Review Article
Mitochondria and arrhythmias

https://doi.org/10.1016/j.freeradbiomed.2014.03.033Get rights and content

Highlights

  • We review the mechanisms by which mitochondrial dysfunction causes arrhythmias.

  • Mitochondrial dysfunction leads to increased reactive oxidative species production.

  • Mitochondrial dysfunction impairs sarcolemmal and sarcoplasmic channel functions.

  • Mitochondrial dysfunction results in impaired intracellular cation homeostasis.

  • The use of mitochondria-targeted antioxidants might be a novel antiarrhythmic therapy.

Abstract

Mitochondria are essential to providing ATP, thereby satisfying the energy demand of the incessant electrical activity and contractile action of cardiac muscle. Emerging evidence indicates that mitochondrial dysfunction can adversely affect cardiac electrical functioning by impairing the intracellular ion homeostasis and membrane excitability through reduced ATP production and excessive reactive oxygen species (ROS) generation, resulting in increased propensity to cardiac arrhythmias. In this review, the molecular mechanisms linking mitochondrial dysfunction to cardiac arrhythmias are discussed with an emphasis on the impact of increased mitochondrial ROS on the cardiac ion channels and transporters that are critical to maintaining normal electromechanical functioning of the cardiomyocytes. The potential of using mitochondria-targeted antioxidants as a novel antiarrhythmia therapy is highlighted.

Introduction

The normal functioning heart requires coordinated, rhythmic electrical activity and contractile action. At rest, the heart pumps about 280 l of blood throughout the human body per hour, and the energy demand to meet this unceasing action consumes nearly 10% of the total body O2 uptake [1]. Over 90% of the cellular ATP consumed in the heart is produced by the mitochondria through oxidative phosphorylation (OXPHOS) [2]. As the predominant energy generator in the heart, mitochondria account for ~30% of the volume of cardiac cells, forming a network surrounding the sarcoplasmic reticulum (SR), myofilaments, and t-tubules [3]. It is estimated that one-third of the cardiac ATP generated by mitochondria is used for sarcolemmal and SR ion channels and transporters, which are required for the electrical activity of the cardiac cells [4]. Therefore, mitochondrial dysfunction readily disrupts the cardiac rhythm by depleting energy supply to these channels and transporters [5], [6].

In addition to producing ATP, mitochondria also generate reactive oxygen species (ROS) as a by-product of OXPHOS. It is now widely accepted that in addition to their critical bioenergetic function, mitochondria function as signaling hubs in large part by regulating redox signaling in the cell [7], [8]. Under physiological conditions, trace amount of ROS establish a network of mitochondria-driven signals that integrate metabolism with gene transcription and enzymatic activity [9], [10]. Short-term increases in ROS signals trigger adaptive responses and facilitate preconditioning, increasing cellular and tissue resistance against insult [11], [12]. On the other hand, persistently elevated ROS levels can trigger maladaptive responses and persistent abnormalities that compromise function at the molecular, cellular, and tissue levels [13], [14], [15]; in this regard, excessive production of ROS elicits pathologic changes by altering cellular function and increasing cell death [16]. Emerging evidence has shown that excessive mitochondrial ROS production can impair cardiac excitability by affecting the function of various channels and transporters through direct interaction such as posttranslational redox modification of cysteine (S-glutathionylation, sulfhydration, and S-nitrosation) or tyrosine (nitration) residues [17], [18], [19]. Excessive mitochondrial ROS can also modulate ion channel/transporter function indirectly via associated signaling molecules, such as ROS-sensitive kinases, including calcium–calmodulin-dependent protein kinase (CaMKII), cSrc, and protein kinase C (PKC), or via redox-sensitive transcription factors, such as NF-κB [20], [21], [22].

Mitochondria are also critically involved in the homeostatic regulation of cellular cations such as Ca2+, Na+, and K+, disturbance of which can have important consequences for cardiac contractility, energetics, and electrical activity [23], [24], [25]. There is a complex interrelationship between sarcolemmal and mitochondrial cation regulation. Mitochondria can take up and extrude Ca2+, for example, modulating cardiomyocyte function by serving as a dynamic buffer for sarcolemmal Ca2+ [26], [27]. Changes in sarcolemmal cation concentration, on the other hand, can influence mitochondrial structure [28], [29], energetics [30], [31], and mitochondria-dependent cell death [32]. Much of the mitochondria–sarcolemma cation interdependence is mediated by the ion channels or transporters located on the inner membrane of mitochondria (see below).

