PMID: 35751378 PMCID: PMC9543494 DOI: 10.1111/bph.15905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 05/16/2022] [Accepted: 06/01/2022] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND AND PURPOSES Several hERG blocking drugs known for their propensity to trigger Torsades de Pointes (TdP) were reported to induce a sympatho-vagal coactivation and to enhance High Frequency heart rate (HFHR) and QT oscillations (HFQT) from telemetric data. The present work aims to characterise the underlying mechanism(s) leading to these autonomic changes. EXPERIMENTAL APPROACH Effects of 15 torsadogenic hERG blocking drugs (astemizole, chlorpromazine, cisapride, droperidol, ibutilide, dofetilide, haloperidol, moxifloxacin, pimozide, quinidine, risperidone, sotalol, sertindole, terfenadine, thioridazine) were assessed by telemetry in beagle dogs. Hemodynamic effects on diastolic and systolic arterial pressure were analysed from the first doses causing QTc prolongation and/or HFQT oscillations enhancement. Autonomic control changes were analysed with the High Frequency Autonomic Modulation (HFAM) model. KEY RESULTS Except moxifloxacin and quinidine, all torsadogenic hERG blockers induced parasympathetic activation or sympatho-vagal coactivation combined with enhancement of HFQT oscillations. These autonomic effects result from reflex compensatory mechanisms in response to mild hemodynamic side effects. These hemodynamic mechanisms were characterised by transient HR acceleration during HF oscillations. A phenomenon of concealed QT prolongation was unmasked for several torsadogenic hERG blockers under β-adrenoceptors blockade by atenolol. Resulting enhancement of HFQT oscillations was shown to contribute directly to triggering of dofetilide induced ventricular arrhythmias. CONCLUSIONS AND IMPLICATIONS This work supports for the first time a contribution of hemodynamic side properties to ventricular arrhythmias triggering by torsadogenic hERG blocking drugs. These hemodynamic side effects may constitute a second component of their arrhythmic profile acting as a trigger alongside their intrinsic arrhythmogenic electrophysiological properties.
Collapse
Affiliation(s)
| | - Raafat Fares
- ERBC France, Chemin de Montifault, Baugy, France
| | - Thierry Bastogne
- CRAN CNRS UMR 7039, Université de Lorraine, Vandœuvre-lès-Nancy, France
| | | | - Jean-Yves Le Guennec
- Laboratoire PHYMEDEXP, Université de Montpellier, INSERM, CNRS, Montpellier cedex, France
| | - Jérôme Thireau
- Laboratoire PHYMEDEXP, Université de Montpellier, INSERM, CNRS, Montpellier cedex, France
| |
Collapse
|
37
|
Gholami S, Mokhtari B, Badalzadeh R. Alpha-lipoic acid potentiates the anti-arrhythmic effects of ischemic postconditioning in the setting of cardiac ischemia/reperfusion injury in diabetic rats. J Diabetes Metab Disord 2022; 21:707-716. [PMID: 35673476 DOI: 10.1007/s40200-022-01034-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 03/15/2022] [Indexed: 01/13/2023]
Abstract
Background Prevention of lethal ventricular arrhythmias induced by myocardial ischemia/reperfusion (I/R) in diabetic patients is the major goal of cardioprotective strategies. Here, we aimed to examine the anti-arrhythmic effect of ischemic postconditioning (IPostC) and alpha-lipoic acid (ALA) in myocardial I/R injury of type-II diabetic rats, focusing on the involvement of connexin-43 and nitric oxide (NO) in this context. Methods Diabetes (duration of 12 weeks) was induced by high-fat diet and low dose of streptozotocin in thirty male Wistar rats (12 weeks old, 200-250 g). After mounting the hearts on the Langendorff apparatus, I/R was induced by the ligation of left anterior descending coronary artery for 35 min, and reperfusion for 60 min. ALA (100 mg/kg/day) was administered orally in diabetic rats for five weeks before I/R. IPostC was applied immediately at early reperfusion. The arrhythmias were evaluated according to the Lambeth convention. Connexin-43 expression and NO levels were assessed by western blotting and Griess calorimetric method, respectively. Results IPostC could not significantly decrease the number, duration, and incidence of premature ventricular contraction, ventricular tachycardia, and ventricular fibrillation, also the severity of arrhythmias in diabetic hearts. However, IPostC in combination with ALA-preconditioning significantly decreased the above mentioned parameters compared with untreated or monotherapies-received diabetic rats (P < 0.05 to P < 0.001). Furthermore, this combination therapy significantly increased connexin-43 expression and NO levels, compared with untreated diabetic rats (P < 0.01). Conclusion Preconditioning with ALA restored anti-arrhythmic effect of IPostC in diabetic hearts. Increased connexin-43 expression and NO levels may be the key players in this cardioprotection.
