1
|
Elmadani M, Raatikainen S, Mattila O, Alakoski T, Piuhola J, Åström P, Tenhunen O, Magga J, Kerkelä R. Dasatinib targets c-Src kinase in cardiotoxicity. Toxicol Rep 2023; 10:521-528. [PMID: 37152411 PMCID: PMC10160240 DOI: 10.1016/j.toxrep.2023.04.013] [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: 03/02/2023] [Revised: 04/04/2023] [Accepted: 04/24/2023] [Indexed: 05/09/2023] Open
Abstract
Dasatinib is a multitargeted kinase inhibitor used for treatment of chronic myeloid leukemia and acute lymphoblastic leukemia. Unfortunately, treatment of cancer patients with some kinase inhibitors has been associated with cardiotoxicity. Cancer treatment with dasatinib has been reported to be associated with cardiotoxic side effects such as left ventricular dysfunction, heart failure, pericardial effusion and pulmonary hypertension. Here we aimed to investigate the molecular mechanisms underlying the cardiotoxicity of dasatinib. We found that among the resident cardiac cell types, cardiomyocytes were most sensitive to dasatinib-induced cell death. Exposure of cardiomyocytes to dasatinib attenuated the activity of extracellular signal-regulated kinase (ERK), which is a downstream target of dasatinib target kinase c-Src. Similar to dasatinib, c-Src depletion in cardiomyocytes compromised cardiomyocyte viability. Overexpression of dasatinib-resistant mutant of c-Src rescued the toxicity of dasatinib on cardiomyocytes, whereas forced expression of wild type c-Src did not have protective effect. Collectively, our results show that c-Src is a key target of dasatinib mediating the toxicity of dasatinib to cardiomyocytes. These findings may influence future drug design and suggest closer monitoring of patients treated with agents targeting c-Src for possible adverse cardiac effects.
Collapse
Affiliation(s)
- Manar Elmadani
- Research Unit of Biomedicine and Internal Medicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu, Finland
| | - Sami Raatikainen
- Research Unit of Biomedicine and Internal Medicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu, Finland
| | - Orvokki Mattila
- Research Unit of Biomedicine and Internal Medicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu, Finland
| | - Tarja Alakoski
- Research Unit of Biomedicine and Internal Medicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu, Finland
| | - Jarkko Piuhola
- Division of Cardiology, Oulu University Hospital, Oulu, Finland
| | - Pirjo Åström
- Research Unit of Biomedicine and Internal Medicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu, Finland
| | - Olli Tenhunen
- Research Unit of Biomedicine and Internal Medicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu, Finland
| | - Johanna Magga
- Research Unit of Biomedicine and Internal Medicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu, Finland
- Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Risto Kerkelä
- Research Unit of Biomedicine and Internal Medicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu, Finland
- Biocenter Oulu, University of Oulu, Oulu, Finland
- Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
- Correspondence to: Research Unit of Biomedicine and Internal Medicine, University of Oulu, P.O.BOX 5000, FIN-90014 Oulu, Finland.
| |
Collapse
|
2
|
Armandeh M, Bameri B, Baeeri M, Haghi-Aminjan H, Rahimifard M, Hassani S, Hooshangi Shayesteh MR, Khalid M, Samadi M, Hosseini R, Masoudi Fard M, Abdollahi M. The role of levosimendan in phosphine-induced cardiotoxicity: evaluation of electrocardiographic, echocardiographic, and biochemical parameters. Toxicol Mech Methods 2021; 31:631-643. [PMID: 34219611 DOI: 10.1080/15376516.2021.1950248] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Aluminum phosphide (AlP) causes serious poisoning in which severe cardiac suppression is the significant lethal consequence. According to evidence, levosimendan can exert outstanding cardiac support and protection in different pathological conditions. This study aimed to investigate the mechanisms by which levosimendan may alleviate cardiovascular toxicity due to AlP intoxication in the rat model. The groups included control group (normal saline only), sole levosimendan groups (12, 24, 48 μg/kg), AlP group (10 mg/kg), and AlP + levosimendan groups receiving 12, 24, 48 μg/kg levosimendan intraperitoneally 30 min after AlP administration. Electrocardiographic (ECG) parameters (QRS and PR duration and ST height), heart rate, and blood pressure were monitored for 180 minutes. Also, after 24 h of poisoning, echocardiography was applied to assess left ventricle function. Evaluation of the biochemical parameters in heart tissue, including mitochondrial complexes I, II, IV activity, ADP/ATP ratio, the rate of apoptosis, malondialdehyde (MDA), lactate, and troponin I levels, were done after 12 and 24 h. AlP-induced ECG abnormalities (PR duration and ST height), reduction in heart rate, blood pressure, cardiac output, ejection fraction, and stroke volume were improved by levosimendan administration. Besides, levosimendan significantly improved complex IV activity, the ADP/ATP ratio, apoptosis, MDA, lactate, and troponin I level following AlP-poisoning. Our results suggest that levosimendan might alleviate AlP-induced cardiotoxicity by modulating mitochondrial activity and improving cardiac function. However, the potential clinical use of levosimendan in this toxicity needs more investigations.
