Therapeutic Advances in Cardiovascular Disease http://tak.sagepub.com/

Exploration of pharmacological interventions to prevent isoproterenol-induced myocardial infarction in experimental models Monika Garg and Deepa Khanna Ther Adv Cardiovasc Dis published online 9 May 2014 DOI: 10.1177/1753944714531638 The online version of this article can be found at: http://tak.sagepub.com/content/early/2014/05/07/1753944714531638 A more recent version of this article was published on - Jun 11, 2014

Published by: http://www.sagepublications.com

Additional services and information for Therapeutic Advances in Cardiovascular Disease can be found at: Email Alerts: http://tak.sagepub.com/cgi/alerts Subscriptions: http://tak.sagepub.com/subscriptions Reprints: http://www.sagepub.com/journalsReprints.nav Permissions: http://www.sagepub.com/journalsPermissions.nav

Version of Record - Jun 11, 2014 >> OnlineFirst Version of Record - May 9, 2014 What is This?

Downloaded from tak.sagepub.com at LOUISIANA STATE UNIV on August 18, 2014

531638 research-article2014

TAK0010.1177/1753944714531638Therapeutic Advances in Cardiovascular DiseaseM Garg and D Khanna

Therapeutic Advances in Cardiovascular Disease

Exploration of pharmacological interventions to prevent isoproterenol-induced myocardial infarction in experimental models

Review

Ther Adv Cardiovasc Dis 1­–15 DOI: 10.1177/ 1753944714531638 © The Author(s), 2014. Reprints and permissions: http://www.sagepub.co.uk/ journalsPermissions.nav

Monika Garg and Deepa Khanna

Abstract:  High incidences of myocardial infarction associated with high morbidity and mortality, are a major concern and economic burden on industrialized nations. Persistent β-adrenergic receptor stimulation with isoproterenol leads to the development of oxidative stress, myocardial inflammation, thrombosis, platelet aggregation and calcium overload, which ultimately cause myocardial infarction. Therapeutic agents that are presently employed for the prevention and management of myocardial infarction are beta-blockers, antithrombotics, thrombolytics, statins, angiotensin converting enzyme inhibitors, angiotensin II type 1 receptor blockers, calcium channel blockers and nitrovasodilators. In spite of effective available interventions, the mortality rate of myocardial infarction is progressively increasing. Thus, there has been a regular need to develop effective therapies for the prevention and management of this insidious disease. In this review, the authors give an overview of the consequences of isoproterenol in the pathogenesis of cardiac disorders and various therapeutic possibilities to prevent these disorders.

Keywords:  atherosclerosis, calcium overload, inflammation, isoproterenol, oxidative stress, reactive oxygen species

Introduction Cardiovascular diseases are the most common cause of death in the world. Myocardial infarction (MI), an insidious condition, plays a major role in this mortality [Yusuf et al. 2004; Iakovlev, 2010; Gerczuk and Kloner, 2012]. MI is characterized by intense chest pain, which may radiate into the neck, jaw and arms, and can cause shortness of breath. Patients suffering from cardiovascular and metabolic disorders such as hypertension, atherosclerosis and diabetes mellitus are at a higher risk of MI [Bianchi et al. 2008; Erbel and Budoff, 2012; Ma et  al. 2012]. The growing list of pathophysiological changes seen during MI is due to the development of oxidative stress, myocardial inflammation, thrombosis and calcium overload, and altered signalling pathways. Recent studies have suggested that increased β-adrenergic receptors play a role in the generation of myocardial oxidative stress,

inflammation, calcium overload and coronary spasm, followed by loss of myocytes [Communal et  al. 1998; Frangogiannis et  al. 2002]. β-adrenergic mediated oxidative stress generates reactive oxygen species (ROS), which lead to the formation of a macrophage-rich atheroma and, as a consequence, the risk of unstable angina, thrombosis and acute MI develops [Stanger and Weger, 2003; Libby and Aikawa, 2002, 2003]. β-adrenergic receptor activation induces the expression of interleukin (IL)-18, a proinflammatory cytokine, in the myocardium and in cardiac-derived endothelial cells via activation of nuclear factor (NF)-κB [Chandrasekar et  al. 2004]. Further, β-adrenergic over-activation increases the concentration of cyclic adenosine monophosphate (cAMP) through activation of adenyl cyclase, resulting in activation of protein kinase A and phosphorylation of L-type calcium channels. This cellular signalling cascade

Correspondence to: Deepa Khanna PhD Department of Pharmacology, Cardiovascular Pharmacology Division, Institute of Pharmacy, Rajendra Institute of Technology and Sciences [RITS], Sirsa-125 055, India, [email protected] Monika Garg, M. Pharm Cardiovascular Pharmacology Division Department of Pharmacology Rajendra Institute of Technology and Sciences India

http://tac.sagepub.com 1

TAK531638.indd 1

Downloaded from tak.sagepub.com at LOUISIANA STATE UNIV on August 18, 2014

06/05/2014 10:50:21 AM

Therapeutic Advances in Cardiovascular Disease  increases intracellular calcium concentration in the heart, followed by the induction of a series of myocardial events such as abnormal gene transcription and loss of myocytes in terms of incidence of apoptosis and necrosis [Mann et  al. 1992; Iwase et al. 1996; Communal et al. 1998; Geng et al. 1999]. Isoproterenol (ISO) is a synthetic nonselective β-adrenergic agonist [Ma et  al. 2009; Nichtova et  al. 2012]. ISO is commonly administered in high doses to induce experimental acute MI in rats [Hussain et  al. 2012; Patel et  al. 2012; Nagoor Meeran and Stanely Mainzen Prince, 2012] and this action is mediated by inducing myocardial oxidative stress, inflammation and calcium overload, through activation of β1-adrenergic receptors in the heart [Mohan and Bloom, 1999; Izem-Meziane et  al. 2012; Vijayan et al. 2012]. Persistent β-adrenergic receptor activation with ISO is associated with deleterious myocardial effects, including left ventricular hypertrophy [Rona et  al. 1959; Taylor and Tang, 1984; Ni et al. 2011; Song et al. 2011], increased ventricular collagen content and a reduced inotropic response to ISO [Vassallo et al. 1988; Chang et  al. 1982; Kenakin and Ferris, 1983]. ISO treatment directly increases cardiac expression and activity of angiotensin converting enzymes (ACE); thus activation of the circulatory as well as the cardiac angiotensin system could be expected under sympathoexcitatory heart failure [Oliveira and Krieger, 2005]. Clinical and experimental studies have tried out a number of therapeutic agents such as betablockers, antithrombotics, statins, ACE inhibitors, calcium channel blockers (CCBs) and nitrovasodilators; these are being employed for the prevention, management and mitigation of MI [Ott and Fenster, 1991; Held and Yusuf, 1993; Spinarová et al. 2011; Burchill et al. 2012; Bhatt, 2012; Carey et al. 2012]. Exploring pharmacological interventions to ameliorate ISOinduced cardiac abnormalities is of potential therapeutic value in preventing the initiation and progression of MI. This article reviews the ISOinduced consequences and various explored pharmacological interventions which ameliorate ISO-induced cardiac abnormalities Consequences of ISO administration ISO mediated oxidative stress Oxidative stress plays a critical role in the development of structural and functional changes in the heart. Oxidative stress is generated due to

ROS and imbalanced antioxidant defence mechanisms [Bagatini et al. 2011]. ROS are generated by an activated nicotinamide adenine dinucleotide phosphate oxidase, xanthine oxidase, autooxidized catecholamines, increased angiotensin-II and aldosterone levels as well as released proinflammatory cytokines [Di Filippo et  al. 2006; Landmesser et al. 2002]. Further, the other cause of ROS generation is signalling alterations. ROSdependent apoptosis [Remondino et  al. 2003], extracellular matrix biosynthesis [Zhang et  al. 2005] and cardiac hypertrophy [Zhang et  al. 2007] following ISO-induced β-adrenergic receptor (AR) activation have been described. The catecholamine-induced cardiotoxicity was explained by direct and indirect formation of free radicals and sulphydryl reactivity through a variety of its oxidation products which inactivates cell functions [Singal et al. 1981]. Chronic as well as acute treatment with a renowned β-agonist catecholamine, ISO enhances cardiac oxidative stress in rats [Zhang et al. 2005]. The occurrence of oxidative stress in ISO-injected rats was indicated by the increase in cardiac malondialdehyde content and formation of conjugated dienes as well as by the low Reduced Glutathione/Oxidized Glutathione (GSH/GSSG) ratio, and by reducing the level of superoxide dismutase (SOD), catalase, glutathione peroxidise [Tappia et  al. 2001; Dhalla et al. 2000]. Excessive stimulation of cardiac β-adrenergic receptors and angiotensin II type 1 receptors by ISO increases oxidative stress and reduces cAMP formation in the ventricle membranes thus causing MI [Zhang et  al. 2005; Singal et al. 1982; Remondino et al. 2003; Zhang et al. 2007; Singh et al. 2000, Tappia et al. 2001]. ISO increases the release of cytochrome-c and activation of c-Jun NH2-terminal kinase (JNK), and extracellular signal-regulated kinase activities which further generate ROS and induces apoptosis [Remondino et  al. 2003; Saadane et  al. 1999] (Figure 1). ISO-mediated accumulation of collagen and phosphorylation of p38 Mitogen activated protein kinase (MAPK) kinase and their upstream elements, Ras-related C3 botulinum toxin substrate 1 (Rac-1), protooncogene serine/threonine-protein kinase (Raf1), Apoptosis signal-regulating kinase 1 (ASK-1), which promote lipid peroxidation. Lipid peroxidation increases membrane permeability and causes cardiac injury [Singal et  al. 1983; Singal et  al. 1982; Zhang et  al. 2007] (Figure 1). ISO decreases coronary blood flow [Somani et  al. 1970] and produces a complete shutdown of myocardial perfusion in the crucial early period