Collapse
Affiliation(s)
- Sanaz Gholami
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Islamic Republic of Iran
| | - Behnaz Mokhtari
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Islamic Republic of Iran.,Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Islamic Republic of Iran
| | - Reza Badalzadeh
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Islamic Republic of Iran.,Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Islamic Republic of Iran.,Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Islamic Republic of Iran
| |
Collapse
|
38
|
Metformin Reduces Potassium Currents and Prolongs Repolarization in Non-Diabetic Heart. Int J Mol Sci 2022; 23:ijms23116021. [PMID: 35682699 PMCID: PMC9181026 DOI: 10.3390/ijms23116021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 05/14/2022] [Accepted: 05/24/2022] [Indexed: 01/27/2023] Open
Abstract
Metformin is the first choice drug for the treatment of type 2 diabetes due to positive results in reducing hyperglycaemia and insulin resistance. However, diabetic patients have higher risk of ventricular arrhythmia and sudden cardiac death, and metformin failed to reduce ventricular arrhythmia in clinical trials. In order to explore the mechanisms responsible for the lack of protective effect, we investigated in vivo the effect of metformin on cardiac electrical activity in non-diabetic rats; and in vitro in isolated ventricular myocytes, HEK293 cells expressing the hERG channel and human induced pluripotent stem cells derived cardiomyocytes (hIPS-CMs). Surface electrocardiograms showed that long-term metformin treatment (7 weeks) at therapeutic doses prolonged cardiac repolarization, reflected as QT and QTc interval duration, and increased ventricular arrhythmia during the caffeine/dobutamine challenge. Patch-clamp recordings in ventricular myocytes isolated from treated animals showed that the cellular mechanism is a reduction in the cardiac transient outward potassium current (Ito). In vitro, incubation with metformin for 24 h also reduced Ito, prolonged action potential duration, and increased spontaneous contractions in ventricular myocytes isolated from control rats. Metformin incubation also reduced IhERG in HEK293 cells. Finally, metformin incubation prolonged action potential duration at 30% and 90% of repolarization in hIPS-CMs, which is compatible with the reduction of Ito and IhERG. Our results show that metformin directly modifies the electrical behavior of the normal heart. The mechanism consists in the inhibition of repolarizing currents and the subsequent decrease in repolarization capacity, which prolongs AP and QTc duration.
Collapse
|
39
|
Blake MR, Parrish DC, Staffenson MA, Sueda S, Woodward WR, Habecker BA. Chondroitin sulfate proteoglycan 4,6 sulfation regulates sympathetic nerve regeneration after myocardial infarction. eLife 2022; 11:e78387. [PMID: 35604022 PMCID: PMC9197393 DOI: 10.7554/elife.78387] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 05/22/2022] [Indexed: 11/13/2022] Open
Abstract
Sympathetic denervation of the heart following ischemia/reperfusion induced myocardial infarction (MI) is sustained by chondroitin sulfate proteoglycans (CSPGs) in the cardiac scar. Denervation predicts risk of sudden cardiac death in humans. Blocking CSPG signaling restores sympathetic axon outgrowth into the cardiac scar, decreasing arrhythmia susceptibility. Axon growth inhibition by CSPGs can depend on the sulfation status of the glycosaminoglycan (CS-GAG) side chains. Tandem sulfation of CS-GAGs at the 4th (4S) and 6th (6S) positions of n-acetyl-galactosamine inhibits outgrowth in several types of central neurons, but we don't know if sulfation is similarly critical during peripheral nerve regeneration. We asked if CSPG sulfation prevented sympathetic axon outgrowth after MI. Reducing 4S with the 4-sulfatase enzyme Arylsulfatase-B (ARSB) enhanced outgrowth of dissociated rat sympathetic neurons over CSPGs. Likewise, reducing 4S with ARSB restored axon outgrowth from mouse sympathetic ganglia co-cultured with cardiac scar tissue. We quantified enzymes responsible for adding and removing sulfation, and found that CHST15 (4S dependent 6-sulfotransferase) was upregulated, and ARSB was downregulated after MI. This suggests a mechanism for production and maintenance of sulfated CSPGs in the cardiac scar. We decreased 4S,6S CS-GAGs in vivo by transient siRNA knockdown of Chst15 after MI, and found that reducing 4S,6S restored tyrosine hydroxylase (TH) positive sympathetic nerve fibers in the cardiac scar. Reinnervation reduced isoproterenol induced arrhythmias. Our results suggest that modulating CSPG-sulfation after MI may be a therapeutic target to promote sympathetic nerve regeneration in the cardiac scar and reduce post-MI cardiac arrhythmias.
