1
|
Maslov LN, Popov SV, Naryzhnaya NV, Mukhomedzyanov AV, Kurbatov BK, Derkachev IA, Boshchenko AA, Prasad NR, Ma H, Zhang Y, Sufianova GZ, Fu F, Pei JM. K ATP channels are regulators of programmed cell death and targets for the creation of novel drugs against ischemia/reperfusion cardiac injury. Fundam Clin Pharmacol 2023; 37:1020-1049. [PMID: 37218378 DOI: 10.1111/fcp.12924] [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] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 04/29/2023] [Accepted: 05/19/2023] [Indexed: 05/24/2023]
Abstract
BACKGROUND The use of percutaneous coronary intervention (PCI) in patients with ST-segment elevation myocardial infarction (STEMI) is associated with a mortality rate of 5%-7%. It is clear that there is an urgent need to develop new drugs that can effectively prevent cardiac reperfusion injury. ATP-sensitive K+ (KATP ) channel openers (KCOs) can be classified as such drugs. RESULTS KCOs prevent irreversible ischemia and reperfusion injury of the heart. KATP channel opening promotes inhibition of apoptosis, necroptosis, pyroptosis, and stimulation of autophagy. KCOs prevent the development of cardiac adverse remodeling and improve cardiac contractility in reperfusion. KCOs exhibit antiarrhythmic properties and prevent the appearance of the no-reflow phenomenon in animals with coronary artery occlusion and reperfusion. Diabetes mellitus and a cholesterol-enriched diet abolish the cardioprotective effect of KCOs. Nicorandil, a KCO, attenuates major adverse cardiovascular event and the no-reflow phenomenon, reduces infarct size, and decreases the incidence of ventricular arrhythmias in patients with acute myocardial infarction. CONCLUSION The cardioprotective effect of KCOs is mediated by the opening of mitochondrial KATP (mitoKATP ) and sarcolemmal KATP (sarcKATP ) channels, triggered free radicals' production, and kinase activation.
Collapse
Affiliation(s)
- Leonid N Maslov
- Cardiology Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russia
| | - Sergey V Popov
- Cardiology Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russia
| | - Natalia V Naryzhnaya
- Cardiology Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russia
| | - Alexandr V Mukhomedzyanov
- Cardiology Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russia
| | - Boris K Kurbatov
- Cardiology Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russia
| | - Ivan A Derkachev
- Cardiology Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russia
| | - Alla A Boshchenko
- Cardiology Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russia
| | - N Rajendra Prasad
- Department of Biochemistry and Biotechnology, Faculty of Science, Annamalai University, Annamalainagar, India
| | - Huijie Ma
- Department of Physiology, Hebei Medical University, Shijiazhuang, China
| | - Yi Zhang
- Department of Physiology, Hebei Medical University, Shijiazhuang, China
| | - Galina Z Sufianova
- Department of Pharmacology, Tyumen State Medical University, Tyumen, Russia
| | - Feng Fu
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Jian-Ming Pei
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
2
|
Gunata M, Parlakpinar H. Experimental heart failure models in small animals. Heart Fail Rev 2023; 28:533-554. [PMID: 36504404 DOI: 10.1007/s10741-022-10286-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/08/2022] [Indexed: 12/14/2022]
Abstract
Heart failure (HF) is one of the most critical health and economic burdens worldwide, and its prevalence is continuously increasing. HF is a disease that occurs due to a pathological change arising from the function or structure of the heart tissue and usually progresses. Numerous experimental HF models have been created to elucidate the pathophysiological mechanisms that cause HF. An understanding of the pathophysiology of HF is essential for the development of novel efficient therapies. During the past few decades, animal models have provided new insights into the complex pathogenesis of HF. Success in the pathophysiology and treatment of HF has been achieved by using animal models of HF. The development of new in vivo models is critical for evaluating treatments such as gene therapy, mechanical devices, and new surgical approaches. However, each animal model has advantages and limitations, and none of these models is suitable for studying all aspects of HF. Therefore, the researchers have to choose an appropriate experimental model that will fully reflect HF. Despite some limitations, these animal models provided a significant advance in the etiology and pathogenesis of HF. Also, experimental HF models have led to the development of new treatments. In this review, we discussed widely used experimental HF models that continue to provide critical information for HF patients and facilitate the development of new treatment strategies.
Collapse
Affiliation(s)
- Mehmet Gunata
- Department of Medical Pharmacology, Faculty of Medicine, Inonu University, Malatya, 44280, Türkiye
| | - Hakan Parlakpinar
- Department of Medical Pharmacology, Faculty of Medicine, Inonu University, Malatya, 44280, Türkiye.
| |
Collapse
|
3
|
Harvey AP, Robinson E, Edgar KS, McMullan R, O’Neill KM, Alderdice M, Amirkhah R, Dunne PD, McDermott BJ, Grieve DJ. Downregulation of PPARα during Experimental Left Ventricular Hypertrophy Is Critically Dependent on Nox2 NADPH Oxidase Signalling. Int J Mol Sci 2020; 21:E4406. [PMID: 32575797 PMCID: PMC7352162 DOI: 10.3390/ijms21124406] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 06/17/2020] [Accepted: 06/18/2020] [Indexed: 12/31/2022] Open
Abstract
Pressure overload-induced left ventricular hypertrophy (LVH) is initially adaptive but ultimately promotes systolic dysfunction and chronic heart failure. Whilst underlying pathways are incompletely understood, increased reactive oxygen species generation from Nox2 NADPH oxidases, and metabolic remodelling, largely driven by PPARα downregulation, are separately implicated. Here, we investigated interaction between the two as a key regulator of LVH using in vitro, in vivo and transcriptomic approaches. Phenylephrine-induced H9c2 cardiomyoblast hypertrophy was associated with reduced PPARα expression and increased Nox2 expression and activity. Pressure overload-induced LVH and systolic dysfunction induced in wild-type mice by transverse aortic constriction (TAC) for 7 days, in association with Nox2 upregulation and PPARα downregulation, was enhanced in PPARα-/- mice and prevented in Nox2-/- mice. Detailed transcriptomic analysis revealed significantly altered expression of genes relating to PPARα, oxidative stress and hypertrophy pathways in wild-type hearts, which were unaltered in Nox2-/- hearts, whilst oxidative stress pathways remained dysregulated in PPARα-/- hearts following TAC. Network analysis indicated that Nox2 was essential for PPARα downregulation in this setting and identified preferential inflammatory pathway modulation and candidate cytokines as upstream Nox2-sensitive regulators of PPARα signalling. Together, these data suggest that Nox2 is a critical driver of PPARα downregulation leading to maladaptive LVH.
