You are on page 1of 17

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/339982261

Redox Signaling in Widespread Health Benefits of Exercise

Article in Antioxidants and Redox Signaling · March 2020


DOI: 10.1089/ars.2019.7949

CITATIONS READS
6 2,424

7 authors, including:

Ruy Andrade Louzada Leonardo Matta


University of Miami Miller School of Medicine Federal University of Rio de Janeiro
36 PUBLICATIONS 559 CITATIONS 8 PUBLICATIONS 66 CITATIONS

SEE PROFILE SEE PROFILE

João Pedro Saar Werneck-de-Castro Corinne Dupuy


Federal University of Rio de Janeiro Institut de Cancérologie Gustave Roussy
66 PUBLICATIONS 1,601 CITATIONS 113 PUBLICATIONS 4,240 CITATIONS

SEE PROFILE SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Genetic susceptibility to radiation induced thyroïd cancer View project

Modulating factors of neurogenesis in the adult CNS View project

All content following this page was uploaded by Leonardo Matta on 27 April 2020.

The user has requested enhancement of the downloaded file.


ANTIOXIDANTS & REDOX SIGNALING
Volume 00, Number 00, 2020
ª Mary Ann Liebert, Inc.
DOI: 10.1089/ars.2019.7949

Redox Signaling in Widespread Health Benefits of Exercise

Ruy A. Louzada,1,2 Jessica Bouviere,1 Leonardo P. Matta,1 Joao Pedro Werneck-de-Castro,3


Corinne Dupuy,2 Denise P. Carvalho,1 and Rodrigo S. Fortunato1
Downloaded by UNIV OF MIAMI MILLER SCH MED Package NERL from www.liebertpub.com at 04/27/20. For personal use only.

Abstract

Significance: Exercise-induced reactive oxygen species (ROS) production activates multiple intracellular
signaling pathways through genomic and nongenomic mechanisms that are responsible for the beneficial effects
of exercise in muscle. Beyond the positive effect of exercise on skeletal muscle cells, other tissues such as white
and brown adipose, liver, central nervous system, endothelial, heart, and endocrine organ tissues are also
responsive to exercise.
Recent Advances: Crosstalk between different cells is essential to achieve homeostasis and to promote the
benefits of exercise through paracrine or endocrine signaling. This crosstalk can be mediated by different
effectors that include the secretion of metabolites of muscle contraction, myokines, and exosomes. During the
past 20 years, it has been demonstrated that contracting muscle cells produce and secrete different classes of
myokines, which functionally link muscle with nearly all other cell types.
Critical Issues: The redox signaling behind this exercise-induced crosstalk is now being decoded. Many of
these widespread beneficial effects of exercise require not only a complex ROS-dependent intramuscular
signaling cascade but simultaneously, an integrated network with many remote tissues.
Future Directions: Strong evidence suggests that the powerful beneficial effect of regular physical activity for
preventing (or treating) a large range of disorders might also rely on ROS-mediated signaling. Within a
contracting muscle, ROS signaling may control exosomes and myokines secretion. In remote tissues, exercise
generates regular and synchronized ROS waves, creating a transient pro-oxidative environment in many cells.
These new concepts integrate exercise, ROS-mediated signaling, and the widespread health benefits of exercise.
Antioxid. Redox Signal. 00, 000–000.

Keywords: ROS, exercise, myokines, redox signaling

Introduction physical exercise (64, 133). A more detailed description of


how physical exercise increases skeletal muscle ROS gen-

R eactive oxygen species (ROS), such as superoxide


(O2-), hydroxyl (OH), and the nonfree radical species,
H2O2, are small radical or nonradical molecules derived from
eration and the role of ROS-mediated signaling for muscular
exercise adaptations has been extensively reviewed by others
(63, 84, 129, 130).
molecular oxygen (O2) (142). Superoxide and nitric oxide In 1961, the term ‘‘exercise factors’’ was used to define a
seem to be the primary free radicals generated by different group of factors delivered by contracting muscles that were
sources in contracting skeletal muscle during and immedi- able to lower glycemia (62). Nowadays, metabolites, lipid
ately after exercise (84, 129, 130). ROS have a critical role as peroxidation products, extracellular ROS, myokines, and
signaling molecules that are necessary for skeletal muscle exosomes are the main suggested mechanisms through which
force development and to achieve the adaptive responses to muscle cells communicate with remote cells. These factors