2 http://tac.sagepub.com

TAK531638.indd 2

Downloaded from tak.sagepub.com at LOUISIANA STATE UNIV on August 18, 2014

06/05/2014 10:50:21 AM

M Garg and D Khanna

Figure 1.  Beta adrenergic receptor over activation mediated cardiac toxicity through oxidative stress, Calcium overload and inflammatory cascades.

Schematic diagram represents the sequences where isoproterenol, a beta adrenergic agonist activates G- protein coupled receptor through β1, β2 and AT-1 adrenergic receptor via Gs and Gi pathways. Overstimulation of beta receptors activates mediatory pathways like oxidative stress, inflammatory reactions and formation of c-AMP which triggered calcium overload. ISO; Isoproterenol, c- AMP; cyclic adenosine monophosphate, PKA; protein kinase-A, ATP; adenosine triphosphate, PI3KY; Phosphatidylinositide 3-kinases, ERK; extracellular-signal-regulated kinases, JNK; c-Jun N-terminal kinases, TNF-α; tumour necrosis factor-α MAPKS; mitogen-activated protein kinase, CAMKs; calmodulin-dependent protein kinases, NF-κB; nuclear factor-κB, IKK; IκB kinase, IL-1β; interlukin-1β, ROS; reactive oxygen species, TBARS; thiobarbituric acid reactive substance, IGF; Insulin-like growth factor.

of ISO cardiotoxicity. Overall these findings suggest that myocardial cells are injured with ROS generated by ISO. ISO mediated calcium overload Calcium maintains the vitality of organs by contraction and relaxation in smooth muscles and endothelial tissues [Orallo, 1996]. Fluctuations in the intracellular free calcium ion concentration control a number of diverse cellular processes (vasa vasorum flow, endothelial contraction, collagen, elastins, proteoglycans synthesis and platelet activity) affecting various proteases and calcium dependent phospholipase enzymes [Henry, 1990]. Studies suggest that early alteration of sarcolemma due to calcium overload

results from exaggerated direct β-receptor stimulation. Several studies also reported that an increased concentration of cAMP by the overactivation of β-adrenergic receptors results in an altered cellular signalling cascade, which increases intracellular calcium concentration in the heart [Mann et al. 1992; Iwase et al. 1996; Geng et al. 1999]. ISO administration releases endogenous myocardial norepinephrine, increases calcium influx into myocardial cells and this increase in calcium influx results in cell necrosis and the breakdown of membrane permeability barriers [Mallov, 1984] (Figure 1). Increase of myocardial Ca2+ content will result in myofilament over-stimulation, increase of contractile force and oxygen requirement as well as excessive adenosine triphosphate (ATP) breakdown; each of these

http://tac.sagepub.com 3

TAK531638.indd 3

Downloaded from tak.sagepub.com at LOUISIANA STATE UNIV on August 18, 2014

06/05/2014 10:50:22 AM

Therapeutic Advances in Cardiovascular Disease  factors contribute to cardiac muscle cell injury [Tokgözoğlu, 2009]. Ca2+ influx followed by its deposition in mitochondria occurs as early as 2 minutes following ISO administration [Ishikawa et al. 2004; Cohen and Fuster, 1990]. ISO-induced intracellular calcium overload affects the membrane by activating calcium-dependent phospholipases and protease enzymes and depletion of high energy phosphates [Nirdlinger and Bramante, 1974; Titus, 1983; Fleckenstein et  al. 1974; Kondo et  al. 1987]. Activation of these calcium dependent enzymes further, inhibits membrane-bound enzymes such as Na+/K+-ATPase and as a result there is an increase in Na+ levels and a loss of cytoplasmic K+ ions. This increased Na+ concentration leads to calcium accumulation through the Na+- Ca2+ exchange system (Figure 1). The summation of these alterations leads to cellular dysfunction and cardiotoxicity [Ramos et al. 1984]. ISO mediated inflammation Inflammation associated metabolic disorders, namely hypertension, hypercholesterolemia, hyperhomocysteinemia, lipid deposition and diabetes, are the major risk factors responsible for the development of cardiovascular diseases [Niessen et al. 2003]. ISO induces IL-18 expression both in vivo and in vitro via activation of NF-κB (Figure 1). This induction mediates progressive left ventricular remodelling in heart failure [Chandrasekar et al. 2004]. NF-κB activation is β-2AR dependent and requires signalling through a cascade involving heteromeric G protein subunit (Gi), Phosphatidylinositol-4,5bisphosphate 3-kinase (PI3Kc), Protein kinase B (Akt) and IKK (Figure 1). Further, these signalling cascades are critical for cardiac cells [Chandrasekar et al. 2004]. IL-18 promotes and activates the migration of inflammatory cells [McInnes et al. 2005]. In addition, ISO induces cardiotoxic effects via other inflammatory cytokines such as tumour necrosis factor (TNF)α, IL-1β, and IL-6 through cAMP [Mann, 1996, 1998; McInnes et al. 2005; Murray et al. 2000]. ISO mediated lipid peroxidation ISO causes oxidative stress in the myocardium resulting in infarct-like necrosis of the heart muscle and an increase in the levels of lipids in the myocardium. The levels of mitochondrial calcium, cholesterol, free fatty acids, and

triglycerides were considerably increased and ATP and phospholipids were considerably decreased in ISO-induced rats [Kumaran and Prince, 2010]. The mitochondrial membrane is rich in polyunsaturated fatty acids (PUFAs) which are present in its phospholipids form and highly susceptible to lipid peroxidation [Halliwell and Gutteridge 1990]. Activated lipid peroxidation is an important pathogenic event in MI and the levels of lipid peroxide reflect the major stages of disease and its complications. Administration of ISO induces lipid peroxidation in the mitochondrial membrane [Tappel 1973]. The process of lipid peroxidation is one of oxidative conversion of PUFAs to products known as malondialdehydes, which are measurable as thiobarbituric acid reactive substances or lipid peroxides [Bakan et  al. 2002]. Kumaran and Prince showed in the results of their study that mitochondrial lipid peroxidation products (thiobarbituric acid reactive substances and plasma lipid hydro peroxides) were increased during ISO administration [Kumaran and Prince, 2010]. An increase in mitochondrial lipid peroxide level indicates enhanced lipid peroxidation by free radicals generated on ISO administration. Increased levels of lipid peroxides injure blood vessels, causing increasing adherence and aggregation of platelets to the injured sites [Grylewski, 1980]. ISO mediated renin–angiotensin release Activation of the renin–angiotensin system plays a crucial role in the progression of hemodynamic alterations and cardiac remodelling. ISO significantly elevates plasma cAMP, plasma renin activity, plasma aldosterone, and cardiac ACE activity with long-term administration [Grimm et  al. 1998]. It has also been suggested that ISO increases left ventricular and right ventricular weight with activation of the circulatory renin– angiotensin system [Nagano and Ogihara, 1994]. Increased plasma levels of atrial natriuretic peptide found a close correlation of infarct size and right atrial and left-ventricular end-diastolic pressures. ACE activity also increases in the heart after infarction [Hirsch et al. 1991], mainly in the scar tissue [Hokimoto et al. 1995; Busatto et al. 1997]. Pharmacological interventions to attenuate ISO-induced MIs As stated in the previous section, the consequences of ISO induction is myocardial injury

4 http://tac.sagepub.com

TAK531638.indd 4

Downloaded from tak.sagepub.com at LOUISIANA STATE UNIV on August 18, 2014

06/05/2014 10:50:22 AM

TAK531638.indd 5

Mibefradil (CCBs)

Metoprolol (betablocker)

Carvedilol (vasodilator and beta-blocker) Amiodarone

 1

 2

 3

Magnesium chloride Verapamil (CCB)

Amlodipine (CCB)

