5161

J Physiol 594.18 (2016) pp 5161–5183

TO P I C A L R E V I E W

Mitochondria in the middle: exercise preconditioning protection of striated muscle John M. Lawler, Dinah A. Rodriguez and Jeffrey M. Hord Redox Biology & Cell Signalling Laboratory, Department of Health and Kinesiology, Graduate Faculty of Nutrition & Food Science, Texas A&M University, College Station, TX, USA Sarcolemma

Nox

The Journal of Physiology

NF-κB ROS

XO

δ-Opioid receptor

Exercise preconditioning (EPC)

Gene expression HIF-1, Nrf2, etc. Nucleus

Post-translational modifications

• Cytosolic antioxidants • Heat shock proteins • Nitric oxide signaling

Mitochondrial adaptations: • MnSOD • Trx2 • ICDH-NADP • mitoKATP channels • HSP70 • SIRT-1/PGC-1α

MAPK

Mitochondria Blood vessel

Abstract Cellular and physiological adaptations to an atmosphere which became enriched in molecular oxygen spurred the development of a layered system of stress protection, including antioxidant and stress response proteins. At physiological levels reactive oxygen and nitrogen species regulate cell signalling as well as intracellular and intercellular communication. Exercise and physical activity confer a variety of stressors on skeletal muscle and the cardiovascular system: mechanical, metabolic, oxidative. Transient increases of stressors during acute bouts of

Dinah A. Rodriguez, John M. Lawler, and Jeffrey M. Hord work in the Department of Health and Kinesiology at Texas A&M University (USA). John Lawler is a Professor in the Department of Health and Kinesiology and Director of the Redox Biology & Cell Signaling Laboratory, and leads the research group on projects focusing on the production of reactive oxygen species (ROS) and the resulting ROS-related cellular signalling pathways in striated muscle. Dinah Rodriguez is currently a Masters student. Jeffrey Hord is currently a Doctoral candidate whose research has primarily focused on understanding the redox-sensitive cell signalling pathways involved in disuse-induced skeletal muscle atrophy.

 C 2016 The Authors. The Journal of Physiology  C 2016 The Physiological Society

DOI: 10.1113/JP270656

5162

J. M. Lawler and others

J Physiol 594.18

exercise or exercise training stimulate enhancement of cellular stress protection against future insults of oxidative, metabolic and mechanical stressors that could induce injury or disease. This phenomenon has been termed both hormesis and exercise preconditioning (EPC). EPC stimulates transcription factors such as Nrf-1 and heat shock factor-1 and up-regulates gene expression of a cadre of cytosolic (e.g. glutathione peroxidase and heat shock proteins) and mitochondrial adaptive or stress proteins (e.g. manganese superoxide dismutase, mitochondrial KATP channels and peroxisome proliferator activated receptor γ coactivator-1 (PGC-1)). Stress response and antioxidant enzyme inducibility with exercise lead to protection against striated muscle damage, oxidative stress and injury. EPC may indeed provide significant clinical protection against ischaemia–reperfusion injury, Type II diabetes and ageing. New molecular mechanisms of protection, such as δ-opioid receptor regulation and mitophagy, reinforce the notion that mitochondrial adaptations (e.g. heat shock proteins, antioxidant enzymes and sirtuin-1/PGC-1 signalling) are central to the protective effects of exercise preconditioning. (Received 29 September 2015; accepted after revision 1 April 2016; first published online 6 April 2016) Corresponding author J. M. Lawler: 213A Heldenfels Hall, Department of Health and Kinesiology, Graduate Faculty of Nutrition, Texas A&M University, College Station, TX 77843-4243, USA. Email: [email protected] Abstract figure legend Mild, non-exhaustive exercise results in a rise in reactive oxygen species (ROS) that leads to cellular adaptations capable of offering defence against ischemia–reperfusion (I/R) injury in striated muscle fibres. These exercise-induced cardiac and skeletal muscle adaptations are referred to as exercise preconditioning (EPC). EPC induces ROS production from mitochondria, NADPH oxidase (Nox) and xanthine oxidase (XO). The ROS produced from these oxidant sources leads to increased expression and post-translational modifications of cytosolic antioxidants and cytosolic HSPs, and enhanced nitric oxide signalling. Mitochondrial adaptations such as increased MnSOD, Trx2, NADP-specific isocitrate dehydrogenase (ICDH-NADP), mitochdonrial KATP (mitoKATP ) channels, HSP70 and SIRT-1/PGC-1α are also outcomes of EPC. Another potential limb of EPC involves mitochondrial adaptations being driven by δ-opiod receptor activation and subsequent P38/MAPK signalling. Abbreviations AMPK, adenosine monophosphate kinase; BNIP3, Bcl2/adenovirus E1B 19 kDa interacting protein 3; Cu/ZnSOD, copper/zinc superoxide dismutase; EC-SOD, extracellular superoxide dismutase; EPC, exercise preconditioning; ETC, electron transport chain; FoxO3a, forkhead box O3a; GAS, general adaptation syndrome; GPX, glutathione peroxidase; GSH, reduced glutathione; GSSG, glutathione disulfide; HSF-1, heat shock factor-1; HSP, heat shock protein; I-κB, inhibitor of kappa B; IPC, ischemic preconditioning; I/R, ischaemia–reperfusion; KATP , ATP-sensitive K+ channel; LC3, microtubule associated protein light chain 3; MAPK, mitogen-activated protein kinases; mdx, mouse model of Duchenne/Becker muscular dystrophy; mitoKATP , mitochondrial ATP-sensitive K+ channel; MnSOD, manganese superoxide dismutase; NF-κB, nuclear factor kappa B; Nox, nicotinamide adenine dinucleotide phosphate oxidase; Nrf, nuclear factor-erythroid-2 p45-related factor; PGC-1, peroxisome proliferator activated receptor γ coactivator-1; PKC, protein kinase C; ROS, reactive oxygen species; sarcKATP , sarcolemmal ATP-sensitive K+ channel; SERCA, sarco/endoplasmic reticulum calcium ATPase; SIRT, sirtuin; SOD, superoxide dismutase; TGF-β, transforming growth factor beta; TLR-4, toll-like receptor 4; TNF-α, tumor necrosis factor-alpha; Trx, thioredoxin reductase; VSMC, vascular smooth muscle cell.

Introduction

The general adaptation syndrome (GAS) described the ability of an organism to cope acutely or chronically with dynamic changes in stressors. The general adaptation syndrome was first articulated by Sir Hans Selye (Selye, 1946), an Austrian-Canadian endocrinologist and physician of Hungarian descent, who published over 1700 articles (Neylan, 1998). GAS is a non-specific physiological response that predicts cells, tissues and organisms can experience three stages of response when a stressor is increased: alarm, resistance or adaptation, and exhaustion. While GAS initially pertained to ‘fight or flight’ as well as cortisol effects in patients recovering

from illness or surgery, the theory broadly explains adaptations to environmental stressors (e.g. cold, heat and altitude), changes in mechanical stress and increased metabolic stress. Research over the past 30 years is consistent with the notion that response and adaptations to oxidative stress also follow the tenets of the GAS (de Magalhaes & Church, 2006; Lawler & Hindle, 2011). For example, cells and integrative biological systems respond to moderate redox challenges by increasing antioxidant enzymes and other protective proteins (e.g. heat shock proteins) (Das et al. 1993; Omar & Pappolla, 1993; Radak et al. 2008). A bout of exercise increases mechanical stress, metabolic stress and oxidative stress, which could be injurious at high  C 2016 The Authors. The Journal of Physiology  C 2016 The Physiological Society

J Physiol 594.18

Exercise preconditioning protection of striated muscle

levels of intensity. Thus it is not surprising that levels of many proteins involved in protection against stressors are increased either acutely or as an adaptation to repeated or habitual bouts of exercise in skeletal muscle and the heart (Locke et al. 1995; Samelman, 2000; Khassaf et al. 2001; Kim et al. 2015b). Biological response and adaptations to stressors experienced with exercise provide protection against potential injury or cellular damage in a future bout of exercise or stress. Thus a cellular objective is to maintain homeostasis within a desired cellular and organism tolerance. ‘Preconditioning’ is a term used in biology and physiology to describe a process where small doses of a potential hazardous stimulus, organism, or compound evoke an adaptation and increased resistance to the noxious stimulus. For example, mild ischaemia provokes production of a cluster of stress response proteins (e.g. hypoxia-inducible factor 1a, endothelial nitric oxide synthase, vascular endothelial growth factor, heat shock proteins, manganese superoxide dismutase) that provide protection against damage and cell death (Heads et al. 1995; Hamilton et al. 2003; Starnes et al. 2003). An acute bout of exercise increases skeletal muscle and cardiac levels of reactive oxygen species (ROS) and produces oxidative stress. The term ‘exercise preconditioning’ was first evident in the literature in an article by Radak et al. (2000), where the authors described resistance to oxidative stress in the myocardium induced by exercise training. tert-Butyl hydroperoxide was used as an oxidant challenge over 3 weeks. Exercise training reduced protein carbonyls as a marker of oxidative stress and peptide activity of the proteasome. DT-diaphorase, which maintains coenzyme Q in its reduced or antioxidant state, was increased by exercise and was responsive to tert-butyl hydroperoxide in the exercise group. Exercise preconditioning is now recognized to be critical in not only reducing oxidative stress during exercise, but also mitigating oxidative stress and damage due to ischaemia–reperfusion, reducing glucose intolerance and insulin resistance, as well as attenuating skeletal muscle atrophy (Radak et al. 2000; Ding et al. 2005; Dupont-Versteegden et al. 2006; Fontana et al. 2010). Therefore, the purpose of this review is to examine endogenous antioxidant systems that are (a) inducible with exercise and (b) integral to exercise preconditioning. The review begins with a brief overview of antioxidant enzyme systems and heat shock proteins, and their up-regulation with exercise. The concept of ‘hormesis’ is visited and applied to oxidative stress induction of a signalling cascade with exercise that leads to enhanced stress protection. Exercise preconditioning appears to be a central mechanism for protection against ischaemia–reperfusion, insulin resistance, ageing and mitophagy.

 C 2016 The Authors. The Journal of Physiology  C 2016 The Physiological Society

5163

Antioxidant and stress protection in skeletal muscle and heart

In order to regulate and manage reactive oxygen species and oxidative stress, a posse of antioxidant enzymes evolved when cyanobacteria or blue-green algae released a poisonous gas, oxygen, in runaway quantities into the oceans about 2.3 billion years ago. As oceans released dissolved oxygen into the atmosphere, the proportion of O2 reached levels approached 20% (0.2 atm) about 800 million years ago (Holland, 2006). While oxygen made extinct the vast majority of anaerobic life, high levels of gaseous molecular oxygen (O2 ) allowed, in time, far more complex multicellular organisms to evolve. Tiny creatures that could harness oxygen for energy, which became mitochondria, were incorporated into cells. Utilizing electrons from oxygen for metabolism, organisms adapted to a new environment where cells ‘play with fire,’ and utilize electrons from oxygen for metabolism, movement, cell signalling and tissue remodelling. Single electron reduction of oxygen from O2 yields superoxide anions via a number of cellular sources. Endogenous sources of ROS in striated muscle include the electron transport chain, NADPH oxidases (e.g. Nox2 and Nox4), xanthine oxidase, 5-lipoxygenase, prostaglandin synthesis, peroxisomes and myeloperoxidase. ROS production in striated muscle. The distribution of

ROS, high reactivity, exposure to antioxidant buffering and specificity of sites of production lead to subcellular redox microenvironments. Mitochondria have been long considered a primary cellular source of ROS in striated muscle. Complex I and complex III of the electron transport chain (ETC) can ‘leak’ 1–3% of electron flow through the ETC as superoxide anions (O2 •– ) in the state 4 or resting state (Boveris & Chance, 1973). Because of the high density of protons abutting the inside membrane of the mitochondria, it is nearly impossible for O2 •– to cross through mitochondrial membrane to the cytosol without becoming protonated (Boveris & Chance, 1973). Thus ROS released from mitochondria do so in the form of hydrogen peroxide (H2 O2 ), or peroxynitrite (OONO− ) when superoxide anion reacts with nitric oxide (•NO). In contrast to ETC leakage of ROS in state 3, active contracting skeletal muscle features a far more coupled electronic flux through the ETC, and thus the production of O2 •– dwindles to a stingy leakage of superoxide anions. Increased mitochondrial H2 O2 release during acute exercise was recently linked with forkhead box O3a (FoxO3a) up-regulation (Wang et al. 2015). However, studies from Jackson and colleagues suggest a minor role for mitochondrial ROS when healthy, unfatigued muscle fibres are contracting (Sakellariou et al. 2013, 2014; Claflin et al. 2015).

5164

J. M. Lawler and others

However, mitochondrial and microsomal release of ROS become predominant with chronic disease and metabolic disorders (Anderson et al. 2009; Kane et al. 2010; Bikman et al. 2010a; Muoio & Neufer, 2012). Insulin-resistance and Type II diabetes result in a profound release of H2 O2 from mitochondria in animal and human studies (Anderson et al. 2009). In addition, high fat meals result in a large spike of ROS release from both mitochondria and microsomes in skeletal muscle from Zucker rats (Noland et al. 2007). A deluge of lipids from a high fat diet into mitochondria tasks lipid transporters and increases oxidative stress in mitochondria. Microsomes attempt to handle the overflow of lipids, but the consequence is a significant release of ROS (Noland et al. 2007). NADPH oxidase (Nox) is a family of membrane-bound oxidoreductases that produce superoxide anions in response to stretch, contractions, hypoxia, insulin signalling and damage in non-phagocytic and phagocytic cells (Fisher, 2009; Khairallah et al. 2012). The membrane-bound Nox2 isoform of NADPH oxidase responds to stretch and skeletal muscle contractions by releasing ROS (Sakellariou et al. 2014). Jackson and colleagues (Sakellariou et al. 2013) demonstrated, using overexpression of a mitochondria-specific catalase and single fibre experiments, that Nox2 is responsible for most of the ROS released in a stretched or contracting fibre. In healthy, young skeletal muscle, mitochondria appear to contribute little to ROS released during muscle contractions (Sakellariou et al. 2013). Nox2 becomes hyper-responsive to stretch in skeletal muscles of mdx (mouse model of Duchenne/Becker muscular dystrophy) mice, producing excessive ROS (termed ‘X-ROS’) (Prosser et al. 2011; Khairallah et al. 2012). Indeed, Nox2 and X-ROS amplified limb muscle pathology via the stretch-activated Ca2+ channel TRPC1 (transient receptor potential channel 1; Whitehead et al. 2010; Prosser et al. 2011; Khairallah et al. 2012; Kim & Lawler, 2012). Hyper-responsiveness to stretch by Nox2 was dependent upon microtubule conformation (Khairallah et al. 2012) via an unknown mechanism. Hyper-responsiveness of Nox2 to stretch may also be a commonality among limb girdle muscular dystrophies when membrane proteins (e.g. dysferlin) are mutated (Kombairaju et al. 2014). Nox2 also appears hyper-sensitive and up-regulated in skeletal muscle during mechanical unloading (Lawler et al. 2014). However, the mechanism of unloading-induced elevation of Nox2 is currently unknown. Nox2 may also activate pro-fibrotic transforming growth factor β (TGF-β) as well as matrix metalloproteinase-9, thus playing a role in remodelling (Wang et al. 2010). NADPH oxidase 4 (Nox4) has recently been shown to be present in both the heart and skeletal muscle. Nox4 has been shown to regulate a pO2 -dependent Ca2+

J Physiol 594.18

release by ryanodine receptor calcium channels in the sarcoplasmic reticulum (Sun et al. 2011). In addition, Nox4 can stimulate overloading-induced hypertrophy with neuronal nitric oxide synthase by activating the non-voltage-dependent Ca2+ channel trpv1 (transient receptor potential channel, subfamily V, type 1) (Ito et al. 2013). Increased levels of Nox4 were recently detected in the heart of dystrophin-deficient mice (Spurney et al. 2008), and appear to be contributory to cardiac fibrosis and pathology with Duchenne muscular dystrophy (Spurney et al. 2008). Superoxide anions and hydrogen peroxide can also function as substrates for Fenton and Haber–Weiss reactions, catalysed by transition metals (iron, copper). The Haber–Weiss reaction generates hydroxyl radicals (•OH), which are highly reactive and promiscuous, oxidizing neighbouring molecules (Halliwell, 1995). Lipid peroxidation, a chain reaction of oxidation in lipid membranes, can be initiated by hydroxyl radicals. Hydroperoxides, lipid radicals and aldehydes are generated via lipid peroxidation and can damage proteins, DNA and carbohydrates. Cellular antioxidant protection. In order to regulate ROS

levels and their participation in cell signalling, an antioxidant system of scavengers and enzymes serves to reduce potential pathological effects of high levels or exposure to ROS. Non-enzymatic, aqueous antioxidant scavengers include reduced glutathione (GSH), urate, coenzyme Q, ubiquinone and ascorbate (vitamin C) (Powers & Hamilton, 1999). Lipid soluble cellular and serum antioxidants include β-carotene, retinol and α-tocopherol (vitamin E) (Powers & Hamilton, 1999). Thus antioxidant capacities of the cytosol and lipid membrane microenvironments (e.g. plasma membrane and mitochondrial) are bolstered via scavenger molecules. An integrative system of antioxidant enzymes serves to maintain reactive species in a range that (a) promotes physiological cell signalling and (b) minimizes oxidative damage and pathology. An imbalance between sources of ROS and antioxidant enzyme and scavengers yields high oxidative stress and oxidative damage. A family of superoxide dismutases (SOD) removes O2 •– as a reactant and produces H2 O2 . A copper, zinc-dependent isoform of superoxide dismutase (Sod1 or Cu/ZnSOD) sequesters cytosolic O2 •– and yields H2 O2 . Sod2 (also termed MnSOD) is a manganese-dependent, mitochondrially localized isoform that catalyses the reduction of O2 •– to H2 O2 . An extracellular superoxide dismutase (Sod3 or EC-SOD) also exists and also uses copper and zinc as a co-factor. EC-SOD is bound to the extracellular matrix of vascular and other tissues. EC-SOD binds to heparin sulfate moieties of proteoglycan aggregates and collagen in the extracellular matrix.  C 2016 The Authors. The Journal of Physiology  C 2016 The Physiological Society