Collapse
Affiliation(s)
- Maryam Armandeh
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.,Toxicology and Diseases Group, Pharmaceutical Sciences Research Center (P SRC), The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Behnaz Bameri
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.,Toxicology and Diseases Group, Pharmaceutical Sciences Research Center (P SRC), The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Baeeri
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.,Toxicology and Diseases Group, Pharmaceutical Sciences Research Center (P SRC), The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Hamed Haghi-Aminjan
- Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Mahban Rahimifard
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.,Toxicology and Diseases Group, Pharmaceutical Sciences Research Center (P SRC), The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Shokoufeh Hassani
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.,Toxicology and Diseases Group, Pharmaceutical Sciences Research Center (P SRC), The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Hooshangi Shayesteh
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.,Toxicology and Diseases Group, Pharmaceutical Sciences Research Center (P SRC), The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Madiha Khalid
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.,Toxicology and Diseases Group, Pharmaceutical Sciences Research Center (P SRC), The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Mahedeh Samadi
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Rohollah Hosseini
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Majid Masoudi Fard
- Department of Surgery & Radiology, Faculty of Veterinary Medicine, Tehran University, Tehran, Iran
| | - Mohammad Abdollahi
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.,Toxicology and Diseases Group, Pharmaceutical Sciences Research Center (P SRC), The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
3
|
Pharmacological Pre- and Postconditioning With Levosimendan Protect H9c2 Cardiomyoblasts From Anoxia/Reoxygenation-induced Cell Death via PI3K/Akt Signaling. J Cardiovasc Pharmacol 2021; 77:378-385. [PMID: 33662980 DOI: 10.1097/fjc.0000000000000969] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 11/17/2020] [Indexed: 12/15/2022]
Abstract
ABSTRACT The calcium sensitizer levosimendan is indicated for the hemodynamic stabilization of patients with acutely decompensated heart failure and has been shown to be protective against reperfusion injury after myocardial infarction. However, affected forms of cell death and underlying signaling pathways remain controversial. Therefore, the aim of this study was to examine the influence of levosimendan preconditioning and postconditioning on anoxia/reoxygenation-induced apoptosis, necrosis, and autophagy in H9c2 myoblasts. To mimic conditions of myocardial ischemia/reperfusion, rat cardiac H9c2 myoblasts were exposed to anoxia/starvation, followed by reoxygenation/refeeding. Apoptosis, necrosis, autophagy, cell viability, survival signaling, and mitochondrial permeability transition pore (mPTP) opening were measured. Both, pharmacological preconditioning and postconditioning with levosimendan were capable to reduce apoptosis as well as necrosis in stressed H9c2 cells. However, preconditioning showed to have the stronger impact compared with postconditioning. Moreover, levosimendan preconditioning increased autophagy, suggesting enhanced repair processes initiated by the early presence of the drug. Underlying mechanisms differ between both interventions: Although both are associated with PI3/Akt activation and reduced mPTP opening, only postconditioning but not preconditioning depended on mKATP activation. This variation might indicate that a pharmacological treatment after the onset of reoxygenation at least in part directly addresses mitochondrial structures for protection. In conclusion, we demonstrate that both pharmacological preconditioning and postconditioning with levosimendan protect anoxia/reoxygenation-stressed cells but differ in the underlying mechanisms. These results are decisive to obtain more insights into the beneficial effects of levosimendan in the treatment of reperfusion-mediated damage.