Nicorandil K(ATP) channel opener Enalapril (ACE inhibitor) Quinidine (class I antiarrhythmic agent)

 6

 8

 9

Downloaded from tak.sagepub.com at LOUISIANA STATE UNIV on August 18, 2014

Imidapril (ACE inhibitor)

14

13

DITPA (Thyroid hormone analogue) Propranolol and labetalol (betablocker)

12

11

10

 7

Fenofibrate (PPAR α)

 5

 4

Drug

S. no

ISO can cause ischaemic myocardial alterations, lipid peroxide generation and procoagulant activity, including areas of myocardial necrosis, contraction band necrosis, increased plasma levels of cardiac necrosis markers (c-troponin I and myoglobin) Alterations in cardiac function, positive inotropic effect of ISO, β1-adrenergic density and ISO stimulated AC activity in the failing heart

Oxidative stress after ISO-induced myocardial injury, decrease in GSH levels and activities of SOD and an increase in MDA level Utilization of free fatty acids was reduced in ISO, i.e. increases the level of fatty acids resulting in myocardial injury ISO administration revealed significant reduction of CK (C Max) activity Increase in calcium level and overexpression of βadrenergic receptors Pathological changes and oxidative stress and calcium overload after ISO administration ISO showed significant changes in antioxidant defence system and lipid profile levels β-receptor sensitization and increase in negative inotropic effect Administration of ISO induced lipid per oxidation in cardiac tissue and exhibited a significantly elevated serum glutamate oxaloacetate transaminase levels. Increased SOD with a concomitant decrease in catalase activity ISO alters endothelial function after myocardial infarction

Prevents changes in β- adrenergic signal transduction in CHF, decreases calcium level and reduces AC activity

Improves endothelial nitric oxide and β-adrenergic receptor mediated vasorelaxation by increasing nitric oxide in the vasculature Prevents myocardial necrosis damage and the associated risk factors.

(Continued)

Sethi et al. [2004]

Pinelli et al. [2004]

Spooner et al. [2004]

Chattopadhyay et al. [2003]

Igawa et al. [2002]

Sympatho inhibition and prevention of β-adrenergic desensitization and also improves ionotropic response Reduction of lipid peroxidation reaction, exerts an 'indirect antioxidant' effect, scavenge superoxide anion and hydrogen peroxide

Maintains lipid profile and antioxidant defence system

Min et al. [1999], Hanafy et al. [2010] Min et al. [1999], Sathish et al. [2003] Sathish et al. [2003]

Naik et al. [1999]

Ide et al. [1999], Albayrak et al. [2009] Yuan et al. [2008]

Yue et al. [1992]

Kalaycioglu et al. [1999]

Sandmann et al. [1999], Min et al. [1999]

Reference

Reduces calcium overload and reduces overexpression of β-adrenergic receptors Reduces lipid peroxidation and reduces calcium level

Protective effects against oxygen radical-mediated injury in cardiac myocytes and maintains the level of biochemical marker enzymes Activates lipoprotein lipase and reduces apoproteins thus increasing lipolysis and eliminating triglyceriderich particles from plasma Maintains ionic balance and cellular enzymes

Protects myocardium against ischaemia-induced Ca2+-overload and increases β-adrenergic response in chronically failing rat hearts, Potent vasodilating effect, improved chronic inotropic response Negative inotropic/chronotropic effects, reduces lipid peroxidation and also reduces the myocardial energy demand by decreasing the heart rate Inhibited Fe(++)-initiated lipid peroxidation by scavenging free radicals

β-adrenergic stimulation with ISO enhanced contractility and Ca2+ availability by increasing calcium overload, contractile dysfunction, and arrhythmias in failing hearts ISO increases negative inotropic/chronotropic effect and lipid peroxidation effect Alters oxidative stress and lipid levels

Preventive mechanism of drug

Mechanism of action of isoproterenol

Table 1.  Mechanisms of action exhibited by isoproterenol in the pathogenesis of cardiac disorders and the role of therapeutic drugs in myocardial protection.

M Garg and D Khanna

http://tac.sagepub.com 5

06/05/2014 10:50:22 AM

TAK531638.indd 6

Atorvastatin

Nimesulide (cyclooxygenase-2 inhibitor) Fluvastatin

15

16

Captopril (ACE inhibitor) Fasudil hydrochloride hydrate Rosuvastatin

Dexrazoxane (DEX)

SMP-300

Aspirin

Cromakalim

20

22

23

24

25

26

Downloaded from tak.sagepub.com at LOUISIANA STATE UNIV on August 18, 2014

ISO increases myocardial calcium overload and influence in fenton reaction that alters ironchelating property ISO-induced ST segment depression and increases Na+/H+ exchange ISO marked increases in cholesterol, free fatty acids, triglycerides and lipid peroxides in heart mitochondria, activity of Na+ K(+)-ATPase decreases, activity of Ca2+-ATPase increases ISO administration alters biochemical markers and decreases blood clotting time

ISO induced myocardial injury by increasing serum cardiac biomarker, inflammatory markers such as TNF-α

Administration of ISO increases inflammatory markers such as CK isozyme and troponin-I in serum ISO-treated rats showed significant rise in STsegment and increase in content of LDH, glutamic oxalacetic transaminase, CK and MDA, as well as a fall in the activities of glutathione peroxidase, SOD and catalase were observed. Oxidative stress has been increased after ISO administration with alterations in biochemical levels ISO-induced cardiotoxicity with a significant reduction in activities of myocardial CK-MB isoenzyme, LDH, SOD, catalase, and reduced GSH level along with increase in MDA content ISO-induced myocardial damage by biochemical enzymatic alterations ISO-induced heart failure in relationship with RhoA/ ROCK to the ERK and the JNK pathways

ISO administration produced severe myocardial damage and oxidative stress in rats

Mechanism of action of isoproterenol

Increase in blood clotting time and also depleted intracytoplasmic glycogen

Promotes angiogenesis and reduces cholesterol level by suppressing the synthesis of mevalonic acid, also reduces tissue, TNF-α and upregulated vascular endothelial growth factor levels Exerts iron-chelating properties in reducing catecholamine toxicity and also inhibits myocardial calcium overload Selective Na+/H+ exchange inhibitor that targets in the prevention of angina Decreases the level of cholesterol, free fatty acids, triglycerides and lipid peroxides

ρ-kinase inhibitor suppresses ISO-induced heart failure in rats via JNK and ERK1/2 pathways

Attenuates the development of acute myocardial infarction by restoring hemodynamic, biochemical, histopathological and ultrastructural changes along with increases in MDA content Potent vasodilating effect

Improves nitric oxide stress and DNA damage by maintaining serum enzymes

Cardioprotective effect by inhibiting HMG-CoA reductase inhibitor via maintaining activities of endogenous antioxidant enzymes

Reduces cholesterol formation by reducing the synthesis of mevalonate acid by inhibiting HMG-CoA reductase Anti-ischaemic effects

Preventive mechanism of drug

Aghi et al. [1992]

Yamamoto et al. [2002] Manjula and Devi [1993]

Zatloukalová et al. [2012]

Zaitone and AboGresha [2012]

Asdaq and Inamdar [2010] Wang et al. [2011]

Goyal et al. [2009, 2010]

Keles et al. [2009]

Zhou et al. [2008]

Saeed and Ahmed [2005, 2006]

Trivedi et al. [2006]

Reference

ISO, isoproterenol; CK, creatine kinase; PPAR, peroxisome proliferator-activated receptors; CCB, calcium channel blocker; K(ATP), ATP-sensitive potassium; ACE, angiotensin 1converting enzyme; AR, adrenergic receptor; AC, adenylyl cyclise; CHF, congestive heart failure; HMG-CoA, 3-hydroxy-3-methyl-glutaryl-CoA; CK-MB, creatine phosphokinase; LDH, lactate dehydrogenase; MDA, malondialdehyde; SOD, superoxide dismutase; GSH, glutathione; ERK, extracellular signal-regulated kinases; JNK, c-jun NH 2-terminal kinase; TNF-α, tumour necrosis factor-α

21

19

Lacidipine, ramipril and valsartan Telmisartan (PPAR-δ)

18

17

Drug

S. no

Table 1. (Continued)