Many central metabolic systems operate totally or partially within the mitochondria. These systems dynamically regulate cellular energetic status and sarcolemmal ATP-sensitive potassium (sarcKATP) currents through oscillating mitochondrial membrane potential (∆Ψm) in response to the changes in the supply of fuel substrates and O2 [33], [34], [35]. In the presence of metabolic stress such as myocardial ischemia, depolarization of ∆Ψm diminishes mitochondrial ATP production, resulting in the opening of the sarcKATP channels, which creates a “current sink” in the myocardium, capable of slowing or blocking cardiac electrical propagation, thereby fomenting arrhythmias (see below) [33], [36].

After a brief review on the ionic basis of cardiac excitability, mitochondrial energetics/ROS production, and mitochondrial/sarcolemmal cation homeostasis, the role of mitochondrial dysfunction in influencing myocyte excitability and cardiac arrhythmogenesis is discussed, with an emphasis on the impact of mitochondrial ROS on sarcolemmal and sarcoplasmic channel/transporter functioning. In addition, the potential antiarrhythmic therapies targeting mitochondrial dysfunction in cardiac diseases are highlighted.

Section snippets

Ionic basis of cardiac excitability and contractile function

The normal contractile function of the mammalian heart depends on proper myocardial electrical activity, including the sequential activation of cells in specialized conducting systems, the normal propagation of electrical activity through the myocardium, and the generation of action potentials in individual cardiomyocytes [37], [38]. The normal cardiac cycle begins with the action potential originating in the sinoatrial node, propagating through the atria to the atrioventricular node. The

Mitochondrial energetics and ROS production

The mitochondria are organelles containing a double-membrane structure (inner and outer membranes) that creates separate compartments, the intermembrane space and the mitochondrial matrix. Mitochondria utilize glucose and fatty acids, the primary metabolic substrates for the myocardium, to generate ATP through OXPHOS. Glucose and fatty acids are sequentially oxidized to produce acetyl-CoA, the metabolic intermediate allowing the production of reducing equivalents of nicotinamide adenine

Interdependent regulation of mitochondrial and sarcolemmal cation homeostasis

As mentioned above, the sarcolemmal cation concentration is tightly controlled in cardiomyocytes by ion channels and transporters located on the plasma membrane and SR. Mitochondria also harbor ion channels and transporters. Mitochondrial cation influx and efflux not only contribute to the dynamic regulation of cytoplasmic ionic homeostasis but also play a critical role in modulating mitochondrial function.

Mitochondrial Ca2+ is crucial for the regulation of energy production, mitochondrial

Mitochondrial ROS and cardiac sodium channels

Cardiac voltage-gated Na+ (Nav) channels consist of heteromeric assembly of a pore-forming α subunit and auxiliary β subunits that modulate channel functions. Nav1.5 (SCN5A) is the major Nav α subunit expressed in the mammalian myocardium, whereas multiple Nav β subunits (Navβ1, β2, β3, β4.1, and β4.2) have been described in the cardiomyocytes [39]. Voltage-gated Na+ channels play a critical role in the membrane excitability of cardiomyocytes by generating the rapid upstroke (phase 0) of the

Cellular redox state, mitochondria, and Ca2+ homeostasis

Calcium ions are important intracellular signaling molecules, responsible for the regulation of numerous cellular processes in cardiomyocytes including excitation–contraction coupling, enzyme activity, transcription regulation, and cell death [116]. The intracellular Ca2+ levels ([Ca2+]i) fluctuate markedly between systole and diastole, yet the changes in [Ca2+]i are highly regulated. Abnormal Ca2+ handling has been implicated in the mechanical dysfunction and arrhythmogenesis observed in

ROS, mitochondria, and cardiac potassium channels

Multiple voltage-gated K+ (Kv) channels and non-voltage-gated inwardly rectifying (Kir) channels contribute to myocardial action potential repolarization [38], [39]. Functional K+ channels are integral membrane protein complexes consisting of pore-forming (α) subunits, multiple accessory (β) subunits, and regulatory proteins [38], [39]. The α subunits of Kv channels are six-transmembrane-spanning-domain (S1–S6) proteins, and functional Kv channels are composed of four α subunits. In addition, a