Collapse
|
4
|
Makkos A, Szántai Á, Pálóczi J, Pipis J, Kiss B, Poggi P, Ferdinandy P, Chatgilialoglu A, Görbe A. A Comorbidity Model of Myocardial Ischemia/Reperfusion Injury and Hypercholesterolemia in Rat Cardiac Myocyte Cultures. Front Physiol 2020; 10:1564. [PMID: 31992989 PMCID: PMC6962358 DOI: 10.3389/fphys.2019.01564] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 12/12/2019] [Indexed: 12/19/2022] Open
Abstract
Introduction The use of comorbidity models is crucial in cardioprotective drug development. Hypercholesterolemia causes endothelial and myocardial dysfunction, as well as aggravates ischemia/reperfusion (I/R)-induced myocardial injury. Endogenous cardioprotective mechanisms against I/R are impaired in hyperlipidemic and hyperglycemic in vivo animal models. Therefore, our aim was to develop a medium throughput comorbidity cell-based test system of myocardial I/R injury, hypercholesterolemia and hyperglycemia that mimics comorbidity conditions. Methods Cardiac myocytes isolated from neonatal or adult rat hearts were cultured in control or in three different hypercholesterolemic media with increasing cholesterol content (hiChol) or hiChol + hyperglycemic medium, respectively. Each group was then subjected to simulated ischemia/reperfusion (SI/R) or corresponding normoxic condition, respectively. Cholesterol uptake was tested by Filipin staining in neonatal cardiac myocytes. Cell viability, total cell count and oxidative stress, i.e., total reactive oxygen species (ROS) and superoxide level were measured by fluorescent assays. Results Neonatal cardiac myocytes took up cholesterol from the different hiChol media at a concentration-dependent manner. In normoxia, viability of hiChol neonatal cardiac myocytes was not significantly changed, however, superoxide levels were increased as compared to vehicle. After SI/R, the viability of hiChol neonatal cardiac myocytes was decreased and total ROS level was increased as compared to vehicle. HiChol combined with hyperglycemia further aggravated cell death and oxidative stress in normoxic as well as in SI/R conditions. Viability of hiChol adult cardiac myocytes was significantly decreased and superoxide level was increased in normoxia and these changes were further aggravated by SI/R. HiChol combined with hyperglycemia further aggravated cell death, however level of oxidative stress increased only in normoxic condition. Conclusion HiChol rat cardiac myocytes showed reduction of cell viability and increased oxidative stress, which were further aggravated by SI/R and with additional hyperglycemia. This is the first demonstration that the combination of the current hypercholesterolemic medium and SI/R in cardiac myocytes mimics the cardiac pathology of the comorbid heart with I/R and hypercholesterolemia.
Collapse
Affiliation(s)
- András Makkos
- Cardiometabolic Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Ágnes Szántai
- Department of Biochemistry, University of Szeged, Szeged, Hungary
| | - János Pálóczi
- Department of Biochemistry, University of Szeged, Szeged, Hungary
| | | | - Bernadett Kiss
- Cardiometabolic Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary.,Department of Biochemistry, University of Szeged, Szeged, Hungary
| | | | - Péter Ferdinandy
- Cardiometabolic Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary.,Department of Biochemistry, University of Szeged, Szeged, Hungary.,Pharmahungary Group, Szeged, Hungary
| | | | - Anikó Görbe
- Cardiometabolic Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary.,Department of Biochemistry, University of Szeged, Szeged, Hungary.,Pharmahungary Group, Szeged, Hungary
| |
Collapse
|
5
|
Rababa'h AM, Khabour OF, Alzoubi KH, Al-Momani D, Ababneh M. Assessment of Genotoxicity of Levosimendan in Human Cultured Lymphocytes. Curr Mol Pharmacol 2019; 12:160-165. [PMID: 30848225 DOI: 10.2174/1874467212666190306164926] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 02/16/2019] [Accepted: 02/20/2019] [Indexed: 01/15/2023]
Abstract
BACKGROUND AND OBJECTIVE Levosimendan is a positive inotropic and a vasodilator agent with pleotropic characteristics that include antioxidation, anti-inflammation and smooth muscle vasodilation. METHODS In this study, the effects of levosimendan (0, 0.1, 1, 10, and 20 µg/ml) on oxidative DNA damage and sister-chromatid exchanges (SCEs) were evaluated in human cultured lymphocytes. RESULTS The results showed that levosimendan increased the frequency of SCEs in all examined concentrations (P<0.01) except for 0.1 µg/ml. On the other hand, levosimendan did not induce oxidative DNA damage as measured by the 8-OHdG biomarker (P > 0.05). In addition, neither mitotic arrest nor proliferation index was affected by levosimendan at all examined doses (P > 0.05). CONCLUSION In conclusion, levosimendan might be associated with increases in sister-chromatid exchanges in cultured human lymphocytes. In vivo studies are required to confirm the present findings.