J Physiol 594.18

Exercise preconditioning protection of striated muscle

Hydrogen peroxide and hydroperoxides are removed by GSH, protein thiol groups and a layered system of enzymes that include catalase, glutathione peroxidases, thioredoxins, peroxiredoxins and glutaredoxins. Reduced glutathione sequesters hydroperoxides and nitric oxide, and regulates cell signalling stimulated by reactive species. Catalase catalyses the decomposition of H2 O2 to O2 and H2 O with high flux. A single catalase molecule converts millions of molecules of hydrogen peroxide to water and oxygen per second (Goodsell, 2004). Catalase is a large tetramer with each subunit powered by a porphyrin haem group, and is usually located in peroxisomes, although peroxiredoxins also scavenge significant H2 O2 . Peroxisomes sequester H2 O2 to protect the remainder of the cell. While genetic ablation of catalase does not adversely affect development, a reflection of the redundant nature of enzymes that remove hydroperoxides, catalase deficiency has been linked to an increased risk of Type II diabetes (Goth, 2008) and appearance of grey hair (Wood et al. 2009). In contrast, a transgenic mouse that expresses a mitochondria-specific catalase has an increased lifespan, suggesting increased H2 O2 accelerates the ageing process (Schriner & Linford, 2006). A family of glutathione peroxidases remove H2 O2 and hydroperoxides; they are located in the cytosol and mitochondria and can be selenium dependent or independent. There are eight isoforms of glutathione peroxidase (GPX), GPX1–8 (Bermingham et al. 2014). The preferred substrate for selenium-dependent GPX1 is H2 O2 . GPX4 resides in a phospholipid environment and is critical to the removal of lipid peroxides. GPXs use for reducing power GSH, which is regenerated from glutathione disulfide (GSSG) by glutathione reductase. In mammalian skeletal muscle and cardiac tissue, NADPH needed for regeneration of GSH via glutathione reductase is produced by NADPH-specific isocitrate dehydrogenase and glyceraldehyde-3-phosphate dehydrogenase (Lawler et al. 1993a). Thioredoxin reductases are similar in structure to glutathione reductase and are important catalytic proteins for reducing thioredoxin; they remove hydroperoxides in the cytosol and mitochondria and participate in cell signalling. In skeletal muscle and heart, thioredoxin reductase (Trx) 1 protects cytosolic components while Trx2 guards mitochondria against damaging hydroperoxides and aldehydes. Thioredoxin contains dithiol-disulfide active sites and facilitates cysteine thiol-disulfide exchange. Overexpression of thioredoxin reduces chronic inflammation and increases lifespan (Yoshida et al. 2005). Peroxiredoxins and glutaredoxins also sequester cellular hydroperoxides that use GSH, ascorbate, or thioredoxin as reducing molecules. There are six peroxiredoxin isoforms that control cytokine-induced peroxides, mediate cell signalling, and regulate circadian rhythms. All  C 2016 The Authors. The Journal of Physiology  C 2016 The Physiological Society

5165

peroxiredoxins target H2 O2 , with peroxiredoxin-4 present specifically in mitochondria (Nonn et al. 2003). Glutaredoxins are small proteins important in supporting hydroperoxide removal by reducing oxidized ascorbate, peroxiredoxins and methionine sulfoxide reductase (Fernandes & Holmgren, 2004). Glutaredoxin’s disulfide bond is reduced via GSH oxidation, and participates in transport of iron–sulfur clusters to target proteins as needed (Rouhier et al. 2008). Glutaredoxin-2 exists in mammals as cytosolic and mitochondrial isoforms. Heat shock proteins. Heat shock proteins (HSPs) are stress-protective proteins that serve (a) as chaperones of nascent or partially oxidized proteins, (b) to reduce skeletal muscle proteolysis, (c) in recycling old proteins, (d) in nitric oxide signalling, (e) in the immune response, and (f) in repair and regeneration (Schlesinger, 1990). Heat shock proteins are responsive or inducible with a variety of stressors including heat, cold, mechanical stress, metabolic stress and exercise (Kregel, 2002). Expression of many of the HSPs is regulated by the transcription factor heat shock factor-1 (HSF-1). Most of the HSPs are named in accordance with their molecular mass in kilodaltons: HSP20, HSP27, HSP60, HSP70 and HSP90. However, αB-Crystallin and other small molecular mass HSPs are increasingly recognized in cell signalling and stress protection (Martindale & Holbrook, 2002). Many of the HSPs are up-regulated with acute and chronic bouts of exercise (Naito et al. 2001; Morton et al. 2006). HSPs protect skeletal muscle and heart against injury and loss of mass. Overexpression of HSP25 and HSP70 prevents up-regulation of FoxO3a activation, ubiquitin ligases and muscle atrophy with mechanical unloading (Senf et al. 2008). Overexpression also reduced skeletal muscle damage and disruption of Ca2+ homeostasis in an mdx mouse. In addition, overexpression of HSP70 also reduces sarcopenia, muscle damage and fibre loss incurred with the ageing process (McArdle et al. 2004; Broome et al. 2006). Interestingly, cardiac muscle is much more thermosensitive than skeletal muscle (Ali et al. 1997). It has also been shown that the inducible form of HSP70 known as HSP72 remains less responsive to exercise performed in cold conditions compared to warm conditions in the myocardium (Hamilton et al. 2001; Quindry et al. 2007). HSP70 also protects the heart against ischaemia–reperfusion injury (Starnes et al. 2003, 2005).

Inducibility of antioxidant and stress response proteins with exercise

It is well documented that ROS levels or ‘oxidative stress’ increases during a bout of acute exercise (Bejma & Ji, 1999). The spike in ROS during exercise is intensity and time dependent (Munoz Marin et al. 2010). In response to the oxidant spike during exercise, many of the antioxidant

5166

J. M. Lawler and others

J Physiol 594.18

Superoxide dismutase and exercise. Isoforms of SOD

(Ji et al. 2007), and AMP kinase (AMPK) (Brandauer et al. 2015). Fibre-type differences in SOD inducibility with exercise are also evident. Slow twitch and Type IIa fibres have higher levels of MnSOD and Cu/ZnSOD than white muscle (Type IIb, IIx) (Powers et al. 1994; Hollander et al. 1999). MnSOD was inducible in red, but not white, skeletal muscle in response to exercise training (Pereira et al. 1994). Antioxidant enzymes in red gastrocnemius were most responsive to exercise training (Powers et al. 1994). Cu/ZnSOD did not increase in the gastrocnemius with moderate levels of exercise intensity (Song et al. 1996). Enhancement of SOD by exercise appears to be intensity dependent. Jump-training increased skeletal muscle levels of SOD and the coupled enzymes glutathione peroxidase and glutathione reductase in human subjects (Ortenblad et al. 1997). Eccentric exercise increased Cu/ZnSOD in the rectus femoris (Radak et al. 2013). High intensity exercise was needed to up-regulate SOD activity in the myocardium (Powers et al. 1993). In contrast, lifelong light-intensity exercise does not increase SOD1 (Cobley et al. 2014), although this may be confounded by interaction with ageing. For example, land and water shrews in the wild are able to up-regulate superoxide dismutase, catalase and/or glutathione peroxidase levels well above young adults (Hindle et al. 2010). However, very long-duration exercise (e.g. 2 h of exercise per day) may not yield increased SOD activity (Laughlin et al. 1990). These data suggest that higher intensity exercise may be necessary to protect against oxidative stress and damage, whether the exercise is acute, short-term, or across the lifespan.

increase in protein content and enzyme activity during exercise. Total activity for SOD increases in skeletal muscle after bouts of acute exercise (Lawler et al. 1993b; Powers & Lennon, 1999; Hollander et al. 2001) and habitual exercise training (Powers et al. 1993, 1999; Vincent et al. 2002; Nakatani et al. 2005; Higashida et al. 2011). SOD activity and GSH are elevated by exercise training (Husain & Somani, 1997). A divergent response among SOD isoforms may exist in skeletal muscle in response to exercise. MnSOD increased in skeletal muscle with swim training (Nakao et al. 2000). MnSOD RNA increased in the vastus lateralis with a single bout of exercise (Hollander et al. 2001). In contrast, Cu/ZnSOD protein levels were inducible without an increase in RNA transcripts (Hollander et al. 2001), suggesting translational or post-translational activation of a quiescent Cu/ZnSOD pool. Moderate levels of exercise training may increase MnSOD, but not Cu/ZnSOD, in skeletal muscles of young rats (Lambertucci et al. 2007). Exercise-induced boosting of MnSOD may protect skeletal muscle against hypoxia (Bo et al. 2014). MnSOD up-regulation with exercise appears to be triggered upstream by nuclear factor-kappaB (NF-κB) (Ji et al. 2006), mitogen-activated protein kinases (MAPK)

Catalase and exercise. Catalase activity appears to be less responsive to acute and habitual exercise training than MnSOD (Powers et al. 1994) in the gastrocnemius. However, catalase was up-regulated in the deep vastus lateralis, with a mix of Types IIa, IIb and I fibres (Luginbuhl et al. 1984), by exercise training, but not in the soleus (Ji et al. 1992) muscle, which is predominantly type I muscle. Alessio & Goldfarb (1988) found that catalase was up-regulated in red and white muscle following acute exercise. In contrast, exhaustive exercise reduced catalase levels (Ohishi et al. 1998). Laughlin and colleagues (Sexton et al. 1990) found that catalase only up-regulated when ischaemia–reperfusion was present. In addition, swim training had little effect on catalase (Toshinai et al. 1997). Food restriction may also impair the exercise effect on catalase and other antioxidant enzymes in the liver, while only impairing catalase in the gastrocnemius (Filaire et al. 2009). Since this study did not alter the nutritional make-up of the chow, but only restricted access to the food from ad libitum to 1 h per day, it is not known whether the impairment was due to a reduction in any specific macronutrient (Filaire et al. 2009). As expected, adenovirus transfection of a

enzymes adapt, or are inducible, with acute and repeated bouts of exercise. Li Li Ji’s and John Holloszy’s laboratories first reported in the late 1980s that skeletal muscles and the heart responded to exercise by increasing their antioxidant enzymes, including superoxide dismutase, catalase and glutathione peroxidase (Higuchi et al. 1985; Alessio & Goldfarb, 1988; Ji et al. 1988a; Ji & Fu, 1992). Heat shock proteins are also increased by exercise (Smolka et al. 2000; Fittipaldi et al. 2014); however, this may be dependent on training status and current antioxidant enzyme abundance and capacity. Subjecting a sedentary animal to exercise causes a dramatic increase in HSP72, whereas subjecting a trained animal to a bout of exercise will result in little to no change in HSP72 expression (Smolka et al. 2000). This exercise response is due in part to the antioxidant enzyme abundance and capacity of the subject prior to the exercise bout, further emphasizing the positive adaptations of exercise-induced preconditioning. In contrast, exhaustive exercise can decrease antioxidant enzyme activity, including catalase and glutathione peroxidase (Smolka et al. 2000). High levels of ROS and oxidative damage are linked to inhibition in antioxidant enzyme function with highly strenuous exercise (Davies et al. 1982; O’Neill et al. 1996). Oxidative stress during strenuous exercise was reduced by administration of a superoxide derivative (Radak et al. 1995). When muscles are able to up-regulate antioxidant enzymes in response to oxidative stress, a barrier against nucleic acid and protein oxidation is raised (Cobley et al. 2015).

 C 2016 The Authors. The Journal of Physiology  C 2016 The Physiological Society

J Physiol 594.18

Exercise preconditioning protection of striated muscle

mitochondrial catalase increased exercise performance (Li et al. 2009). Regulation of catalase and integration into stress protection and mitochondrial function are emerging. Activation of the nuclear factor-erythroid-2 p45-related factor (Nrf) 2/antioxidant response element is believed to regulate exercise induced up-regulation of catalase, SOD and the glutathione peroxidase family (Li et al. 2015). Peroxisome proliferator activated receptor γ coactivator-1 (PGC-1), important in mitochondrial biogenesis, is responsive to exercise and believed to be upstream of Nrf-1 and thus catalase and other antioxidant enzymes (Kang & Li Ji, 2012). Regulation of cellular H2 O2 triggers p(66Shc) and FoxO3a, and elicits downstream signalling (Wang et al. 2015). The glutathione peroxidase system and exercise. The family of glutathione peroxidases and/or GSH is fairly consistently up-regulated in skeletal muscle and heart in response to acute and chronic exercise (Leeuwenburgh et al. 1997). Selenium-dependent and -independent GPX have been shown to be inducible with exercise. In addition, exercise has also increased glutathione peroxidase, responsible for regenerating GSH from GSSG, in some skeletal muscle studies (Ji et al. 1988b). Chandan Sen’s and Li Li Ji’s laboratories demonstrated that reduced and total glutathione levels increased in skeletal muscle in response to exercise training (Leeuwenburgh et al. 1994; Sen, 1999). Acute exercise also stimulated the liver to release glutathione as a reinforcement for blood and contracting skeletal muscle glutathione (Lew et al. 1985). Strenuous exercise bouts increased glutathione peroxidase and glutathione reductase in rat skeletal muscle (Ji & Fu, 1992). Glutathione peroxidase and glutathione reductase activities also increased in humans with intense exercise (Ortenblad et al. 1997). Glutathione peroxidase is increased with acute and long-term exercise training in young animals, but inducibility may be lost with ageing (Lawler et al. 1993b; Gunduz et al. 2004). Alessio and Goldfarb first showed that exercise-induced enhancement of antioxidant enzymes was linked to protection against exercise-induced lipid peroxidation. The glutathione peroxidase family enhancement is regulated in part by the transcription factor Nrf-2 and PGC-1 (Kang & Li Ji, 2012; Li et al. 2015). NADPH is used as a reducing equivalent by glutathione reductase to power the conversion of GSSG to GSH. In skeletal muscle, the NADP-specific isocitrate dehydrogenase, malic enzyme and the pentose phosphate shunt enzyme glucose-6-phosphate dehydrogenase produce NADPH, which can be used for the glutathione peroxidase system, nitric oxide synthases, lipid synthesis and immune response. While glucose-6-phosphate dehydrogenase is inconsistently  C 2016 The Authors. The Journal of Physiology  C 2016 The Physiological Society