1
Institut of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.
2
Université Paris-Sud, Orsay, UMR 8200 CNRS and Institut Gustave Roussy, Villejuif, France.
3
Division of Endocrinology, Diabetes and Metabolism, Miller School of Medicine, University of Miami, Miami, Florida, USA.

1
2 LOUZADA ET AL.

that are modulated by exercise, in a chronic perspective, are


directly related to the benefits of physical exercise under
different physiological and pathophysiological conditions. In
this review, we will focus on the effects of physical exercise
beyond skeletal muscle, providing an overview about the
communication between contracting muscles and remote
tissues through ROS-mediated mechanisms.

Hypothetical Model of ROS Waves


The ROS waves hypothesis is based on the ability of
physical exercise to increase ROS production in contracting
muscles and its later spread to remote tissues. A hypothetical
model is proposed in Figure 1 to summarize the probable
Downloaded by UNIV OF MIAMI MILLER SCH MED Package NERL from www.liebertpub.com at 04/27/20. For personal use only.

mechanisms through which contracting skeletal muscle


communicates with remote tissues, leading to changes in
redox-mediated signaling pathways that operate the wide-
spread health beneficial effects of exercise. A first phase of
ROS waves after exercise occurs in contracting muscle and
promotes multiple post-translational modifications of ami-
noacids in many kinases and phosphatases (e.g., AMPK,
CaMK, PTEN, PPIA, etc.) that are crucial for the local
stimulus of muscular glucose uptake, mitochondrial biogen-
esis, and antioxidant capacity. Further, skeletal muscles also
produce ROS in the extracellular milieu that leads to macro-
molecules oxidation, such as lipid peroxidation. Interestingly,
lipid peroxidation products can act as signaling molecules.
Myokines and exosomes might be released from skeletal
muscle in response to intracellular ROS stimulated by phys-
ical exercise. Once in blood circulation, these mediators
change the function of remote tissues during and after exercise FIG. 1. Exercise-induced ROS waves illustration. (A)
periods. Further, the second phase of ROS waves might Contracting muscle creates an oxidative environment through
stimulate many similar redox-sensible signaling pathways in consecutive ROS spikes. The first ROS wave is produced
noncontracting tissues that promote widespread responses to mainly by skeletal muscle Nox2, induces activation of ROS-
physical exercise (Fig. 1B). mediated signaling pathways, and promotes correct folding of
In 1978, Dillard et al. found that expired pentane, an index newly born proteins. Skeletal muscle releases signals to re-
of lipid peroxidation, increased during aerobic physical ex- mote tissues in many ways during muscle contraction. For
ercise (42). One year later, Brady et al. demonstrated that example, metabolites released by skeletal muscle can be ta-
lipid peroxidation levels were higher in the skeletal muscle ken up and metabolized in remote tissues, an event that can
stimulate ROS production. Moreover, myokines can also
and liver of rats immediately after swimming (22). Probably, control the production of ROS in remote tissues. Products of
this was the first indirect demonstration of the ability of lipid peroxidation (HODE and HNE) produced during skel-
physical exercise to increase ROS generation, but the tissue etal muscle contraction are capable of binding to nuclear
involved in their production was only revealed 3 years later. receptors in remote cells in which these compounds can then
Davies et al. showed by using electron paramagnetic resonance regulate gene expression. Finally, exosomes released by the
that skeletal muscle and liver ROS production are increased muscles undergoing contraction might stimulate signaling
immediately after exhaustive exercise (36). Nowadays, it is pathways in remote tissues. ROS production in remote tissues
well known that skeletal muscle contractile activity leads to after contraction is proposed as a second ROS wave. (B) First
ROS generation, which is related to biomolecule oxidation, not and second ROS waves induced by physical exercise in a
only in skeletal muscle but also in other tissues (114, 130). temporal perspective. During skeletal muscle contraction, the
first ROS wave within skeletal muscle occurs during exercise
Electrical stimulation of the hind limb muscle in adult mice and can spread to remote tissues. At the end of exercise,
was found to increase malonaldehyde levels in serum and liver contracting muscle causes a decrease in ROS production,
and decrease liver glutathione and protein thiol content (34), whereas remote tissues present the second ROS wave, leading
suggesting that the ROS generated by skeletal muscle could to activation of ROS-mediated signaling pathways. 13-
oxidize extracellular molecules also in noncontracting tissues, HODE, 13-hydroxyoctadecadienoic acid; 4-HNE, 4-hydro-
such as the liver. In humans, the venous concentration of ox- xynonenal; Nox2, NADPH oxidase 2; ROS, reactive oxygen
idized glutathione was found to increase after physical exercise species. Color images are available online.
in an intensity-dependent manner (35, 114).
The redox communication between contracting muscle myokines; (iv) ROS gradient reaching adjacent cells; (v) lipid
and remote tissues can be mediated by several mechanisms: peroxidation products; and (vi) exosomes secretion. The
(i) lactate conversion to pyruvate in almost every cell, which oxidative environment induced in remote tissues by physical
increases NADH levels; (ii) purines released by contracting exercise is probably related to protein modification in their
muscle that serve as substrates for xanthine oxidase; (iii) cysteine residues, which cause structural and functional
REDOX SIGNALING IN WIDESPREAD EFFECTS OF EXERCISE 3