Therapeutic Advances in Cardiovascular Disease 

6 http://tac.sagepub.com

06/05/2014 10:50:22 AM

M Garg and D Khanna due to high myocardial oxidative stress, lipid deposition, release of angiotensin and aldosterone, calcium overload and exaggerated myocardial inflammatory events, which are followed by induction of apoptosis and necrosis with loss of myocardial cells. This section of the review updates numerous pharmacological interventions, which halt the progression of myocardial injury and could be utilised to ameliorate the toxicity in ISO treated animals. Lipid-lowering agents Fibrates are lipid-lowering agents, which on long treatment significantly ameliorate myocardial injury. Fenofibrate confers its benefits on endothelial function, inflammatory cytokines, cardiac hypertrophy and vascular dysfunction by showing increased expression of peroxisome proliferatoractivated receptor-α in ISO-induced acute MI with reduction of cardiac marker enzymes [lactate dehydrogenase (LDH) and creatine phosphokinase isoenzyme (CK-MB)] (Table 1) [Devchand et al. 1996; Yuan et al. 2008]. The protective effect of clofibrate in ISO-induced MI is due to its ability to change corticosterone and serum lipid levels in the circulation [Wexler and Greenberg, 1978]. Statins (3-hydroxy-3-methylglutaryl-CoA reductase inhibitors) exhibit cardioprevention by their lipid lowering action and by increasing the level of deprived enzymes in myocardial-infarct rats. Specifically, rosuvastatin reduces inflammatory cytokines, tissue TNF-α and upregulates vascular endothelial growth factor level (Table 1). Further, fluvastatin and atorvastatin also attenuate acute MI by maintaining the antioxidant defence system and ATP activity [Zaitone and Abo-Gresha, 2012; Akila et al. 2007; Trivedi et al. 2006]. Moreover, combined therapy of atorvastatin and coenzyme Q10 improves the left ventricular function in ISOinduced heart failure in rats [Garjani et al. 2011]. Antioxidative agents Quinidine, a Na+ channel blocker with free radical scavenging effects, strengthens the antioxidant defence system by elevating SOD and catalase activity (Table 1) [Chattopadhyay et  al. 2003]. Dhalla and colleagues [Dhalla et al.2000] experimentally demonstrated that free radical scavenging enzymes are the first-line cellular defence against oxidative stress by decomposing O2 and H2O2 before interacting to form more reactive hydroxyl radicals. In addition to this, Akila and

colleagues [Akila et  al. 2007] showed significant increases in catalase activity after reperfusion, suggesting that the antioxidant defence system protects the cells against reactive species. Administration of telmisartan and olmesartan reduces this increased level of malondialdehyde (Table 1) [Goyal et al. 2009; Zhang et al. 2007]. The occurrence of oxidative stress in the ISOinjected rats was indicated by the increase of cardiac malondialdehyde content. Pretreatment with monoamine oxidase inhibitors significantly ameliorates the severity of myocardial injury produced by ISO treatment. This action was reflected by improved biochemical events [Stanton et al. 1970; Singh et  al. 1980]. Aspirin treatment decreases the biochemical lesions which occur due to the activation of lipid peroxidation and also normalizes the antioxidant defence enzymes such as α-glycerophosphate dehydrogenase (GDP), glutathione S-transferase, SOD, and catalase (Table 1) [Manjula et al. 1994]. ACE inhibitors and AT1 receptor blockers Angiotensin receptor blockers treatment was found to be more effective for reducing cardiac mass enlargement, oxidative stress and collagen accumulation in ISO-infused mice [Brown et al. 2005; Kass et  al. 2004]. ACE inhibitors directly prevent the inappropriate growth and hypertrophy stimulated by angiotensin II and other growth factors in rat myocardial tissue [Weber, 1997; Lindpaintner et  al. 1993]. Asdaq and Inamdar [Asdaq and Inamdar, 2010] showed that longterm use of captopril (an ACE inhibitor) effectively reduces the size of myocardial infarcts (Table 1). Captopril dislodges the effect of ISO by retaining back the activity of superoxide dismutase, catalase, LDH and CK-MB [Asdaq and Inamdar, 2010]. Further, long term captopril treatment resulted in a significant reduction in left ventricular end-systolic volume index, and increase in stroke volume index and ejection fraction [French et  al. 1999]. The protective role of carvedilol (α- and β-adrenoreceptor blocker) in preventing MI was associated with attenuation by scavenging free radicals and inhibiting lipid peroxidation [Yue et al. 1992] (Table 1). β-adrenoreceptor antagonist Beta-blockers are more beneficial than ACE inhibitors in cardioprotection. Metoprolol exerts a cardioprotective effect by showing a significant reduction in ventricular fibrillation, the event

http://tac.sagepub.com 7

TAK531638.indd 7

Downloaded from tak.sagepub.com at LOUISIANA STATE UNIV on August 18, 2014

06/05/2014 10:50:22 AM

Therapeutic Advances in Cardiovascular Disease  causing myocardial ischaemia and infarction [Coram et  al. 1987, Kalaycioglu et  al. 1999] (Table 1). Propanolol shows myocardial protective action mediated through hormonal (corticosterone, aldosterone) and metabolic changes (blood pressure). Further, it brings biochemical changes such as reduction of blood serum creatine phosphokinase, LDH and triglyceride levels [Wexler, 1985]. Calcium antagonists The rational use of combination treatment provides more powerful results in preclinical studies as suggested by the use of verapamil (a calcium antagonist) and magnesium chloride. The two drugs reveal significant cardioprotective potential by reducing CK-MB activity in ISO-induced MI [Naik et al. 1999] (Table 1). Further, Sathish and colleagues [Sathish et  al. 2003] found that the combined dose of nicorandil and amlodipine, proved to be more protective in experimentally-induced MI in rats. The extent of lysosomal membrane damage was reduced and lysosomal membrane integrity was preserved by these drugs. Calcium antagonists have a variety of actions to overcome excitation contractions, tending to produce vasodilatation and reduce myocardial contractility. On the other hand, it also limits the calcium entry into cardiac and smooth muscle cells, thereby reducing the splitting of adenosine triphosphate and myocardial oxygen demand. Dexrazoxane probably decreases the mortality rate in ISO-treated animals by reducing myocardial calcium overload and improving histological impairment as well as peripheral hemodynamic changes [Zatloukalová et  al. 2012] (Table 1). Moreover, verapamil exerts protection against myocardial lesions, electrocardiographic alterations, high plasma cardiac necrosis marker c-troponin I levels, and prevents the hemodynamic and procoagulation changes. Therefore verapamil seems to be suitable for preventing myocardial ischaemic lesions induced by ISO and vasopressin [Pinelli et  al. 2004]. Nimodipine is used in the management of patients with stroke or subarachnoid haemorrhage [Towart and Kazda, 1979]. Diltiazem has potent coronary vasodilator activity, which proves to be beneficial in the treatment of stable and unstable angina, ultimately reducing intracellular Ca2 + accumulation and MI [Strauss et  al. 1982; Hossack et  al. 1984; Boden et  al. 1985; Clozel et  al. 1983]. The cardioprotective action of CCB (verapamil, amlodipine and

diltiazem) therapies on ISO-treated animals have been augmented and determined by diagnostic enzymes such as LDH and CK-MB in serum and heart tissue homogenate [Kumar et al. 2009]. Mibefradil protects the myocardium against ischaemia-induced Ca2+-overload and improves the cardiac function by increasing βadrenergic responsiveness in chronically failing rat hearts [Sandmann et  al. 1999]. Oxodipine and nitrenidipine, both by exhibiting CCB properties, reduce the infarct size lesions located in the subendocardial areas of the left ventricle intramural at the apex and ventricular septum [Pérez-Cao et  al. 1994]. CCBs provoke spasm and are useful in improving the prognosis in patients with acute myocardial infarction (AMI) after stent implantation by suppressing coronary spasms [Katoh et al. 2012]. Other synthetic treatments Spironolactone treatment in ISO-treated rats reduces elevated mRNA levels of transforming growth factor β, connective tissue growth factor, matrix metalloprotease 2, matrix metalloprotease inhibitor 2, TNF-α, IL-1β, p22phox (subunit of NADPH oxidase) and xanthine dehydrogenase [Martín-Fernández et  al. 2012]. Propanolol and its combination therapy with nifedipine and guggulsterone exhibit cardiac protection against an ISO-induced marked increase of lipid peroxides, xanthine oxidase, creatine phosphokinase, phospholipase and superoxide dismutase enzymes [Kaul and Kapoor, 1989]. Together, lacidipine (a CCB), ramipril (an ACE inhibitor) and valsartan (an AT1 receptor blocker) reduce the severity of MI as indicated by routine biochemistry indicators, alanine aminotransferase, aspartate aminotransferase, LDH, creatine kinase (CK), CK-MB, troponin I (TnI) and nitric oxide. 7, 8-Dihydro-8-oxo-guanine, which is an indicator of DNA damage, was decreased by lacidipine, ramipril and valsartan. [Bayir et  al. 2012; Keles et  al. 2009]. Cromakalim, a potassium channel opener seems to be beneficial on serum LDH, serum glutamic oxaloacetic transaminase and depleted intracytoplasmic glycogen in ISOtreated animals [Aghi et al. 1992] (Table 1). The thyroid hormone analogue, 3, 5 diiodothyropropionic acid, improves the endothelial function by enhancing endothelial nitric oxide and β- adrenergic-mediated vasorelaxation. Thus, this analogue shows a protective effect against inflammatory reactions induced in MI [Spooner et al. 2004] (Table 1).