Collapse
Affiliation(s)
- Matthew R Blake
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science UniversityPortlandUnited States
| | - Diana C Parrish
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science UniversityPortlandUnited States
| | - Melanie A Staffenson
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science UniversityPortlandUnited States
| | - Shanice Sueda
- Portland State University EXITO Scholars Program, Portland State UniversityPortlandUnited States
| | - William R Woodward
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science UniversityPortlandUnited States
| | - Beth A Habecker
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science UniversityPortlandUnited States
| |
Collapse
|
40
|
Singhanat K, Apaijai N, Sumneang N, Maneechote C, Arunsak B, Chunchai T, Chattipakorn SC, Chattipakorn N. Therapeutic potential of a single-dose melatonin in the attenuation of cardiac ischemia/reperfusion injury in prediabetic obese rats. Cell Mol Life Sci 2022; 79:300. [PMID: 35588335 PMCID: PMC11072751 DOI: 10.1007/s00018-022-04330-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 04/24/2022] [Accepted: 04/27/2022] [Indexed: 01/05/2023]
Abstract
Although acute melatonin treatment effectively reduces cardiac ischemia/reperfusion (I/R) injury in lean rats by modulating melatonin receptor 2 (MT2), there is no information regarding the temporal effects of melatonin administration during cardiac I/R injury in prediabetic obese rats. Prediabetic obese rats induced by chronic consumption of a high-fat diet (HFD) were used. The rats underwent a cardiac I/R surgical procedure (30-min of ischemia, followed by 120-min of reperfusion) and were randomly assigned to receive either vehicle or melatonin treatment. In the melatonin group, rats were divided into 3 different subgroups: (1) pretreatment, (2) treatment during ischemic period, (3) treatment at the reperfusion onset. In the pretreatment subgroup either a nonspecific MT blocker (Luzindole) or specific MT2 blocker (4-PPDOT) was also given to the rats prior to melatonin treatment. Pretreatment with melatonin (10 mg/kg) effectively reduced cardiac I/R injury by reducing infarct size, arrhythmia, and LV dysfunction. Reduction in impaired mitochondrial function, mitochondrial dynamic balance, oxidative stress, defective autophagy, and apoptosis were observed in rats pretreated with melatonin. Unfortunately, the cardioprotective benefits were not observed when 10-mg/kg of melatonin was acutely administered to the rats after cardiac ischemia. Thus, we increased the dose of melatonin to 20 mg/kg, and it was administered to the rats during ischemia or at the onset of reperfusion. The results showed that 20-mg/kg of melatonin effectively reduced cardiac I/R injury to a similar extent to the 10-mg/kg pretreatment regimen. The MT2 blocker inhibited the protective effects of melatonin. Acute melatonin treatment during cardiac I/R injury exerted protective effects in prediabetic obese rats. However, a higher dose of melatonin is required when given after the onset of cardiac ischemia. These effects of melatonin were mainly mediated through activation of MT2.
Collapse
Affiliation(s)
- Kodchanan Singhanat
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Nattayaporn Apaijai
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Natticha Sumneang
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Chayodom Maneechote
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Busarin Arunsak
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Titikorn Chunchai
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Siriporn C Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
- Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Nipon Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand.
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand.
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand.
| |
Collapse
|
41
|
Hesketh LM, Sikkel MB, Mahoney-Sanchez L, Mazzacuva F, Chowdhury RA, Tzortzis KN, Firth J, Winter J, MacLeod KT, Ogrodzinski S, Wilder CDE, Patterson LH, Peters NS, Curtis MJ. OCT2013, an ischaemia-activated antiarrhythmic prodrug, devoid of the systemic side effects of lidocaine. Br J Pharmacol 2022; 179:2037-2053. [PMID: 34855992 DOI: 10.1111/bph.15764] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 09/28/2021] [Accepted: 11/04/2021] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND AND PURPOSE Sudden cardiac death (SCD) caused by acute myocardial ischaemia and ventricular fibrillation (VF) is an unmet therapeutic need. Lidocaine suppresses ischaemia-induced VF, but its utility is limited by side effects and a narrow therapeutic index. Here, we characterise OCT2013, a putative ischaemia-activated prodrug of lidocaine. EXPERIMENTAL APPROACH The rat Langendorff-perfused isolated heart, anaesthetised rat and rat ventricular myocyte preparations were utilised in a series of blinded and randomised studies to investigate the antiarrhythmic effectiveness, adverse effects and mechanism of action of OCT2013, compared with lidocaine. KEY RESULTS In isolated hearts, OCT2013 and lidocaine prevented ischaemia-induced VF equi-effectively, but OCT2013 did not share lidocaine's adverse effects (PR widening, bradycardia and negative inotropy). In anaesthetised rats, i.v. OCT2013 and lidocaine suppressed VF and increased survival equi-effectively; OCT2013 had no effect on cardiac output even at 64 mg·kg-1 i.v., whereas lidocaine reduced it even at 1 mg·kg-1 . In adult rat ventricular myocytes, OCT2013 had no effect on Ca2+ handling, whereas lidocaine impaired it. In paced isolated hearts, lidocaine caused rate-dependent conduction slowing and block, whereas OCT2013 was inactive. However, during regional ischaemia, OCT2013 and lidocaine equi-effectively hastened conduction block. Chromatography and MS analysis revealed that OCT2013, detectable in normoxic OCT2013-perfused hearts, became undetectable during global ischaemia, with lidocaine becoming detectable. CONCLUSIONS AND IMPLICATIONS OCT2013 is inactive but is bio-reduced locally in ischaemic myocardium to lidocaine, acting as an ischaemia-activated and ischaemia-selective antiarrhythmic prodrug with a large therapeutic index, mimicking lidocaine's benefit without adversity.
