Review Article
Cardioprotective Function of Inducible Nitric Oxide Synthase and Role of Nitric Oxide in Myocardial Ischemia and Preconditioning: an Overview of a Decade of Research

https://doi.org/10.1006/jmcc.2001.1462Get rights and content

Abstract

Over the past decade, an enormous number of studies (>100) have focused on the role of nitric oxide (NO) in myocardial ischemia. It is important to distinguish the function of NO in unstressed (non-preconditioned) myocardium from its function in preconditioned myocardium (i.e. myocardium that has shifted to a defensive phenotype in response to stress). Of the 92 studies that have examined the role of NO in modulating the severity of ischemia/reperfusion injury in non-preconditioned myocardium, the vast majority [67 (73%)] have concluded that NO (either endogenous or exogenous) has a protective effect and only 11 (12%) found a detrimental effect. The proportion of studies supporting a cytoprotective role of NO is similar in vivo[35 (71%) out of 49] and in vitro[32 (74%) out of 43]. With regard to the delayed acquisition of tolerance to ischemia [late preconditioning (PC)], overwhelming evidence indicates a critical role of NO in this phenomenon. Specifically, enhanced biosynthesis of NO by eNOS is essential to trigger the late phase of ischemia-induced and exercise-induced PC, and enhanced NO production by iNOS is obligatorily required to mediate the anti-stunning and anti-infarct actions of late PC elicited by five different stimuli (ischemia, adenosine A1agonists, opioidδ1 agonists, endotoxin derivatives and exercise). Thus, NO plays a dual role in the pathophysiology of the late phase of PC, acting initially as the trigger and subsequently as the mediator of this adaptive response (“NO hypothesis of late PC”). The diversity of the PC stimuli that converge on iNOS implies that the upregulation of this enzyme is a central mechanism whereby the myocardium protects itself from ischemia. The NO hypothesis of late PC has thus revealed a cytoprotective function of iNOS in the heart, a novel paradigm which has recently been extended to other tissues, including kidney and intestine. Other corollaries of this hypothesis are that the heart responds to stress in a biphasic manner, utilizing eNOS as an immediate but short-term response and iNOS as a delayed but long-term defense, and that the fundamental difference between non-preconditioned and late preconditioned myocardium is the tissue level of iNOS-derived NO, which is tonically higher in the latter compared with the former. Hence, late PC can be viewed as a state of enhanced NO synthesis. The NO hypothesis of late PC has important therapeutic implications. In experimental animals, administration of NO donors in lieu of ischemia can faithfully reproduce the molecular and functional aspects of ischemia-induced late PC, indicating that NO is not only necessary but also sufficient to induce late PC. The recent demonstration that nitroglycerin also induces late PC in patients provides proof-of-principle for the concept that nitrates could be used as a PC-mimetic therapy for the prophylaxis of ischemic injury in the clinical arena. This novel application of nitrates could be as important as, or perhaps even more important than, their current use as antianginal and preload-reducing agents. In addition, gene transfer of either eNOS or iNOS has been shown to replicate the infarct-sparing actions of ischemic PC, suggesting that NOS gene therapy could be an effective strategy for alleviating ischemia/reperfusion injury. Ten years of research have demonstrated that NO plays a fundamental biological role in protecting the heart against ischemia/reperfusion injury. The time has come to translate this enormous body of experimental evidence into clinically useful therapies by harnessing the cytoprotective properties of NO.

References (187)

  • A Mizuno et al.

    Endothelial stunning and myocyte recovery after reperfusion of jeopardized muscle: a role ofl-arginine blood cardioplegia

    J Thorac Cardiovasc Surg

    (1997)
  • R Lee et al.

    Retrograde infusion of lidocaine orl-arginine before reperfusion reduces myocardial infarct size

    Ann Thorac Surg

    (1998)
  • A Kis et al.