Collapse
Affiliation(s)
- Abeer M Rababa'h
- Department of Clinical Pharmacy; College of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - Omar F Khabour
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, Jordan University of Science and Technology, Irbid, Jordan
| | - Karem H Alzoubi
- Department of Clinical Pharmacy; College of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - Dua'a Al-Momani
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, Jordan University of Science and Technology, Irbid, Jordan
| | - Mera Ababneh
- Department of Clinical Pharmacy; College of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| |
Collapse
|
6
|
Tawfik MK, El-Kherbetawy MK, Makary S. Cardioprotective and Anti-Aggregatory Effects of Levosimendan on Isoproterenol-Induced Myocardial Injury in High-Fat-Fed Rats Involves Modulation of PI3K/Akt/mTOR Signaling Pathway and Inhibition of Apoptosis. J Cardiovasc Pharmacol Ther 2018; 23:456-471. [DOI: 10.1177/1074248418763957] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Hyperlipidemia and hypercoagulability states are linked with the increased risks of myocardial infarction (MI). Levosimendan has vasorelaxant and anti-aggregatory properties. The present study evaluated the anti-aggregatory and cardioprotective effects of levosimendan versus cilostazol in high-fat diet (HFD)-fed rats subjected to isoproterenol-induced MI. Rats were assigned to normal, HFD, HFD + isoproterenol, HFD + isoproterenol + cilostazol, and HFD + isoproterenol + levosimendan. The present study investigated the anti-aggregatory effect of both levosimendan and cilostazol and revealed that both drugs attenuated the severity of platelet aggregation. Moreover, both levosimendan and cilostazol revealed effectiveness in attenuating the severity of HFD/isoproterenol-induced myocardial injury as revealed by electrocardiogram signs, apoptotic markers, and histopathological score via counteracting the oxidative stress burden, increments in the expression of inflammatory mediators, and modulating nuclear factor kappa-B (NF-κB) and phosphatidylinositide 3-kinases (PI3K)/protein kinase B (Akt)/ mechanistic target of rapamycin (mTOR) pathway. It was obvious that levosimendan offered more cardioprotective properties than cilostazol. The study showed the relations between hyperlipedemia, hyperaggregability state, and myocardial injury with the modulation of NF-κB and PI3K/Akt/mTOR pathway.
Collapse
Affiliation(s)
- Mona K. Tawfik
- Department of Pharmacology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | | | - Samy Makary
- Department of Physiology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| |
Collapse
|
7
|
Yin C, Chen Y, Wu H, Xu D, Tan W. Attenuation of ischemia/reperfusion-induced inhibition of the rapid component of delayed rectifier potassium current by Isosteviol through scavenging reactive oxygen species. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2017; 1859:2447-2453. [DOI: 10.1016/j.bbamem.2017.09.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 08/18/2017] [Accepted: 09/05/2017] [Indexed: 11/30/2022]
|
8
|
Inhibition of rapid delayed rectifier potassium current (I Kr) by ischemia/reperfusion and its recovery by vitamin E in ventricular myocytes. J Electrocardiol 2017. [PMID: 28646979 DOI: 10.1016/j.jelectrocard.2017.03.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Ischemia/reperfusion (I/R) induces prolongation of QT interval and action potential duration (APD), which is a major cardiac electrical disorder in patients with arrhythmias. However, the mechanism of QT interval prolongation induced by I/R remains unclear. In the present study, we hypothesized that the rapid component of delayed rectifier potassium (IKr) channel plays an important role in I/R-induced QT interval prolongation. We observed a marked attenuation of IKr and a significant prolongation of action potential duration (APD) in a simulated I/R system with sodium dithionite (Na2S2O4) in ventricular myocytes of guinea pigs. The IKr current density was inhibited by 64% and APD increased by 87% respectively. Moreover, the inhibition of IKr is primarily ascribed to overproduction of reactive oxygen species (ROS) by I/R, which can be partly reversed by antioxidant vitamin E (100μmol/L). The value of IKr tail current density increased from 0.516±0.040 pA/pF in I/R to 0.939±0.091 pA/pF when treated with vitamin E. Moreover, we also demonstrated that QTc interval was increased by I/R and reversed by Vitamin E in isolated guinea pig hearts. In conclusion, the inhibition of IKr is one of the underlying mechanisms of prolongation of QT interval and APD in I/R. Vitamin E might have a benefit in coronary reperfusion therapy.
Collapse
|
9
|
Feng J, Liu H, Chen J, Wang J, Liu Z, Ge S. Levosimendan Reduces Lung Injury in a Canine Model of Cardiopulmonary Bypass. Korean Circ J 2016; 46:402-7. [PMID: 27275177 PMCID: PMC4891605 DOI: 10.4070/kcj.2016.46.3.402] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 11/05/2015] [Accepted: 11/12/2015] [Indexed: 11/22/2022] Open
Abstract
Background and Objectives To explore the lung-protective effect of levosimendan (LS) during cardiopulmonary bypass in a canine model by determining the wet/dry weight (W/D) ratio of lung tissue, malonaldehyde (MDA) and superoxide dismutase (SOD) concentrations, and performing a histological evaluation. Materials and Methods Thirty-two canines were divided randomly into four groups and underwent a routine aortic cross-clamping cardiopulmonary bypass procedure for 1 h, followed by recovery for 2 h. Animals were handled as follows: group C (means control group), no special treatment after aortic cross clamping; group P (means pulmonary artery perfusion group), pulmonary artery perfusion with cold oxygenated blood after aortic cross clamping; group LSIV (means intravenous injection of LS group), intravenous injection of LS (65 µg/kg) before thoracotomy, and the rest of the procedure was identical to the control group; group LPS (means pulmonary perfusion with LS group), pulmonary perfusion with cold oxygenated blood combined with LS (65 µg/kg) after aortic cross clamping. Lung tissues were removed and subjected to evaluation of pathological alterations, W/D ratio and MDA and SOD concentrations. Results In group C, the W/D ratio and MDA concentration were higher, while the SOD concentrations were lower (p<0.05). Compared with groups P and LSIV, the MDA concentration was lower in group LPS, while that of SOD was higher (p<0.05); Light and electron microscopy indicated that LS intervention reduced impairment of lung tissues. Conclusion Our findings suggest that LS plays an important role in protecting lung tissues.