Collapse
Affiliation(s)
- Louise M Hesketh
- Cardiovascular Division, Faculty of Life Sciences and Medicine, The Rayne Institute, St Thomas' Hospital, King's College London, London, UK
| | - Markus B Sikkel
- National Heart and Lung Institute, Faculty of Medicine, ICTEM, The Hammersmith Hospital, Imperial College London, London, UK
| | - Laura Mahoney-Sanchez
- Cardiovascular Division, Faculty of Life Sciences and Medicine, The Rayne Institute, St Thomas' Hospital, King's College London, London, UK
| | | | - Rasheda A Chowdhury
- National Heart and Lung Institute, Faculty of Medicine, ICTEM, The Hammersmith Hospital, Imperial College London, London, UK
| | - Konstantinos N Tzortzis
- National Heart and Lung Institute, Faculty of Medicine, ICTEM, The Hammersmith Hospital, Imperial College London, London, UK
| | - Jahn Firth
- National Heart and Lung Institute, Faculty of Medicine, ICTEM, The Hammersmith Hospital, Imperial College London, London, UK
| | - James Winter
- Cardiovascular Division, Faculty of Life Sciences and Medicine, The Rayne Institute, St Thomas' Hospital, King's College London, London, UK
| | - Kenneth T MacLeod
- National Heart and Lung Institute, Faculty of Medicine, ICTEM, The Hammersmith Hospital, Imperial College London, London, UK
| | | | - Catherine D E Wilder
- Cardiovascular Division, Faculty of Life Sciences and Medicine, The Rayne Institute, St Thomas' Hospital, King's College London, London, UK
| | | | - Nicholas S Peters
- National Heart and Lung Institute, Faculty of Medicine, ICTEM, The Hammersmith Hospital, Imperial College London, London, UK
| | - Michael J Curtis
- Cardiovascular Division, Faculty of Life Sciences and Medicine, The Rayne Institute, St Thomas' Hospital, King's College London, London, UK
| |
Collapse
|
42
|
Chen J, Liu Z, Deng F, Liang J, Fan B, Zhen X, Tao R, Sun L, Zhang S, Cong Z, Li X, Du W. Mechanisms of Lian-Gui-Ning-Xin-Tang in the treatment of arrhythmia: Integrated pharmacology and in vivo pharmacological assessment. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 99:153989. [PMID: 35272242 DOI: 10.1016/j.phymed.2022.153989] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 01/27/2022] [Accepted: 02/10/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND Lian-Gui-Ning-Xin-Tang (LGNXT), a classical traditional Chinese medicine (TCM) formula, has been widely used in clinical practice and has shown satisfactory efficacy in the treatment of arrhythmias. However, its mechanism of action in the treatment of arrhythmias is still unknown. Moreover, the complex chemical composition and therapeutic targets of LGNXT pose a challenge in pharmacological research. PURPOSE To analyze the active compounds and action mechanisms of LGNXT for the treatment of arrhythmias. METHODS Here, we used an integrated pharmacology approach to identify the potential active compounds and mechanisms of action of LGNXT in treating arrhythmias. Potential active compounds in LGNXT were identified using ultra-performance liquid chromatography-quadrupole-time-of-flight mass spectrometry (UPLC-Q-TOF/MS) and the potential related targets of these compounds were predicted using an integrated in silico approach. The obtained targets were mapped onto relevant databases to identify their corresponding pathways, following the experiments that were conducted to confirm whether the presumptive results of systemic pharmacology were correct. RESULTS Eighty-three components were identified in herbal materials and in animal plasma using UPLC-Q-TOF/MS and were considered the potential active components of LGNXT. Thirty key targets and 57 Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways were identified as possible targets and pathways involved in LGNXT-mediated treatment using network pharmacology, with the cyclic adenosine monophosphate (cAMP)/protein kinase A (PKA)/Ca2+ system pathway being the most significantly affected. This finding was validated using an adrenaline (Adr)-induced rat model of arrhythmias. Pretreatment with LGNXT delayed the occurrence, shortened the duration, and reduced the severity of arrhythmias. LGNXT exerted antiarrhythmic effects by inhibiting cAMP, PKA, CACNA1C, and RyR2. CONCLUSIONS The findings of this study revealed that preventing intracellular Ca2+ overload and maintaining intracellular Ca2+ homeostasis may be the primary mechanisms of LGNXT in alleviating arrhythmias. Thus, we suggest that the β-adrenergic receptor (AR)/cAMP/PKA/Ca2+ system signaling hub may constitute a promising molecular target for the development of novel antiarrhythmic therapeutic interventions. Additionally, we believe that the approach of investigation of the biological effects of a multi-herbal formula by the combination of metabolomics and network pharmacology, as used in this study, could serve as a systematic model for TCM research.
Collapse
Affiliation(s)
- Jinhong Chen
- Graduate School, Tianjin University of TCM, Tianjin 301617, China
| | - Zhichao Liu
- Graduate School, Tianjin University of TCM, Tianjin 301617, China
| | - Fangjun Deng
- Graduate School, Tianjin University of TCM, Tianjin 301617, China
| | - Jiayu Liang
- Graduate School, Tianjin University of TCM, Tianjin 301617, China
| | - Boya Fan
- Graduate School, Tianjin University of TCM, Tianjin 301617, China
| | - Xin Zhen
- Graduate School, Tianjin University of TCM, Tianjin 301617, China
| | - Rui Tao
- Department of TCM, Tianjin University of TCM, Tianjin, 301617, China
| | - Lili Sun
- Department of TCM, Tianjin University of TCM, Tianjin, 301617, China
| | - Shaoqiang Zhang
- Department of Cardiology, The Second Affiliated Hospital of Tianjin University of TCM, Tianjin 300150, China
| | - Zidong Cong
- Department of Cardiology, The Second Affiliated Hospital of Tianjin University of TCM, Tianjin 300150, China
| | - Xiaofeng Li
- Department of Cardiology, The Second Affiliated Hospital of Tianjin University of TCM, Tianjin 300150, China.
| | - Wuxun Du
- Department of Cardiology, The Second Affiliated Hospital of Tianjin University of TCM, Tianjin 300150, China.