Collapse
Affiliation(s)
- Adam P. Harvey
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast BT9 7AE, UK; (A.P.H.); (E.R.); (K.S.E.); (R.M.); (K.M.O.); (B.J.M.)
| | - Emma Robinson
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast BT9 7AE, UK; (A.P.H.); (E.R.); (K.S.E.); (R.M.); (K.M.O.); (B.J.M.)
| | - Kevin S. Edgar
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast BT9 7AE, UK; (A.P.H.); (E.R.); (K.S.E.); (R.M.); (K.M.O.); (B.J.M.)
| | - Ross McMullan
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast BT9 7AE, UK; (A.P.H.); (E.R.); (K.S.E.); (R.M.); (K.M.O.); (B.J.M.)
| | - Karla M. O’Neill
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast BT9 7AE, UK; (A.P.H.); (E.R.); (K.S.E.); (R.M.); (K.M.O.); (B.J.M.)
| | - Matthew Alderdice
- Patrick G Johnston Centre for Cancer Research, Queen’s University Belfast, Belfast BT7 1NN, UK; (M.A.); (R.A.); (P.D.D.)
| | - Raheleh Amirkhah
- Patrick G Johnston Centre for Cancer Research, Queen’s University Belfast, Belfast BT7 1NN, UK; (M.A.); (R.A.); (P.D.D.)
| | - Philip D. Dunne
- Patrick G Johnston Centre for Cancer Research, Queen’s University Belfast, Belfast BT7 1NN, UK; (M.A.); (R.A.); (P.D.D.)
| | - Barbara J. McDermott
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast BT9 7AE, UK; (A.P.H.); (E.R.); (K.S.E.); (R.M.); (K.M.O.); (B.J.M.)
| | - David J. Grieve
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast BT9 7AE, UK; (A.P.H.); (E.R.); (K.S.E.); (R.M.); (K.M.O.); (B.J.M.)
| |
Collapse
|
4
|
Gomes AC, Falcão-Pires I, Pires AL, Brás-Silva C, Leite-Moreira AF. Rodent models of heart failure: an updated review. Heart Fail Rev 2013; 18:219-49. [PMID: 22446984 DOI: 10.1007/s10741-012-9305-3] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Heart failure (HF) is one of the major health and economic burdens worldwide, and its prevalence is continuously increasing. The study of HF requires reliable animal models to study the chronic changes and pharmacologic interventions in myocardial structure and function and to follow its progression toward HF. Indeed, during the past 40 years, basic and translational scientists have used small animal models to understand the pathophysiology of HF and find more efficient ways of preventing and managing patients suffering from congestive HF (CHF). Each species and each animal model has advantages and disadvantages, and the choice of one model over another should take them into account for a good experimental design. The aim of this review is to describe and highlight the advantages and drawbacks of some commonly used HF rodents models, including both non-genetically and genetically engineered models, with a specific subchapter concerning diastolic HF models.
Collapse
Affiliation(s)
- A C Gomes
- Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319, Porto, Portugal
| | | | | | | | | |
Collapse
|
5
|
Antiadrenergic effect of adenosine involves connexin 43 turn-over in H9c2 cells. Eur J Pharmacol 2013; 715:56-61. [DOI: 10.1016/j.ejphar.2013.06.019] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Revised: 06/05/2013] [Accepted: 06/19/2013] [Indexed: 01/06/2023]
|
6
|
Cannavo A, Rengo G, Liccardo D, Pagano G, Zincarelli C, De Angelis MC, Puglia R, Di Pietro E, Rabinowitz JE, Barone MV, Cirillo P, Trimarco B, Palmer TM, Ferrara N, Koch WJ, Leosco D, Rapacciuolo A. β1-adrenergic receptor and sphingosine-1-phosphate receptor 1 (S1PR1) reciprocal downregulation influences cardiac hypertrophic response and progression to heart failure: protective role of S1PR1 cardiac gene therapy. Circulation 2013; 128:1612-22. [PMID: 23969695 DOI: 10.1161/circulationaha.113.002659] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND The sphingosine-1-phosphate receptor 1 (S1PR1) and β1-adrenergic receptor (β1AR) are G-protein-coupled receptors expressed in the heart. These 2 receptors have opposing actions on adenylyl cyclase because of differential G-protein coupling. Importantly, both of these receptors can be regulated by the actions of G-protein-coupled receptor kinase-2, which triggers desensitization and downregulation processes. Although classic signaling paradigms suggest that simultaneous activation of β1ARs and S1PR1s in a myocyte would simply result in opposing action on cAMP production, in this report we have uncovered a direct interaction between these 2 receptors, with regulatory involvement of G-protein-coupled receptor kinase-2. METHODS AND RESULTS In HEK (human embryonic kidney) 293 cells overexpressing both β1AR and S1PR1, we demonstrated that β1AR downregulation can occur after stimulation with sphingosine-1-phosphate (an S1PR1 agonist), whereas S1PR1 downregulation can be triggered by isoproterenol (a β-adrenergic receptor agonist) treatment. This cross talk between these 2 distinct G-protein-coupled receptors appears to have physiological significance, because they interact and show reciprocal regulation in mouse hearts undergoing chronic β-adrenergic receptor stimulation and in a rat model of postischemic heart failure. CONCLUSIONS We demonstrate that restoration of cardiac plasma membrane levels of S1PR1 produces beneficial effects that counterbalance the deleterious β1AR overstimulation in heart failure.