Mitochondrial ROS and cardiac gap junction remodeling

Gap junctions, the membrane channels formed by the assembly of a pair of hemichannels, consist of six connexin proteins, mediate the cell-to-cell communication of small metabolites and ions, and play a critical role in cardiac impulse conduction [189]. There are three major connexin isoforms expressed in the heart: connexin (Cx) 40, Cx43, and Cx45. Whereas Cx43 is extensively expressed in both the atrial and the ventricular cardiomyocytes, Cx40 is predominantly expressed in the atria and

Conclusion

In summary, mitochondrial dysfunction is prevalent in arrhythmogenic cardiac diseases, including cardiac hypertrophy, heart failure, and myocardial ischemia. Reduced ATP synthesis and increased ROS production associated with mitochondrial dysfunction can lead to malfunction of various cellular mechanisms that are required to maintain normal electrical functioning and intracellular ionic homeostasis in cardiomyocytes. As summarized in Fig. 2 and Table 1, mitochondrial dysfunction can lead to

Acknowledgment

This work was funded by National Institutes of Health Grants RO1 HL104025 (S.C.D.) and HL106592 (S.C.D.), Veterans Affairs MERIT Grant BX000859 (S.C.D.), and American Heart Association Midwest Affiliation Postdoctoral Fellowship AHA13POST14380029 (K.C.Y.); Dr. Bonini is funded by the American Heart Association (13GRNT16400010; 09SDG2250933) and the U.S. Department of Defense W911NF-12-1-0493). Dr. Dudley is an inventor of 13/551,790 “A Method for Ameliorating or Preventing Arrhythmic Risk

References (210)

  • A.J. Tompkins et al.

    Mitochondrial dysfunction in cardiac ischemia–reperfusion injury: ROS from complex I, without inhibition

    Biochim. Biophys. Acta

    (2006)
  • M. Liu et al.

    Mitochondrial dysfunction causing cardiac sodium channel downregulation in cardiomyopathy

    J. Mol. Cell. Cardiol.

    (2013)
  • D.B. Zorov et al.

    Mitochondrial ROS-induced ROS release: an update and review

    Biochim. Biophys. Acta

    (2006)
  • S.W. Werns et al.

    Myocardial glutathione depletion impairs recovery of isolated blood-perfused hearts after global ischaemia

    J. Mol. Cell. Cardiol.

    (1992)
  • J. Murray et al.

    Oxidative damage to mitochondrial complex I due to peroxynitrite: identification of reactive tyrosines by mass spectrometry

    J. Biol. Chem.

    (2003)
  • G. Paradies et al.

    The effect of reactive oxygen species generated from the mitochondrial electron transport chain on the cytochrome c oxidase activity and on the cardiolipin content in bovine heart submitochondrial particles

    FEBS Lett.

    (2000)
  • G. Szabadkai et al.

    Mitochondrial dynamics and Ca2+ signaling

    Biochim. Biophys. Acta

    (2006)
  • T. Przygodzki et al.

    Calcium ionophore A23187 action on cardiac myocytes is accompanied by enhanced production of reactive oxygen species

    Biochim. Biophys. Acta

    (2005)
  • A.J. Kowaltowski et al.

    Ca2+-stimulated mitochondrial reactive oxygen species generation and permeability transition are inhibited by dibucaine or Mg2+

    Arch. Biochem. Biophys.

    (1998)
  • L. Buntinas et al.

    The rapid mode of calcium uptake into heart mitochondria (RaM): comparison to RaM in liver mitochondria

    Biochim. Biophys. Acta

    (2001)
  • G. Beutner et al.

    Identification of a ryanodine receptor in rat heart mitochondria

    J. Biol. Chem.

    (2001)
  • B.A. Altschafl et al.

    The mitochondrial ryanodine receptor in rat heart: a pharmaco-kinetic profile

    Biochim. Biophys. Acta

    (2007)
  • R. Rizzuto et al.

    Ca2+ transfer from the ER to mitochondria: when, how and why

    Biochim. Biophys. Acta

    (2009)
  • E.K. Seppet et al.

    Functional complexes of mitochondria with Ca,MgATPases of myofibrils and sarcoplasmic reticulum in muscle cells

    Biochim. Biophys. Acta

    (2001)
  • P. Bernardi et al.

    The permeability transition pore as a Ca2+ release channel: new answers to an old question

    Cell Calcium

    (2012)
  • M. Zoratti et al.