| |
Collapse
|
43
|
Norvik A, Unneland E, Bergum D, Buckler D, Bhardwaj A, Eftestøl T, Aramendi E, Nordseth T, Abella B, Kvaløy J, Skogvoll E. Pulseless Electrical Activity in In-Hospital Cardiac Arrest – A crossroad for decisions. Resuscitation 2022; 176:117-124. [DOI: 10.1016/j.resuscitation.2022.04.024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 04/20/2022] [Accepted: 04/21/2022] [Indexed: 02/05/2023]
|
44
|
Qin X, Liu B, Gao F, Hu Y, Chen Z, Xu J, Zhang X. Gluconolactone Alleviates Myocardial Ischemia/Reperfusion Injury and Arrhythmias via Activating PKCε/Extracellular Signal-Regulated Kinase Signaling. Front Physiol 2022; 13:856699. [PMID: 35360251 PMCID: PMC8964113 DOI: 10.3389/fphys.2022.856699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 02/22/2022] [Indexed: 11/26/2022] Open
Abstract
Gluconolactone (D-glucono-1,5-lactone or GDL) is a food additive which presents in dietary products such as tofu, yogurt, cheese, bread, wine, etc. GDL has long been considered as a free radical scavenger; however, its role in cardioprotection remains elusive. In this study, using a mouse model of myocardial ischemia/reperfusion (I/R) injury and a model of hypoxia/reoxygenation (H/R) in neonatal rat cardiomyocytes (NRCM), we explored the role of GDL in I/R injury. We found that GDL (5 mg/kg, i.p.) attenuated myocardial I/R injury as evidenced by decreased infarct size, release of cardiac injury markers and apoptosis. Additionally, GDL decreased reperfusion-induced arrhythmias and oxidative stress. These effects were also observed in parallel in vitro studies. Mechanistically, we found that GDL treatment was strongly associated with activation of pro-survival extracellular signal-regulated kinase (ERK) signaling both in vivo and in vitro, and pharmacological inhibition of ERK signaling via U0126 attenuated GDL-induced cardioprotection against H/R injury in NRCM cells. To reveal how GDL regulates ERK signaling, we predicted the putative targets of GDL by Swiss Target Prediction, and protein kinase C (PKC) emerged as the most promising target for GDL. By pharmacological intervention and immunofluorescence, we found that PKCε, an important member of the PKC family, was activated after GDL treatment in heart, thereby leading to ERK activation and cardioprotection against I/R injury. Taken together, our results demonstrated that GDL acts as a potent activator of PKCε and, thus, provides cardioprotection against I/R injury via activation of ERK signaling.
Collapse
Affiliation(s)
- Xinghua Qin
- School of Life Sciences, Northwestern Polytechnical University, Xi’an, China
| | - Binghua Liu
- School of Life Sciences, Northwestern Polytechnical University, Xi’an, China
| | - Feng Gao
- School of Life Sciences, Northwestern Polytechnical University, Xi’an, China
| | - Yuanyuan Hu
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi’an, China
| | - Ziwei Chen
- Research Center for Prevention and Treatment of Respiratory Disease, School of Clinical Medicine, Xi’an Medical University, Xi’an, China
| | - Jie Xu
- Department of Cardiology, 986th Hospital, Fourth Military Medical University, Xi’an, China
- *Correspondence: Jie Xu,
| | - Xing Zhang
- School of Aerospace Medicine, Fourth Military Medical University, Xi’an, China
- Xing Zhang,
| |
Collapse
|
45
|
Anti-inflammatory, antioxidant, antihypertensive, and antiarrhythmic effect of indole-3-carbinol, a phytochemical derived from cruciferous vegetables. Heliyon 2022; 8:e08989. [PMID: 35243102 PMCID: PMC8866897 DOI: 10.1016/j.heliyon.2022.e08989] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 10/29/2021] [Accepted: 02/15/2022] [Indexed: 11/20/2022] Open
Abstract
Background Cardiovascular inflammation and oxidative stress are determining factors in high blood pressure and arrhythmias. Indole-3-carbinol is a cruciferous-derived phytochemical with potential anti-inflammatory and antioxidant effects. However, its implications on the modulation of cardiovascular inflammatory-oxidative markers are unknown. Objectives To establish the effects of indole-3-carbinol on the oxidative-inflammatory-proarrhythmic conditions associated with hypertension. Materials Histological, biochemical, molecular, and functional aspects were evaluated in 1) Culture of mouse BV-2 glial cells subjected to oxidative-inflammatory damage by lipopolysaccharides (100 ng/mL) in the presence or absence of 40 μM indole-3-carbinol (n = 5); 2) Male spontaneously hypertensive rats (SHR) and Wistar Kyoto rats receiving indole-3-carbinol (2000 ppm/day, orally) during the first 8 weeks of life (n = 15); 3) Isolated rat hearts were submitted to 10 min regional ischemia and 10 min reperfusion. Results 1) lipopolysaccharides induced oxidative stress and increased inflammatory markers; indole-3-carbinol reversed both conditions (interleukin 6, tumor necrosis factor α, the activity of nicotinamide adenine dinucleotide phosphate oxidase, nitric oxide, inducible nitric oxide synthase, heat shock protein 70, all p < 0.01 vs lipopolysaccharides). 2) SHR rats showed histological, structural, and functional changes with increasing systolic blood pressure (154 ± 8 mmHg vs. 122 ± 7 mmHg in Wistar Kyoto rats, p < 0.01); Inflammatory-oxidative markers also increased, and nitric oxide and heat shock protein 70 decreased. Conversely, indole-3-carbinol reduced oxidative-inflammatory markers and systolic blood pressure (133 ± 8 mmHg, p < 0.01 vs. SHR). 3) indole-3-carbinol reduced reperfusion arrhythmias from 8/10 in SHR to 0/10 (p = 0.0007 by Fisher's exact test). Conclusions Indole-3-carbinol reduces the inflammatory-oxidative-proarrhythmic process of hypertension. The nitric oxide and heat shock protein 70 are relevant mechanisms of indole-3-carbinol protective actions. Further studies with this pleiotropic phytochemical as a promising cardioprotective are guaranteed. Indole-3-carbinol, a cruciferous-derived compound, has cardioprotective potential. We confirmed its anti-inflammatory and antioxidant effects in vitro and in vivo. Oral administration reduced blood pressure and cardiac remodeling. In isolated hearts from hypertensive rats prevented ischemia-reperfusion arrhythmias. Heat shock protein 70 and NO contribute to indole-3-carbinol protective actions.