8 http://tac.sagepub.com

TAK531638.indd 8

Downloaded from tak.sagepub.com at LOUISIANA STATE UNIV on August 18, 2014

06/05/2014 10:50:22 AM

M Garg and D Khanna Nutraceuticals treatment The beneficial effects of nutraceuticals represent a great impact on nutritional therapy. Recently reported nutritional therapy seems beneficial in dislodging the effect of ISO-induced myocardial injuries. Myocardial injuries have occurred following mechanisms such as mitochondrial dysfunction, calcium overload, lipid peroxidation, alteration in membrane bound enzymes, apoptosis and cell necrosis. Mitochondria play a central role in the energy-generating process within the cell. Apart from this important function, mitochondria are involved in complex processes such as apoptosis. The cardioprotective actions of potassium channel openers have revealed that cardiac mitochondria are more important as the primary targets of these drugs than the plasma membrane. Murugesan and Manju reported that luteolin ameliorates mitochondrial damage in ISO- induced MI by maintaining lipid peroxidation metabolism due to its free radical scavenging properties [Murugesan and Manju, 2013]. The mechanism for the protective effect of p-coumaric acid is attributed to antilipid peroxidative, antioxidant and antiapoptotic properties. p-Coumaric acid pretreatment showed protective effects on apoptosis by inhibiting oxidative stress [Prince and Jyoti, 2013]. The cardioprotective effect of Crocus sativus L. (saffron) aqueous extract and safranal in ISO-induced MI was through modulation of oxidative stress in such a way that they maintain the redox status of the cell, by maintaining functional and structural damage through reduction of lipid peroxidation [Mehdizadeh et al. 2013]. Korean red ginseng extract significantly protects against cardiac injury and ISO-induced cardiac infarction by bolstering the antioxidant action in myocardial tissues. Moreover, Korean red ginseng extract also protects the secretion of inflammatory cells by suppressing caspase-3 activity and TNF-α protein production [Lsim et al. 2013]. In the context of the aforementioned studies, it is apparent that any given synthetic and natural products extend their protective effects through assorted mechanisms. It has been found that most of the products act through one or other of their antioxidant potential and many among them give protection from lipid peroxidation to the myocardium. Moreover, natural and synthetic drugs have also been able to reduce or even prevent the inflammation that arises due to myocardial necrosis produced by ISO intoxication. Synthetic drugs

are the preferable treatment to overcome the above problems as treatment with natural products requires high doses, long treatment, the characterization of chemical constituents and the difficulty in extrapolating the findings to clinical research. Natural products still hold great potential among them as a first-line therapy for myocardial injuries because the chances of occurrence of adverse events in natural products were much less compared with synthetic products. So, a future prospective could be the exploration of nutraceutical products, their activity and the targeting of the mechanisms involved in prevention against ISO-induced toxicity. Conclusion In summary, it has been concluded that ISO, a synthetic non-selective β-adrenergic agonist induced pathophysiological changes, which were seen during MI. ISO develops oxidative stress, myocardial inflammation, thrombosis, calcium overload and also alters signalling pathways. The rational use of certain pharmacological interventions alone or in combination with a few nutraceuticals helps in attenuating these ISO-induced changes. These interventions halt the progression of myocardial injury and could be utilised to ameliorate ISO- induced toxicity. Acknowledgement We express our thanks to Dr Rajendar Singh, Chairman, Shri Om Parkash, Director and Mr Sanjeev Kalra, Administrator, Rajendra Institute of Technology and Sciences, Sirsa, India, for their inspiration and constant support. Gratitude is also extended to Dr Pitchai Balakumar for his expert suggestions. Funding This research received no specific grant from any funding agency in the public, commercial, or notfor-profit sectors. Conflict of interest statement The authors have no conflicts of interest to declare.

References Aghi, M., Husain, Z., Pillai, K., Qadry, J., Balani, D. and Imran, M. (1992) Cardioprotective effect of cromakalim (potassium channel opener) in isoproterenol induced myocardial infarction in rats. Indian J Exp Biol 30: 611–614.

http://tac.sagepub.com 9

TAK531638.indd 9

Downloaded from tak.sagepub.com at LOUISIANA STATE UNIV on August 18, 2014

06/05/2014 10:50:22 AM

Therapeutic Advances in Cardiovascular Disease  Akila., D’souza, B., Vishwanath, P. and D’souza, V. (2007) Oxidative injury and antioxidants in coronary artery bypass graft surgery: off-pump CABG significantly reduces oxidative stress. Clin Chim Acta 375: 147–152.

Chandrasekar, B., Marelli–Berg, F., Tone, M., Bysani, S., Prabhu, S. and Murray, D. (2004) Beta-adrenergic stimulation induces interleukin-18 expression via beta2–AR, PI3K, Akt, IKK, and NF– kappaB. Biochem Biophys Res Commun 319: 304–311.

Asdaq, S. and Inamdar, M. (2010) Pharmacodynamic interaction of captopril with garlic in isoproterenolinduced myocardial damage in rat. Phytother Res 4: 720–725.

Chang, H., Klein, R. and Kunos, G. (1982) Selective desensitization of cardiac beta adrenoceptors by prolonged in vivo infusion of catecholamines in rats. J Pharmacol Exp Ther 221: 784–789.

Bagatini, M., Martins, C., Battisti, V., Gasparetto, D., da Rosa, C., Spanevello, R. et al. (2011) Oxidative stress versus antioxidant defenses in patients with acute myocardial infarction. Heart Vessels 26: 55–63.

Chattopadhyay, A., Biswas, S., Bandyopadhyay, D., Sarkar, C. and Datta, A. (2003) Effect of isoproterenol on lipid peroxidation and antioxidant enzymes of myocardial tissue of mice and protection by quinidine. Mol Cell Biochem 245: 43–49.

Bakan, E., Taysi, S. and Polat, M. F. (2002) Nitric oxide levels and lipid peroxidation in plasma of patients with gastric cancer. Jpn J Clin Oncol 32: 162–166. Bayir, Y., Halici, Z., Karakus, E., Albayrak, A., Ferah, I., Kabalar, E. et al. (2012) Comparing effects of lacidipine, ramipril, and valsartan against experimentally induced myocardial infarcted rats. Cardiovasc Toxicol 12: 166–174. Bhatt, D. (2012) Antiplatelet therapy following myocardial infarction in patients with diabetes. J Am Med Assoc 308: 921–922. Bianchi, C., Miccoli, R., Penno, G. and Del Prato, S. (2008) Primary prevention of cardiovascular disease in people with dysglycemia. Diabetes Care 31: S208– S214. Boden, W., Bough, E., Reichman, M., Rich, V., Young, P., Korr, K. et al. (1985) Beneficial effects of high-dose diltiazem in patients with persistent effort angina on beta-blockers and nitrates: a randomized, double–blind, placebo–controlled cross–over study. Circulation 71: 1197–1205. Brown, R., Ambler, S., Mitchell, M. and Long, C. (2005) The cardiac fibroblast: therapeutic target in myocardial remodeling and failure. Annu Rev Pharmacol Toxicol 45: 657–687. Burchill, L., Velkoska, E., Dean, R., Griggs, K., Patel, S. and Burrell, L. (2012) Combination renin-angiotensin system blockade and angiotensinconverting enzyme 2 in experimental myocardial infarction: implications for future therapeutic directions. Clin Sci (Lond) 123: 649–658. Busatto, V., Cicilini, M. and Mill, J. (1997) Increased angiotensin-converting enzyme activity in the left ventricle after infarction. Braz J Med Biol Res 30: 679–687. Carey, I., DeWilde, S., Shah, S., Harris, T., Whincup, P. and Cook, D. (2012) Statin use after first myocardial infarction in UK men and women from 1997 to 2006: Who started and who continued treatment? Nutr Metab Cardiovasc Dis 22: 400–408.