Collapse
Affiliation(s)
- Junbo Feng
- The First Ward of Cardiovascular Surgery Department of the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Haiyuan Liu
- The Oncology Department of the Hefei Second People's Hospital, Hefei, China
| | - Jiayi Chen
- The First Ward of Cardiovascular Surgery Department of the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jiyuan Wang
- The First Ward of Cardiovascular Surgery Department of the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zhuang Liu
- The First Ward of Cardiovascular Surgery Department of the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Shenglin Ge
- The First Ward of Cardiovascular Surgery Department of the First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
10
|
Okada M, Yamawaki H. Levosimendan inhibits interleukin-1β-induced apoptosis through activation of Akt and inhibition of inducible nitric oxide synthase in rat cardiac fibroblasts. Eur J Pharmacol 2015; 769:86-92. [DOI: 10.1016/j.ejphar.2015.10.056] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 10/23/2015] [Accepted: 10/30/2015] [Indexed: 10/22/2022]
|
11
|
Angelis D, Fontánez Nieves TD, Delivoria-Papadopoulos M. Temporal Changes in Caspase-1 and Caspase-8 Activities Following Brain Hypoxia With and Without Src kinase Inhibition in a Piglet Animal Model. Neurochem Res 2015; 40:2270-9. [PMID: 26342830 DOI: 10.1007/s11064-015-1717-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2015] [Revised: 08/08/2015] [Accepted: 09/01/2015] [Indexed: 12/16/2022]
Abstract
The Src family kinases are a family of intracellular, non-receptor tyrosine kinases that are involved in a variety of cellular functions including the regulation of inflammation and apoptosis after brain hypoxia. Caspase-1 (C1) activates IL-1β through the formation of complex structures, the inflammasomes, while caspase-8 (C8) is part of the extrinsic apoptotic pathway. C8 has been found to directly activate the production of IL-1β. Previously, we observed that C1 and IL-1β are increased in the acute phase after hypoxia in the brain of piglets, but they follow a different pattern long term, with C1 remaining activated throughout the period of observation, while IL-1β returning to baseline at 15 days. Src kinase inhibition ameliorated the activation of C1 and IL-1β early, but did not appear to have any effect long term. Prompted by these findings, we assessed the changes that occur over time (1 h and 15 days) in C1 and C8 activities after brain hypoxia as well as the effect of pretreatment with a Src kinase inhibitor, PP2 on these biochemical markers. Enzymatic activities were determined by spectrophotometry with measurements of C1 and C8 in each cytosolic brain sample (N = 4 in each group). We found that C1 and C8 activities increase in the acute phase following hypoxia in the brain of newborn piglets, with C8 relatively more than C1 (C8/C1 ratio increased from 2:1 as baseline to 3:1 in hypoxia). Fifteen days after hypoxia C8/C1 ratio decreased to about 1:1. In piglets that were pretreated with a Src kinase selective inhibitor (PP2) and then subjected to hypoxia, the C8/C1 ratio early increase was not observed. Immediately after hypoxia C8 and C1 follow a similar pattern of increase while long term this appears to dissociate. We propose that following this experimental methodology, the previously observed IL-1β production after hypoxia might be associated with C8 rather than C1 and that Src kinase is involved in the above process.
Collapse
Affiliation(s)
- Dimitrios Angelis
- Division of Neonatology, Department of Pediatrics, Texas Tech University Health Sciences Center, Odessa, TX, 79763, USA.