Collapse
Affiliation(s)
- Alessandro Cannavo
- Division of Geriatrics, Department of Translational Medical Sciences (A.C., G.R., D.L., G.P., N.F., D.L.), Department of Pediatrics and European Laboratory for the Investigation of Food-Induced Diseases (M.V.B.), and Division of Cardiology, Department of Advanced Biomedical Sciences (M.C.D.A., R.P., E.D.P., P.C., B.T., A.R.), Federico II University, Naples, Italy; Center of Translational Medicine, Temple University, Philadelphia, PA (A.C., J.E.R., W.J.K.); Division of Cardiology, Salvatore Maugeri Foundation, IRCCS, Scientific Institute of Telese Terme (BN), Italy (G.R., C.Z., N.F.); and the Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (T.M.P.)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Early growth response 1 is an early signal inducing Cav3.2 T-type calcium channels during cardiac hypertrophy. Cardiovasc Res 2013; 100:222-30. [DOI: 10.1093/cvr/cvt190] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
|
8
|
Kamolonol suppresses angiotensin II-induced stress fiber formation and cellular hypertrophy through inhibition of Rho-associated kinase 2 activity. Biochem Biophys Res Commun 2013; 438:318-23. [PMID: 23891689 DOI: 10.1016/j.bbrc.2013.07.069] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2013] [Accepted: 07/17/2013] [Indexed: 01/07/2023]
Abstract
Kamolonol (7-[[(1R,2R,4R,4aS,5R,8aS)-4-hydroxy-1,2,4a,5-tetramethyl-6-oxo-3,4,5,7,8,8a-hexahydro-2H-naphthalen-1-yl]methoxy]chromen-2-one) is a sesquiterpene coumarin and an active component of gum extracts from Ferulaassafoetida. The aim of this study was to investigate the anti-fibrotic and anti-cellular hypertrophic effects of kamolonol, and further to explore its possible mechanism. Kamolonol (3-30μM) significantly inhibited stress fiber formation induced by angiotensin II (Ang II) in rat heart-derived H9c2 cells. Furthermore, kamolonol (3-30μM) showed a potent inhibitory effect on Ang II-induced cellular hypertrophy in H9c2 cells. Next, a Rho-associated kinase (ROCK) activity was measured because actin stress fiber formation and/or cellular hypertrophy are usually induced by the activation of ROCK. Rho-associated kinase 2 (ROCK2) studies using a time-resolved fluorescence resonance energy transfer (TR-FRET) showed that kamolonol possesses a potent ROCK2 inhibitory activity with IC50 values of 2.27μM, and has an ATP-competitive inhibitory mode. In validation study, pretreatment of kamolonol (3-30μM) for 2h decreased the Ang II-induced phosphorylation of myosin phosphatase 1 (MYPT1) and myosin light chain 2 (MLC2). Taken together, these results indicate that kamolonol suppresses Ang II-induced stress fiber formation and cellular hypertrophy, and propose that one mechanism underlying these anti-fibrotic and anti-cellular hypertrophic effects involves inhibition of the ROCK-MLC pathway.
Collapse
|
9
|
Cardiovascular effects of a novel selective Rho kinase inhibitor, 2-(1H-indazole-5-yl)amino-4-methoxy-6-piperazino triazine (DW1865). Eur J Pharmacol 2013; 702:218-26. [PMID: 23376156 DOI: 10.1016/j.ejphar.2013.01.027] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2012] [Revised: 01/08/2013] [Accepted: 01/16/2013] [Indexed: 11/23/2022]
Abstract
The arising critical implications of Rho kinase signaling in cardiovascular diseases have been attracting attention in the pharmacological potential of Rho kinase inhibitors. We identified a novel inhibitor of Rho kinase (2-(1H-indazole-5-yl)amino-4-methoxy-6-piperazino triazine; DW 1865) and characterized its effects in biochemical, cellular, tissue and animal based assays. DW 1865 potently inhibited the kinase activity of both Rho kinase 1 and Rho kinase 2 in vitro, and behaved as an ATP-competitive inhibitor. Interestingly, DW1865 was 10 times more potent in inhibiting Rho kinase activities than fasudil as a selective Rho kinase inhibitor. The activity of DW1865 was shown to be highly selective for Rho kinase in the panel assay of 13 other kinases. In the isolated vascular tissue study, DW1865 exerted vasorelaxation in phenylephrine- or 5-hydroxytriptamine-induced contraction in a concentration-dependent manner manner. In spontaneously hypertensive rats, administration of DW1865 caused a significant and dose-related reduction in blood pressure. Furthermore, DW1865 blocked angiotensin II-induced stress fiber formation and cellular hypertrophy in rat heart-derived H9c2 cells. Taken together, these results suggest that DW1865 is a highly selective and potent Rho kinase inhibitor that will alleviate the pathophysiological actions of Rho kinase such as stress fiber formation, cellular hypertrophy, and hypertension.