Collapse
|
46
|
Kui P, Polyák A, Morvay N, Tiszlavicz L, Nagy N, Ördög B, Takács H, Leprán I, Farkas A, Papp JG, Jost N, Varró A, Baczkó I, Farkas AS. Long-Term Endurance Exercise Training Alters Repolarization in a New Rabbit Athlete’s Heart Model. Front Physiol 2022; 12:741317. [PMID: 35237176 PMCID: PMC8882986 DOI: 10.3389/fphys.2021.741317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 10/22/2021] [Indexed: 11/13/2022] Open
Abstract
In the present study, the effect of long-term exercise training was investigated on myocardial morphological and functional remodeling and on proarrhythmic sensitivity in a rabbit athlete’s heart model. New-Zealand white rabbits were trained during a 12-week long treadmill running protocol and compared with their sedentary controls. At the end of the training protocol, echocardiography, in vivo and in vitro ECG recordings, proarrhythmic sensitivity with dofetilide (nM) were performed in isolated hearts, and action potential duration (APD) measurements at different potassium concentrations (4.5 and 2 mM) were made in the isolated papillary muscles. Expression levels of the slow component of delayed rectifier potassium current and fibrosis synthesis and degradation biomarkers were quantified. Echocardiography showed a significantly dilated left ventricle in the running rabbits. ECG PQ and RR intervals were significantly longer in the exercised group (79 ± 2 vs. 69 ± 2 ms and 325 ± 11 vs. 265 ± 6 ms, p < 0.05, respectively). The in vivo heart rate variability (HRV) (SD of root mean square: 5.2 ± 1.4 ms vs. 1.4 ± 0.2 ms, p < 0.05) and Tpeak-Tend variability were higher in the running rabbits. Bradycardia disappeared in the exercised group in vitro. Dofetilide tended to increase the QTc interval in a greater extent, and significantly increased the number of arrhythmic beats in the trained animals in vitro. APD was longer in the exercised group at a low potassium level. Real-time quantitative PCR (RT-qPCR) showed significantly greater messenger RNA expression of fibrotic biomarkers in the exercised group. Increased repolarization variability and higher arrhythmia incidences, lengthened APD at a low potassium level, increased fibrotic biomarker gene expressions may indicate higher sensitivity of the rabbit “athlete’s heart” to life-threatening arrhythmias.
Collapse
Affiliation(s)
- Péter Kui
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Alexandra Polyák
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
- Department of Internal Medicine, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
- ELKH-SZTE Working Group of Cardiovascular Pharmacology, Szeged, Hungary
| | - Nikolett Morvay
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - László Tiszlavicz
- Department of Pathology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Norbert Nagy
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
- ELKH-SZTE Working Group of Cardiovascular Pharmacology, Szeged, Hungary
| | - Balázs Ördög
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Hedvig Takács
- Department of Internal Medicine, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - István Leprán
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - András Farkas
- Department of Internal Medicine, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Julius Gy. Papp
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
- ELKH-SZTE Working Group of Cardiovascular Pharmacology, Szeged, Hungary
| | - Norbert Jost
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
- ELKH-SZTE Working Group of Cardiovascular Pharmacology, Szeged, Hungary
| | - András Varró
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
- ELKH-SZTE Working Group of Cardiovascular Pharmacology, Szeged, Hungary
- Department of Pharmacology and Pharmacotherapy, Interdisciplinary Excellence Centre, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
- *Correspondence: András Varró,
| | - István Baczkó
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
- Department of Pharmacology and Pharmacotherapy, Interdisciplinary Excellence Centre, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Attila S. Farkas
- Department of Internal Medicine, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| |
Collapse
|
47
|
Kaya ST, Agan K, Fulden-Agan A, Agyar-Yoldas P, Ozarslan TO, Kekecoglu M, Kaya A. Protective effect of propolis on myocardial ischemia/reperfusion injury in males and ovariectomized females but not in intact females. J Food Biochem 2022; 46:e14109. [PMID: 35142377 DOI: 10.1111/jfbc.14109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 12/12/2021] [Accepted: 01/22/2022] [Indexed: 11/30/2022]
Abstract