modifications (Fig. 1) and increased oxidation markers that almost all other cellular locations that have reducing redox
are detected after exercise in noncontracting tissues. More- potentials, the typical ER has the oxidative redox potential
over, several myokines released during physical exercise that is necessary to form disulfide bonds. It has been reported
control the expression of genes in an ROS-dependent man- that the ER of insulin-resistant rodents contains much higher
ner, as well as the activity of transcription factors, such as proportions of unfolded polypeptides and fewer disulphide
peroxisome proliferator-activated receptor gamma coacti- bonds than normal endoplasmic reticula, which is associated
vator 1-alpha (PGC-1a), nuclear factor erythroid 2-related with the loss of protein folding regulation (116).
factor 2 (Nrf2), and nuclear factor-kappa B (33, 90, 123, 142). Interestingly, exercise reduces negative outcomes in Alz-
heimer’s disease, including the accumulation of misfolded
proteins (159). The unfolded protein response (UPR) is in-
Adaptations and Benefits of Redox Signaling
duced within contracting muscle cells and plays an important
Due to Exercise
role in cell protection from stress, mainly through an essential
The expression of PGC-1a during physical exercise is chaperoning system (50). Exercise seems to improve many
dependent on a tightly, fine-tuned redox-mediated signal ERS-related pathologies, such as diabetes mellitus, neurode-
Downloaded by UNIV OF MIAMI MILLER SCH MED Package NERL from www.liebertpub.com at 04/27/20. For personal use only.