    Repeated cardiac pacing extends the time during which canine hearts are protected against ischaemia-induced arrhythmias: role of nitric oxide

    J Mol Cell Cardiol

    (1999)
  • D Wang et al.

    Reduction of myocardial infarct size by inhibition of inducible nitric oxide synthase

    Am J Hypertens

    (1999)
  • H Post et al.

    No involvement of endogenous nitric oxide in classical ischemic preconditioning in swine

    J Mol Cell Cardiol

    (2000)
  • SA Naseem et al.

    Sustained inhibition of nitric oxide by NG-nitro-l-arginine improves myocardial function following ischemia/reperfusion in isolated perfused rat heart

    J Mol Cell Cardiol

    (1995)
  • K Takeuchi et al.

    Direct detrimental effects ofl-arginine upon ischemia-reperfusion injury to myocardium

    J Mol Cell Cardiol

    (1995)
  • T Hiramatsu et al.

    Effect ofl-arginine cardioplegia on recovery of neonatal lamb hearts after 2 hours of cold ischemia

    Ann Thorac Surg

    (1995)
  • T Hiramatsu et al.

    Additive effects ofl-arginine infusion and leukocyte depletion on recovery after hypothermic ischemia in neonatal lamb hearts

    J Thorac Cardiovasc Surg

    (1995)
  • FS Wanna et al.

    Simultaneous manipulation of the nitric oxide and prostanoid pathways reduces myocardial reperfusion injury

    J Thorac Cardiovasc Surg

    (1995)
  • DT Engelman et al.

    Constitutive nitric oxide release is impaired after ischemia and reperfusion

    J Thorac Cardiovasc Surg

    (1995)
  • E Bugge et al.

    Bradykinin protects against infarction but does not mediate ischemic preconditioning in the isolated rat heart

    J Mol Cell Cardiol

    (1996)
  • P Ferdinandy et al.

    Loss of pacing-induced preconditioning in rat hearts: role of nitric oxide and cholesterol-enriched diet

    J Mol Cell Cardiol

    (1997)
  • P Massoudy et al.

    Cardioprotection by cyclosporine A in experimental ischemia and reperfusion—evidence for a nitric oxide-dependent mechanism mediated by endothelin

    J Mol Cell Cardiol

    (1997)
  • Z Luo et al.

    Vascular endothelial growth factor attenuates myocardial ischemia-reperfusion injury

    Ann Thorac Surg

    (1997)
  • F Brunner

    Interaction of nitric oxide and endothelin-1 in ischemia/reperfusion injury of rat heart

    J Mol Cell Cardiol

    (1997)
  • U Flogel et al.

    Contribution of NO to ischemia-reperfusion injury in the saline-perfused heart: a study in endothelial NO synthase knockout mice

    J Mol Cell Cardiol

    (1999)
  • A Nakano et al.

    Exogenous nitric oxide can trigger a preconditioned state through a free radical mechanism, but endogenous nitric oxide is not a trigger of classical ischemic preconditioning

    J Mol Cell Cardiol

    (2000)
  • MS Sumeray et al.

    Infarct size and nitric oxide synthase in murine myocardium

    J Mol Cell Cardiol

    (2000)
  • H Horimoto et al.

    The role of nitric oxide, K+(ATP) channels, and cGMP in the preconditioning response of the rabbit

    J Surg Res

    (2000)
  • RL Hannan et al.

    Deletion of endothelial nitric oxide synthase exacerbates myocardial stunning in an isolated mouse heart model

    J Surg Res

    (2000)
  • MV Cohen et al.

    Ischemic preconditioning: from adenosine receptor to KATPchannel

    Annu Rev Physiol

    (2000)
  • R Bolli

    The late phase of preconditioning

    Circ Res

    (2000)
  • G 3rd Johnson et al.

    Cardioprotective effects of acidified sodium nitrite in myocardial ischemia with reperfusion

    J Pharmacol Exp Ther

    (1990)
  • G 3rd Johnson et al.