- Department of Pediatrics, Drexel University and St. Christopher's Hospital for Children, Philadelphia, PA, USA.
| | - Tania D Fontánez Nieves
- Department of Pediatrics, Miami Miller School of Medicine, Jackson Memorial Hospital, Miami, FL, USA
- Department of Pediatrics, Drexel University and St. Christopher's Hospital for Children, Philadelphia, PA, USA
| | | |
Collapse
|
12
|
Abstract
Acute heart failure (AHF) emerges as a major and growing epidemiological concern with high morbidity and mortality rates. Current therapies in patients with acute heart failure rely on different strategies. Patients with hypotension, hypoperfusion, or shock require inotropic support, whereas diuretics and vasodilators are recommended in patients with systemic or pulmonary congestion. Traditionally inotropic agents, referred to as Ca2+ mobilizers load the cardiomyocyte with Ca2+ and thereby increase oxygen consumption and risk for arrhythmias. These limitations of traditional inotropes may be avoided by sarcomere targeted agents. Direct activation of the cardiac sarcomere may be achieved by either sensitizing the cardiac myofilaments to Ca2+ or activating directly the cardiac myosin. In this review, we focus on sarcomere targeted inotropic agents, emphasizing their mechanisms of action and overview the most relevant clinical considerations.
Collapse
|
13
|
Kumar A, Jaggi AS, Singh N. Pharmacology of Src family kinases and therapeutic implications of their modulators. Fundam Clin Pharmacol 2015; 29:115-30. [PMID: 25545125 DOI: 10.1111/fcp.12097] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Revised: 11/18/2014] [Accepted: 12/02/2014] [Indexed: 12/23/2022]
Abstract
Src family kinases (SFKs), the largest family of nonreceptor tyrosine kinases, include 10 members. Src was the first gene product discovered to have intrinsic protein tyrosine kinase activity. Src is widely expressed in many cell types and can have different locations within a cell; the subcellular location of Src can affect its function. Src can associate with cellular membranes, such as the plasma membrane, the perinuclear membrane, and the endosomal membrane. SFKs actions on mammalian cells are pleiotropic and include effect on cell morphology, adhesion, migration, invasion, proliferation, differentiation, and survival. SFKs at one end have been documented to play some important physiological functions; on the other end, they have been described in the pathophysiology of some disorders. In this review article, an exhaustive attempt has been made to unearth pharmacology of SFKs and therapeutic implications of SFKs modulators.
Collapse
Affiliation(s)
- Amit Kumar
- CNS and CVS Research Laboratory, Pharmacology Division, Department of Pharmaceutical Sciences and Drug Research, Faculty of Medicine, Punjabi University, Patiala, 147002, Punjab, India
| | | | | |
Collapse
|
14
|
Aceros H, Farah G, Noiseux N, Mukaddam-Daher S. Moxonidine modulates cytokine signalling and effects on cardiac cell viability. Eur J Pharmacol 2014; 740:168-182. [PMID: 25036265 DOI: 10.1016/j.ejphar.2014.06.047] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Revised: 06/23/2014] [Accepted: 06/25/2014] [Indexed: 10/25/2022]
Abstract
Regression of left ventricular hypertrophy and improved cardiac function in SHR by the centrally acting imidazoline I1-receptor agonist, moxonidine, are associated with differential actions on circulating and cardiac cytokines. Herein, we investigated cell-type specific I1-receptor (also known as nischarin) signalling and the mechanisms through which moxonidine may interfere with cytokines to affect cardiac cell viability. Studies were performed on neonatal rat cardiomyocytes and fibroblasts incubated with interleukin (IL)-1β (5 ng/ml), tumor necrosis factor (TNF)-α (10 ng/ml), and moxonidine (10(-7) and 10(-5) M), separately and in combination, for 15 min, and 24 and 48 h for the measurement of MAPKs (ERK1/2, JNK, and p38) and Akt activation and inducible NOS (iNOS) expression, by Western blotting, and cardiac cell viability/proliferation and apoptosis by flow cytometry, MTT assay, and Live/Dead assay. Participation of imidazoline I1-receptors and the signalling proteins in the detected effects was identified using imidazoline I1-receptor antagonist and signalling protein inhibitors. The results show that IL-1β, and to a lower extent, TNF-α, causes cell death and that moxonidine protects against starvation- as well as IL-1β -induced mortality, mainly by maintaining membrane integrity, and in part, by improving mitochondrial activity. The protection involves activation of Akt, ERK1/2, p38, JNK, and iNOS. In contrast, moxonidine stimulates basal and IL-1β-induced fibroblast mortality by mechanisms that include inhibition of JNK and iNOS. Thus, apart from their actions on the central nervous system, imidazoline I1-receptors are directly involved in cardiac cell growth and death, and may play an important role in cardiovascular diseases associated with inflammation.