(87), and boosting PGC-1a has been related to protection generative diseases, sarcopenia, or cardiovascular alterations
against several pathological conditions, such as sarcopenia, (81, 91, 169) and PGC-1a, the redox sensible master gene
insulin resistance, and cancer-induced cachexia (68). PGC- involved in the beneficial effects of exercise, interacts with
1a is responsible for many of the beneficial effects of ATF6a to regulate the UPR in muscle (169). Many proteins
exercise in muscle due to its ability to orchestrate the tran- included in the axis that regulates UPR activation (e.g.,
scription of both nuclear and mitochondrial DNA, promoting PERK, eIF2a, ATF4, CHOP, IRE1, XBP1, BiP, and ATF-6)
a balanced expression of mitochondrial proteins and anti- are regulated in both muscle and remote tissues after ex-
oxidant defenses against exacerbated ROS production. Im- ercise training. For example, exercise training upregulates
portantly, an accumulation of PGC-1a after each bout of ER chaperones, such as binding immunoglobulin protein
exercise is indispensable to achieve these beneficial effects (BiP) in mouse muscle (39, 110), liver (31), and brain (89,
(47, 53, 70). 96). In summary, both acute and chronic exercise seems to
Many of the intracellular actors involved in obtaining the activate UPR signaling to alleviate ERS-related patholo-
beneficial effects of exercise contain redox sensible regions gies (50).
that are directly affected by ROS. The crucial requirement of A tight regulation of UPR is also crucial to the correct
optimal ROS signaling is clearly observed by the effect of folding of tumor suppressor proteins (79, 166). An exercise-
antioxidants in hampering many important adaptations that mediated control of tumor suppressor proteins has been
are related to endurance training, such as an increase in ex- proposed to be one plausible mechanism for the lower inci-
ercise capacity (21, 64), mitochondrial biogenesis (153), dence of cancer in regular physically active individuals (113,
muscular glucose uptake, improvement in insulin sensitivity 166). Indeed, a tumor suppressor protein might be delivered
(133), and the increase of lean mass after resistance training from contracting muscle or synthetized directly in different
(17). tissues by ROS-mediated signaling that is induced by phys-
Several chronic exercise adaptations are dependent on ical exercise (Fig. 1A). Another possible mechanism is re-
ROS produced by skeletal muscle in response to an acute lated to decreased insulin resistance in trained individuals,
exercise session. Transient increases in ROS induced through since insulin resistance seem to be positively related to tumor
bouts of acute exercise stimulate Nrf2 activation. Nrf2 is the development (3). Once again, notably, such hypotheses fit
master regulator of antioxidant defenses, a transcription well with the association of being actively healthy and the
factor that regulates the expression of more than 200 cyto- lower incidence of cancer (83).
protective genes (44). Regular aerobic exercise in rodent
models has consistently been shown to activate Nrf2 sig-
Lipid Peroxidation as a Mediator of Crosstalk
naling in multiple tissues, including skeletal muscle (111),
Between Different Tissues
kidney (122), brain (162), liver (135), and myocardium (145),
which leads to the upregulation of endogenous antioxidant Over the past three decades, an extensive body of literature
defenses and an overall greater ability to counteract the has shown the role of lipid peroxidation, not only in the
damaging effects of oxidative stress. pathophysiology of diseases but also in cell biology and
Regarding protein quality control, exercise might be im- human health (9). Lipid peroxidation is a process in which
portant to the correct folding of proteins, due to the creation oxidants attack lipids containing carbon–carbon double
of an oxidative redox potential needed to oxidize the free bond(s) and involve hydrogen abstraction from a carbon,
sulfhydryl groups of cysteine leading to disulfide bonds for- with oxygen insertion resulting in lipid peroxyl radicals and
mation that stabilize the protein conformation (Fig. 1) (166). hydroperoxides (27). Increased ROS production can lead to
Antioxidant administration before exercise (64, 133) pre- membrane lipid peroxidation and different cell fate, de-
vents some of the most important exercise responses, prob- pending on the cellular metabolic state and repair ability (9).
ably by maintaining the intracellular environment in an Products of lipid peroxidation (at low rates) can mediate
inappropriate reduced state. This reduced state may impair cell to cell communication during exercise. NADPH oxidase
the correct folding of born proteins (166). Loss of protein 2 (NOX2) is expressed at the sarcolemma of skeletal muscle,
folding regulation leads to an accumulation of unfolded or and it seems to be the main source of ROS during contractile
misfolded proteins inside the endoplasmic reticulum (ER) activities that produce superoxide in the extracellular me-
lumen and drives ER stress (ERS), which accentuates the dium (74, 77, 138). It is believed that superoxide is rapidly
progression of many diseases, including Alzheimer’s. Unlike converted into H2O2, which is more stable and membrane
4 LOUZADA ET AL.