5167

affected by exercise (Schulpis et al. 2008), NADP-specific isocitrate dehydrogenase is substantially responsive to exercise training (Lawler et al. 1994; Lawler & Demaree, 2001). Conversely, disuse or mechanical unloading significantly impairs NADP-specific isocitrate dehydrogenase, in concert with changes in glutathione peroxidase (Lawler et al. 2006b). Thus the glutathione peroxidase family responds in a coordinated manner to both increased and decreased use in skeletal muscle. During disuse, alterations in antioxidant and HSP levels are permissive or exacerbate oxidative stress in unloaded skeletal muscle (Lawler & Hindle, 2011). Howlett & Willis (1998) proposed that NADP-specific isocitrate dehydrogenase and mitochondrial transhydrogenase were more important in red muscle, where basal levels of ROS and hydroperoxides are higher, than in white skeletal muscle. Indeed, fibre-type differences between slow- and fast-twitch fibres between glutathione peroxidase and NADP-specific isocitrate dehydrogenase are similar. Thioredoxin, peroxiredoxin, glutaredoxin reductases and adaptation to exercise. Thioredoxins, peroxiredoxins

and glutaredoxins provide significant support to the glutathione peroxidase system in removing and regulating hydroperoxides in the cytosol, peroxisomes, cell membranes and mitochondria. Exercise stress induction of thioredoxin was first reported in plasma mononuclear cells (Sumida et al. 2004). Thioredoxin 1 up-regulation with exercise was linked to protection against the risk of Type II diabetes (Lappalainen et al. 2009). Elevation of thioredoxin and thioredoxin reductases with exercise regulated reversible oxidative, cytoprotection and thiol protein–protein interactions (Radak et al. 2013). α-Lipoic acid enhanced the exercise-induced increase in thioredoxin reductase (Kinnunen et al. 2009; Lappalainen et al. 2010). Up-regulation of mitochondrial Trx2 appears to be a critical response in the heart to exercise, by reducing mitochondrial H2 O2 flux when rats were fed a high fat + high sugar diet (Fisher-Wellman et al. 2013). Interestingly, mitochondrial Trx2 in the red gastrocnemius was only up-regulated with exercise in the absence of the high fat + high sugar diet (Fisher-Wellman et al. 2013). Glutaredoxins may respond to exercise, but evidence is limited (Lappalainen et al. 2010). Induction of peroxiredoxin-5 (PRDX5) with exercise was first reported in Type II diabetic men, with minimal response from other peroxiredoxins (Brinkmann et al. 2012). However, peroxidredoxins can over-oxidize in pathological conditions during strenuous exercise (Brinkmann & Brixius, 2013). In addition, over-oxidation of peroxiredoxins was also noted after ultra-marathon races, which persisted for a week (Turner et al. 2013). Heat shock proteins in exercise. Exercise produces thermal stress, mechanical stress, metabolic stress and

5168

J. M. Lawler and others

oxidative stress, all of which can stimulate a family of inducible heat shock proteins (Fehrenbach & Niess, 1999). HSP70, HSP25, αB-crystallin and glucose-related protein 94 (grp94) levels are elevated with acute and chronic exercise training (Huey & Meador, 2008; Ogata et al. 2009). For example, HSP70 and HSP90 respond profoundly (200–1000%) to exhaustive exercise in human skeletal muscle, peaking at 3–6 days after exercise (Khassaf et al. 2001). Resistance training also stimulates an increase in HSP70 and HSP25 by over 900% in skeletal muscle (Murlasits et al. 2006). Uphill running induced higher levels of HSP70 than downhill running in slow-twitch muscles (Lollo et al. 2013). HSPs in slow-twitch and fast-twitch skeletal muscle respond to exercise training (Bombardier et al. 2013), although red muscle may experience a greater magnitude of protein abundance (Lewis et al. 2013). HSP25 and αB-crystallin may be more responsive to endurance training than resistance training, whereas HSP70 is induced by high and moderate intensity exercise (Folkesson et al. 2013). Recently, lifelong wheel running was shown to elevate HSP25, associated with protection against apoptosis and sarcopenia (Kim et al. 2015a). HSP70 and HSP25 are also up-regulated in cardiac muscle with exercise training (Lawler et al. 2006a; Lollo et al. 2013). Elevations of protein levels for HSP70 and other heat shock proteins in response to exercise occur rapidly, progressing from transcription and translation to active protein in approximately 60 min (Cumming et al. 2014). Rapid synthesis of viable heat shock protein provides swift stress protection against damage to proteins, lipids and nucleic acids. Up-regulation of HSPs during exercise provides protection against partially oxidized proteins and proteins undergoing unfolding, and protects against oxidative stress (Smolka et al. 2000). Tim Koh’s laboratory noticed that HSP25 and phosphorylated HSP25 translocated to the extracellular matrix (endomysium) when mechanical loading was altered (Koh & Escobedo, 2004). HSP25 and αB-crystallin also shift to the cytoskeleton with resistive exercise in humans, responding within 1 h and subsiding in 24–48 h (Cumming et al. 2014). Mechanical stretch, heat and contractile force are believed to activate HSPs. Many of the heat shock factors are regulated by the transcription factor HSF-1. Exercise training also up-regulated HSF-1, as an upstream mechanism behind exercise-induced increases in HSPs (Atalay et al. 2004). HSF-1 protein levels and activity are increased in the heart with exercise training (Demirel et al. 2003). HSF-1 induction can occur with a loss of reduced glutathione, signifying that increased expression of HSP is triggered by insufficient antioxidant protection (Paroo et al. 2002b). Oestrogen can up-regulate HSP70 in the heart via NF-κB activation of HSF-1 (Hamilton et al. 2004). However, oestrogen may also blunt exercise-induced up-regulation of HSP70 (Paroo

J Physiol 594.18

et al. 2002a; Bombardier et al. 2013). Sex differences have been observed in the expression levels of HSPs. In cardiac muscle, females express higher endogenous levels of HSP72, but evidence suggests that males and ovariectomized rats may be more sensitive to stressors, such as exercise, enabling them to better enhance their expression of HSPs (Paroo et al. 2002a; Bupha-Intr & Wattanapermpool, 2004). Conversely, disuse or mechanical unloading (bedrest, casting, spaceflight) causes a down-regulation of HSP70, HSP25 and HSF-1 (Lawler et al. 2006b; Senf et al. 2008; Lawler & Hindle, 2011). Thus HSPs are highly sensitive to both increased and decreased mechanical unloading. With increased oxidative stress during unloading (Lawler et al. 2003), rapid proteolysis is essential to remove partially oxidized proteins, particularly because of loss of protective HSPs. Overexpression of HSP70 and HSP25 protect against FoxO3a activation and proteolysis (Senf et al. 2008). Reducing oxidative stress during disuse also protects against FoxO3a activation and muscle fibre atrophy (Lawler et al. 2014). Up-regulation of HSPs including HSP70 indeed has been postulated to reduce oxidative stress, particularly during high intensity exercise when antioxidant status is impaired (Smolka et al. 2000). Thus heat shock proteins can be conceived as a second line of defence against oxidative damage. Hormesis – stress response to exercise. Allan Goldfarb,

John Holloszy and Li Li Ji were among the first investigators to characterize adaptation of antioxidant enzymes to mild to moderate oxidative stress that occurs during exercise (Higuchi et al. 1985; Ji, 2002; Bloomer et al. 2005). Training reduced oxidative stress in response to an acute bout of exercise (Alessio & Goldfarb, 1988; Ji et al. 1988a, 1992; Bloomer et al. 2005). Adaptations of antioxidant enzymes to exercise in skeletal muscle and heart were dependent upon exercise intensity, duration and age (Hammeren et al. 1992; Powers et al. 1993). Hormesis is commonly defined as a beneficial effect conferred by a low dose of an agent or phenomenon that would cause injury or toxicity at high levels. Li Li Ji’s laboratory first used the term hormesis to describe this adaptation in 2006 (Ji et al. 2006). The pro-inflammatory transcription factor NF-κB and MAPK were identified as part of a signalling cascade stimulated by exercise which up-regulated MnSOD and inducible nitric oxide synthase (Ji et al. 2006). Exercise-induced signalling was initiated by xanthine oxidase, as use of allopurinol blunted exercise-induced activation of NF-κB and MnSOD. ROS activate NF-κB by phosphorylating serine residues on the inhibitor of κB (I-κB), an inhibitor usually bound to quiescent NF-κB. Phosphorylation of I-κB releases NF-κB, which then translocates to the nucleus. Hormesis was also found to be beneficial in ageing skeletal muscle as well  C 2016 The Authors. The Journal of Physiology  C 2016 The Physiological Society

J Physiol 594.18

Exercise preconditioning protection of striated muscle

(Ji et al. 2010). Thus exercise-induced enhancement of antioxidant enzymes and heat shock proteins fortify a line of defence against oxidative stress and damage. In addition, hormesis allows ROS to function at physiological levels where they regulate important cell signalling related to stress protection, protein turnover, metabolism, glucose uptake, modulation of the immune system, regulation of blood flow, hypertrophy and atrophy. Furthermore, hormesis could be viewed as a process that explains the exercise preconditioning effect articulated by Zsolt Radak (Radak et al. 2000). Mild oxidative stress induced by exercise stimulates increased protection against oxidative stress, metabolic stress, mechanical stress, hypoxia, ischaemia, heat, cold and other perturbations of homeostasis. More recently, the notion of mitochondrial hormesis and exercise has been proposed (Ristow & Zarse, 2010). Up-regulation of mitochondrial antioxidant enzymes, sirtuins and PGC-1 with exercise is thought to provide protection against Type II diabetes and ageing (Balestrieri et al. 2015). Mitohormesis via NADPH producing pathways (e.g. NADP-specific isocitrate dehydrogenase) and glutathione homeostasis may stimulate a biosynthetic serine–1-carbon–glycine pathway and the longevity-promoting polyamine spermidine (Ost et al. 2015). Because pro-oxidant and pro-inflammatory mediators are highly integrated into normal skeletal muscle and cardiac function, including remodelling and resisting thermal, metabolic, hypoxic and mechanical stressors, exercise hormesis is becoming an active focus of research (Peake et al. 2015). Recently, NADPH oxidases were identified as requisite for exercise preconditioning in the heart (Frasier et al. 2013). Identification of transcription factors, antioxidant enzymes, ROS sources, heat shock proteins, kinases, mechanotransduction and other signalling pathways involved will be crucial to our understanding of striated muscle adaptations (Alleman et al. 2014; Peake et al. 2015). What doesn’t kill you, indeed makes you stronger. The following portion of this review will focus on translational benefits of hormesis or exercise preconditioning and clinical benefits. Exercise preconditioning and ischaemia–reperfusion.

The notion of exercise preconditioning is that transient, mild to moderate oxidative stress during exercise stimulates the up-regulation of protective antioxidant and stress proteins. Enhancing antioxidant and other stress proteins fortify cellular resistance to oxidative damage, injury, inflammation and chronic disease. Ischaemia, and in particular ischaemia followed by profound perfusion, results in tissue damage (Farb et al. 1993). Tissue damage can actually be far greater during the reperfusion phase than during ischaemia itself, due to high levels of oxidative stress and Ca2+ influx (Starkov et al. 2004). Indeed, the surprisingly large damage that occurs  C 2016 The Authors. The Journal of Physiology  C 2016 The Physiological Society

5169

during reperfusion has been termed ‘the oxygen paradox’ and ‘the calcium paradox’ (Hearse et al. 1978). However, ischaemia may prime reperfusion with dire consequences (ischaemia–reperfusion; I/R), as a dramatic reduction in levels of superoxide dismutase and catalase has been reported after I/R (Laughlin et al. 1990). In addition, I/R suppresses sirtuin (SIRT) 1, a protective histone deacetylase linked to longevity involved in elevation of AMPK phosphorylation and PGC-1 (Cattelan et al. 2015). During ischaemia, mitochondria are starved of oxygen, and cardiac cells shift metabolism to glycolysis (Buja, 2005). Hydrogen ions and lactate levels increase, decreasing pH below 6.5. Na+ /H+ exchange and Na+ /K+ pumps are compromised as ATP levels are challenged (Borges & Lessa, 2015). Reperfusion causes a burst of ROS and greatly exacerbates Ca2+ influx and damage. Sarcoplasmic reticulum Ca2+ pumps (sarco/endoplasmic reticulum calcium ATPase, SERCA) are impaired by oxidative stress and lower ATP, thus contributing to calcium overload (Grover & Samson, 1988; Grover et al. 1992; Ermak & Davies, 2002). As oxidized SERCA re-sequesters less Ca2+ , calcium levels rise and lead to damage and proteolysis (Murachi et al. 1981; Takahashi, 1990). There is a compensatory increase in Na+ /Ca2+ exchange as an adaptation for high internal Na+ caused when Na+ /K+ pumping is impaired during ischaemia, and the Na+ /K+ exchanger is increased during reperfusion (Hoffman et al. 2004). ROS and high cytosolic [Ca2+ ] then trigger mitochondrial permeability transition pore opening and myofibrillar hypercontractility as pH rises (Razvickas et al. 2013). In addition, cardiac ischaemia also down-regulates HSP70, SOD, catalase and glutathione peroxidase in skeletal muscle (Lawler et al. 2006a), thus reducing cardioprotection. Reduction of MnSOD in heart expose to I/R has been cited as contributory to damage (French et al. 2008). Influence of exercise preconditioning in striated muscle

As discussed in the previous section, skeletal and cardiac muscles are highly responsive to single and regular bouts of exercise. Increased activity due to stressors such as exercise can precondition the striated muscle tissues, thus building up defence mechanisms. This section will provide an overview of the cellular adaptations that lead to exercise preconditioning. Antioxidant enzymes and exercise preconditioning.

Exercise that is performed prior to ischaemia and reperfusion provides defence for the myocardium and skeletal muscle against I/R injury, a phenomenon often termed exercise preconditioning (Radak et al. 2000). Exercise-induced cardioprotection and skeletal muscle

5170

J. M. Lawler and others

resistance against ischaemia–reperfusion injury appear to be conferred by (a) up-regulation of antioxidant capacity, (b) elevation of critical heat shock proteins, (c) increase in nitric oxide signalling, (d) up-regulation of KATP channels, and (e) increased δ-opioid receptors. In skeletal muscle MnSOD, catalase and glutathione peroxidase are up-regulated with exercise training and are linked to prevention of I/R injury (Pereira et al. 1994; Quindry et al. 2005). In the heart MnSOD and catalase are consistently inducible with exercise training and have been linked to cardioprotection (Hamilton et al. 2001; Harris & Starnes, 2001; Lennon et al. 2004b; Moran et al. 2005; French et al. 2008; Kavazis et al. 2008; Quindry et al. 2010, 2012; Esposito et al. 2011). Exercise training increased antioxidant enzymes in concert with improvement of mitochondrial function during I/R (Kavazis et al. 2008). Powers and colleagues demonstrated that exercise-induced up-regulation of MnSOD is causal in cardioprotection against I/R injury by using antisense oligonucleotides to knockdown MnSOD (French et al. 2008; McClung et al. 2010). Indeed, knockdown of the MnSOD gene removed most of the cardioprotection against infarct size, impairment of cardiac function and oxidative damage. Interestingly, the exercise cardioprotective effects appear to be acute rather than chronic, meaning that habitual exercise is needed to retain the beneficial effects of exercise preconditioning. Each bout of exercise confers a cardioprotective benefit that lasts approximately 72 h (Yamashita et al. 1999; Lennon et al. 2004a). Heat shock proteins and exercise preconditioning. Many of the members of the heat shock protein family are inducible with exercise training, and have been linked to cardioprotection: αB-crystallin, HSP10, HSP60, HSP70 and HSP90. Higher intensity exercise appears to induce a greater increase in HSP70 than moderate intensity exercise (Lennon et al. 2004b). Starnes et al. (2005) also found that moderate intensity exercise (50–60% V˙ O2 max ) resulted in a small up-regulation of HSP70. Long-term exercise training (24 weeks) increased HSP70 protein expression, without an increase in MnSOD and glutathione reductase (Moran et al. 2005). Antioxidant supplementation appears to lead to a reduction in myocardial HSP72, but this reduction does not attenuate the enhanced MnSOD activity or the overall exercise-induced protection of the myocardium (Hamilton et al. 2003). Harris & Starnes (2001) modulated body temperature to tease out the most important heat shock proteins in exercise preconditioning. Exercise, while allowing body temperature rise, increased HSP70 protein abundance in the heart and skeletal muscle. In the wetted, cold exercise groups, αB-crystallin and HSP90 were significantly elevated, but HSP70 remained unchanged. Suppressed inducibility of HSP70 to exercise when

J Physiol 594.18

internal temperature was thermally neutral was linked to a dramatic attenuation of protection of cardiac function during ischaemia–reperfusion. Hamilton et al. (2001) and Quindry et al. (2007) found that HSP70 was elevated by exercise training at 70% max for 3–5 days/week under warm, but not cold, conditions. In contrast, glutathione peroxidase was up-regulated during exercise in the cold, and linked to cardioprotection (Hamilton et al. 2001). Protection against apoptosis after I/R was retained by cold exposure during exercise (Quindry et al. 2007) suggesting a independent mechanism for regulating apoptosis. Consistent with the above observations, heat stress up-regulates HSP70 and enhances myocardial recovery after ischaemia–reperfusion injury (Currie et al. 1988). HSP70 can be localized in the cytosol and mitochondria, but it is not clear whether mitochondrial HSP70 is more cardioprotective against I/R. However, Williamson et al. (2008) found that overexpression of a mitochondria-specific HSP70 provided protection against mitochondria and electron-transport chain damage. In short, HSP70 induction by exercise training appears to be most effective when body temperature rises, when exercise intensity or duration are high, when effects are on mitochondria, and when supplemented with antioxidants. KATP channels and exercise preconditioning. ATPsensitive K+ (KATP ) channels in cardiomyocyte and vascular smooth muscle cells (VSMCs) have recently been found to be protective against damage and infarct size in the heart. Exercise training has been found to enhance re-oxygenation of mitochondria. Exercise training increased protein expression of Kir6.1, a subunit of VSMC sarcolemmal KATP (sarcKATP ) channels (Li et al. 2014). Increased KATP channels in VSMCs was linked to better blood flow and oxygenation. Inhibition of KATP channels via gene transfer of the dominant negative Kir6.1AAA delayed recovery and enlarged the size of the infarct (Li et al. 2014). Quindry et al. (2010) used specific inhibitors of mitochondrial KATP (mitoKATP ) and sarcKATP channels to demonstrate a causal relationship of exercise-related cardioprotection against ischaemia–reperfusion injury. Inhibition of mitoKATP channels abrogated the exercise training effect. In addition, up-regulation of mitoKATP channels during exercise was linked to a reduction of cardiac arrhythmias during I/R (Quindry et al. 2012). An emerging role for the KATP channel is its ability to regulate and reduce oxidative stress. Diazoxide, a mitoKATP channel agonist, reduces oxidative stress by protecting protein thiols and increasing levels of reduced glutathione, abrogated by KATP channel blockers (Lemos Caldas et al. 2015). Indeed, diazoxide has a significant preconditioning effect on the heart, up-regulating pro-survival genes (e.g. VEGF, Bcl-2, PCNA, SDF-1a) and Akt phosphorylation (Mehmood et al. 2015). Progenitor cell survival is  C 2016 The Authors. The Journal of Physiology  C 2016 The Physiological Society