Collapse
Affiliation(s)
- Henry Aceros
- Centre Hospitalier de L'Université de Montréal Research Center (CRCHUM), Montreal, Québec, Canada; Department of Pharmacology, Université de Montréal, Montreal, Québec, Canada
| | - Georges Farah
- Centre Hospitalier de L'Université de Montréal Research Center (CRCHUM), Montreal, Québec, Canada; Department of Pharmacology, Université de Montréal, Montreal, Québec, Canada
| | - Nicolas Noiseux
- Centre Hospitalier de L'Université de Montréal Research Center (CRCHUM), Montreal, Québec, Canada; Department of Medicine, Université de Montréal, Montreal, Québec, Canada
| | - Suhayla Mukaddam-Daher
- Centre Hospitalier de L'Université de Montréal Research Center (CRCHUM), Montreal, Québec, Canada; Department of Pharmacology, Université de Montréal, Montreal, Québec, Canada; Department of Medicine, Université de Montréal, Montreal, Québec, Canada.
| |
Collapse
|
15
|
Role of soluble adenylyl cyclase in cell death and growth. Biochim Biophys Acta Mol Basis Dis 2014; 1842:2646-55. [PMID: 25010002 DOI: 10.1016/j.bbadis.2014.06.034] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 06/26/2014] [Accepted: 06/27/2014] [Indexed: 12/13/2022]
Abstract
cAMP signaling is an evolutionarily conserved intracellular communication system controlling numerous cellular functions. Until recently, transmembrane adenylyl cyclase (tmAC) was considered the major source for cAMP in the cell, and the role of cAMP signaling was therefore attributed exclusively to the activity of this family of enzymes. However, increasing evidence demonstrates the role of an alternative, intracellular source of cAMP produced by type 10 soluble adenylyl cyclase (sAC). In contrast to tmAC, sAC produces cAMP in various intracellular microdomains close to specific cAMP targets, e.g., in nucleus and mitochondria. Ongoing research demonstrates involvement of sAC in diverse physiological and pathological processes. The present review is focused on the role of cAMP signaling, particularly that of sAC, in cell death and growth. Although the contributions of sAC to the regulation of these cellular functions have only recently been discovered, current data suggest that sAC plays key roles in mitochondrial bioenergetics and the mitochondrial apoptosis pathway, as well as cell proliferation and development. Furthermore, recent reports suggest the importance of sAC in several pathologies associated with apoptosis as well as in oncogenesis. This article is part of a Special Issue entitled: The role of soluble adenylyl cyclase in health and disease.
Collapse
|
16
|
Abstract
The field of mitochondrial ion channels has recently seen substantial progress, including the molecular identification of some of the channels. An integrative approach using genetics, electrophysiology, pharmacology, and cell biology to clarify the roles of these channels has thus become possible. It is by now clear that many of these channels are important for energy supply by the mitochondria and have a major impact on the fate of the entire cell as well. The purpose of this review is to provide an up-to-date overview of the electrophysiological properties, molecular identity, and pathophysiological functions of the mitochondrial ion channels studied so far and to highlight possible therapeutic perspectives based on current information.
Collapse
|
17
|
Erb J, Beutlhauser T, Feldheiser A, Schuster B, Treskatsch S, Grubitzsch H, Spies C. Influence of levosimendan on organ dysfunction in patients with severely reduced left ventricular function undergoing cardiac surgery. J Int Med Res 2014; 42:750-64. [PMID: 24781725 DOI: 10.1177/0300060513516293] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Accepted: 11/19/2013] [Indexed: 11/16/2022] Open
Abstract
OBJECTIVES Levosimendan is an inotropic drug with organ-protective properties due to its activation of mitochondrial K(ATP) channels. This prospective, randomized, double-blind, placebo-controlled study investigated whether administration of levosimendan prior to cardiopulmonary bypass could reduce organ dysfunction and influence subsequent secondary endpoints. PATIENTS AND METHODS Patients with left ventricular ejection fraction <30% scheduled for elective coronary artery bypass surgery (with or without valve surgery) received either levosimendan (12.5 mg, 0.1 µg kg(-1) per min; n = 17) or placebo (n = 16) central venous infusion, immediately after anaesthesia induction, as add-on medication to a goal-orientated treatment algorithm. RESULTS A total of 33 patients completed the study. There were no statistically significant differences in Sequential Organ Failure Assessment scores, survival, haemodynamic parameters, time to extubation, time in intensive care unit, need for haemodialysis or health-related quality-of-life at 6 months post operation. The levosimendan group compared with the placebo group had significantly lower use of epinephrine (35% versus 81%) and nitroglycerine (6% versus 44%) 24 h postoperation, and significantly less frequent serious adverse events (13% versus 47%). CONCLUSIONS These preliminary results show that timely perioperative levosimendan treatment is feasible, has a favourable safety profile safe and may help to prevent low cardiac output syndrome. However, organ function was not preserved. Further studies, using larger sample sizes, are required.