    The mitochondrial permeability transition

    Biochim. Biophys. Acta

    (1995)
  • J. Herrington et al.

    Dominant role of mitochondria in clearance of large Ca2+ loads from rat adrenal chromaffin cells

    Neuron

    (1996)
  • B. O׳Rourke et al.

    Mitochondrial Ca2+ uptake: tortoise or hare?

    J. Mol. Cell. Cardiol.

    (2009)
  • L.A. Jelicks et al.

    Effects of hypertrophy and heart failure on [Na+]i in pressure-overloaded guinea pig heart

    Am. J. Hypertens.

    (1995)
  • P. Paucek et al.

    Reconstitution and partial purification of the glibenclamide-sensitive, ATP-dependent K+ channel from rat liver and beef heart mitochondria

    J. Biol. Chem.

    (1992)
  • B. O׳Rourke et al.

    Mitochondrial ion channels: gatekeepers of life and death

    Physiology (Bethesda)

    (2005)
  • D.A. Harris et al.

    Control of mitochondrial ATP synthesis in the heart

    Biochem. J.

    (1991)
  • J. Schaper et al.

    Ultrastructural morphometric analysis of myocardium from dogs, rats, hamsters, mice, and from human hearts

    Circ. Res.

    (1985)
  • M. Schramm et al.

    The energy expenditure of actomyosin-ATPase, Ca2+-ATPase and Na+,K+-ATPase in guinea-pig cardiac ventricular muscle

    J. Physiol.

    (1994)
  • C.L. Overend et al.

    Altered cardiac sarcoplasmic reticulum function of intact myocytes of rat ventricle during metabolic inhibition

    Circ. Res.

    (2001)
  • H.S. Silverman et al.

    Ionic basis of ischaemic cardiac injury: insights from cellular studies

    Cardiovasc. Res.

    (1994)
  • N.S. Zinkevich et al.

    ROS-induced ROS release in vascular biology: redox–redox signaling

    Am. J. Physiol. Heart Circ. Physiol

    (2011)
  • Y.F. Pung et al.

    Mitochondrial oxidative stress corrupts coronary collateral growth by activating adenosine monophosphate activated kinase-alpha signaling

    Arterioscler. Thromb. Vasc. Biol.

    (2013)
  • S.R. Sripathi et al.

    Mitochondrial–nuclear communication by prohibitin shuttling under oxidative stress

    Biochemistry

    (2011)
  • J.Z. Sun et al.

    Evidence for an essential role of reactive oxygen species in the genesis of late preconditioning against myocardial stunning in conscious pigs

    J. Clin. Invest.

    (1996)
  • A.A. Sovari et al.

    Mitochondria oxidative stress, connexin43 remodeling, and sudden arrhythmic death

    Circ. Arrhythmia Electrophysiol

    (2013)
  • P.N. Sanders et al.

    CaMKII is essential for the proasthmatic effects of oxidation

    Sci. Transl. Med.

    (2013)
  • A.M. Zafari et al.

    Free radicals in heart failure: therapeutic targets for old and new drugs

    Congest. Heart Fail

    (2002)
  • D. Sorescu et al.

    Reactive oxygen species, mitochondria, and NAD(P)H oxidases in the development and progression of heart failure

    Congest. Heart Fail

    (2002)
  • N.T. Aggarwal et al.

    Redox control of cardiac excitability

    Antioxid. Redox Signaling

    (2013)
  • M. Kawakami et al.

    Superoxide anion radical-triggered Ca2+ release from cardiac sarcoplasmic reticulum through ryanodine receptor Ca2+ channel

    Mol. Pharmacol.

    (1998)
  • L. Xu et al.

    Activation of the cardiac calcium release channel (ryanodine receptor) by poly-S-nitrosylation

    Science

    (1998)
  • A. Clerk et al.

    Stimulation of multiple mitogen-activated protein kinase sub-families by oxidative stress and phosphorylation of the small heat shock protein, HSP25/27, in neonatal ventricular myocytes

    Biochem. J.

    (1998)
  • L.L. Shang et al.

    NF-kappaB-dependent transcriptional regulation of the cardiac scn5a sodium channel by angiotensin II

    Am. J. Physiol. Cell Physiol

    (2008)
  • E. Murphy et al.

    Regulation of intracellular and mitochondrial sodium in health and disease

    Circ. Res.

    (2009)
  • Cited by (0)

    View full text