Collapse
|
10
|
Hirose M, Takeishi Y, Nakada T, Shimojo H, Kashihara T, Nishio A, Suzuki S, Mende U, Matsumoto K, Matsushita N, Taira E, Sato F, Yamada M. Nicorandil prevents Gαq-induced progressive heart failure and ventricular arrhythmias in transgenic mice. PLoS One 2012; 7:e52667. [PMID: 23285142 PMCID: PMC3527603 DOI: 10.1371/journal.pone.0052667] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2011] [Accepted: 11/19/2012] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Beneficial effects of nicorandil on the treatment of hypertensive heart failure (HF) and ischemic heart disease have been suggested. However, whether nicorandil has inhibitory effects on HF and ventricular arrhythmias caused by the activation of G protein alpha q (Gα(q)) -coupled receptor (GPCR) signaling still remains unknown. We investigated these inhibitory effects of nicorandil in transgenic mice with transient cardiac expression of activated Gα(q) (Gα(q)-TG). METHODOLOGY/PRINCIPAL FINDINGS Nicorandil (6 mg/kg/day) or vehicle was chronically administered to Gα(q)-TG from 8 to 32 weeks of age, and all experiments were performed in mice at the age of 32 weeks. Chronic nicorandil administration prevented the severe reduction of left ventricular fractional shortening and inhibited ventricular interstitial fibrosis in Gα(q)-TG. SUR-2B and SERCA2 gene expression was decreased in vehicle-treated Gα(q)-TG but not in nicorandil-treated Gα(q)-TG. eNOS gene expression was also increased in nicorandil-treated Gα(q)-TG compared with vehicle-treated Gα(q)-TG. Electrocardiogram demonstrated that premature ventricular contraction (PVC) was frequently (more than 20 beats/min) observed in 7 of 10 vehicle-treated Gα(q)-TG but in none of 10 nicorandil-treated Gα(q)-TG. The QT interval was significantly shorter in nicorandil-treated Gα(q)-TG than vehicle-treated Gα(q)-TG. Acute nicorandil administration shortened ventricular monophasic action potential duration and reduced the number of PVCs in Langendorff-perfused Gα(q)-TG mouse hearts. Moreover, HMR1098, a blocker of cardiac sarcolemmal K(ATP) channels, significantly attenuated the shortening of MAP duration induced by nicorandil in the Gα(q)-TG heart. CONCLUSIONS/SIGNIFICANCE These findings suggest that nicorandil can prevent the development of HF and ventricular arrhythmia caused by the activation of GPCR signaling through the shortening of the QT interval, action potential duration, the normalization of SERCA2 gene expression. Nicorandil may also improve the impaired coronary circulation during HF.
Collapse
Affiliation(s)
- Masamichi Hirose
- Department of Molecular and Cellular Pharmacology, Iwate Medical University School of Pharmaceutical Sciences, Shiwa, Iwate, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Passariello CL, Gottardi D, Cetrullo S, Zini M, Campana G, Tantini B, Pignatti C, Flamigni F, Guarnieri C, Caldarera CM, Stefanelli C. Evidence that AMP-activated protein kinase can negatively modulate ornithine decarboxylase activity in cardiac myoblasts. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2011; 1823:800-7. [PMID: 22230191 DOI: 10.1016/j.bbamcr.2011.12.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2011] [Revised: 12/21/2011] [Accepted: 12/22/2011] [Indexed: 01/11/2023]
Abstract
The responses of AMP-activated protein kinase (AMPK) and Ornithine decarboxylase (ODC) to isoproterenol have been examined in H9c2 cardiomyoblasts, AMPK represents the link between cell growth and energy availability whereas ODC, the key enzyme in polyamine biosynthesis, is essential for all growth processes and it is thought to have a role in the development of cardiac hypertrophy. Isoproterenol rapidly induced ODC activity in H9c2 cardiomyoblasts by promoting the synthesis of the enzyme protein and this effect was counteracted by inhibitors of the PI3K/Akt pathway. The increase in enzyme activity became significant between 15 and 30min after the treatment. At the same time, isoproterenol stimulated the phosphorylation of AMPKα catalytic subunits (Thr172), that was associated to an increase in acetyl coenzyme A carboxylase (Ser72) phosphorylation. Downregulation of both α1 and α2 isoforms of the AMPK catalytic subunit by siRNA to knockdown AMPK enzymatic activity, led to superinduction of ODC in isoproterenol-treated cardiomyoblasts. Downregulation of AMPKα increased ODC activity even in cells treated with other adrenergic agonists and in control cells. Analogue results were obtained in SH-SY5Y neuroblastoma cells transfected with a shRNA construct against AMPKα. In conclusion, isoproterenol quickly activates in H9c2 cardiomyoblasts two events that seem to contrast one another. The first one, an increase in ODC activity, is linked to cell growth, whereas the second, AMPK activation, is a homeostatic mechanism that negatively modulates the first. The modulation of ODC activity by AMPK represents a mechanism that may contribute to control cell growth processes.
Collapse
|
12
|
ATP-Sensitive Potassium Channel Currents in Eccentrically Hypertrophied Cardiac Myocytes of Volume-Overloaded Rats. Int J Cell Biol 2011; 2011:838951. [PMID: 21845191 PMCID: PMC3154573 DOI: 10.1155/2011/838951] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2011] [Revised: 04/24/2011] [Accepted: 05/20/2011] [Indexed: 11/18/2022] Open
Abstract
ATP-sensitive potassium channels (K(ATP)) protect the myocardium from hypertrophy induced by pressure-overloading. In this study, we determined the effects of these channels in volume-overloading. We compared the effects of a K(ATP) agonist and a K(ATP) antagonist on sarcolemmal transmembrane current density (pA/pF) clamped at 20 mV increments of membrane potential from -80 to +40 mV in ventricular cardiac myocytes. The basal outward potassium pA/pF in myocytes of volume-overloaded animals was significantly smaller than that in the myocytes of sham-operated controls. Treatment of the control myocytes with the K(ATP) agonist cromakalim increased pA/pF significantly. This increase was blocked by the K(ATP) antagonist glibenclamide. Treatment of the hypertrophied myocytes from volume-overloaded animals with cromakalim and in the presence and absence of glibenclamide did not change pA/pF significantly. These findings suggest that eccentrically hypertrophied cardiac myocytes from volume-overloading may be unresponsive to specific activation/inactivation of K(ATP) and that dysfunctional K(ATP) may fail to protect the myocardium from left ventricular hypertrophy associated with volume-overloading.