J Physiol 594.18

Exercise preconditioning protection of striated muscle

enhanced via mitoKATP channel activators (Mehmood et al. 2015). Conversely, hyperlipidaemia can attenuate mitoKATP channels and thus impair oxidative stress and increase oxidative stress in the heart (Csonka et al. 2014). Because H2 O2 can act on KATP channels (Melo et al. 2015), it can be argued that ROS initiate a preconditioning effect on cardioprotection in part through KATP channels. Thus enhancement of mitochondrial oxygen flux and KATP channels may be central to managing oxidative stress and exercise preconditioning in the heart. δ-Opioid receptors and exercise preconditioning. Opioid peptide receptors are part of the G-protein coupled receptor family. Recent and exciting literature has also provided evidence that opioid receptors may be another limb of exercise preconditioning. δ-Opioid receptors have been shown to reduce mitochondrial electron chain damage and induce neuroprotection (Zhu et al. 2009). In addition, δ-opioid receptors reduced oxidative stress through a protein kinase C (PKC) pathway in glial cells (Zhou et al. 2013). δ-Opioid receptor agonists reduce mitochondrial permeability transition pore opening and morphological changes in mitochondria (Zeng et al. 2011). P38/MAPK signalling has been implicated in δ-opioid receptor cardioprotection (Peart et al. 2007). Epicatechins, found in high concentrations in a variety of flavanol-rich foods and beverages such as green tea, dark chocolate, cocoa, black grapes, cherries and apples, are potent antioxidant and anti-inflammatory nutraceuticals. Recently, epicatechin was shown to have δ-opioid receptor binding activity and to improve mitochondrial state 3 respiration, thus stimulating preconditioning (Panneerselvam et al. 2013). Exercise training up-regulates δ-opioid receptors in the heart, and that effect contributes to cardioprotection. Michelsen et al. (2012) showed that blockade of δ-opioid receptors dampened exercise preconditioning of the heart. Furthermore, δ-Opioid receptor blockade limits exercise preconditioning in reducing infarct size (Dickson et al. 2008; Galvao et al. 2011). Exercise attenuation of necrosis, but not apoptosis, was linked to δ-opioid receptor signalling (Miller et al. 2015). Mitophagy and exercise preconditioning. Regular bouts

of physical activity result in an expansion of the mitochondrial pool in skeletal muscle in an effort to meet the metabolic demands and improve endurance performance (i.e. reduce fatigability). Exercise-induced remodelling and turnover of the mitochondrial pool improves the overall quality of the mitochondria through turnover via degradation and biogenesis. Maintaining quality of the mitochondrial pool requires the elimination of damaged and/or dysfunctional mitochondrial fragments. Impaired portions of the

 C 2016 The Authors. The Journal of Physiology  C 2016 The Physiological Society

5171

reticulum are isolated from the healthy portions via fission, and are then tagged for selective degradation by macro-autophagy. Organelle recycling via degradation of damaged mitochondrial fragments by macro-autophagy is a process often referred to as ‘mitophagy’. Mitophagy is thus a quality control system that optimizes highly functioning mitochondria. Autophagy is regulated by several autophagy-related genes (e.g. atrogins), including Atg6, which is better known as Beclin. Tagging of the fissioned mitochondrial fragments for degradation occurs through mitophagy-specific receptors, such as Bcl2/adenovirus E1B 19 kDa interacting protein 3 (BNIP3) (Lee et al. 2011; Rikka et al. 2011), or by ubiquitination of outer membrane proteins of the mitochondria by E3 ligases such as Parkin (Narendra et al. 2008; Geisler et al. 2010). Tagged mitochondria are eventually engulfed by the autophagosome before being degraded by the lysosome. Autophagy flux is often estimated by the ratio of microtubule associated protein light chain 3 (LC3) II to LC3-I, where a shift in the LC3-II:LC3-I ratio indicates increased flux, and lower levels of p62/sequestosome 1, which is degraded when it escorts tagged cellular fragments to the autophagosome (Mizushima et al. 2008). In skeletal muscle, voluntary wheel running has been shown to elevate levels of Beclin protein, reflective of an improved ability to induce autophagy (Lira et al. 2013; Greene et al. 2015). Aerobic exercise increases levels of mitochondrially localized LC3-II, p62 and ubiquitination (Saleem et al. 2014). In addition, Vainshtein et al. (2015) found that a single bout of exercise is capable of enhancing mitochondrial localized LC3-II, Parkin and elevated ubiquitination of the mitochondrial fraction. PGC-1 has been cited as an important upstream regulator of mitochondrial quality (Greene et al. 2015). From these findings it appears that mitophagy-related signalling and flux are enhanced after just a single bout of exercise. Of primary interest is whether a regular exercise regimen can elicit resting state adaptations in skeletal muscle tissue. Lira et al. (2013) determined that 4–5 weeks of voluntary wheel running was capable of enhancing basal autophagy and mitophagy protein expression. Overall, the limited data available suggest that regular exercise has an additive effect in skeletal muscle that eventually leads to improved autophagy and mitophagy capacity during exercise itself, as well as improved clearance at rest. The role of autophagy in cardiac muscle is often assumed to protect the myocardium against the accumulation of aggregates and dysfunctional mitochondria that can emit cell death (apoptosis) signals. In contrast, findings from skeletal muscle suggest that autophagy and mitophagy also contribute to regulation of energy homeostasis and metabolic stress in response to exercise (He et al. 2012a; Jamart et al. 2013; Lira et al. 2013;

5172

J. M. Lawler and others

Greene et al. 2015). Studies have shown that in response to acute (Ogura et al. 2011; He et al. 2012b) and chronic (Willis et al. 2013) exercise, autophagy is increased in cardiomyocytes. While data examining the influence of exercise-induced mitophagy in the myocardium are scarce, evidence from ischaemic preconditioning (IPC) can offer indications of what might occur. Following a period of cardiac ischaemia, ridding the cardiomyocytes of damaged and/or dysfunctional mitochondria is thought to protect the cells. IPC leads to mitochondrially localized Parkin, tagging the mitochondria for mitophagic degradation (Huang et al. 2011; Kubli et al. 2013), whereas Parkin-deficient mice accumulated dysfunctional and/or damaged mitochondria (Huang et al. 2011; Kubli et al. 2013). Parkin knockout mice exhibited decreased p62 protein, localized to the mitochondria in response to infarction (Huang et al. 2011). In response to ischaemic injury, elevated levels of tumour suppressor p53 are capable of reducing mitophagy by functioning as a negative regulator of BNIP3 and ROS signalling (Hoshino et al. 2012). Together, these findings suggest that the remodelling that occurs post-infarction involves clearance of damaged and/or dysfunctional mitochondria in an effort to protect against apoptosis and cardiomyocyte loss. Evidence presented here indicates it is likely that the stress of regular exercise can precondition the myocardium similarly to what has been seen in IPC and exercise models. The potential role(s) of ROS during exercise-induced mitophagy remain unclear. Exercise is known to increase the expression of regulators of mitochondrial remodelling (Ding et al. 2010; Perry et al. 2010; He et al. 2012b; Lira et al. 2013), possibly through contraction-induced ROS release (Fan et al. 2010). Recent findings suggest that ROS signalling is crucial for the promotion of mitophagy following ischaemia (Hoshino et al. 2012) and the enhancement of autophagy following exercise (Lo Verso et al. 2014). Lo Verso et al. (2014) found that chronic antioxidant treatment inhibits autophagy, and is therefore detrimental to the exercise-induced adaptations. Prolonged use of N-acetyl cysteine, as well as short-term use of the mitochondria-targeted antioxidant Mito-TEMPO, led to impaired performance and mitochondrial function in exercised mice (Lo Verso et al. 2014). These results highlight the importance of mild levels of oxidative stress, including that of mitochondria-derived ROS, produced during exercise, which lead to improved quality of the mitochondrial pool via mitophagy-driven remodelling. Future studies should examine the impact and mechanisms of exercise-induced ROS signalling in the mitochondrial remodelling process, including mitophagy. Other avenues of interest include the intensity and/or duration of exercise needed to elicit remodelling and mitophagy flux, as well as the identification of the various potential sources of ROS.

J Physiol 594.18

Exercise preconditioning and clinical benefits in striated muscle

The many benefits derived from exercise preconditioning can significantly impact chronic disorders that negatively affect striated muscle. Throughout this section we will highlight two conditions known to lead to muscle dysfunction – ageing and Type II diabetes. Ageing, skeletal muscle wasting, and exercise preconditioning. The ability of novel exercise training to

up-regulate stress proteins including antioxidant enzymes may be compromised as age progresses. Cu/ZnSOD, GPX and catalase become unresponsive to exercise stimulus in old rats, compared with young, and MnSOD levels were significantly reduced by exercise (Lambertucci et al. 2007). Stress intolerance increases with ageing, impairing mitochondrial function, caveolae number, MAPK signalling and PKC (Peart et al. 2014). Ageing also suppressed δ-opioid receptor cardioprotection through failure of MAPK signalling (Peart et al. 2007). Thus the risk of injury from ischaemia–reperfusion is raised, while exercise protection diminishes. Indeed, infarct area increases when stressed by I/R in the ageing heart (McCully et al. 2006). Glutathione peroxides, glutathione reductase and superoxide dismutase increase in skeletal muscle with ageing, yet become unresponsive to exercise training (Leeuwenburgh et al. 1994). Navarro-Arevalo et al. (1999) found that antioxidant enzymes responded to exhaustive exercise in the heart, but not skeletal muscle. However, exercise was able to up-regulate SIRT-1 in the ageing heart, which could protect mitochondrial function (Ferrara et al. 2008). Ageing also attenuates the ability of ROS to increase and modulate low-frequency contractility in skeletal muscle (Lawler et al. 1997). Mice overexpressing MnSOD do not live longer but have reduced age-associated disease or pathology. Exercise training does increase superoxide dismutase in skeletal muscle in senescent rats, with a marked increase in cardiac MnSOD (Starnes et al. 2003; Parise et al. 2005; Lawler et al. 2009). Overexpression of MnSOD was recently found to reduce apoptosis and fibrosis in the ageing heart, acting as an exercise mimetic (Kwak et al. 2015). One year of swimming also increased SOD in the heart (Gunduz et al. 2004). Starnes et al. (2003) showed that exercise does improve post-ischaemic function, possibly related to a reduction in mitochondrial ROS production (Starnes et al. 2007). Protection of mitochondria may be the key to preserving cardioprotection and exercise preconditioning. A new study suggested that protection of mitochondrial function during I/R by exercise preconditioning in the ageing heart may be conferred by redox-sensitive ornithine decarboxylase/polyamine signalling (Smirnova et al. 2012; Wang et al. 2014).  C 2016 The Authors. The Journal of Physiology  C 2016 The Physiological Society

J Physiol 594.18

Exercise preconditioning protection of striated muscle

Knockout of cytosolic Cu/ZnSOD results in a premature ageing or dystrophic-like phenotype (Muller et al. 2007). However, some of the myopathy with the global knockout is due to pathology at the neuromuscular junction (Shi et al. 2014). Conversely, overexpression of Cu/ZnSOD yields an amyotrophic lateral sclerosis-like phenotype (Shi et al. 2014). A large up-regulation of Cu/ZnSOD, without a compensatory increase in catalase or glutathione peroxidase, has also been observed during unloading-induced oxidative stress and atrophy (Lawler et al. 2006b). However, glutathione peroxidase and reduced glutathione are less inducible with exercise in skeletal muscles from old rats (Leeuwenburgh et al. 1994). Indeed, Radak et al. (2005) proposed that exercise hormesis in ageing muscle is linked to a reduced response to ROS produced during exercise. Heat shock response is also impaired with ageing. There was a 47% reduction in HSF-1 activation in the heart when Fischer 344 rats were exposed to heat stress at 41°C (Locke & Tanguay, 1996). Inducibility of HSP70 was also suppressed in old hearts when subjected to heat stress (Locke & Tanguay, 1996). High intensity, resistance exercise training also increased HSP25 and HSP70 in skeletal muscle of old rats (Murlasits et al. 2006). Concomitant with a reduced heat stress response, protection against I/R-induced damage was also lost. Reduction in HSP25 has also been linked to impaired δ-opioid signalling and cardioprotection against I/R injury (Locke & Tanguay, 1996). Lifelong exercise protects HSP25 in skeletal muscle (Kim et al. 2015a). Ageing, however, suppressed inducibility of HSP70 to heat stress and exercise in the ageing heart in a study conducted by Scott Powers’s laboratory (Demirel et al. 2003), but daily exercise increased HSP70 and HSP25 in the ageing heart in another study (Rinaldi et al. 2006). Induction of HSP in ageing skeletal muscle was dependent upon duration and frequency of exercise (Kim et al. 2015b). For example, exercise 5 days a weeks was superior in up-regulating HSP25, HSP60, HSP70, HSP90, MnSOD, Cu/ZnSOD and EC-SOD vs. 3 days a week, possibly linked to increased MAPK (Kim et al. 2015b). Heat shock proteins were inducible in slow and fast muscle with the exception of HSP27, which was responsive to exercise in slow-twitch muscle exclusively. Given that the risk of ischaemia–reperfusion injury increases with ageing, clearly far more research is needed to further our understanding of compromised mechanisms of exercise preconditioning in ageing heart and skeletal muscle. It has been suggested that exercise when combined with caloric restriction my enhance exercise preconditioning (Abete et al. 2010). In addition, the tandem of caloric restriction and exercise protected against ischaemia–reperfusion injury (Abete et al. 2005).