permeable, acting as a signaling molecule. However, super- oxidation during muscle contraction that has been suggested
oxide production near the sarcolema might increase the as a candidate molecule to communicate muscle with remote
oxidation of extracellular lipids/lipoproteins, converting cells. Formerly, it was believed that lactate was a waste
linonelic acid into 13-hydroxyoctadecadienoic acid (13- product of glycolysis, even referred to be the cause of fa-
HODE) and cholesterol into oxysterol, which is able to tiguing muscle during physical exercise (24). The role of
activate transcription factors such as peroxisome proliferator- lactate has changed from a possibly bad intermediate mole-
activated receptor gamma and liver X receptor alpha, cule in muscle physiology to an important messenger mole-
respectively (41). Apart from this, 4-Hydroxynonenal (4-HNE) cule during physical exercise (1). Nowadays, many
is another product of lipid peroxidation that can activate physiological functions are attributed to lactate, such as
transcription factors. It was demonstrated that 4-HNE acti- slowing muscular fatigue (117), increasing force production
vates Nrf2, increasing mitochondriogenesis and antioxidant (92), and serving as a high-energy rich carbon skeleton for
defense (95). Interestingly, Nrf2 is activated after physical many crucial biochemical processes within cells (24, 140).
exercise in several tissues, including the brain, kidney, and The biochemical aspect of lactate production and its
testis (44). Once products of lipid peroxidation are increased physiological role were extensively reviewed by Brooks (24).
Downloaded by UNIV OF MIAMI MILLER SCH MED Package NERL from www.liebertpub.com at 04/27/20. For personal use only.

in the blood after physical exercise (66), it is possible that In brief, there are three main proposed functions for lactate,
they can communicate with skeletal muscle and with other as follows: (i) as an energy source, (ii) a major gluconeogenic
distant cells in the body (Fig. 2). precursor after exercise, mainly due to a shuttle between the
skeletal muscle and the liver (the Cori cycle), and (iii) a
‘‘lactormone,’’ able to mediate autocrine, paracrine, and
Metabolites Acting as Communicating Molecules
endocrine communication (24). New information regarding a
Through ROS-Dependent Mechanisms
shuttle between contracting muscle and microbiota has just
A growing body of evidence suggests that different me- become available, which suggests that intestinal microbiota
tabolites produced during muscle contraction are secreted are able to metabolize lactate and, thus, influence the exercise
during physical exercise. Interestingly, these metabolic capacity (143). Besides, a potential mechanism showing that
products can act like chemical mediators between cells (20). lactate might be able to stimulate ROS-mediated signaling
Lactate is a very well-known metabolite produced by glucose has also been proposed (Fig. 3).

FIG. 2. Lipid peroxidation


as a mediator of skeletal
muscle-remote tissue cross-
talk. NADPH oxidase is pro-
posed as the main source of
ROS during contractile activity
and due to its transmembrane
location, ROS production is
very close to the phospholipids
of the membrane. Circulating
lipids and membrane phos-
pholipids can be attacked by
extracellular-produced ROS to
form lipid peroxidation inter-
mediates that can interact with
nuclear receptors in remote
tissues and modulate gene ex-
pression. Color images are
available online.
REDOX SIGNALING IN WIDESPREAD EFFECTS OF EXERCISE 5
Downloaded by UNIV OF MIAMI MILLER SCH MED Package NERL from www.liebertpub.com at 04/27/20. For personal use only.

FIG. 3. Crosstalk between muscle and other tissues. Skeletal muscle contraction increases glucose oxidation rates in an
exercise intensity-dependent manner and stimulates the conversion of pyruvate into lactate via LDH. Lactate is mainly
produced by fast-twitch muscles during exercise and is released in muscles undergoing contraction to the blood circulation
through specific transporters (such as MCT). Once in circulation, lactate can be taken up by many tissues, such as liver,
WAT, brain, heart, and kidney. In the liver, lactate is converted into pyruvate by PDH, causes an increase in the levels of
NADH+, and stimulates several intracellular metabolic pathways. Robust ATP hydrolysis produces ADP, which is con-
verted into ATP and AMP by AK. AMP deamination yields hypoxanthine, which is released into the circulation. Xanthine
oxidase converts hypoxanthine into urate and also produces superoxide, leading to redox-mediated signaling in the liver.
Urate is a potent antioxidant molecule that is released into the circulation. The proposed crosstalk between muscle and liver
involves spatially and temporally synchronized ROS production and ROS scavenging to ensure that correct ROS-mediated
signaling occurs within the whole body during and after exercise. In the left panel, intestinal NADH-dependent ROS
production is proposed as a mechanism that may use NADH derived from lactate oxidation by NAD(P)H oxidase to produce
ROS in a microbiota-dependent way. ADP, adenosine diphosphate; AK, adenylate kinase; AMP, adenosine monophosphate;
ATP, adenosine triphosphate; LDH, lactate dehydrogenase; MCT, monocarboxylate transporter; PDH, pyruvate dehydro-
genase; WAT, white adipose tissue. Color images are available online.