Collapse
|
13
|
Passariello CL, Zini M, Nassi PA, Pignatti C, Stefanelli C. Upregulation of SIRT1 deacetylase in phenylephrine-treated cardiomyoblasts. Biochem Biophys Res Commun 2011; 407:512-6. [PMID: 21414296 DOI: 10.1016/j.bbrc.2011.03.049] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2011] [Accepted: 03/10/2011] [Indexed: 01/30/2023]
Abstract
The sirtuin SIRT1 is an ubiquitous NAD(+) dependent deacetylase that plays a role in biological processes such as longevity and stress response. In cardiac models, SIRT1 is associated to protection against many stresses. However, the link between SIRT1 and heart hypertrophy is complex and not fully understood. This study focuses specifically on the response of SIRT1 to the α-adrenergic agonist phenylephrine in H9c2 cardiac myoblasts, a cell model of cardiac hypertrophy. After 24 and 48h of phenylephrine treatment, SIRT1 expression and deacetylase activity were significantly increased. SIRT1 upregulation by phenylephrine was not associated to changes in NAD(+) levels, but was blocked by inhibitors of AMP-activated Protein Kinase (AMPK) or by AMPK knockdown by siRNA. When SIRT1 was inhibited with sirtinol or downregulated by siRNA, H9c2 cell viability was significantly decreased following phenylephrine treatment, showing that SIRT1 improves cell survival under hypertrophic stress. We so then propose that the increase in SIRT1 activity and expression in H9c2 cells treated with phenylephrine is an adaptive response to the hypertrophic stress, suggesting that adrenergic stimulation of heart cells activates hypertrophic programming and at the same time also promotes a self-protecting and self-regulating mechanism.
Collapse
|
14
|
Flores-Muñoz M, Smith NJ, Haggerty C, Milligan G, Nicklin SA. Angiotensin1-9 antagonises pro-hypertrophic signalling in cardiomyocytes via the angiotensin type 2 receptor. J Physiol 2010; 589:939-51. [PMID: 21173078 DOI: 10.1113/jphysiol.2010.203075] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The renin–angiotensin system (RAS) regulates blood pressure mainly via the actions of angiotensin (Ang)II, generated via angiotensin converting enzyme (ACE). The ACE homologue ACE2 metabolises AngII to Ang1-7, decreasing AngII and increasing Ang1-7, which counteracts AngII activity via the Mas receptor. However, ACE2 also converts AngI to Ang1-9, a poorly characterised peptide which can be further converted to Ang1-7 via ACE. Ang1-9 stimulates bradykinin release in endothelium and has antihypertrophic actions in the heart, attributed to its being a competitive inhibitor of ACE, leading to decreased AngII, rather than increased Ang1-7. To date no direct receptor-mediated effects of Ang1-9 have been described. To further understand the role of Ang1-9 in RAS function we assessed its action in cardiomyocyte hypertrophy in rat neonatal H9c2 and primary adult rabbit left ventricular cardiomyocytes, compared to Ang1-7. Cardiomyocyte hypertrophy was stimulated with AngII or vasopressin, significantly increasing cell size by approximately 1.2-fold (P < 0.05) as well as stimulating expression of the hypertrophy gene markers atrial natriuretic peptide, brain natriuretic peptide, β-myosin heavy chain and myosin light chain (2- to 5-fold, P < 0.05). Both Ang1-9 and Ang1-7 were able to block hypertrophy induced by either agonist (control, 186.4 μm; AngII, 232.8 μm; AngII+Ang1-7, 198.3 μm; AngII+Ang1-9, 195.9 μm; P < 0.05). The effects of Ang1-9 were not inhibited by captopril, supporting previous evidence that Ang1-9 acts independently of Ang1-7. Next, we investigated receptor signalling via angiotensin type 1 and type 2 receptors (AT1R, AT2R) and Mas. The AT1R antagonist losartan blocked AngII-induced, but not vasopressin-induced, hypertrophy. Losartan did not block the antihypertrophic effects of Ang1-9, or Ang1-7 on vasopressin-stimulated cardiomyocytes. The Mas antagonist A779 efficiently blocked the antihypertrophic effects of Ang1-7, without affecting Ang1-9. Furthermore, Ang1-7 activity was also inhibited in the presence of the bradykinin type 2 receptor antagonist HOE140, without affecting Ang1-9. Moreover, we observed that the AT2R antagonist PD123,319 abolished the antihypertrophic effects of Ang1-9, without affecting Ang1-7, suggesting Ang1-9 signals via the AT2R. Radioligand binding assays demonstrated that Ang1-9 was able to bind the AT2R (pKi = 6.28 ± 0.1). In summary, we ascribe a direct biological role for Ang1-9 acting via the AT2R. This has implications for RAS function and identifying new therapeutic targets in cardiovascular disease.