 C 2016 The Authors. The Journal of Physiology  C 2016 The Physiological Society

5173

Type II diabetes and exercise preconditioning. Exercise consistently improves glucose tolerance and reduces the risk of Type II diabetes (Tuomilehto et al. 2001; Mendham et al. 2015). Skeletal muscle and liver show improvement of lipid handling (Cortright et al. 2006; Jordy et al. 2015). Reduction in pro-inflammatory, pro-oxidant signalling is believed to be the primary mechanism (Barakat et al. 1983; Pilegaard et al. 2000; Cortright et al. 2006; Bikman et al. 2010b). Exercise training reduces the incidence of insulin resistance, and is linked to (a) reduced oxidative stress, (b) elevation of stress proteins (antioxidant, heat shock proteins), and (c) elevation of SIRT-1/PGC1 pathways and AMPK phosphorylation (Hawley & Lessard, 2008; Lambert et al. 2008). Indeed, prior exercise improves insulin signalling (Wojtaszewski et al. 2003) and may be protective against periodic or intermittent meals of calorically dense food. Thus the positive benefits of exercise on insulin signalling go beyond weight loss (Lambert et al. 2008). Impaired ability to handle fatty acids by mitochondria appears to be critical in elevating oxidative stress and contributing to insulin resistance (Kim et al. 2000; Boyle et al. 2012). Toll-like receptor 4 (TLR-4) has been shown to mediate oxidative stress and metabolic changes with high fat diet that leads to poor fatty acid oxidation and insulin resistance (McMillan et al. 2015). Zanchi et al. (2010) showed that resistive training suppresses TLR-4 and cytokine tumour necrosis factor-α (TNF-α) in skeletal muscle. Exercise down-regulates TLR-4 and is associated with increases in HSP70 in skeletal muscle (Rodriguez-Miguelez et al. 2014). Indeed, up-regulation of HSP70 is causal in reduction of TLR-4 in glial cells (Sinha et al. 2015). Exercise may delay or attenuate pro-inflammatory signalling by raising levels of heat shock proteins (Hooper & Hooper, 2009). Exercise also increases SIRT-1 and SIRT-3 in the heart and reduces apoptosis (Sack, 2011; Lai et al. 2014), similar to exercise preconditioning against I/R injury, the effects of exercise on glucose tolerance and GLUT4 availability are acute. Obesity and type II diabetes blunt exercise preconditioning, associated with poor inducibility of antioxidant enzymes, heat shock proteins and SIRT/PGC-1 signalling (Cappel et al. 2015). Cholesteryl ester transfer protein (CETP) shuttles lipids between serum lipoproteins and tissues, which may be a mechanism of impairment of PGC-1α and exercise response with obesity and Type II diabetes (Cappel et al. 2015). Cholesteryl ester transfer protein is reduced with exercise (Seip et al. 1993; Wilund et al. 2002) and polymorphisms have been linked to differences in high density lipoprotein cholesterol levels (Spielmann et al. 2007; Zhang et al. 2013). Large release of H2 O2 from mitochondria and mitochondrial defects are relieved by aerobic exercise

5174

J. M. Lawler and others

training (Konopka et al. 2015). Nox2 inhibition by sesamin was recently found to protect against impaired mitochondrial function and reduced exercise capacity with a high fat diet (Takada et al. 2015). Thus Nox2 and mitochondrial ROS may act together as an amplifier to chronically elevate oxidative stress and impair skeletal muscle function with insulin resistance. In a highly novel study, Strobel et al. (2014) demonstrated that depletion of glutathione levels with diethyl maleate amplified the ability of exercise training to increase PGC-1α. Increased oxidative stress with exercise and diethyl maleate plus exercise was confirmed using F2-isoprostanes as a marker. Exercise training improves artery relaxation and superoxide dismutase levels in rats fed a high fat diet (de Moraes et al. 2008). These data are consistent with the hypothesis that exercise preconditioning increases mitochondrial biogenesis and protection via transient oxidative stress during each bout of exercise. Summary and conclusions

Evolution and adaptations to an atmosphere that became rich in molecular oxygen led to the development of antioxidant and stress response proteins and pathways, which confer cellular protection against a host of stressors and insults. High energy reactive oxygen and nitrogen species are indeed utilized for cell signalling and communication. Transient increases in oxidative stress and inflammatory signalling that occur during exercise are important stimuli to produce cellular adaptations that protect against higher levels of oxidative stress. This is a process known as hormesis or exercise preconditioning. Cell-protective antioxidant enzymes (e.g. MnSOD and glutathione peroxidase), heat shock proteins and SIRT/PGC-1 pathways are frequently up-regulated by exercise in the heart and skeletal muscle in response to acute or chronic bouts of exercise. A central player to exercise preconditioning appears to be the organelle captured over a billion years ago by single celled organisms to handle and harness the power of oxygen: mitochondria. In this model, protection against oxidative stress, ischaemia–reperfusion, insulin resistance and ageing depends upon adaptation of mitochondrial morphology, function and stress-protective proteins with mitochondrial location or impact (MnSOD, Trx2, HSP70, SIRT-1, PGC-1α). Recent evidence suggests that Na+ /K+ -ATPase channels and δ-opioid receptors are involved in mitochondrial adaptations to oxidative stress and exercise preconditioning. Exercise preconditioning is blunted with obesity, Type II diabetes and ageing, but also serves as a powerful and preventative therapeutic agent against those conditions. Emerging research is identifying novel molecular mechanisms that could restore and enhance exercise preconditioning during chronic disease.

J Physiol 594.18

References Abete P, Cacciatore F, Testa G, Della-Morte D, Galizia G, de Santis D, Calabrese C, Cioppa A, Ferrara N & Rengo F (2010). Ischemic preconditioning in the aging heart: from bench to bedside. Ageing Res Rev 9, 153–162. Abete P, Testa G, Galizia G, Mazzella F, Della Morte D, de Santis D, Calabrese C, Cacciatore F, Gargiulo G, Ferrara N, Rengo G, Sica V, Napoli C & Rengo F (2005). Tandem action of exercise training and food restriction completely preserves ischemic preconditioning in the aging heart. Exp Gerontol 40, 43–50. Alessio HM & Goldfarb AH (1988). Lipid peroxidation and scavenger enzymes during exercise: adaptive response to training. J Appl Physiol 64, 1333–1336. Ali A, Fernando P, Smith WL, Ovsenek N, Lepock JR & Heikkila JJ (1997). Preferential activation of HSF-binding activity and hsp70 gene expression in Xenopus heart after mild hyperthermia. Cell Stress Chaperones 2, 229–237. Alleman RJ, Katunga LA, Nelson MA, Brown DA & Anderson EJ (2014). The “Goldilocks Zone” from a redox perspective—Adaptive vs. deleterious responses to oxidative stress in striated muscle. Front Physiol 5, 358. Anderson EJ, Lustig ME, Boyle KE, Woodlief TL, Kane DA, Lin CT, Price JW 3rd, Kang L, Rabinovitch PS, Szeto HH, Houmard JA, Cortright RN, Wasserman DH & Neufer PD (2009). Mitochondrial H2 O2 emission and cellular redox state link excess fat intake to insulin resistance in both rodents and humans. J Clin Invest 119, 573–581. Atalay M, Oksala NK, Laaksonen DE, Khanna S, Nakao C, Lappalainen J, Roy S, Hanninen O & Sen CK (2004). Exercise training modulates heat shock protein response in diabetic rats. J Appl Physiol 97, 605–611. Balestrieri ML, Rizzo MR, Barbieri M, Paolisso P, D’Onofrio N, Giovane A, Siniscalchi M, Minicucci F, Sardu C, D’Andrea D, Mauro C, Ferraraccio F, Servillo L, Chirico F, Caiazzo P, Paolisso G & Marfella R (2015). Sirtuin 6 expression and inflammatory activity in diabetic atherosclerotic plaques: effects of incretin treatment. Diabetes 64, 1395–1406. Barakat HA, Kasperek GJ & Dohm GL (1983). Progressive changes in fatty acid metabolism in rat liver and muscle during exercise. Biochem Med 29, 298–306. Bejma J & Ji LL (1999). Aging and acute exercise enhance free radical generation in rat skeletal muscle. J Appl Physiol (1985) 87, 465–470. Bermingham EN, Hesketh JE, Sinclair BR, Koolaard JP & Roy NC (2014). Selenium-enriched foods are more effective at increasing glutathione peroxidase (GPx) activity compared with selenomethionine: a meta-analysis. Nutrients 6, 4002–4031. Bikman BT, Zheng D, Kane DA, Anderson EJ, Woodlief TL, Price JW, Dohm GL, Neufer PD & Cortright RN (2010a). Metformin improves insulin signaling in obese rats via reduced IKKβ action in a fiber-type specific manner. J Obes 2010, 970865. Bikman BT, Zheng D, Reed MA, Hickner RC, Houmard JA & Dohm GL (2010b). Lipid-induced insulin resistance is prevented in lean and obese myotubes by AICAR treatment. Am J Physiol Regul Integr Comp Physiol 298, R1692–R1699.

 C 2016 The Authors. The Journal of Physiology  C 2016 The Physiological Society

J Physiol 594.18

Exercise preconditioning protection of striated muscle

Bloomer RJ, Goldfarb AH, Wideman L, McKenzie MJ & Consitt LA (2005). Effects of acute aerobic and anaerobic exercise on blood markers of oxidative stress. J Strength Cond Res 19, 276–285. Bo H, Li L, Duan FQ & Zhu J (2014). [Exercise training in hypoxia prevents hypoxia induced mitochondrial DNA oxidative damage in skeletal muscle]. Sheng Li Xue Bao 66, 597–604. Bombardier E, Vigna C, Bloemberg D, Quadrilatero J, Tiidus PM & Tupling AR (2013). The role of estrogen receptor-α in estrogen-mediated regulation of basal and exercise-induced Hsp70 and Hsp27 expression in rat soleus. Can J Physiol Pharmacol 91, 823–829. Borges JP & Lessa MA (2015). Mechanisms involved in exercise-induced cardioprotection: a systematic review. Arq Bras Cardiol 105, 71–81. Boveris A & Chance B (1973). The mitochondrial generation of hydrogen peroxide. General properties and effect of hyperbaric oxygen. Biochem J 134, 707–716. Boyle KE, Zheng D, Anderson EJ, Neufer PD & Houmard JA (2012). Mitochondrial lipid oxidation is impaired in cultured myotubes from obese humans. Int J Obes (Lond) 36, 1025–1031. Brandauer J, Andersen MA, Kellezi H, Risis S, Frosig C, Vienberg SG & Treebak JT (2015). AMP-activated protein kinase controls exercise training- and AICAR-induced increases in SIRT3 and MnSOD. Front Physiol 6, 85. Brinkmann C & Brixius K (2013). Peroxiredoxins and sports: new insights on the antioxidative defense. J Physiol Sci 63, 1–5. Brinkmann C, Chung N, Schmidt U, Kreutz T, Lenzen E, Schiffer T, Geisler S, Graf C, Montiel-Garcia G, Renner R, Bloch W & Brixius K (2012). Training alters the skeletal muscle antioxidative capacity in non-insulin-dependent type 2 diabetic men. Scand J Med Sci Sports 22, 462–470. Broome CS, Kayani AC, Palomero J, Dillmann WH, Mestril R, Jackson MJ & McArdle A (2006). Effect of lifelong overexpression of HSP70 in skeletal muscle on age-related oxidative stress and adaptation after nondamaging contractile activity. FASEB J 20, 1549–1551. Buja LM (2005). Myocardial ischemia and reperfusion injury. Cardiovasc Pathol 14, 170–175. Bupha-Intr T & Wattanapermpool J (2004). Cardioprotective effects of exercise training on myofilament calcium activation in ovariectomized rats. J Appl Physiol 96, 1755–1760. Cappel DA, Lantier L, Palmisano BT, Wasserman DH & Stafford JM (2015). CETP Expression protects female mice from obesity-induced decline in exercise capacity. PLoS One 10, e0136915. Cattelan A, Ceolotto G, Bova S, Albiero M, Kuppusamy M, De Martin S, Semplicini A, Fadini GP, de Kreutzenberg SV & Avogaro A (2015). NAD+ -dependent SIRT1 deactivation has a key role on ischemia-reperfusion-induced apoptosis. Vascul Pharmacol 70, 35–44. Claflin DR, Jackson MJ & Brooks SV (2015). Age affects the contraction-induced mitochondrial redox response in skeletal muscle. Front Physiol 6, 21.

 C 2016 The Authors. The Journal of Physiology  C 2016 The Physiological Society

5175

Cobley JN, Moult PR, Burniston JG, Morton JP & Close GL (2015). Exercise improves mitochondrial and redox-regulated stress responses in the elderly: better late than never! Biogerontology 16, 249–264. Cobley JN, Sakellariou GK, Owens DJ, Murray S, Waldron S, Gregson W, Fraser WD, Burniston JG, Iwanejko LA, McArdle A, Morton JP, Jackson MJ & Close GL (2014). Lifelong training preserves some redox-regulated adaptive responses after an acute exercise stimulus in aged human skeletal muscle. Free Radic Biol Med 70, 23–32. Cortright RN, Sandhoff KM, Basilio JL, Berggren JR, Hickner RC, Hulver MW, Dohm GL & Houmard JA (2006). Skeletal muscle fat oxidation is increased in African-American and white women after 10 days of endurance exercise training. Obesity (Silver Spring) 14, 1201–1210. Csonka C, Kupai K, Bencsik P, Gorbe A, Paloczi J, Zvara A, Puskas LG, Csont T & Ferdinandy P (2014). Cholesterol-enriched diet inhibits cardioprotection by ATP-sensitive K+ channel activators cromakalim and diazoxide. Am J Physiol Heart Circ Physiol 306, H405–H413. Cumming KT, Paulsen G, Wernbom M, Ugelstad I & Raastad T (2014). Acute response and subcellular movement of HSP27, αB-crystallin and HSP70 in human skeletal muscle after blood-flow-restricted low-load resistance exercise. Acta Physiol (Oxf) 211, 634–646. Currie RW, Karmazyn M, Kloc M & Mailer K (1988). Heat-shock response is associated with enhanced postischemic ventricular recovery. Circ Res 63, 543–549. Das DK, Engelman RM & Kimura Y (1993). Molecular adaptation of cellular defences following preconditioning of the heart by repeated ischaemia. Cardiovasc Res 27, 578–584. Davies KJ, Quintanilha AT, Brooks GA & Packer L (1982). Free radicals and tissue damage produced by exercise. Biochem Biophys Res Commun 107, 1198–1205. de Magalhaes JP & Church GM (2006). Cells discover fire: employing reactive oxygen species in development and consequences for aging. Exp Gerontol 41, 1–10. de Moraes C, Davel AP, Rossoni LV, Antunes E & Zanesco A (2008). Exercise training improves relaxation response and SOD-1 expression in aortic and mesenteric rings from high caloric diet-fed rats. BMC Physiol 8, 12. Demirel HA, Hamilton KL, Shanely RA, Tumer N, Koroly MJ & Powers SK (2003). Age and attenuation of exercise-induced myocardial HSP72 accumulation. Am J Physiol Heart Circ Physiol 285, H1609–H1615. Dickson EW, Hogrefe CP, Ludwig PS, Ackermann LW, Stoll LL & Denning GM (2008). Exercise enhances myocardial ischemic tolerance via an opioid receptor-dependent mechanism. Am J Physiol Heart Circ Physiol 294, H402–H408. Ding H, Jiang N, Liu H, Liu X, Liu D, Zhao F, Wen L, Liu S, Ji LL & Zhang Y (2010). Response of mitochondrial fusion and fission protein gene expression to exercise in rat skeletal muscle. Biochim Biophys Acta 1800, 250–256. Ding YH, Young CN, Luan X, Li J, Rafols JA, Clark JC, McAllister JP 2nd & Ding Y (2005). Exercise preconditioning ameliorates inflammatory injury in ischemic rats during reperfusion. Acta Neuropathol 109, 237–246.

5176

J. M. Lawler and others

Dupont-Versteegden EE, Fluckey JD, Knox M, Gaddy D & Peterson CA (2006). Effect of flywheel-based resistance exercise on processes contributing to muscle atrophy during unloading in adult rats. J Appl Physiol 101, 202–212. Ermak G & Davies KJ (2002). Calcium and oxidative stress: from cell signaling to cell death. Mol Immunol 38, 713–721. Esposito F, Ronchi R, Milano G, Margonato V, Di Tullio S, Marini M, Veicsteinas A & Samaja M (2011). Myocardial tolerance to ischemia-reperfusion injury, training intensity and cessation. Eur J Appl Physiol 111, 859–868. Fan X, Hussien R & Brooks GA (2010). H2 O2 -induced mitochondrial fragmentation in C2C12 myocytes. Free Radic Biol Med 49, 1646–1654. Farb A, Kolodgie FD, Jenkins M & Virmani R (1993). Myocardial infarct extension during reperfusion after coronary artery occlusion: pathologic evidence. J Am Coll Cardiol 21, 1245–1253. Fehrenbach E & Niess AM (1999). Role of heat shock proteins in the exercise response. Exerc Immunol Rev 5, 57–77. Fernandes AP & Holmgren A (2004). Glutaredoxins: glutathione-dependent redox enzymes with functions far beyond a simple thioredoxin backup system. Antioxid Redox Signal 6, 63–74. Ferrara N, Rinaldi B, Corbi G, Conti V, Stiuso P, Boccuti S, Rengo G, Rossi F & Filippelli A (2008). Exercise training promotes SIRT1 activity in aged rats. Rejuvenation Res 11, 139–150. Filaire E, Rouveix M, Massart A, Gladine C, Davicco MJ & Durand D (2009). Lipid peroxidation and antioxidant status in rat: effect of food restriction and wheel running. Eur J Appl Physiol 107, 243–250. Fisher AB (2009). Redox signaling across cell membranes. Antioxid Redox Signal 11, 1349–1356. Fisher-Wellman KH, Mattox TA, Thayne K, Katunga LA, La Favor JD, Neufer PD, Hickner RC, Wingard CJ & Anderson EJ (2013). Novel role for thioredoxin reductase-2 in mitochondrial redox adaptations to obesogenic diet and exercise in heart and skeletal muscle. J Physiol 591, 3471–3486. Fittipaldi S, Dimauro I, Mercatelli N & Caporossi D (2014). Role of exercise-induced reactive oxygen species in the modulation of heat shock protein response. Free Radic Res 48, 52–70. Folkesson M, Mackey AL, Langberg H, Oskarsson E, Piehl-Aulin K, Henriksson J & Kadi F (2013). The expression of heat shock protein in human skeletal muscle: effects of muscle fibre phenotype and training background. Acta Physiol (Oxf) 209, 26–33. Fontana L, Klein S & Holloszy JO (2010). Effects of long-term calorie restriction and endurance exercise on glucose tolerance, insulin action, and adipokine production. Age (Dordr) 32, 97–108. Frasier CR, Moukdar F, Patel HD, Sloan RC, Stewart LM, Alleman RJ, La Favor JD & Brown DA (2013). Redox-dependent increases in glutathione reductase and exercise preconditioning: role of NADPH oxidase and mitochondria. Cardiovasc Res 98, 47–55.