Intracellular lactate levels increase during physical ex- in remote tissues. Recently, it was demonstrated that lactate
ercise in an intensity dependent way. Lactate can cross the was able to upregulate transforming growth factor beta2
sarcolemma and reach blood circulation through mono- (TGF-b2) in white adipose tissue. Further, TGF-b2 was se-
carboxylate transporters (MCTs). Serum lactate can be creted by adipose tissue in response to physical exercise,
uptaken by a wide range of cells such as contracting skel- which is related to the improvement of glucose tolerance.
etal muscles themselves (16), heart (59), brain (132), liver Interestingly, the administration of a lactate-lowering agent
(48), kidney (112), astrocytes, or neurons (126). It is be- dichloroacetate during exercise training decreased circulat-
lieved that slow-twitch skeletal muscle fibers and liver are ing TGF-b2 levels and reduced exercise-stimulated im-
the most important tissues for the removal of lactate from provements in glucose tolerance (154).
blood (Fig. 3) (60), and the expression of MCTs is crucial A hypothetical model is proposed in Figure 1, which shows
for the uptake of lactate from circulation. Moreover, the probable redox-mediated mechanisms through which
functional mitochondria are fundamental for lactate to be contracting skeletal muscle communicates with remote tis-
oxidized (12, 24). sues. ROS might be involved in lactate action in different
As proposed by Hashimoto and Brooks, lactate may act as tissues. Indeed, lactate increases the expression of genes in-
a communicating mediator called ‘‘lactormone’’ that couples volved in lactate transport and mitochondrial oxidation in an
contracting muscle to other tissues (69). Supporting this hy- ROS-dependent manner (69), although the source of ROS has
pothesis, intraperitoneal l-lactate administration was found not been defined. Recently, it has been shown that lactate-
to increase mitochondrial content in diverse mouse tissues mediated increase in PGC1-a expression was blunted by
such as liver and brain (46), as well as PGC-1a and un- N-acetyl cysteine pretreatment in skeletal muscle (90, 115).
coupling protein 3 expression in gastrocnemius (90). Because Figure 3 illustrates the hypothetical sources of ROS that can
these events are also observed after physical exercise, the be activated by lactate: mitochondria, xanthine oxidase, and
transitory increase in blood lactate during physical exercise NAD(P)H oxidase. During lactate oxidation, NADH is gen-
can contribute to the widespread health adaptations that occur erated, increasing mitochondrial oxidative phosphorylation,
6 LOUZADA ET AL.

which can increase ROS production. Another potential signaling can lead to impaired function in many tissues and
mechanism may be the use of NADH derived from lactate blunt the widespread health benefits of exercise.
oxidation by NAD(P)H oxidase to produce ROS. This hy-
pothesis is supported by experiments in Drosophila in which H2O2 Gradient as a Chemoattracting Agent
lactate stimulates intestinal NADH-dependent ROS produc- Supporting Skeletal Muscle Regeneration
tion (82). A dysbiosis marked by overgrowth of Lactobacillus
In skeletal muscle, cooperation between immune and
plantarum in the fly gut increases lactate production and
muscular cells is observed during muscular regeneration. The
promotes over-proliferation of intestinal stem cells. Using
recruitment of neutrophils and monocytes from blood cir-
this model, the authors proposed that lactate is a key element
culation is mediated by chemoattracting molecules that are
for communication between microbes and intestinal cells,
secreted by many cells (e.g., endothelial cells, satellites cells,
and that NADH produced by oxidation of lactate derived
and immune-resident cells) at the lesion site. Multiple factors
from bacteria could be a substrate for NAD(P)H oxidase to
such as C-C motif ligand 2/monocyte chemoattractant
increase ROS production (82). In this context, it would be
protein-1 (MCP1) and tumor necrosis factor alpha are known
interesting to test whether the same process occurs after ex-
Downloaded by UNIV OF MIAMI MILLER SCH MED Package NERL from www.liebertpub.com at 04/27/20. For personal use only.