    Cardioprotective effects of authentic nitric oxide in myocardial ischemia with reperfusion

    Crit Care Med

    (1991)
  • AS Weyrich et al.

    The role ofl-arginine in ameliorating reperfusion injury after myocardial ischemia in the cat

    Circulation

    (1992)
  • MR Siegfried et al.

    Cardioprotection and attenuation of endothelial dysfunction by organic nitric oxide donors in myocardial ischemia-reperfusion

    J Pharmacol Exp Ther

    (1992)
  • K Nakanishi et al.

    Intracoronaryl-arginine during reperfusion improves endothelial function and reduces infarct size

    Am J Physiol

    (1992)
  • G Matheis et al.

    Role ofl-arginine-nitric oxide pathway in myocardial reoxygenation injury

    Am J Physiol

    (1992)
  • Z Yao et al.

    Role of nitric oxide, muscarinic receptors, and the ATP-sensitive K+ channel in mediating the effects of acetylcholine to mimic preconditioning in dogs

    Circ Res

    (1993)
  • DJ Lefer et al.

    Endothelial and myocardial cell protection by a cysteine-containing nitric oxide donor after myocardial ischemia and reperfusion

    J Cardiovasc Pharmacol

    (1993)
  • DJ Lefer et al.

    Antineutrophil and myocardial protecting actions of a novel nitric oxide donor after acute myocardial ischemia and reperfusion of dogs

    Circulation

    (1993)
  • N Hasebe et al.

    Inhibition of endothelium-derived relaxing factor enhances myocardial stunning in conscious dogs

    Circulation

    (1993)
  • JC Hartman et al.

    Inhibition of nitric oxide synthase prevents myocardial protection by ramiprilat

    J Pharmacol Exp Ther

    (1994)
  • J Pernow et al.

    The protective effect ofl-arginine on myocardial injury and endothelial function following ischaemia and reperfusion in the pig

    Eur Heart J

    (1994)
  • T Ehring et al.

    Attenuation of myocardial stunning by the ACE inhibitor ramiprilat through a signal cascade of bradykinin and prostaglandins but not nitric oxide

    Circulation

    (1994)
  • V Richard et al.

    Myocardial and coronary endothelial protective effects of acetylcholine after myocardial ischaemia and reperfusion in rats: role of nitric oxide

    Br J Pharmacol

    (1995)
  • S Hoshida et al.

    Nitric oxide synthase protects the heart against ischemia-reperfusion injury in rabbits

    J Pharmacol Exp Ther

    (1995)
  • MW Williams et al.

    Endogenous nitric oxide (NO) protects against ischaemia-reperfusion injury in the rabbit

    Cardiovasc Res

    (1995)
  • R Pabla et al.

    Intracoronary nitric oxide improves postischemic coronary blood flow and myocardial contractile function

    Am J Physiol

    (1995)
  • Cited by (570)

    • Exercise-induced late preconditioning in mice is triggered by eNOS-dependent generation of nitric oxide and activation of PKCε and is mediated by increased iNOS activity

      2021, International Journal of Cardiology
      Citation Excerpt :

      This adaptation consists of an early phase, which begins within minutes but abates after 1–2 h, and a late phase, which begins 12–24 h after the stimulus and lasts for ~72 h. [1–4] Several studies have shown that a delayed cardioprotective effect similar to that induced by ischemia can be elicited by administration of various pharmacologic agents. [5–15] Although PC could be an attractive mechanism to protect myocardium in humans, [3,16–20] ischemic PC is not clinically feasible while pharmacologic PC is limited by side effects and practical considerations. [3,19,20] Recent evidence indicates that brief exposure to physical exercise also elicits a late PC effect. [21–30]

    View all citing articles on Scopus

    Please address all correspondence to: Roberto Bolli, MD, Division of Cardiology, University of Louisville, Louisville, Kentucky 40292, USA. Fax: (502) 852-6454;E-mail: [email protected]

    View full text