J Physiol 594.18

French JP, Hamilton KL, Quindry JC, Lee Y, Upchurch PA & Powers SK (2008). Exercise-induced protection against myocardial apoptosis and necrosis: MnSOD, calcium-handling proteins, and calpain. FASEB J 22, 2862–2871. Galvao TF, Matos KC, Brum PC, Negrao CE, Luz PL & Chagas AC (2011). Cardioprotection conferred by exercise training is blunted by blockade of the opioid system. Clinics (Sao Paulo) 66, 151–157. Geisler S, Holmstrom KM, Skujat D, Fiesel FC, Rothfuss OC, Kahle PJ & Springer W (2010). PINK1/Parkin-mediated mitophagy is dependent on VDAC1 and p62/SQSTM1. Nat Cell Biol 12, 119–131. Goodsell D (2004). Catalase. RCSB Protein Data Bank. Goth L (2008). Catalase deficiency and type 2 diabetes. Diabetes Care 31, e93. Greene NP, Lee DE, Brown JL, Rosa ME, Brown LA, Perry RA, Henry JN & Washington TA (2015). Mitochondrial quality control, promoted by PGC-1α, is dysregulated by Western diet-induced obesity and partially restored by moderate physical activity in mice. Physiol Rep 3, e12470. Grover AK & Samson SE (1988). Effect of superoxide radical on Ca2+ pumps of coronary artery. Am J Physiol Cell Physiol 255, C297–C303. Grover AK, Samson SE & Fomin VP (1992). Peroxide inactivates calcium pumps in pig coronary artery.Am J Physiol Heart Circ Physiol 263, H537–H543. Gunduz F, Senturk UK, Kuru O, Aktekin B & Aktekin MR (2004). The effect of one year’s swimming exercise on oxidant stress and antioxidant capacity in aged rats. Physiol Res 53, 171–176. Halliwell B (1995). How to characterize an antioxidant: an update. Biochem Soc Symp 61, 73–101. Hamilton KL, Gupta S & Knowlton AA (2004). Estrogen and regulation of heat shock protein expression in female cardiomyocytes: cross-talk with NFκB signaling. J Mol Cell Cardiol 36, 577–584. Hamilton KL, Powers SK, Sugiura T, Kim S, Lennon S, Tumer N & Mehta JL (2001). Short-term exercise training can improve myocardial tolerance to I/R without elevation in heat shock proteins. Am J Physiol Heart Circ Physiol 281, H1346–H1352. Hamilton KL, Staib JL, Phillips T, Hess A, Lennon SL & Powers SK (2003). Exercise, antioxidants, and HSP72: protection against myocardial ischemia/reperfusion. Free Radic Biol Med 34, 800–809. Hammeren J, Powers S, Lawler J, Criswell D, Martin D, Lowenthal D & Pollock M (1992). Exercise training-induced alterations in skeletal muscle oxidative and antioxidant enzyme activity in senescent rats. Int J Sports Med 13, 412–416. Harris MB & Starnes JW (2001). Effects of body temperature during exercise training on myocardial adaptations. Am J Physiol Heart Circ Physiol 280, H2271–H2280. Hawley JA & Lessard SJ (2008). Exercise training-induced improvements in insulin action. Acta Physiol (Oxf) 192, 127–135.

 C 2016 The Authors. The Journal of Physiology  C 2016 The Physiological Society

J Physiol 594.18

Exercise preconditioning protection of striated muscle

He C, Bassik MC, Moresi V, Sun K, Wei Y, Zou Z, An Z, Loh J, Fisher J, Sun Q, Korsmeyer S, Packer M, May HI, Hill JA, Virgin HW, Gilpin C, Xiao G, Bassel-Duby R, Scherer PE & Levine B (2012a). Exercise-induced BCL2-regulated autophagy is required for muscle glucose homeostasis. Nature 481, 511–515. He C, Sumpter R Jr & Levine B (2012b). Exercise induces autophagy in peripheral tissues and in the brain. Autophagy 8, 1548–1551. Heads RJ, Yellon DM & Latchman DS (1995). Differential cytoprotection against heat stress or hypoxia following expression of specific stress protein genes in myogenic cells. J Mol Cell Cardiol 27, 1669–1678. Hearse DJ, Humphrey SM & Bullock GR (1978). The oxygen paradox and the calcium paradox: two facets of the same problem? J Mol Cell Cardiol 10, 641–668. Higashida K, Kim SH, Higuchi M, Holloszy JO & Han DH (2011). Normal adaptations to exercise despite protection against oxidative stress. Am J Physiol Endocrinol Metab 301, E779–E784. Higuchi M, Cartier LJ, Chen M & Holloszy JO (1985). Superoxide dismutase and catalase in skeletal muscle: adaptive response to exercise. J Gerontol 40, 281–286. Hindle AG, Lawler JM, Campbell KL & Horning M (2010). Muscle aging and oxidative stress in wild-caught shrews. Comp Biochem Physiol B Biochem Mol Biol 155, 427–434. Hoffman JW Jr, Gilbert TB, Poston RS & Silldorff EP (2004). Myocardial reperfusion injury: etiology, mechanisms, and therapies. J Extra Corpor Technol 36, 391–411. Holland HD (2006). The oxygenation of the atmosphere and oceans. Philos Trans R Soc Lond B Biol Sci 361, 903–915. Hollander J, Fiebig R, Gore M, Bejma J, Ookawara T, Ohno H & Ji LL (1999). Superoxide dismutase gene expression in skeletal muscle: fiber-specific adaptation to endurance training. Am J Physiol Regul Integr Comp Physiol 277, R856–R862. Hollander J, Fiebig R, Gore M, Ookawara T, Ohno H & Ji LL (2001). Superoxide dismutase gene expression is activated by a single bout of exercise in rat skeletal muscle. Pflugers Arch 442, 426–434. Hooper PL & Hooper PL (2009). Inflammation, heat shock proteins, and type 2 diabetes. Cell Stress Chaperones 14, 113–115. Hoshino A, Matoba S, Iwai-Kanai E, Nakamura H, Kimata M, Nakaoka M, Katamura M, Okawa Y, Ariyoshi M, Mita Y, Ikeda K, Ueyama T, Okigaki M & Matsubara H (2012). p53-TIGAR axis attenuates mitophagy to exacerbate cardiac damage after ischemia. J Mol Cell Cardiol 52, 175–184. Howlett RA & Willis WT (1998). Fiber-type-related differences in the enzymes of a proposed substrate cycle. Biochim Biophys Acta 1363, 224–230. Huang C, Andres AM, Ratliff EP, Hernandez G, Lee P & Gottlieb RA (2011). Preconditioning involves selective mitophagy mediated by Parkin and p62/SQSTM1. PloS One 6, e20975. Huey KA & Meador BM (2008). Contribution of IL-6 to the Hsp72, Hsp25, and αB-crystallin [corrected] responses to inflammation and exercise training in mouse skeletal and cardiac muscle. J Appl Physiol 105, 1830–1836.

 C 2016 The Authors. The Journal of Physiology  C 2016 The Physiological Society

5177

Husain K & Somani SM (1997). Response of cardiac antioxidant system to alcohol and exercise training in the rat. Alcohol 14, 301–307. Ito N, Ruegg UT, Kudo A, Miyagoe-Suzuki Y & Takeda S (2013). Activation of calcium signaling through Trpv1 by nNOS and peroxynitrite as a key trigger of skeletal muscle hypertrophy. Nat Med 19, 101–106. Jamart C, Naslain D, Gilson H & Francaux M (2013). Higher activation of autophagy in skeletal muscle of mice during endurance exercise in the fasted state. Am J Physiol Endocrinol Metab 305, E964–E974. Ji LL (2002). Exercise-induced modulation of antioxidant defense. Ann N Y Acad Sci 959, 82–92. Ji LL, Dickman JR, Kang C & Koenig R (2010). Exercise-induced hormesis may help healthy aging. Dose Response 8, 73–79. Ji LL & Fu R (1992). Responses of glutathione system and antioxidant enzymes to exhaustive exercise and hydroperoxide. J Appl Physiol 72, 549–554. Ji LL, Fu R & Mitchell EW (1992). Glutathione and antioxidant enzymes in skeletal muscle: effects of fiber type and exercise intensity. J Appl Physiol 73, 1854–1859. Ji LL, Gomez-Cabrera MC & Vina J (2006). Exercise and hormesis: activation of cellular antioxidant signaling pathway. Ann N Y Acad Sci 1067, 425–435. Ji LL, Gomez-Cabrera MC & Vina J (2007). Role of nuclear factor κB and mitogen-activated protein kinase signaling in exercise-induced antioxidant enzyme adaptation. Appl Physiol Nutr Metab 32, 930–935. Ji LL, Stratman FW & Lardy HA (1988a). Antioxidant enzyme systems in rat liver and skeletal muscle. Influences of selenium deficiency, chronic training, and acute exercise. Arch Biochem Biophys 263, 150–160. Ji LL, Stratman FW & Lardy HA (1988b). Enzymatic down regulation with exercise in rat skeletal muscle. Arch Biochem Biophys 263, 137–149. Jordy AB, Kraakman MJ, Gardner T, Estevez E, Kammoun HL, Weir JM, Kiens B, Meikle PJ, Febbraio MA & Henstridge DC (2015). Analysis of the liver lipidome reveals insights into the protective effect of exercise on high-fat diet-induced hepatosteatosis in mice. Am J Physiol Endocrinol Metab 308, E778–E791. Kane DA, Anderson EJ, Price JW 3rd, Woodlief TL, Lin CT, Bikman BT, Cortright RN & Neufer PD (2010). Metformin selectively attenuates mitochondrial H2 O2 emission without affecting respiratory capacity in skeletal muscle of obese rats. Free Radic Biol Med 49, 1082–1087. Kang C & Li JiL (2012). Role of PGC-1α signaling in skeletal muscle health and disease. Ann NY Acad Sci 1271, 110–117. Kavazis AN, McClung JM, Hood DA & Powers SK (2008). Exercise induces a cardiac mitochondrial phenotype that resists apoptotic stimuli. Am J Physiol Heart Circ Physiol 294, H928–H935. Khairallah RJ, Shi G, Sbrana F, Prosser BL, Borroto C, Mazaitis MJ, Hoffman EP, Mahurkar A, Sachs F, Sun Y, Chen YW, Raiteri R, Lederer WJ, Dorsey SG & Ward CW (2012). Microtubules underlie dysfunction in duchenne muscular dystrophy. Sci Signal 5, ra56.

5178

J. M. Lawler and others

Khassaf M, Child RB, McArdle A, Brodie DA, Esanu C & Jackson MJ (2001). Time course of responses of human skeletal muscle to oxidative stress induced by nondamaging exercise. J Appl Physiol 90, 1031–1035. Kim JH & Lawler JM (2012). Amplification of proinflammatory phenotype, damage, and weakness by oxidative stress in the diaphragm muscle of mdx mice. Free Radic Biol Med 52, 1597–1606. Kim JH, Lee Y, Kwak HB & Lawler JM (2015a). Lifelong wheel running exercise and mild caloric restriction attenuate nuclear EndoG in the aging plantaris muscle. Exp Gerontol 69, 122–128. Kim JS, Lee YH, Choi DY & Yi HK (2015b). Expression of heat shock proteins (HSPs) in aged skeletal muscles depends on the frequency and duration of exercise training. J Sports Sci Med 14, 347–353. Kim JY, Hickner RC, Cortright RL, Dohm GL & Houmard JA (2000). Lipid oxidation is reduced in obese human skeletal muscle. Am J Physiol Endocrinol Metab 279, E1039–E1044. Kinnunen S, Oksala N, Hyyppa S, Sen CK, Radak Z, Laaksonen DE, Szabo B, Jakus J & Atalay M (2009). α-Lipoic acid modulates thiol antioxidant defenses and attenuates exercise-induced oxidative stress in standardbred trotters. Free Radic Res 43, 697–705. Koh TJ & Escobedo J (2004). Cytoskeletal disruption and small heat shock protein translocation immediately after lengthening contractions. Am J Physiol Cell Physiol 286, C713–C722. Kombairaju P, Kerr JP, Roche JA, Pratt SJ, Lovering RM, Sussan TE, Kim JH, Shi G, Biswal S & Ward CW (2014). Genetic silencing of Nrf2 enhances X-ROS in dysferlin-deficient muscle. Front Physiol 5, 57. Konopka AR, Asante A, Lanza IR, Robinson MM, Johnson ML, Dalla Man C, Cobelli C, Amols MH, Irving BA & Nair KS (2015). Defects in mitochondrial efficiency and H2 O2 emissions in obese women are restored to a lean phenotype with aerobic exercise training. Diabetes 64, 2104–2115. Kregel KC (2002). Heat shock proteins: modifying factors in physiological stress responses and acquired thermotolerance. J Appl Physiol 92, 2177–2186. Kubli DA, Zhang X, Lee Y, Hanna RA, Quinsay MN, Nguyen CK, Jimenez R, Petrosyan S, Murphy AN & Gustafsson AB (2013). Parkin protein deficiency exacerbates cardiac injury and reduces survival following myocardial infarction. J Biol Chem 288, 915–926. Kwak HB, Lee Y, Kim JH, Van Remmen H, Richardson AG & Lawler JM (2015). MnSOD overexpression reduces fibrosis and pro-apoptotic signaling in the aging mouse heart. J Gerontol A Biol Sci Med Sci 70, 533–544. Lai CH, Ho TJ, Kuo WW, Day CH, Pai PY, Chung LC, Liao PH, Lin FH, Wu ET & Huang CY (2014). Exercise training enhanced SIRT1 longevity signaling replaces the IGF1 survival pathway to attenuate aging-induced rat heart apoptosis. Age (Dordr) 36, 9706. Lambert CP, Wright NR, Finck BN & Villareal DT (2008). Exercise but not diet-induced weight loss decreases skeletal muscle inflammatory gene expression in frail obese elderly persons. J Appl Physiol 105, 473–478.

J Physiol 594.18

Lambertucci RH, Levada-Pires AC, Rossoni LV, Curi R & Pithon-Curi TC (2007). Effects of aerobic exercise training on antioxidant enzyme activities and mRNA levels in soleus muscle from young and aged rats. Mech Ageing Dev 128, 267–275. Lappalainen Z, Lappalainen J, Laaksonen DE, Oksala KJ, Khanna S, Sen CK & Atalay M (2010). Acute exercise and thioredoxin-1 in rat brain, and α-lipoic acid and thioredoxin-interacting protein response, in diabetes. Int J Sport Nutr Exerc Metab 20, 206–215. Lappalainen Z, Lappalainen J, Oksala NK, Laaksonen DE, Khanna S, Sen CK & Atalay M (2009). Diabetes impairs exercise training-associated thioredoxin response and glutathione status in rat brain. J Appl Physiol 106, 461–467. Laughlin MH, Simpson T, Sexton WL, Brown OR, Smith JK & Korthuis RJ (1990). Skeletal muscle oxidative capacity, antioxidant enzymes, and exercise training. J Appl Physiol 68, 2337–2343. Lawler JM, Cline CC, Hu Z & Coast JR (1997). Effect of oxidant challenge on contractile function of the aging rat diaphragm. Am J Physiol Endocrinol Metab 272, E201–E207. Lawler JM & Demaree SR (2001). Relationship between NADP-specific isocitrate dehydrogenase and glutathione peroxidase in aging rat skeletal muscle. Mech Ageing Dev 122, 291–304. Lawler JM & Hindle A (2011). Living in a box or call of the wild? Revisiting lifetime inactivity and sarcopenia. Antioxid Redox Signal 15, 2529–2541. Lawler JM, Kunst M, Hord JM, Lee Y, Joshi K, Botchlett RE, Ramirez A & Martinez DA (2014). EUK-134 ameliorates nNOSmu translocation and skeletal muscle fiber atrophy during short-term mechanical unloading. Am J Physiol Regul Integr Comp Physiol 306, R470–R482. Lawler JM, Kwak HB, Kim JH & Suk MH (2009). Exercise training inducibility of MnSOD protein expression and activity is retained while reducing prooxidant signaling in the heart of senescent rats. Am J Physiol Regul Integr Comp Physiol 296, R1496–R1502. Lawler JM, Kwak HB, Song W & Parker JL (2006a). Exercise training reverses downregulation of HSP70 and antioxidant enzymes in porcine skeletal muscle after chronic coronary artery occlusion. Am J Physiol Regul Integr Comp Physiol 291, R1756–R1763. Lawler JM, Powers SK & Criswell DS (1993a). Inducibility of NADP-specific isocitrate dehydrogenase with endurance training in skeletal muscle. Acta Physiol Scand 149, 177–181. Lawler JM, Powers SK, Van Dijk H, Visser T, Kordus MJ & Ji LL (1994). Metabolic and antioxidant enzyme activities in the diaphragm: effects of acute exercise. Respir Physiol 96, 139–149. Lawler JM, Powers SK, Visser T, Van Dijk H, Kordus MJ & Ji LL (1993b). Acute exercise and skeletal muscle antioxidant and metabolic enzymes: effects of fiber type and age. Am J Physiol Regul Integr Comp Physiol 265, R1344–R1350. Lawler JM, Song W & Demaree SR (2003). Hindlimb unloading increases oxidative stress and disrupts antioxidant capacity in skeletal muscle. Free Radic Biol Med 35, 9–16.