Collapse
Affiliation(s)
- M Flores-Muñoz
- Institute of Cardiovascular and Medical Sciences, BHF GCRC, University of Glasgow, 126 University Place, Glasgow G12 8TA, UK
| | | | | | | | | |
Collapse
|
15
|
Ichikawa H, Yagi H, Tanaka T, Cyong JC, Masaki T. Lagerstroemia speciosa extract inhibit TNF-induced activation of nuclear factor-kappaB in rat cardiomyocyte H9c2 cells. JOURNAL OF ETHNOPHARMACOLOGY 2010; 128:254-256. [PMID: 20045454 DOI: 10.1016/j.jep.2009.12.033] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2009] [Revised: 12/05/2009] [Accepted: 12/17/2009] [Indexed: 05/28/2023]
Abstract
AIM OF THE STUDY Lagerstroemia speciosa has been used as a folk medicine among people with diabetes in the Philippines. It is known to exhibit antidiabetic, antiobesity, and glucose transport activities through mechanisms not well defined. Diabetes leads to cardiomyocyte hypertrophy in association with an upregulation of vasoactive factors and activation of nuclear factor (NF)-kappaB and activating protein-1. We therefore investigated the effect of Lagerstroemia speciosa on the activation of NF-kappaB as a key mediator of cardiomyocyte hypertrophy, in rat cardiomyocyte H9c2 cells. MATERIALS AND METHODS Water extract of Lagerstroemia speciosa (Lythraceae family) was prepared. H9c2 cells were used for treatment of Lagerstroemia speciosa extract with/without tumor necrosis factor (TNF). To examine NF-kappaB's activation, we performed an electrophoretic mobility shift assay (EMSA). RESULTS The activation of NF-kappaB by TNF was completely blocked by a Lagerstroemia speciosa extract in a dose- and time-dependent manner in H9c2 cells. CONCLUSION Overall, our results indicate that Lagerstroemia speciosa can inhibit DNA-binding of NF-kappaB. This may explain its possible inhibition of diabetes-induced cardiomyocyte hypertrophy.
Collapse
Affiliation(s)
- Haruyo Ichikawa
- International Research and Educational Institute for Integrated Medical Sciences, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo 162-8666, Japan.
| | | | | | | | | |
Collapse
|
16
|
Calderone V, Testai L, Martelli A, Rapposelli S, Digiacomo M, Balsamo A, Breschi MC. Anti-ischemic properties of a new spiro-cyclic benzopyran activator of the cardiac mito-KATP channel. Biochem Pharmacol 2010; 79:39-47. [DOI: 10.1016/j.bcp.2009.07.017] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2009] [Revised: 07/14/2009] [Accepted: 07/27/2009] [Indexed: 11/16/2022]
|
17
|
Rokutan H, Anker SD, Springer J. In vivomodels of cardiac diseases: application to drug development and screening. Expert Opin Drug Discov 2009; 5:65-78. [DOI: 10.1517/17460440903460299] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
18
|
Guo WG, Su FF, Yuan LJ, Yang GD, Shi XQ, Li RY, Shu Q, Liu XT, Lu ZF, Zheng QS. Simvastatin inhibits angiotensin II-induced cardiac cell hypertrophy: role of Homer 1a. Clin Exp Pharmacol Physiol 2009; 37:40-5. [PMID: 19515066 DOI: 10.1111/j.1440-1681.2009.05221.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
1. The scaffolding protein Homer 1a is constitutively expressed in the myocardium, although its function in cardiomyocytes remains poorly understood. The aim of the present study was to investigate Homer 1a expression in hypertrophic cardiac cells and its role in angiotensin (Ang) II-induced cardiac hypertrophy. 2. After serum starvation for 24 h, cells were treated with 1 micromol/L simvastatin, 100 nmol/L angiotensin (Ang) II or their combination added to Dulbecco's modified Eagle's medium containing 0.5% serum. For combination treatment with AngII plus simvastatin, cells were exposed to simvastatin 12 h before the addition of AngII to the medium and cells were then incubated in the presence of both drugs for a further 24 h. Western blotting was used to determine Homer 1a protein expression. Hypertrophy was evaluated by determining the protein content per cell. 3. Homer 1a protein levels were upregulated following AngII-induced hypertrophy in H9C2 cells and neonatal rat cardiomyocytes, and these increases were augmented by simvastatin pretreatment. Concomitantly, simvastatin pretreatment inhibited extracellular signal-regulated kinase (ERK) 1/2 phosphorylation and AngII-induced hypertrophy. 4. The inhibitory effects of simvastatin against AngII-induced hypertrophy were attenuated by Homer 1a silencing, suggesting that simvastatin suppresses cardiac hypertrophy in a Homer 1a-dependent manner. Furthermore, AngII-induced hypertrophy and ERK1/2 phosphorylation in neonatal rat cardiomyocytes were significantly inhibited following the overexpression of Homer 1a using an adenovirus. 5. These results suggest a possible role for Homer 1a in inhibiting cardiac hypertrophy perhaps in part through inhibition of ERK1/2 activation.
Collapse
Affiliation(s)
- Wan-Gang Guo
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Jeong K, Kwon H, Min C, Pak Y. Modulation of the caveolin-3 localization to caveolae and STAT3 to mitochondria by catecholamine-induced cardiac hypertrophy in H9c2 cardiomyoblasts. Exp Mol Med 2009; 41:226-35. [PMID: 19299911 DOI: 10.3858/emm.2009.41.4.025] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
We investigated the effect of phenylephrine (PE)- and isoproterenol (ISO)-induced cardiac hypertrophy on subcellular localization and expression of caveolin-3 and STAT3 in H9c2 cardiomyoblast cells. Caveolin-3 localization to plasma membrane was attenuated and localization of caveolin-3 to caveolae in the plasma membrane was 24.3% reduced by the catecholamine- induced hypertrophy. STAT3 and phospho-STAT3 were up-regulated but verapamil and cyclosporin A synergistically decreased the STAT3 and phospho- STAT3 levels in PE- and ISO-induced hypertrophic cells. Both expression and activation of STAT3 were increased in the nucleus by the hypertrophy. Immunofluorescence analysis revealed that the catecholamine- induced hypertrophy promoted nuclear localization of pY705-STAT3. Of interest, phosphorylation of pS727- STAT3 in mitochondria was significantly reduced by catecholamine-induced hypertrophy. In addition, mitochondrial complexes II and III were greatly down- regulated in the hypertrophic cells. Our data suggest that the alterations in nuclear and mitochondrial activation of STAT3 and caveolae localization of caveolin-3 are related to the development of the catecholamine-induced cardiac hypertrophy.