The aim of this study is to investigate the effect of propolis, which may have estrogenic effects, on myocardial ischemia/reperfusion (mI/R) injury not only in male rats but also in intact and ovariectomized (ovx) female rats. Six groups were formed: untreated males (n = 8), treated males (n = 9), untreated intact females (n = 9), treated intact females (n = 10), untreated ovx females (n = 10), and treated ovx females (n = 8). An alcoholic extract of a single dose of propolis (200 mg/kg) was administered orally daily for 14 days. Thirty minutes of ischemia and 120 min of reperfusion were performed. Blood pressure, heart rate, arrhythmias (ventricular premature contraction [VPC], ventricular tachycardia [VT], ventricular fibrillation [VF]), and myocardial infarct size were evaluated. Total antioxidant status (TAS), total oxidant status (TOS), superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPx), and 17 beta-estradiol (E2) were measured. The untreated females showed more resistance to mI/R injury than the untreated males, as evidenced by lower duration, incidence, and score of arrhythmias, and smaller infarct size (p < .05). After ovx, this resistance disappeared. Propolis improved these values in treated males and treated ovx females (p < .05). Propolis increased TAS in treated males and decreased TOS in treated ovx females as well as elevated SOD in all treated groups (p < .05). Propolis decreased E2 level in treated intact females; however, it increased E2 level in treated ovx females (p < .05). The results revealed that propolis could protect the heart against mI/R injury in males and ovx females. PRACTICAL APPLICATIONS: It is known that the female heart has an increased sensitivity to myocardial ischemia/reperfusion (mI/R) injury due to estrogen deficiency and/or estrogen deprivation following menopause or surgical removal of the ovaries. Propolis has the potential to mimic estrogen under physiological and pathophysiological conditions, as well as its antioxidant property. The results indicated that propolis decreased myocardial infarct size, arrhythmia score, arrhythmia duration, and incidence in ovariectomized female rats and male rats. In addition, the present results demonstrated that an alcoholic extract of propolis as a natural product can effectively maintain the resistance of female heart to mI/R injury after estrogen deficiency.
Collapse
Affiliation(s)
- Salih Tunc Kaya
- Faculty of Arts and Science, Department of Biology, Duzce University, Duzce, Turkey
| | - Kagan Agan
- Coordination Unit of Healthy and Environmental, Duzce University, Duzce, Turkey
| | - Aydan Fulden-Agan
- Beekeeping Research, Development and Application Centre, Duzce University, Duzce, Turkey
| | - Pınar Agyar-Yoldas
- Coordination Unit of Healthy and Environmental, Duzce University, Duzce, Turkey
| | - Talat Ogulcan Ozarslan
- Faculty of Medicine, Department of Infectious Diseases and Clinical Microbiology, Bolu Abant Izzet Baysal University, Bolu, Turkey
| | - Meral Kekecoglu
- Faculty of Arts and Science, Department of Biology, Duzce University, Duzce, Turkey.,Beekeeping Research, Development and Application Centre, Duzce University, Duzce, Turkey
| | - Adnan Kaya
- Faculty of Medicine, Department of Internal Medicine, Cardiology Section, Duzce University, Duzce, Turkey
| |
Collapse
|
48
|
Diaz-Maue L, Steinebach J, Richter C. Patterned Illumination Techniques in Optogenetics: An Insight Into Decelerating Murine Hearts. Front Physiol 2022; 12:750535. [PMID: 35087413 PMCID: PMC8787046 DOI: 10.3389/fphys.2021.750535] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 12/02/2021] [Indexed: 11/21/2022] Open
Abstract
Much has been reported about optogenetic based cardiac arrhythmia treatment and the corresponding characterization of photostimulation parameters, but still, our capacity to interact with the underlying spatiotemporal excitation patterns relies mainly on electrical and/or pharmacological approaches. However, these well-established treatments have always been an object of somehow heated discussions. Though being acutely life-saving, they often come with potential side-effects leading to a decreased functionality of the complex cardiac system. Recent optogenetic studies showed the feasibility of the usage of photostimulation as a defibrillation method with comparatively high success rates. Although, these studies mainly concentrated on the description as well as on the comparison of single photodefibrillation approaches, such as locally focused light application and global illumination, less effort was spent on the description of excitation patterns during actual photostimulation. In this study, the authors implemented a multi-site photodefibrillation technique in combination with Multi-Lead electrocardiograms (ECGs). The technical connection of real-time heart rhythm measurements and the arrhythmia counteracting light control provides a further step toward automated arrhythmia classification, which can lead to adaptive photodefibrillation methods. In order to show the power effectiveness of the new approach, transgenic murine hearts expressing channelrhodopsin-2 ex vivo were investigated using circumferential micro-LED and ECG arrays. Thus, combining the best of two methods by giving the possibility to illuminate either locally or globally with differing pulse parameters. The optical technique presented here addresses a number of challenges of technical cardiac optogenetics and is discussed in the context of arrhythmic development during photostimulation.