 C 2016 The Authors. The Journal of Physiology  C 2016 The Physiological Society

J Physiol 594.18

Exercise preconditioning protection of striated muscle

Lawler JM, Song W & Kwak HB (2006b). Differential response of heat shock proteins to hindlimb unloading and reloading in the soleus. Muscle Nerve 33, 200–207. Lee Y, Lee HY, Hanna RA & Gustafsson AB (2011). Mitochondrial autophagy by Bnip3 involves Drp1-mediated mitochondrial fission and recruitment of Parkin in cardiac myocytes. Am J Physiol Heart Circ Physiol 301, H1924–H1931. Leeuwenburgh C, Fiebig R, Chandwaney R & Ji LL (1994). Aging and exercise training in skeletal muscle: responses of glutathione and antioxidant enzyme systems. Am J Physiol Regul Integr Comp Physiol 267, R439–R445. Leeuwenburgh C, Hollander J, Leichtweis S, Griffiths M, Gore M & Ji LL (1997). Adaptations of glutathione antioxidant system to endurance training are tissue and muscle fiber specific. Am J Physiol Regul Integr Comp Physiol 272, R363–R369. Lemos Caldas FR, Rocha Leite IM, Tavarez Filgueiras AB, de Figueiredo Junior IL, Gomes Marques de Sousa TA, Martins PR, Kowaltowski AJ & Fernandes Facundo H (2015). Mitochondrial ATP-sensitive potassium channel opening inhibits isoproterenol-induced cardiac hypertrophy by preventing oxidative damage. J Cardiovasc Pharmacol 65, 393–397. Lennon SL, Quindry JC, Hamilton KL, French JP, Hughes J, Mehta JL & Powers SK (2004b). Elevated MnSOD is not required for exercise-induced cardioprotection against myocardial stunning. Am J Physiol Heart Circ Physiol 287, H975–H980. Lennon SL, Quindry J, Hamilton KL, French J, Staib J, Mehta JL & Powers SK (2004a). Loss of exercise-induced cardioprotection after cessation of exercise. J Appl Physiol 96, 1299–1305. Lew H, Pyke S & Quintanilha A (1985). Changes in the glutathione status of plasma, liver and muscle following exhaustive exercise in rats. FEBS Lett 185, 262–266. Lewis EJ, Ramsook AH, Locke M & Amara CE (2013). Mild eccentric exercise increases Hsp72 content in skeletal muscles from adult and late middle-aged rats. Cell Stress Chaperones 18, 667–673. Li D, Lai Y, Yue Y, Rabinovitch PS, Hakim C & Duan D (2009). Ectopic catalase expression in mitochondria by adeno-associated virus enhances exercise performance in mice. PLoS One 4, e6673. Li T, He S, Liu S, Kong Z, Wang J & Zhang Y (2015). Effects of different exercise durations on Keap1-Nrf2-ARE pathway activation in mouse skeletal muscle. Free Radic Res 49, 1269–1274. Li Y, Cai M, Cao L, Qin X, Zheng T, Xu X, Sandvick TM, Hutchinson K, Wold LE, Hu K, Sun Q, Thomas DP, Ren J & He G (2014). Endurance exercise accelerates myocardial tissue oxygenation recovery and reduces ischemia reperfusion injury in mice. PLoS One 9, e114205. Lira VA, Okutsu M, Zhang M, Greene NP, Laker RC, Breen DS, Hoehn KL & Yan Z (2013). Autophagy is required for exercise training-induced skeletal muscle adaptation and improvement of physical performance. FASEB J 27, 4184–4193.

 C 2016 The Authors. The Journal of Physiology  C 2016 The Physiological Society

5179

Locke M, Noble EG, Tanguay RM, Feild MR, Ianuzzo SE & Ianuzzo CD (1995). Activation of heat-shock transcription factor in rat heart after heat shock and exercise. Am J Physiol Cell Physiol 268, C1387–C1394. Locke M & Tanguay RM (1996). Diminished heat shock response in the aged myocardium. Cell Stress Chaperones 1, 251–260. Lollo PC, Moura CS, Morato PN & Amaya-Farfan J (2013). Differential response of heat shock proteins to uphill and downhill exercise in heart, skeletal muscle, lung and kidney tissues. J Sports Sci Med 12, 461–466. Lo Verso F, Carnio S, Vainshtein A & Sandri M (2014). Autophagy is not required to sustain exercise and PRKAA1/AMPK activity but is important to prevent mitochondrial damage during physical activity. Autophagy 10, 1883–1894. Luginbuhl AJ, Dudley GA & Staron RS (1984). Fiber type changes in rat skeletal muscle after intense interval training. Histochemistry 81, 55–58. Martindale JL & Holbrook NJ (2002). Cellular response to oxidative stress: signaling for suicide and survival. J Cell Physiol 192, 1–15. McArdle A, Dillmann WH, Mestril R, Faulkner JA & Jackson MJ (2004). Overexpression of HSP70 in mouse skeletal muscle protects against muscle damage and age-related muscle dysfunction. FASEB J 18, 355–357. McClung JM, Deruisseau KC, Whidden MA, Van Remmen H, Richardson A, Song W, Vrabas IS & Powers SK (2010). Overexpression of antioxidant enzymes in diaphragm muscle does not alter contraction-induced fatigue or recovery. Exp Physiol 95, 222–231. McCully JD, Toyoda Y, Wakiyama H, Rousou AJ, Parker RA & Levitsky S (2006). Age- and gender-related differences in ischemia/reperfusion injury and cardioprotection: effects of diazoxide. Ann Thorac Surg 82, 117–123. McMillan RP, Wu Y, Voelker K, Fundaro G, Kavanaugh J, Stevens JR, Shabrokh E, Ali M, Harvey M, Anderson AS, Boutagy NE, Mynatt RL, Frisard MI & Hulver MW (2015). Selective overexpression of Toll-like receptor-4 in skeletal muscle impairs metabolic adaptation to high-fat feeding. Am J Physiol Regul Integr Comp Physiol 309, R304–R313. Mehmood A, Ali M, Khan SN & Riazuddin S (2015). Diazoxide preconditioning of endothelial progenitor cells improves their ability to repair the infarcted myocardium. Cell Biol Int 39, 1251–1263. Melo MR, Menani JV, Colombari E & Colombari DS (2015). Hydrogen peroxide attenuates the dipsogenic, renal and pressor responses induced by cholinergic activation of the medial septal area. Neuroscience 284, 611–621. Mendham AE, Duffield R, Marino F & Coutts AJ (2015). A 12-week sports-based exercise programme for inactive Indigenous Australian men improved clinical risk factors associated with type 2 diabetes mellitus. J Sci Med Sport 18, 438–443. Michelsen MM, Stottrup NB, Schmidt MR, Lofgren B, Jensen RV, Tropak M, St-Michel EJ, Redington AN & Botker HE (2012). Exercise-induced cardioprotection is mediated by a bloodborne, transferable factor. Basic Res Cardiol 107, 260.

5180

J. M. Lawler and others

Miller LE, McGinnis GR, Peters BA, Ballmann CG, Nanayakkara G, Amin R & Quindry JC (2015). Involvement of the δ-opioid receptor in exercise-induced cardioprotection. Exp Physiol 100, 410–421. Mizushima N, Levine B, Cuervo AM & Klionsky DJ (2008). Autophagy fights disease through cellular self-digestion. Nature 451, 1069–1075. Moran M, Blazquez I, Saborido A & Megias A (2005). Antioxidants and ecto-5 -nucleotidase are not involved in the training-induced cardioprotection against ischaemia-reperfusion injury. Exp Physiol 90, 507–517. Morton JP, MacLaren DP, Cable NT, Bongers T, Griffiths RD, Campbell IT, Evans L, Kayani A, McArdle A & Drust B (2006). Time course and differential responses of the major heat shock protein families in human skeletal muscle following acute nondamaging treadmill exercise. J Appl Physiol 101, 176–182. Muller FL, Song W, Jang YC, Liu Y, Sabia M, Richardson A & Van Remmen H (2007). Denervation-induced skeletal muscle atrophy is associated with increased mitochondrial ROS production. Am J Physiol Regul Integr Comp Physiol 293, R1159–R1168. Munoz Marin D, Olcina G, Timon R, Robles MC, Caballero MJ & Maynar M (2010). Effect of different exercise intensities on oxidative stress markers and antioxidant response in trained cyclists. J Sports Med Phys Fitness 50, 93–98. Muoio DM & Neufer PD (2012). Lipid-induced mitochondrial stress and insulin action in muscle. Cell Metab 15, 595–605. Murachi T, Tanaka K, Hatanaka M & Murakami T (1981). Intracellular Ca2+ -dependent protease (calpain) and its high-molecular-weight endogenous inhibitor (calpastatin). Adv Enzyme Regul 19, 407–424. Murlasits Z, Cutlip RG, Geronilla KB, Rao KM, Wonderlin WF & Alway SE (2006). Resistance training increases heat shock protein levels in skeletal muscle of young and old rats. Exp Gerontol 41, 398–406. Naito H, Powers SK, Demirel HA & Aoki J (2001). Exercise training increases heat shock protein in skeletal muscles of old rats. Med Sci Sports Exerc 33, 729–734. Nakao C, Ookawara T, Kizaki T, Oh-Ishi S, Miyazaki H, Haga S, Sato Y, Ji LL & Ohno H (2000). Effects of swimming training on three superoxide dismutase isoenzymes in mouse tissues. J Appl Physiol 88, 649–654. Nakatani K, Komatsu M, Kato T, Yamanaka T, Takekura H, Wagatsuma A, Aoyama K, Xu B, Hirano T, Kasai H, Ando S & Takeuchi T (2005). Habitual exercise induced resistance to oxidative stress. Free Radic Res 39, 905–911. Narendra D, Tanaka A, Suen DF & Youle RJ (2008). Parkin is recruited selectively to impaired mitochondria and promotes their autophagy. J Cell Biol 183, 795–803. Navarro-Arevalo A, Canavate C & Sanchez-del-Pino MJ (1999). Myocardial and skeletal muscle aging and changes in oxidative stress in relationship to rigorous exercise training. Mech Ageing Dev 108, 207–217. Neylan TC (1998). Hans Selye and the field of stress research. J Neuropsychiatry Clin Neurosci 10, 230–230.

J Physiol 594.18

Noland RC, Woodlief TL, Whitfield BR, Manning SM, Evans JR, Dudek RW, Lust RM & Cortright RN (2007). Peroxisomal-mitochondrial oxidation in a rodent model of obesity-associated insulin resistance. Am J Physiol Endocrinol Metab 293, E986–E1001. Nonn L, Berggren M & Powis G (2003). Increased expression of mitochondrial peroxiredoxin-3 (thioredoxin peroxidase-2) protects cancer cells against hypoxia and drug-induced hydrogen peroxide-dependent apoptosis. Mol Cancer Res 1, 682–689. O’Neill CA, Stebbins CL, Bonigut S, Halliwell B & Longhurst JC (1996). Production of hydroxyl radicals in contracting skeletal muscle of cats. J Appl Physiol 81, 1197–1206. Ogata T, Oishi Y, Higashida K, Higuchi M & Muraoka I (2009). Prolonged exercise training induces long-term enhancement of HSP70 expression in rat plantaris muscle. Am J Physiol Regul Integr Comp Physiol 296, R1557–R1563. Ogura Y, Iemitsu M, Naito H, Kakigi R, Kakehashi C, Maeda S & Akema T (2011). Single bout of running exercise changes LC3-II expression in rat cardiac muscle. Biochem Biophys Res Commun 414, 756–760. Ohishi S, Kizaki T, Ookawara T, Toshinai K, Haga S, Karasawa F, Satoh T, Nagata N, Ji LL & Ohno H (1998). The effect of exhaustive exercise on the antioxidant enzyme system in skeletal muscle from calcium-deficient rats. Pflugers Arch 435, 767–774. Omar R & Pappolla M (1993). Oxygen free radicals as inducers of heat shock protein synthesis in cultured human neuroblastoma cells: relevance to neurodegenerative disease. Eur Arch Psychiatry Clin Neurosci 242, 262–267. Ortenblad N, Madsen K & Djurhuus MS (1997). Antioxidant status and lipid peroxidation after short-term maximal exercise in trained and untrained humans. Am J Physiol Regul Integr Comp Physiol 272, R1258–R1263. Ost M, Keipert S, van Schothorst EM, Donner V, van der Stelt I, Kipp AP, Petzke KJ, Jove M, Pamplona R, Portero-Otin M, Keijer J & Klaus S (2015). Muscle mitohormesis promotes cellular survival via serine/glycine pathway flux. FASEB J 29, 1314–1328. Panneerselvam M, Ali SS, Finley JC, Kellerhals SE, Migita MY, Head BP, Patel PM, Roth DM & Patel HH (2013). Epicatechin regulation of mitochondrial structure and function is opioid receptor dependent. Mol Nutr Food Res 57, 1007–1014. Parise G, Phillips SM, Kaczor JJ & Tarnopolsky MA (2005). Antioxidant enzyme activity is up-regulated after unilateral resistance exercise training in older adults. Free Radic Biol Med 39, 289–295. Paroo Z, Dipchand ES & Noble EG (2002a). Estrogen attenuates postexercise HSP70 expression in skeletal muscle. Am J Physiol Cell Physiol 282, C245–C251. Paroo Z, Meredith MJ, Locke M, Haist JV, Karmazyn M & Noble EG (2002b). Redox signaling of cardiac HSF1 DNA binding. Am J Physiol Cell Physiol 283, C404–C411. Peake JM, Markworth JF, Nosaka K, Raastad T, Wadley GD & Coffey VG (2015). Modulating exercise-induced hormesis: Does less equal more? J Appl Physiol 119, 172–189.

 C 2016 The Authors. The Journal of Physiology  C 2016 The Physiological Society

J Physiol 594.18

Exercise preconditioning protection of striated muscle

Peart JN, Gross ER, Headrick JP & Gross GJ (2007). Impaired p38 MAPK/HSP27 signaling underlies aging-related failure in opioid-mediated cardioprotection. J Mol Cell Cardiol 42, 972–980. Peart JN, Pepe S, Reichelt ME, Beckett N, See Hoe L, Ozberk V, Niesman IR, Patel HH & Headrick JP (2014). Dysfunctional survival-signaling and stress-intolerance in aged murine and human myocardium. Exp Gerontol 50, 72–81. Pereira B, Costa Rosa LF, Safi DA, Medeiros MH, Curi R & Bechara EJ (1994). Superoxide dismutase, catalase, and glutathione peroxidase activities in muscle and lymphoid organs of sedentary and exercise-trained rats. Physiol Behav 56, 1095–1099. Perry CG, Lally J, Holloway GP, Heigenhauser GJ, Bonen A & Spriet LL (2010). Repeated transient mRNA bursts precede increases in transcriptional and mitochondrial proteins during training in human skeletal muscle. J Physiol 588, 4795–4810. Pilegaard H, Ordway GA, Saltin B & Neufer PD (2000). Transcriptional regulation of gene expression in human skeletal muscle during recovery from exercise. Am J Physiol Endocrinol Metab 279, E806–E814. Powers SK, Criswell D, Lawler J, Ji LL, Martin D, Herb RA & Dudley G (1994). Influence of exercise and fiber type on antioxidant enzyme activity in rat skeletal muscle. Am J Physiol Regul Integr Comp Physiol 266, R375–R380. Powers SK, Criswell D, Lawler J, Martin D, Lieu FK, Ji LL & Herb RA (1993). Rigorous exercise training increases superoxide dismutase activity in ventricular myocardium. Am J Physiol Heart Circ Physiol 265, H2094–H2098. Powers SK & Hamilton K (1999). Antioxidants and exercise. Clin Sports Med 18, 525–536. Powers SK, Ji LL & Leeuwenburgh C (1999). Exercise training-induced alterations in skeletal muscle antioxidant capacity: a brief review. Med Sci Sports Exerc 31, 987–997. Powers SK & Lennon SL (1999). Analysis of cellular responses to free radicals: focus on exercise and skeletal muscle. Proc Nutr Soc 58, 1025–1033. Prosser BL, Ward CW & Lederer WJ (2011). X-ROS signaling: rapid mechano-chemo transduction in heart. Science 333, 1440–1445. Quindry J, French J, Hamilton K, Lee Y, Mehta JL & Powers S (2005). Exercise training provides cardioprotection against ischemia-reperfusion induced apoptosis in young and old animals. Exp Gerontol 40, 416–425. Quindry JC, Hamilton KL, French JP, Lee Y, Murlasits Z, Tumer N & Powers SK (2007). Exercise-induced HSP-72 elevation and cardioprotection against infarct and apoptosis. J Appl Physiol 103, 1056–1062. Quindry JC, Miller L, McGinnis G, Kliszczewicz B, Irwin JM, Landram M, Urbiztondo Z, Nanayakkara G & Amin R (2012). Ischemia reperfusion injury, KATP channels, and exercise-induced cardioprotection against apoptosis. J Appl Physiol 113, 498–506. Quindry JC, Schreiber L, Hosick P, Wrieden J, Irwin JM & Hoyt E (2010). Mitochondrial KATP channel inhibition blunts arrhythmia protection in ischemic exercised hearts. Am J Physiol Heart Circ Physiol 299, H175–H183.