Collapse
Affiliation(s)
- Kyuho Jeong
- Department of Biochemistry, Gyeongsang National University, Jinju 660-701, Korea
| | | | | | | |
Collapse
|
20
|
Yang MK, Lee SH, Seo HW, Yi KY, Yoo SE, Lee BH, Chung HJ, Won HS, Lee CS, Kwon SH, Choi WS, Shin HS. KR-31761, a novel K+(ATP)-channel opener, exerts cardioprotective effects by opening both mitochondrial K+(ATP) and Sarcolemmal K+(ATP) channels in rat models of ischemia/reperfusion-induced heart injury. J Pharmacol Sci 2009; 109:222-32. [PMID: 19234365 DOI: 10.1254/jphs.08132fp] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
The cardioprotective effects of KR-31761, a newly synthesized K+(ATP) opener, were evaluated in rat models of ischemia/reperfusion (I/R) heart injury. In isolated rat hearts subjected to 30-min global ischemia/30-min reperfusion, KR-31761 perfused prior to ischemia significantly increased both the left ventricular developed pressure (% of predrug LVDP: 17.8, 45.1, 54.2, and 62.6 for the control, 1 microM, 3 microM, and 10 microM, respectively) and double product (DP: heart rate x LVDP; % of predrug DP: 17.5, 44.9, 56.2, and 64.5 for the control, 1 microM, 3 microM, and 10 microM, respectively) at 30-min reperfusion while decreasing the left ventricular end-diastolic pressure (LVEDP). KR-31761 (10 microM) significantly increased the time to contracture during the ischemic period, whereas it concentration-dependently decreased the lactate dehydrogenase release during reperfusion. All these parameters were significantly reversed by 5-hydroxydecanoate (5-HD, 100 microM) and glyburide (1 microM), selective and nonselective blockers of the mitochondrial K+(ATP) (mitoK+(ATP)) channel and K+(ATP) channel, respectively. In anesthetized rats subjected to 30-min occlusion of left anterior descending coronary artery/2.5-h reperfusion, KR-31761 administered 15 min before the onset of ischemia significantly decreased the infarct size (72.2%, 55.1%, and 47.1% for the control, 0.3 mg/kg, i.v., and 1.0 mg/kg, i.v., respectively); and these effects were completely and almost completely abolished by 5-HD (10 mg/kg, i.v.) and HMR-1098, a selective blocker of sarcolemmal K+(ATP) (sarcK+(ATP)) channel (6 mg/kg, i.v.) administered 5 min prior to KR-31761 (72.3% and 67.9%, respectively). KR-31761 only slightly relaxed methoxamine-precontracted rat aorta (IC50: > 30.0 microM). These results suggest that KR-31761 exerts potent cardioprotective effects through the opening of both mitoK+(ATP) and sarcK+(ATP) channels in rat hearts with a minimal vasorelaxant effect.
Collapse
Affiliation(s)
- Min-Kyu Yang
- Department of Applied Biochemistry, Division of Life Science, College of Biomedical and Health Science, Konkuk University, Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Huang TH, He L, Qin Q, Yang Q, Peng G, Harada M, Qi Y, Yamahara J, Roufogalis BD, Li Y. Salacia oblonga root decreases cardiac hypertrophy in Zucker diabetic fatty rats: inhibition of cardiac expression of angiotensin II type 1 receptor. Diabetes Obes Metab 2008; 10:574-85. [PMID: 17645561 DOI: 10.1111/j.1463-1326.2007.00750.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
AIMS We investigated the effect of the water extract of Salacia oblonga (SOE), an ayurvedic antidiabetic and antiobesity medicine, on obesity and diabetes-associated cardiac hypertrophy and discuss the role of modulation of cardiac angiotensin II type 1 receptor (AT(1)) expression in the effect. METHODS SOE (100 mg/kg) was given orally to male Zucker diabetic fatty (ZDF) rats for 7 weeks. At the end-point of the treatment, the hearts and left ventricles were weighed, cardiomyocyte cross-sectional areas were measured, and cardiac gene profiles were analysed. On the other hand, angiotensin II-stimulated embryonic rat heart-derived H9c2 cells and neonatal rat cardiac fibroblasts were pretreated with SOE and one of its prominent components mangiferin (MA), respectively. Atrial natriuretic peptide (ANP) mRNA expression and protein synthesis and [(3)H]thymidine incorporation were determined. RESULTS SOE-treated ZDF rats showed less cardiac hypertrophy (decrease in weights of the hearts and left ventricles and reduced cardiomyocyte cross-sectional areas). SOE treatment suppressed cardiac overexpression of ANP, brain natriuretic peptide (BNP) and AT(1) mRNAs and AT(1) protein in ZDF rats. SOE (50-100 microg/ml) and MA (25 micromol) suppressed angiotensin II-induced ANP mRNA overexpression and protein synthesis in H9c2 cells. They also inhibited angiotensin II-stimulated [(3)H]thymidine incorporation by cardiac fibroblasts. CONCLUSIONS Our findings demonstrate that SOE decreases cardiac hypertrophy in ZDF rats, at least in part by inhibiting cardiac AT(1) overexpression. These studies provide insights into a potential cardioprotective role of a traditional herb, which supports further clinical evaluation in obesity and diabetes-associated cardiac hypertrophy.