Collapse
Affiliation(s)
- Joachim Erb
- Department of Anaesthesia, Surgical Intensive Care, Prehospital Emergency Medicine and Pain Therapy, University Hospital of Basel, Basel, Switzerland
| | - Torsten Beutlhauser
- Department of Anaesthesiology and Intensive Care Medicine, Campus Charité Mitte and Campus Virchow-Klinikum, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Aarne Feldheiser
- Department of Anaesthesiology and Intensive Care Medicine, Campus Charité Mitte and Campus Virchow-Klinikum, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Birgit Schuster
- Department of Anaesthesiology and Intensive Care Medicine, Campus Charité Mitte and Campus Virchow-Klinikum, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Sascha Treskatsch
- Department of Anaesthesiology and Intensive Care Medicine, Campus Charité Mitte and Campus Virchow-Klinikum, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Herko Grubitzsch
- Department of Cardiovascular Surgery, Campus Charité Mitte, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Claudia Spies
- Department of Anaesthesiology and Intensive Care Medicine, Campus Charité Mitte and Campus Virchow-Klinikum, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
18
|
Kolseth SM, Rolim NPL, Salvesen Ø, Nordhaug DO, Wahba A, Høydal MA. Levosimendan improves contractility in vivo and in vitro in a rodent model of post-myocardial infarction heart failure. Acta Physiol (Oxf) 2014; 210:865-74. [PMID: 24495280 DOI: 10.1111/apha.12248] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Revised: 11/18/2013] [Accepted: 01/30/2014] [Indexed: 01/09/2023]
Abstract
AIM As few studies have presented a thorough analysis of the effect of levosimendan (LEV) on contractility, our purpose was to investigate in vivo cardiac function as well as in vitro cardiomyocyte function and calcium (Ca(2+) ) handling following LEV treatment. METHODS Rats with post-myocardial infarction heart failure (HF) induced by ligation of the left anterior descending coronary artery and sham-operated animals were randomized to the infusion of LEV (2.4 μg kg(-1) min(-1) ) or vehicle for 40 min. Echocardiographic examination was coupled to pressure-volume sampling in the left ventricle before (B) and after (40 min) infusion. Isolated left ventricular cardiomyocytes were studied in an epifluorescence microscope. RESULTS HF LEV (n = 6), HF vehicle (n = 7), sham LEV (n = 5) and sham vehicle (n = 6) animals were included. LEV infusion compared to vehicle in HF animals reduced left ventricular end-diastolic pressure and mean arterial pressure (both P < 0.001) and improved the slope of the preload-recruitable stroke work (P < 0.05). Administrating LEV to HF cardiomyocytes in vitro improved fractional shortening and Ca(2+) sensitivity index ratio, and increased the diastolic Ca(2+) (all P < 0.01). CONCLUSION In HF animals, LEV improved the contractility by increasing the Ca(2+) sensitivity. Furthermore loading conditions were changed, and LEV could consequently change organ perfusion. An observed increase in diastolic Ca(2+) following LEV treatment and clinical implications of this should be further addressed.
Collapse
Affiliation(s)
- S. M. Kolseth
- Department of Circulation and Medical Imaging; Norwegian University of Science and Technology; Trondheim Norway
| | - N. P. L. Rolim
- Department of Circulation and Medical Imaging; Norwegian University of Science and Technology; Trondheim Norway
- K.G. Jebsen Center of Exercise in Medicine; Trondheim Norway
| | - Ø. Salvesen
- Department of Circulation and Medical Imaging; Norwegian University of Science and Technology; Trondheim Norway
| | - D. O. Nordhaug
- Department of Circulation and Medical Imaging; Norwegian University of Science and Technology; Trondheim Norway
- Department of Cardiothoracic Surgery; St Olav's University Hospital; Trondheim Norway
| | - A. Wahba
- Department of Circulation and Medical Imaging; Norwegian University of Science and Technology; Trondheim Norway
- K.G. Jebsen Center of Exercise in Medicine; Trondheim Norway
- Department of Cardiothoracic Surgery; St Olav's University Hospital; Trondheim Norway
| | - M. A. Høydal
- Department of Circulation and Medical Imaging; Norwegian University of Science and Technology; Trondheim Norway
- K.G. Jebsen Center of Exercise in Medicine; Trondheim Norway
- Norwegian Council on Cardiovascular Disease; Trondheim Norway
| |
Collapse
|
19
|
Kumar A, Jaggi AS, Singh N. Pharmacological investigations on possible role of Src kinases in neuroprotective mechanism of ischemic postconditioning in mice. Int J Neurosci 2014; 124:777-86. [DOI: 10.3109/00207454.2013.879869] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|