 C 2016 The Authors. The Journal of Physiology  C 2016 The Physiological Society

5181

Radak Z, Asano K, Inoue M, Kizaki T, Oh-Ishi S, Suzuki K, Taniguchi N & Ohno H (1995). Superoxide dismutase derivative reduces oxidative damage in skeletal muscle of rats during exhaustive exercise. J Appl Physiol 79, 129–135. Radak Z, Chung HY & Goto S (2005). Exercise and hormesis: oxidative stress-related adaptation for successful aging. Biogerontology 6, 71–75. Radak Z, Chung HY & Goto S (2008). Systemic adaptation to oxidative challenge induced by regular exercise. Free Radic Biol Med 44, 153–159. Radak Z, Sasvari M, Nyakas C, Pucsok J, Nakamoto H & Goto S (2000). Exercise preconditioning against hydrogen peroxide-induced oxidative damage in proteins of rat myocardium. Arch Biochem Biophys 376, 248–251. Radak Z, Zhao Z, Koltai E, Ohno H & Atalay M (2013). Oxygen consumption and usage during physical exercise: the balance between oxidative stress and ROS-dependent adaptive signaling. Antioxid Redox Signal 18, 1208–1246. Razvickas CV, Borges FT, de Oliveira AS, Schor N & Boim MA (2013). The effect of hypoxia and reoxygenation in the response of mesangial cells to angiotensin II in vitro. J Bras Nefrol 35, 259–264. Rikka S, Quinsay MN, Thomas RL, Kubli DA, Zhang X, Murphy AN & Gustafsson AB (2011). Bnip3 impairs mitochondrial bioenergetics and stimulates mitochondrial turnover. Cell Death Differ 18, 721–731. Rinaldi B, Corbi G, Boccuti S, Filippelli W, Rengo G, Leosco D, Rossi F, Filippelli A & Ferrara N (2006). Exercise training affects age-induced changes in SOD and heat shock protein expression in rat heart. Exp Gerontol 41, 764–770. Ristow M & Zarse K (2010). How increased oxidative stress promotes longevity and metabolic health: The concept of mitochondrial hormesis (mitohormesis). Exp Gerontol 45, 410–418. Rodriguez-Miguelez P, Fernandez-Gonzalo R, Almar M, Mejias Y, Rivas A, de Paz JA, Cuevas MJ & Gonzalez-Gallego J (2014). Role of Toll-like receptor 2 and 4 signaling pathways on the inflammatory response to resistance training in elderly subjects. Age (Dordr) 36, 9734. Rouhier N, Lemaire SD & Jacquot JP (2008). The role of glutathione in photosynthetic organisms: emerging functions for glutaredoxins and glutathionylation. Annu Rev Plant Biol 59, 143–166. Sack MN (2011). Emerging characterization of the role of SIRT3-mediated mitochondrial protein deacetylation in the heart. Am J Physiol Heart Circ Physiol 301, H2191–H2197. Sakellariou GK, Jackson MJ & Vasilaki A (2014). Redefining the major contributors to superoxide production in contracting skeletal muscle. The role of NAD(P)H oxidases. Free Radic Res 48, 12–29. Sakellariou GK, Vasilaki A, Palomero J, Kayani A, Zibrik L, McArdle A & Jackson MJ (2013). Studies of mitochondrial and nonmitochondrial sources implicate nicotinamide adenine dinucleotide phosphate oxidase(s) in the increased skeletal muscle superoxide generation that occurs during contractile activity. Antioxid Redox Signal 18, 603–621.

5182

J. M. Lawler and others

Saleem A, Carter HN & Hood DA (2014). p53 is necessary for the adaptive changes in cellular milieu subsequent to an acute bout of endurance exercise. Am J Physiol Cell Physiol 306, C241–C249. Samelman TR (2000). Heat shock protein expression is increased in cardiac and skeletal muscles of Fischer 344 rats after endurance training. Exp Physiol 85, 92–102. Schlesinger MJ (1990). Heat shock proteins. J Biol Chem 265, 12111–12114. Schriner SE & Linford NJ (2006). Extension of mouse lifespan by overexpression of catalase. Age (Dordr) 28, 209–218. Schulpis KH, Tsironi M, Skenderi K, Lazaropoulou C, Parthimos N, Reclos G, Goussetis E, Tsakiris S & Papassotiriou I (2008). Dramatic reduction of erythrocyte glucose-6-phosphate dehydrogenase activity in athletes participating in the ultradistance foot race “Spartathlon”. Scand J Clin Lab Invest 68, 228–232. Seip RL, Moulin P, Cocke T, Tall A, Kohrt WM, Mankowitz K, Semenkovich CF, Ostlund R & Schonfeld G (1993). Exercise training decreases plasma cholesteryl ester transfer protein. Arterioscler Thromb 13, 1359–1367. Selye H (1946). The general adaptation syndrome and the diseases of adaptation. J Clin Endocrinol Metab 6, 117–230. Sen CK (1999). Glutathione homeostasis in response to exercise training and nutritional supplements. Mol Cell Biochem 196, 31–42. Senf SM, Dodd SL, McClung JM & Judge AR (2008). Hsp70 overexpression inhibits NF-κB and Foxo3a transcriptional activities and prevents skeletal muscle atrophy. FASEB J 22, 3836–3845. Sexton WL, Korthuis RJ & Laughlin MH (1990). Microvascular injury after ischemia and reperfusion in skeletal muscle of exercise-trained rats. J Appl Physiol 68, 2329–2336. Shi Y, Ivannikov MV, Walsh ME, Liu Y, Zhang Y, Jaramillo CA, Macleod GT & Van Remmen H (2014). The lack of CuZnSOD leads to impaired neurotransmitter release, neuromuscular junction destabilization and reduced muscle strength in mice. PLoS One 9, e100834. Sinha D, Ghosh AK, Mukherjee S, Biswas R & Biswas T (2015). Porin differentiates TLR mediated proinflammatory response of follicular zone B cell from TLR-unresponsive IL-10 expressing marginal zone B cell. Cytokine 76, 193–205. Smirnova OA, Isaguliants MG, Hyvonen MT, Keinanen TA, Tunitskaya VL, Vepsalainen J, Alhonen L, Kochetkov SN & Ivanov AV (2012). Chemically induced oxidative stress increases polyamine levels by activating the transcription of ornithine decarboxylase and spermidine/spermine-N1-acetyltransferase in human hepatoma HUH7 cells. Biochimie 94, 1876–1883. Smolka MB, Zoppi CC, Alves AA, Silveira LR, Marangoni S, Pereira-Da-Silva L, Novello JC & Macedo DV (2000). HSP72 as a complementary protection against oxidative stress induced by exercise in the soleus muscle of rats. Am J Physiol Regul Integr Comp Physiol 279, R1539–R1545. Song Y, Igawa S & Horii A (1996). Antioxidant enzymes response to endurance exercise training and dietary proteins in rat skeletal muscle and liver. Appl Human Sci 15, 219–225.

J Physiol 594.18

Spielmann N, Leon AS, Rao DC, Rice T, Skinner JS, Bouchard C & Rankinen T (2007). CETP genotypes and HDL-cholesterol phenotypes in the HERITAGE Family Study. Physiol Genom 31, 25–31. Spurney CF, Knoblach S, Pistilli EE, Nagaraju K, Martin GR & Hoffman EP (2008). Dystrophin-deficient cardiomyopathy in mouse: expression of Nox4 and Lox are associated with fibrosis and altered functional parameters in the heart. Neuromuscul Disord 18, 371–381. Starkov AA, Chinopoulos C & Fiskum G (2004). Mitochondrial calcium and oxidative stress as mediators of ischemic brain injury. Cell Calcium 36, 257–264. Starnes JW, Barnes BD & Olsen ME (2007). Exercise training decreases rat heart mitochondria free radical generation but does not prevent Ca2+ -induced dysfunction. J Appl Physiol 102, 1793–1798. Starnes JW, Taylor RP & Ciccolo JT (2005). Habitual low-intensity exercise does not protect against myocardial dysfunction after ischemia in rats. Eur J Cardiovasc Prev Rehabil 12, 169–174. Starnes JW, Taylor RP & Park Y (2003). Exercise improves postischemic function in aging hearts. Am J Physiol Heart Circ Physiol 285, H347–H351. Strobel NA, Matsumoto A, Peake JM, Marsh SA, Peternelj TT, Briskey D, Fassett RG, Coombes JS & Wadley GD (2014). Altering the redox state of skeletal muscle by glutathione depletion increases the exercise-activation of PGC-1α. Physiol Rep 2, e12224. Sumida S, Nakamura H & Yodoi J (2004). Thioredoxin induction of peripheral blood mononuclear cells in mice in response to a single bout of swimming exercise. Gen Physiol Biophys 23, 241–249. Sun QA, Hess DT, Nogueira L, Yong S, Bowles DE, Eu J, Laurita KR, Meissner G & Stamler JS (2011). Oxygen-coupled redox regulation of the skeletal muscle ryanodine receptor-Ca2+ release channel by NADPH oxidase 4. Proc Natl Acad Sci USA 108, 16098–16103. Takada S, Kinugawa S, Matsushima S, Takemoto D, Furihata T, Mizushima W, Fukushima A, Yokota T, Ono Y, Shibata H, Okita K & Tsutsui H (2015). Sesamin prevents decline in exercise capacity and impairment of skeletal muscle mitochondrial function in mice with high-fat diet-induced diabetes. Exp Physiol 100, 1319–1330. Takahashi K (1990). Calpain substrate specificity. In Intracellular Calcium-Dependent Proteolysis, ed. Mellgren R & Murachi T, pp. 55–74. CRC Press, Boca Raton, FL. Toshinai K, Oh-ishi S, Kizaki T, Ookawara T, Haga S & Ohno H (1997). Effect of swimming training on antioxidant enzymes in kidney of young and old mice. Res Commun Mol Pathol Pharmacol 95, 259–274. Tuomilehto J, Lindstrom J, Eriksson JG, Valle TT, Hamalainen H, Ilanne-Parikka P, Keinanen-Kiukaanniemi S, Laakso M, Louheranta A, Rastas M, Salminen V, Uusitupa M; Finnish Diabetes Prevention Study Group (2001). Prevention of type 2 diabetes mellitus by changes in lifestyle among subjects with impaired glucose tolerance. N Engl J Med 344, 1343–1350.

 C 2016 The Authors. The Journal of Physiology  C 2016 The Physiological Society

J Physiol 594.18

Exercise preconditioning protection of striated muscle

Turner JE, Bennett SJ, Campbell JP, Bosch JA, Aldred S & Griffiths HR (2013). The antioxidant enzyme peroxiredoxin-2 is depleted in lymphocytes seven days after ultra-endurance exercise. Free Radic Res 47, 821–828. Vainshtein A, Tryon LD, Pauly M & Hood DA (2015). Role of PGC-1α during acute exercise-induced autophagy and mitophagy in skeletal muscle. Am J Physiol Cell Physiol 308, C710–C719. Vincent HK, Shanely RA, Stewart DJ, Demirel HA, Hamilton KL, Ray AD, Michlin C, Farkas GA & Powers SK (2002). Adaptation of upper airway muscles to chronic endurance exercise. Am J Respir Crit Care Med 166, 287–293. Wang M, Zhang J, Walker SJ, Dworakowski R, Lakatta EG & Shah AM (2010). Involvement of NADPH oxidase in age-associated cardiac remodeling. J Mol Cell Cardiol 48, 765–772. Wang W, Zhang H, Xue G, Zhang L, Zhang W, Wang L, Lu F, Li H, Bai S, Lin Y, Lou Y, Xu C & Zhao Y (2014). Exercise training preserves ischemic preconditioning in aged rat hearts by restoring the myocardial polyamine pool. Oxid Med Cell Longev 2014, 457429. Wang Y, Zhao J, Yang W, Bi Y, Chi J, Tian J & Li W (2015). High-dose alcohol induces reactive oxygen species-mediated apoptosis via PKC-β/p66Shc in mouse primary cardiomyocytes. Biochem Biophys Res Commun 456, 656–661. Whitehead NP, Yeung EW, Froehner SC & Allen DG (2010). Skeletal muscle NADPH oxidase is increased and triggers stretch-induced damage in the mdx mouse. PLoS One 5, e15354. Williamson CL, Dabkowski ER, Dillmann WH & Hollander JM (2008). Mitochondria protection from hypoxia/reoxygenation injury with mitochondria heat shock protein 70 overexpression. Am J Physiol Heart Circ Physiol 294, H249–H256. Willis MS, Min JN, Wang S, McDonough H, Lockyer P, Wadosky KM & Patterson C (2013). Carboxyl terminus of Hsp70-interacting protein (CHIP) is required to modulate cardiac hypertrophy and attenuate autophagy during exercise. Cell Biochem Funct 31, 724–735. Wilund KR, Ferrell RE, Phares DA, Goldberg AP & Hagberg JM (2002). Changes in high-density lipoprotein-cholesterol subfractions with exercise training may be dependent on cholesteryl ester transfer protein (CETP) genotype. Metabolism 51, 774–778. Wojtaszewski JF, Jorgensen SB, Frosig C, MacDonald C, Birk JB & Richter EA (2003). Insulin signalling: effects of prior exercise. Acta Physiol Scand 178, 321–328. Wood JM, Decker H, Hartmann H, Chavan B, Rokos H, Spencer JD, Hasse S, Thornton MJ, Shalbaf M, Paus R & Schallreuter KU (2009). Senile hair graying: H2 O2 -mediated oxidative stress affects human hair color by blunting methionine sulfoxide repair. FASEB J 23, 2065–2075.

 C 2016 The Authors. The Journal of Physiology  C 2016 The Physiological Society

5183

Yamashita N, Hoshida S, Otsu K, Asahi M, Kuzuya T & Hori M (1999). Exercise provides direct biphasic cardioprotection via manganese superoxide dismutase activation. J Exp Med 189, 1699–1706. Yoshida T, Nakamura H, Masutani H & Yodoi J (2005). The involvement of thioredoxin and thioredoxin binding protein-2 on cellular proliferation and aging process. Ann NY Acad Sci 1055, 1–12. Zanchi NE, Lira FS, de Siqueira Filho MA, Rosa JC, de Oliveira Carvalho CR, Seelaender M, Santos RV & Lancha AH Jr (2010). Chronic low frequency/low volume resistance training reduces pro-inflammatory cytokine protein levels and TLR4 mRNA in rat skeletal muscle. Eur J Appl Physiol 109, 1095–1102. Zeng X, Zhao X, Yang Y, Kuai J, Gao C, Yu D, Zhao H, Chai W & Yao L (2011). Opioid δ1 and δ2 receptor agonist attenuate myocardial injury via mPTP in rats with acute hemorrhagic shock. J Surg Res 169, 267–276. Zhang B, Kawachi E, Miura S, Uehara Y, Matsunaga A, Kuroki M & Saku K (2013). Therapeutic approaches to the regulation of metabolism of high-density lipoprotein. Novel HDL-directed pharmacological intervention and exercise. Circ J 77, 2651–2663. Zhou L, Guo X, Chen M, Fu S, Zhou J, Ren G, Yang Z & Fan W (2013). Inhibition of δ-opioid receptors induces brain glioma cell apoptosis through the mitochondrial and protein kinase C pathways. Oncol Lett 6, 1351–1357. Zhu M, Li MW, Tian XS, Ou XM, Zhu CQ & Guo JC (2009). Neuroprotective role of δ-opioid receptors against mitochondrial respiratory chain injury. Brain Res 1252, 183–191.

Additional information Competing interests None declared. Funding Support has been provided by grants to J.M.L. from the National Aeronautics and Space Administration (NASA; NNX13AE45G), National Institutes of Health (NIH; AR054084), National Science Foundation (NSF; 055185F), American Heart Association (AHA; 0855158F), and the Huffines Institute at Texas A&M University. J.M.H. is funded through the Strategic Research Award from the College of Education and Human Development (CEHD). Acknowledgements We wish to thank members of the Redox Biology and Cell Signaling Laboratory for thoughtful and constructive discussions concerning the manuscript.

Mitochondria in the middle: exercise preconditioning protection of striated muscle.

Cellular and physiological adaptations to an atmosphere which became enriched in molecular oxygen spurred the development of a layered system of stres...
535KB Sizes 0 Downloads 9 Views