Clozel, J., Théroux, P. and Bourassa, M. (1983) Effects of diltiazem on experimental myocardial ischemia and on left ventricular performance. Circ Res 52: I120–1128. Cohen, M. and Fuster, V. (1990) Insights into the pathogenetic mechanisms of unstable angina. Haemostasis 20: 102–112. Communal, C., Singh, K., Pimentel, D. and Colucci, W. (1998) Norepinephrine stimulates apoptosis in adult rat ventricular myocytes by activation of the beta-adrenergic pathway. Circulation 98: 1329–1334. Coram, W., Olson, R., Beil, M., Cabot, C. and Weiss, G. (1987) Effects of metoprolol, alone and in combination with lidocaine, on ventricular fibrillation threshold: comparison with atenolol, propranolol, and pindolol. J Cardiovasc Pharmacol 9: 611–621. Devchand, P., Keller, H., Peters, J., Vazquez, M., Gonzalez, F. and Wahli, W. (1996) The PPAR alpha– leukotriene B4 pathway to inflammation control. Nature 384: 39–43. Dhalla, N., Elmoselhi, A., Hata, T. and Makino, N. (2000) Status of myocardial antioxidants in ischemiareperfusion injury. Cardiovasc Res 47: 446–456. Di Filippo, C., Cuzzocrea, S., Rossi, F., Marfella, R. and D’Amico, M. (2006) Oxidative stress as the leading cause of acute myocardial infarction in diabetics. Cardiovasc Drug Rev 24: 77–87. Erbel, R. and Budoff, M. (2012) Improvement of cardiovascular risk prediction using coronary imaging: subclinical atherosclerosis: the memory of lifetime risk factor exposure. Eur Heart J 33: 1201–1213. Fleckenstein, A., Janke, J., Döring, H. and Leder, O. (1974) Myocardial fiber necrosis due to intracellular Ca overload - a new principle in cardiac pathophysiology. Recent Adv Stud Cardiac Struct Metab 4: 563–580. Frangogiannis, N., Smith, C. and Entman, M. (2002) The inflammatory response in myocardial infarction. Cardiovasc Res 53: 31–47.

10 http://tac.sagepub.com

TAK531638.indd 10

Downloaded from tak.sagepub.com at LOUISIANA STATE UNIV on August 18, 2014

06/05/2014 10:50:22 AM

M Garg and D Khanna French, J., Amos, D., Williams, B., Cross, D., Elliott, J., Hart, H. et al. (1999) Effects of early captopril administration after thrombolysis on regional wall motion in relation to infarct artery blood flow. J Am Coll Cardiol 33: 139–145. Garjani, A., Andalib, S., Biabani, S., Soraya, H., Doustar, Y., Garjani, A. et al. (2011) Combined atorvastatin and coenzyme Q10 improve the left ventricular function in isoproterenol-induced heart failure in rat. Eur J Pharmacol 666: 135–141. Geng, Y., Ishikawa, Y., Vatner, D., Wagner, T., Bishop, S., Vatner, S. et al. (1999) Apoptosis of cardiac myocytes in Gsalpha transgenic mice. Circ Res 84: 34–42. Gerczuk, P. and Kloner, R. (2012) An update on cardioprotection: a review of the latest adjunctive therapies to limit myocardial infarction size in clinical trials. J Am Coll Cardiol 59: 969–978. Grimm, D., Elsner, D., Schunkert, H., Pfeifer, M., Griese, D., Bruckschlegel, G. et al. (1998) Development of heart failure following isoproterenol administration in the rat: role of the renin-angiotensin system. Cardiovasc Res 37: 91–100. Grylewski, R. (1980) Prostaglandins platelets and atherosclerosis. CRC Crit Rev Biochem 7: 291–338. Goyal, S., Arora, S., Mittal, R., Joshi, S., Nag, T., Ray, R. et al. (2009) Myocardial salvaging effect of telmisartan in experimental model of myocardial infarction. Eur J Pharmacol 619: 75–84. Halliwell, B. and Gutteridge, J. (1990) Role of free radicals and catalytic metal ions in human disease: an overview. Methods Enzymol 186: 1–85. Hanafy, S., El-Kadi, AO. and Jamali, F. (2010) Drug-disease interaction: reduced verapamil response in isoproterenol-induced myocardial injury in rats. Pharmacology 86(4): 196–202. Held, P. and Yusuf, S. (1993) Effects of beta–blockers and calcium channel blockers in acute myocardial infarction. Eur Heart J 14: 18–25. Henry, P. (1990) Atherogenesis, calcium and calcium antagonists. Am J Cardiol 66: 3I–6I. Hirsch, A., Talness, C., Heribert, S., Paul, M. and Dzau, V. (1991) Tissue-specific activation of cardiac angiotensin converting enzyme in the experimental heart failure. Circ Res 69: 475–482. Hokimoto, S., Yasue, H., Fujimoto, K., Sakata, R. and Miyamoto, E. (1995) Increasedangiotensin converting enzyme activity in left ventricular aneurysms of patients after myocardial infarction. Cardiovasc Res 29: 664–669. Hossack, K., Brown, B., Stewart, D. and Dodge, H. (1984) Diltiazem-induced blockade of sympathetically mediated constriction of normal and diseased

coronary arteries: lack of epicardial coronary dilatory effect in humans. Circulation 70: 465–471. Hussain Shaik, A., Rasool, S., Abdul Kareem, M., Krushna, G., Akhtar, P. and Devi, K. (2012) Maslinic acid protects against isoproterenol–induced cardiotoxicity in albino Wistar rats. J Med Food 15: 741–746. Iakovlev, V. (2010) Risk factors and features of the course of primary and recurrent myocardial infarction in elderly and aged men. Adv Gerontol 23: 274–280. Ishikawa, K., Miyataka, M., Kimura, A., Takeda, N., Hirano, Y., Hayashi, T. et al. (2004) Beta-blockers prevent cardiac events in Japanese patients with myocardial infarction. Circ J 68: 59–67. Iwase, M., Bishop, S., Uechi, M., Vatner, D., Shannon, R., Kudej, R. et al. (1996) Adverse effects of chronic endogenous sympathetic drive induced by cardiac GS alpha overexpression. Circ Res 78: 517–24. Izem-Meziane, M., Djerdjouri, B., Rimbaud, S., Caffin, F., Fortin, D., Garnier, A. et al. (2012) Catecholamineinduced cardiac mitochondrial dysfunction and mPTP opening: protective effect of curcumin. Am J Physiol Heart Circ Physiol 302: H665–H674. Kass, D., Bronzwaer, J. and Paulus, W. (2004) What mechanisms underlie diastolic dysfunction in heart failure? Circ Res 94: 1533–1542. Katoh, D., Mizuno, Y., Harada, E., Ito, T., Morikawa, Y., Nakagawa, H. et al. (2012) High incidence of provoked coronary spasm in the presence of a stent after myocardial infarction: therapeutic and prognostic implications. Coron Artery Dis 23: 141–145. Kaul, S. and Kapoor, N. (1989) Reversal of changes of lipid peroxide, xanthine oxidase and superoxide dismutase by cardio-protective drugs in isoproterenol induced myocardial necrosis in rats. Indian J Exp Biol 27: 625–627. Kalaycioglu, S., Sinci, V., Imren, Y. and Oz, E. (1999) Metoprolol prevents ischemia-reperfusion injury by reducing lipid peroxidation. Jpn Circ J 63(9): 718–721. Keles, M., Bayir, Y., Suleyman, H. and Halici, Z. (2009) Investigation of effects of Lacidipine, Ramipril and Valsartan on DNA damage and oxidative stress occurred in acute and chronic periods following isoproterenol–induced myocardial infarct in rats. Mol Cell Biochem 328: 109–117. Kenakin, T. and Ferris, R. (1983) Effects of in vivo beta-adrenoceptor down-regulation on cardiac responses to prenalterol and pirbuterol. J Cardiovasc Pharmacol 5: 90–97. Kondo, T., Ogawa, Y., Sugiyama, S., Ito, T., Satake, T. and Ozawa, T. (1987) Mechanism of isoproterenol

http://tac.sagepub.com 11

TAK531638.indd 11

Downloaded from tak.sagepub.com at LOUISIANA STATE UNIV on August 18, 2014

06/05/2014 10:50:22 AM

Therapeutic Advances in Cardiovascular Disease  induced myocardial damage. Cardiovasc Res 21: 248–254.

Mann, D. (1996) Stress activated cytokines and the heart. Cytokine Growth Factor Rev 7: 341–354.

Kumaran, K. and Prince, P. (2010) Caffeic acid protects rat heart mitochondria against isoproterenol– induced oxidative damage. Cell Stress Chaperones 15: 791–806.

Mann, D. (1998) Basic mechanisms of disease progression in the failing heart: the role of excessive adrenergic drive. Prog Cardiovasc Dis 41: 1–8.

Kumar, V., Asdaq, S. and Asad, M. (2009) Influence of betel quid on effect of calcium channel blockers on isoprenaline induced myocardial necrosis in mice. Indian J Exp Biol 47: 730–736.

Mann, D., Kent, R., Parsons, B. and Cooper, G. (1992) Adrenergic effects on the biology of the adult mammalian cardiocyte. Circulation 85: 790–804.

Landmesser, U., Spiekermann, S., Dikalov, S., Tatge, H., Wilke, R., Kohler, C. et al. (2002) Vascular oxidative stress and endothelial dysfunction in patients with chronic heart failure: role of xanthine-oxidase and extracellular superoxide dismutase. Circulation 106: 3073–3078.

Martín–Fernández, B., de las Heras, N., Miana, M., Ballesteros, S., Valero–Muñoz, M., Vassallo, D. et al. (2012) Spironolactone prevents alterations associated with cardiac hypertrophy produced by isoproterenol in rats: involvement of serum- and glucocorticoid-regulated kinase type 1. Exp Physiol 97: 710–718.