Collapse
Affiliation(s)
- Laura Diaz-Maue
- Department of Research Electronics, Max-Planck-Institute for Dynamics and Self-Organization, Göttingen, Germany.,Biomedical Physics Research Group, Max-Planck-Institute for Dynamics and Self-Organization, Göttingen, Germany.,German Center for Cardiovascular Research (DZHK e., V.), Göttingen, Germany
| | - Janna Steinebach
- Biomedical Physics Research Group, Max-Planck-Institute for Dynamics and Self-Organization, Göttingen, Germany
| | - Claudia Richter
- German Center for Cardiovascular Research (DZHK e., V.), Göttingen, Germany.,Laboratory Animal Science Unit, German Primate Center, Leibniz-Institute for Primate Research, Göttingen, Germany
| |
Collapse
|
49
|
Polyakova EA, Mikhaylov EN, Galagudza MM, Shlyakhto EV. Hyperleptinemia results in systemic inflammation and the exacerbation of ischemia-reperfusion myocardial injury. Heliyon 2021; 7:e08491. [PMID: 34901513 PMCID: PMC8640453 DOI: 10.1016/j.heliyon.2021.e08491] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 09/09/2021] [Accepted: 11/24/2021] [Indexed: 12/01/2022] Open
Abstract
Aim Hyperleptinemia potentiates the effects of many atherogenic factors, such as inflammation, platelet aggregation, migration, hypertrophy, proliferation of vascular smooth muscle cells, and endothelial cell dysfunction. The present study analysed the effects of long-term hyperleptinemia in an in vivo myocardial ischemia-reperfusion model to demonstrate whether the in vivo deleterious effect also affects cardiac structure and function. Main methods Rats were subcutaneously administered leptin for 8 days to estimate the involvement of the JAK/STAT pathway. Data from 58 male Wistar rats were included in the final analysis. Myocardial infarction (MI) was modelled by the 30-minute ligation of the main left coronary artery followed by 120-minute reperfusion. Hemodynamic measurements, electrocardiography monitoring, echocardiography, myocardial infarct size and area at risk, blood biochemical parameters, leptin, IL-6, TNF-alpha, FGF-21, and cardiomyocyte morphology were measured. The expression of JAK2, p-JAK2, STAT3, p-STAT3 was assessed by Western Blot analysis. Statistical analyses were performed using IBM SPSS Statistics v.26. Key findings Eight-day hyperleptinemia in rats leads to an increase in blood pressure and heart rate, myocardial hypertrophy, impaired LV function, the frequency of ischemic arrhythmias, dyslipidemia, systemic inflammation, and the size of induced myocardial infarction. Significance: The blockade of the JAK/STAT signalling pathway effectively reverses the negative effects of leptin, including increased blood pressure and total cholesterol.
Collapse
Affiliation(s)
- Ekaterina A Polyakova
- Almazov National Medical Research Centre, Institute of Experimental Medicine, Saint-Petersburg, Russian Federation
| | - Evgeny N Mikhaylov
- Almazov National Medical Research Centre, Institute of Experimental Medicine, Saint-Petersburg, Russian Federation
| | - Michael M Galagudza
- Almazov National Medical Research Centre, Institute of Experimental Medicine, Saint-Petersburg, Russian Federation
| | - Evgeny V Shlyakhto
- Almazov National Medical Research Centre, Institute of Experimental Medicine, Saint-Petersburg, Russian Federation
| |
Collapse
|
50
|
Long Y, Hou J, Tang F, Lin Z, Huang X, Li W, Chen Y, Li Z, Wu Z. Proarrhythmic effects induced by benzethonium chloride and domiphen bromide in vitro and in vivo. Toxicol Appl Pharmacol 2021; 431:115731. [PMID: 34592322 DOI: 10.1016/j.taap.2021.115731] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 09/13/2021] [Accepted: 09/23/2021] [Indexed: 12/31/2022]
Abstract
Benzethonium chloride (BZT) and domiphen bromide (DMP) are widely used as antimicrobials in drugs, vaccines and industry. However, no cardiac safety data has been developed on both compounds. Previously we reported BZT and DMP as high-affinity human ether-a-go-go related gene (HERG) channel inhibitors with unknown proarrhythmic risk. Here, we investigate the cardiotoxicity of BZT and DMP in vitro and in vivo, aiming to improve the safety-in-use of both antimicrobials. In the present study, human iPSC derived cardiomyocytes (hiPSC-CMs) were generated and rabbit models were used to examine the proarrhythmic potential of BZT and DMP. Our results found that BZT and DMP induced time- and dose-dependent decrease in the contractile parameters of hiPSC-CMs, prolonged FPDc (≥ 0.1 μM), caused tachycardia/fibrillation-like oscillation (0.3-1 μM), ultimately progressing to irreversible arrest of beating (≥ 1 μM). The IC50 values of BZT and DMP derived from normalized beat rate were 0.13 μM and 0.10 μM on hiPSC-CMs at 76 days. Moreover, in vivo rabbit ECG data demonstrated that 12.85 mg/kg BZT and 3.85 mg/kg DMP evoked QTc prolongation, noncomplex arrhythmias and ventricular tachycardias. Our findings support the cardiac safety of 0.01 μM BZT/DMP in vitro and the intravenous infusion of 3.85 mg/kg BZT and 1.28 mg/kg DMP in vivo, whereas higher concentrations of both compounds cause mild to moderate cardiotoxicity that should not be neglected during medical and industrial applications.
Collapse
Affiliation(s)
- Yan Long
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China; Central Laboratory, Shenzhen Samii Medical Center, Shenzhen, China
| | - Jian Hou
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Feng Tang
- Key Laboratory of Regenerative Biology, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Zuoxian Lin
- Key Laboratory of Regenerative Biology, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Xiaolin Huang
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Wei Li
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yili Chen
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Zhiyuan Li
- Key Laboratory of Regenerative Biology, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.
| | - Zhongkai Wu
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|