Collapse
Affiliation(s)
- T H Huang
- Faculty of Pharmacy, University of Sydney, Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
KR-31762, a novel KATP channel opener, exerts cardioprotective effects by opening SarcKATP channels in rat models of ischemia/reperfusion-induced heart injury. Arch Pharm Res 2008; 31:482-9. [DOI: 10.1007/s12272-001-1182-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2007] [Indexed: 11/25/2022]
|
23
|
Lee TM, Lin MS, Chang NC. Effect of ATP-sensitive potassium channel agonists on ventricular remodeling in healed rat infarcts. J Am Coll Cardiol 2008; 51:1309-18. [PMID: 18371564 DOI: 10.1016/j.jacc.2007.11.067] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2007] [Revised: 10/29/2007] [Accepted: 11/19/2007] [Indexed: 11/30/2022]
Abstract
OBJECTIVES The purpose of this study was to determine whether ATP-sensitive potassium (K(ATP)) channel agonists exert a beneficial effect on the structural, functional, and molecular features of the remodeling heart in infarcted rats. BACKGROUND Myocardial K(ATP) channels have been implicated in the ventricular remodeling after myocardial infarction by inhibition of 70-kDa S6 (p70S6) kinase. METHODS Male Wistar rats after induction of myocardial infarction were randomized to either vehicle, agonists of K(ATP) channels nicorandil and pinacidil, an antagonist of K(ATP) channels glibenclamide, or a combination of nicorandil and glibenclamide or pinacidil and glibenclamide for 4 weeks. To verify the role of p70S6 kinase in ventricular remodeling, rapamycin was also assessed. RESULTS Significant ventricular hypertrophy was detected by increased myocyte size at the border zone isolated by enzymatic dissociation after infarction. Increased synthesis of p70S6 kinase messenger ribonucleic acid after infarction in vehicle-treated rats was confirmed by reverse transcription-polymerase chain reaction, consistent with the results of immunohistochemistry and Western blot for phosphorylated p70S6 kinase. Rats in the nicorandil- and pinacidil-treated groups significantly attenuated cardiomyocyte hypertrophy and phosphorylated p70S6 kinase expression with similar potency, as compared with vehicle. The beneficial effects of nicorandil and pinacidil were abolished by administering either glibenclamide or 5-hydroxydecanoate. Addition of rapamycin attenuated ventricular remodeling and did not have additional beneficial effects compared with those seen in rats treated with either nicorandil or pinacidil alone. CONCLUSIONS Activation of K(ATP) channels by either nicorandil or pinacidil can attenuate ventricular remodeling, probably through a p70S6 kinase-dependent pathway after infarction.
Collapse
Affiliation(s)
- Tsung-Ming Lee
- Cardiology Section, Department of Medicine, Taipei Medical University and Chi-Mei Medical Center, Tainan, Taiwan
| | | | | |
Collapse
|
24
|
Lee JH, Jung IS, Lee SH, Yang MK, Hwang JH, Lee HD, Cho YS, Song MJ, Yi KY, Yoo SE, Kwon SH, Kim B, Lee CS, Shin HS. Cardioprotective effects of BMS-180448, a prototype mitoK(ATP) channel opener, and the role of salvage kinases, in the rat model of global ischemia and reperfusion heart injury. Arch Pharm Res 2007; 30:634-40. [PMID: 17615684 DOI: 10.1007/bf02977659] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
To investigate the involvement of reperfusion-induced salvage kinases (RISK) as possible signaling molecules for the cardioprotective effects of BMS-180448, a prototype mitochondrial ATP-sensitive K+ (mitoK(ATP)) channel opener, we measured its cardioprotective effects in a rat model of ischemia/reperfusion (I/R) heart injury, together with western blotting analysis of five different signaling proteins. In isolated rat hearts subjected to 30-min global ischemia followed by 30-min reperfusion, BMS-180448 (1, 3 and 10 microM) significantly increased reperfusion left ventricular developed pressure (LVDP) and 30-min reperfusion double product (heart rate x LVDP) in a concentration-dependent manner, while decreasing left ventricular end-diastolic pressure (LVEDP) throughout reperfusion period in a concentration-dependent manner. SDS-PAGE/western blotting analysis of left ventricle reperfused for 30 min revealed that BMS-180448 significantly decreased phospho-GSK3beta at high concentration, whereas it tended to increase slightly phospho-eNOS and phospho-p70S6K with concentration. However, BMS-180448 had no effect on phospho-Akt and phospho-Bad. These results suggest that the cardioprotective effects of BMS-180448 against I/R heart injury may result from direct activation of mitoK(ATP) channel in cardiomyocytes, with the minimal role of RISK pathway in the activation of this channel and the cardioprotective effects of BMS-180448.
Collapse
Affiliation(s)
- Ju-Han Lee
- Department of Applied Biochemistry, Division of Life Science, College of Biomedical and Health Science, Konkuk University, Chungju 380-701, Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Rodent models of heart failure. J Pharmacol Toxicol Methods 2007; 56:1-10. [PMID: 17391988 DOI: 10.1016/j.vascn.2007.01.003] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2006] [Accepted: 01/31/2007] [Indexed: 11/28/2022]
Abstract
Heart failure, a complex disorder with heterogeneous aetiologies remains one of the most threatening diseases known. It is a clinical syndrome attributable to a multitude of factors that begins with the compensatory response known as hypertrophy, followed by a decompensated state that finally results in heart failure. Given the lack of a unified theory of heart failure, future research efforts are required to unify and synthesize our current understanding of the multiple mechanisms that control remodelling in heart under various stress conditions. During the past few decades, use of animal models has provided new insights into the complex pathogenesis of this syndrome. Rodents have contributed significantly in the understanding of the pathogenesis and progression of heart failure. With the advent of the transgenic era, rodent models have revolutionized preclinical research associated with heart failure. These models combined with physiological measurements of cardiac hemodynamics, are expected to yield more valuable information regarding the molecular mechanisms of heart failure and aid in the discovery of novel therapeutic targets. However, all animal models used have advantages and limitations, and the issues determining transfer from preclinical to clinical require critical evaluation. The present review focuses upon rodent models of heart failure.
Collapse
|