Libby, P. and Aikawa, M. (2002) Stabilization of atherosclerotic plaques: new mechanisms and clinical targets. Nat Med 8: 1257–1262.

McInnes, I., Liew, F. and Gracie, J. (2005) Interleukin–18: a therapeutic target in rheumatoid arthritis? Arthritis Res Ther 7: 38–41.

Libby, P. and Aikawa, M. (2003) Effects of statins in reducing thrombotic risk and modulating plaque vulnerability. Clin Cardiol 26: I11–114.

Mehdizadeh, R., Parizadeh, MR., Khooei, AR., Mehri, S. and Hosseinzadeh, H. (2013) Cardioprotective effect of saffron extract and safranal in isoproterenol-induced myocardial infarction in wistar rats. Iran J Basic Med Sci 16(1): 56–63.

Lim, KH., Ko, D. and Kim, JH. (2013) Cardioprotective potential of Korean Red Ginseng extract on isoproterenol-induced cardiac injury in rats. J Ginseng Res 37(3): 273–282. Lindpaintner, K., Lu, W., Neidermajer, N., Schieffer, B., Just, H., Ganten, D. et al. (1993) Selective activation of cardiac angiotensinogen gene expression in post-infarction ventricular remodeling in the rat. J Mol Cell Cardiol 25:133–143. Ma, C., Cao, J., Lu, X., Guo, X., Gao, Y., Liu, X. et al. (2012) Cardiovascular and cerebrovascular outcomes in elderly hypertensive patients treated with either ARB or ACEI. J Geriatr Cardiol 9: 252–257. Ma, Y., Cheng, W., Wu, S. and Wong, T. (2009) Oestrogen confers cardioprotection by suppressing Ca2+/calmodulin–dependent protein kinase II. Br J Pharmacol 157: 705–715. Madhesh., Murugesan., Vaiyapuri. and Manju. (2013) Luteolin promotes mitochondrial protection during acute and chronic periods of isoproterenol induced myocardial infarction in rats. The Egyptian Heart Journal 6(4): 319–327. Mallov, S. (1984) Effect of cardiotoxic concentrations of catecholamines on Na+-Ca2+ exchange in cardiac sarcolemmal vesicles. Exp Mol Pathol 40: 206–213.

Mohan, P. and Bloom, S. (1999) Lipolysis is an important determinant of isoproterenol-induced myocardial necrosis. Cardiovasc Pathol 8: 255–261. Murray, D., Prabhu, S. and Chandrasekar, B. (2000) Chronic beta-adrenergic stimulation induces myocardial proinflammatory cytokine expression. Circulation 101: 2338–2341. Nagano, M., Higaki, J., Nakamura, F., Higashimori, K., Nagano, N., Mikami, H. and Ogihara, T. (1992) Role of cardiac angiotensin II in isoproterenolinduced left ventricular hypertrophy. Hypertension 19: 708–712. Nagoor Meeran, M. and Stanely Mainzen Prince, P. (2012) Protective effects of N-acetyl cysteine on membrane-bound adenosine triphosphatases and minerals in isoproterenol-induced myocardialinfarcted rats: an in vivo and in vitro study. J Biochem Mol Toxicol 26: 276–281. Naik, P., Malati, T., Ratnakar, K., Naidu, M. and Rajasekhar, A. (1999) Cardioprotective effect of magnesium chloride in experimental acute myocardial infarction. Indian J Exp Biol 37: 131–137.

Manjula, T., Deepa, R. and Shyamala Devi, C. (1994) Effect of aspirin on lipid peroxidation in experimental myocardial infarction in rats. J Nutr Biochem 5: 95–98.

Ni, L., Zhou, C., Duan, Q., Lv, J., Fu, X., Xia, Y. et al. (2011) β–AR blockers suppresses ER stress in cardiac hypertrophy and heart failure. PLoS One 6: e27294.

Manjula, TS. and Devi, CS. (1993) Effect of aspirin on isoproterenol induced changes in lipid metabolism in rats. Indian J Med Res 98: 30–33.

Nichtova, Z., Novotova, M., Kralova, E. and Stankovicova, T. (2012) Morphological and functional characteristics of models of experimental

12 http://tac.sagepub.com

TAK531638.indd 12

Downloaded from tak.sagepub.com at LOUISIANA STATE UNIV on August 18, 2014

06/05/2014 10:50:22 AM

M Garg and D Khanna myocardial injury induced by isoproterenol. Gen Physiol Biophys 31: 141–151.

toxic manifestations produced by isoproterenol in the rat. AMA Arch Pathol 67: 443–455.

Niessen, H., Krijnen, P., Visser, C., Meijer, C. and Erik, H. (2003) Type II secretory phospholipase A2 in cardiovascular disease: a mediator in atherosclerosis and ischemic damage to cardiomyocytes? Cardiovasc Res 60: 68–77.

Saadane, N., Alpert, L. and Chalifour, L. (1999) Expression of immediate early genes, GATA-4, and Nkx-2.5 in adrenergic–induced cardiac hypertrophy and during regression in adult mice. Br J Pharmacol 127: 1165–1176.

Nirdlinger, E. and Bramante, P. (1974) Subcellular myocardial ionic shifts and mitochondrial alterations in the course of isoproterenol-induced cardiopathy of the rat. J Mol Cell Cardiol 6: 49–60.

Saeed, SA. and Ahmed, S. (2006) Anti-ischemic effects of nimesulide, a cyclooxygenase-2 inhibitor on the ischemic model of rabbit induced by isoproterenol. Arch Pharm Res 29(11): 977–983.

Oliveira, E. and Krieger, J. (2005) Chronic beta– adrenoceptor stimulation and cardiac hypertrophy with no induction of circulating renin. Eur J Pharmacol 520: 135–141.

Sandmann, S., Min, J., Meissner, A. and Unger, T. (1999) Effects of the calcium channel antagonist mibefradil on haemodynamic parameters and myocardial Ca(2+)-handling in infarct-induced heart failure in rats. Cardiovasc Res 44: 67–80.

Orallo, F. (1996) Regulation of cytosolic calcium levels in vascular smooth muscle. Pharmacol Ther 69: 153–171. Ott, P. and Fenster, P. (1991) Combining thrombolytic agents to treat acute myocardial infarction. Am Heart J 121: 1583–1584. Patel, D., Desai, S., Gandhi, H., Devkar, R. and Ramachandran, A. (2012) Cardio protective effect of Coriandrum sativum L. on isoproterenol induced myocardial necrosis in rats. Food Chem Toxicol 50: 3120–3125. Patel, V., Upaganlawar, A., Zalawadia, R. and Balaraman, R. (2010) Cardioprotective effect of melatonin against isoproterenol induced myocardial infarction in rats: A biochemical, electrocardiographic and histoarchitectural evaluation. Eur J Pharmacol 644: 160–168. Pérez–Cao, A., Gil–Loyzaga, P., Merchàn–Pérez, A. and Tamargo, J. (1994) Effects of oxodipine and nitrendipine on the size of experimental myocardial infarct in the rat. Pharmacol Toxicol 74: 321–329. Pinelli, A., Trivulzio, S., Tomasoni, L., Bertolini, B., Brenna, S., Bonacina, E. et al. (2004) Myocardial infarction non-invasively induced in rabbits by administering isoproterenol and vasopressin: protective effects exerted by verapamil. Fundam Clin Pharmacol 18: 657–667. Ramos, K., Combs, A. and Acosta, D. (1984) Role of calcium in isoproterenol cytotoxicity to cultured myocardial cells. Biochem Pharmacol 33: 1989–1992. Remondino, A., Kwon, S., Communal, C., Pimentel, D., Sawyer, D., Singh, K. et al. (2003) Beta– adrenergic receptor-stimulated apoptosis in cardiac myocytes is mediated by reactive oxygen species/c-Jun NH2-terminal kinase-dependent activation of the mitochondrial pathway. Circ Res 92: 136–138. Rona, G., Chappel, C., Balazs, T. and Gaudry, R. (1959) An infarct–like myocardial lesion and other

Sathish, V., Ebenezar, K. and Devaki, T. (2003) Synergistic effect of Nicorandil and Amlodipine on tissue defense system during experimental myocardial infarction in rats. Mol Cell Biochem 243: 133–138. Sethi, R., Shaom, Q., Ren, B., Saini, HK., Takeda, N. and Dhalla, NS. (2004) Changes in betaadrenoceptors in heart failure due to myocardial infarction are attenuated by blockade of reninangiotensin system. Mol Cell Biochem 263(1-2): 11–20. Singal, P., Yates, J., Beamish, R. and Dhalla, N. (1981) Influence of reducing agents on adrenochrome-induced changes in the heart. Arch Pathol Lab Med 105: 664–669. Singal, P., Kapur, N., Dhillon, K., Beamish, R. and Dhalla, N. (1982) Role of free radicals in catecholamine-induced cardiomyopathy. Can J Physiol Pharmacol 60: 1390–1397. Singal, PK., Beamish, RE. and Dhalla, NS. (1983) Potential oxidative pathways of catecholamines in the formation of lipid peroxidase and genesis of heart disease. In: myocardial injury, j. J. Spitzer (ed). Plenum publ. Corp., 391: 491. Singh, K., Communal, C., Sawyer, D. and Colucci, W. (2000) Adrenergic regulation of myocardial apoptosis. Cardiovasc Res 45: 713–719. Singh, K., Joshi, K., Somani, K. and Tariq, M. (1980) Effect of phenelzine, a monoamine oxidase inhibitor, on isoproterenol-induced myocardial damage. Adv Myocardiol 2:165–170. Somani, P., Laddu, A. and Hardm, H. (1970) Nutritional circulation in the heart. 3. Effect of isoproterenol and beta adrenergic blockade on myocardial hemodynamics and rubicium-86 extraction in the isolated supported heart preparation. J Pharmacol Exp Ther 175: 577–592.

http://tac.sagepub.com 13

TAK531638.indd 13

Downloaded from tak.sagepub.com at LOUISIANA STATE UNIV on August 18, 2014

06/05/2014 10:50:22 AM

Therapeutic Advances in Cardiovascular Disease  Song, L., Jiang, W., Qing, Y., Hu, X., Li, Y., Tong, Q. et al. (2011) The antagonistic effect of PI3K-gamma inhibitor AS605240 on cardiac hypertrophy and cardiac fibrosis induced by isoproterenol in rats. Sichuan da xue xue bao. Yi xue ban 42: 471–474. Spinarová, L., Spinar, J. and Vítovec, J. (2011) Pharmacotherapy following myocardial infarction. Vnitr Lek 57: 966–969. Spooner, P., Thai, H., Goldman, S. and Gaballa, M. (2004) Thyroid hormone analog, DITPA, improves endothelial nitric oxide and beta-adrenergic mediated vasorelaxation after myocardial infarction. J Cardiovasc Pharmacol 44: 453–459. Stanely Mainzen Prince, P. and Roy, AJ (2013) p-Coumaric acid attenuates apoptosis in isoproterenol-induced myocardial infarcted rats by inhibiting oxidative stress. Int J Cardiol 168(4): 3259–3266. Stanger, O. and Weger, M. (2003) Interactions of homocysteine, nitric oxide, folate and radicals in the progressively damaged endothelium. Clin Chem Lab Med 41:1444–1454. Stanton, H., Bowman, Z. and Cooper, C. (1970) Effects of monoamine oxidase inhibitors on isoproterenol-induced cardiomegaly in rats. Toxicol Appl Pharmacol 16: 256–263. Strauss, W., McIntyre, K., Parisi, A. and Shapiro, W. (1982) Safety and efficacy of diltiazem hydrochloride for the treatment of stable angina pectoris: report of a cooperative clinical trial. Am J Cardiol 49: 560–566. Tappia, P., Hata, T., Hozaima, L., Sandhu, M., Panagia, V. and Dhalla, N. (2001) Role of oxidative stress in catecholamine-induced changes in cardiac sarcolemmal Ca2+ transport. Arch Biochem Biophys 387: 85–92. Tappel, A. (1973) Lipid peroxidation damage to cell components. Fed Proc 32: 1870–1874. Taylor, P.B. and Tang, Q. (1984) Development of isoproterenol-induced cardiac hypertrophy. Can J Physiol Pharmacol 62: 384–389. Titus, E. (1983) A molecular biologic approach to cardiac toxicology. Adv Exp Med Biol 161: 509–18. Tokgözoğlu, L. (2009) Atherosclerosis and the role of inflammation. Turk Kardiyol Dern Ars 37: 1–6. Towart, R. and Kazda, S. (1979) The cellular mechanism of action of nimodipine (BAY e 9736), a new calcium antagonist proceeding. Br J Pharmacol 67: 409P–410P. Trivedi, C., Balaraman, R., Majithiya, J. and Bothara, S. (2006) Effect of atorvastatin treatment on

isoproterenol–induced myocardial infarction in rats. Pharmacology 77: 25–32. Vassallo, D., Vasquez, E. and Cabral, A. (1988) Contractile performance of papillary muscles of renovascular hypertensive and isoproterenolpretreated rats. Pharmacol Res Commun 20: 61–72. Vijayan, N., Thiruchenduran, M. and Devaraj, S. (2012) Anti-inflammatory and anti-apoptotic effects of Crataegus oxyacantha on isoproterenol-induced myocardial damage. Mol Cell Biochem 367: 1–8. Wang, N., Guan, P., Zhang, JP., Li, YQ., Chang, YZ., Shi, ZH., Wang, FY. and Chu, L. (2011) Fasudil hydrochloride hydrate, a Rho-kinase inhibitor, suppresses isoproterenol-induced heart failure in rats via JNK and ERK1/2 pathways. J Cell Biochem 112(7): 1920–1929. Weber, K. (1997) Extracellular matrix remodeling in heart failure: a role for de novo angiotensin II generation. Circulation 96: 4065–4082. Wexler, B. (1985) Prolonged protective effects following propranolol withdrawal against isoproterenol-induced myocardial infarction in normotensive and hypertensive rats. Br J Exp Pathol 66: 143–154. Wexler, B. and Greenberg, B. (1978) Protective effects of clofibrate on isoproterenol-induced myocardial infarction in arteriosclerotic and nonarteriosclerotic rats. Atherosclerosis 29: 373–395. Yamamoto, S., Matsui, K., Sasabe, M. and Ohashi, N. (2002) Effect of an orally active Na+/H+ exchange inhibitor, SMP-300, on experimental angina and myocardial infarction models in rats. J Cardiovasc Pharmacol 39(2): 234–241. Yuan, J., Wu, J., Hang, Z.G., Zhong, X., Zhou, L. and Yu, B. (2008) Role of peroxisome proliferatoractivated receptor alpha activation in acute myocardial damage induced by isoproterenol in rats. Chin Med J (Engl) 121: 1569–1573. Yue, T., Cheng, H., Lysko, P., McKenna, P., Feuerstein, R., Gu, J. et al. (1992) Carvedilol, a new vasodilator and beta adrenoceptor antagonist, is an antioxidant and free radical scavenger. J Pharmacol Exp Ther 263: 92–98. Yusuf, S., Hawken, S., Ounpuu, S., Dans, T., Avezum, A., Lanas, F. et al. (2004) Effect of potentially modifiable risk factors associated with myocardial infarction in 52 countries (the INTERHEART study): case-control study. Lancet 364: 937–952. Zaitone, S. and Abo–Gresha, N. (2012) Rosuvastatin promotes angiogenesis and reverses isoproterenolinduced acute myocardial infarction in rats: role of iNOS and VEGF. Eur J Pharmacol 691: 134–142.

14 http://tac.sagepub.com

TAK531638.indd 14

Downloaded from tak.sagepub.com at LOUISIANA STATE UNIV on August 18, 2014

06/05/2014 10:50:22 AM

M Garg and D Khanna Zatloukalová, L., Filipský, T., Mladěnka, P., Semecký, V., Macáková, K., Holečková, M. et al. (2012) Dexrazoxane provided moderate protection in a catecholamine model of severe cardiotoxicity. Can J Physiol Pharmacol 90: 473–484.

Zhang, G., Ohmori, K., Nagai, Y., Fujisawa, Y., Nishiyama, A., Abe, Y. et al. (2007) Role of AT1 receptor in isoproterenol-induced cardiac hypertrophy and oxidative stress in mice. J Mol Cell Cardiol 42: 804–811.

Zhang, G., Kimura, S., Nishiyama, A., Shokoji, T., Rahman, M., Yao, L. et al. (2005) Cardiac oxidative stress in acute and chronic isoproterenol–infused rats. Cardiovasc Res 65: 230–238.

Zhou, R., Xu, Q., Zheng, P., Yan, L., Zheng, J. and Dai, G. (2008) Cardioprotective effect of fluvastatin on isoproterenol-induced myocardial infarction in rat. Eur J Pharmacol 586(1-3): 244–250.

Visit SAGE journals online http://tac.sagepub.com

SAGE journals

http://tac.sagepub.com 15

TAK531638.indd 15

Downloaded from tak.sagepub.com at LOUISIANA STATE UNIV on August 18, 2014

06/05/2014 10:50:22 AM

Exploration of pharmacological interventions to prevent isoproterenol-induced myocardial infarction in experimental models.

High incidences of myocardial infarction associated with high morbidity and mortality, are a major concern and economic burden on industrialized natio...
717KB Sizes 0 Downloads 3 Views