to attract monocytes and to differentiate them into macro-


ercise.
phages, respectively (128), orchestrating the process of
Lactate also seems to exert a free radical scavenger func-
myogenesis (32, 94, 137). H2O2 gradient also serves as a
tion, able to act as an antioxidant in some specific conditions
chemoattracting agent. In the zebrafish model, an H2O2
as heart ischemia, since the perfusion with a lactate solution
gradient generated by the DUOX enzyme (after tissue injury)
protects the tissue from oxidative damage (65). These newly
recruits leucocytes to the site of the lesion to promote wound
proposed functions of lactate, acting as a free radical scav-
healing (118). Moreover, H2O2 production seems to be cru-
enger and as a signaling molecule, are illustrated in Figure 3.
cial to promote progenitor cell proliferation during acute
A purine cycle also intermediates the crosstalk between
kidney lesions (33). In these two examples, the inhibition of
contracting muscle and liver. Mostly in the liver, inosine and
H2O2 generation abolished the regenerative process. In
hypoxanthine derived from the muscle can be converted into
mammals, the role of DUOX1 in the recruitment of immune
uric acid by xanthine oxidase (71). This cycle might be a
cells was demonstrated in lung inflammatory response, which
potential mechanism by which exercise promotes (paradox-
was found to be crucial to the regenerative process (30, 67).
ically) a pro-oxidant effect due to xanthine oxidase in the
Thus, one can speculate that exercise-induced H2O2 gener-
liver, and an increase of urate, a powerful antioxidant in
ation in skeletal muscle cells could create a gradient to attract
circulation (114). In fact, increased plasma total antioxidant
supportive cells to participate in exercise responses.
capacity has been observed after high-intensity exercise (71).
After eccentric exercise, there is a peak in pro-
The circulating urate can be taken up by different cells and,
inflammatory CD68+ macrophages infiltration at 48 h (100,
after a recovery period after exercise, converted into allantoin
102) that switches to anti-inflammatory CD163+ macro-
(73). During this process, there is a scavenger of the hydroxyl
phages after 72 h (57, 131). Given that ROS are produced and
radical, hydroperoxide, and lipid peroxidation (37). Inter-
released into the blood from skeletal muscle (11, 34, 84), one
estingly, urate is taken up preferentially by the contracting
can hypothesize that these molecules participate in the re-
muscle, and not by the resting ones (72). Although the
cruitment of immune cells to support cellular regeneration
mechanism is not well understood, urate might be useful for
after damaging exercise. In fact, DUOX1 is expressed in
protecting the contracting muscle from oxidative damage
muscular progenitor cells and its expression declines during
once its exogenous administration reduces exercise-induced
myogenesis (139), suggesting that DUOX1 could be a source
oxidative stress (165).
of ROS from satellite muscular progenitor cells to commu-
Based on this intimate co-operation between muscle and
nicate to other cells through an H2O2 gradient, as observed
liver, we propose synchronized and tightly regulated ROS-
during zebrafish and mammalian lung and kidney injury.
mediated signaling in remote tissues that might be dependent
on the products of muscle contraction and liver processing.
Myokines and Its ROS-Dependent Regulation:
This crosstalk is a well-organized network that synchronizes
Communicating the Contracting Muscle
contracting muscle and other remote tissues to ensure optimal
to the Whole Body
redox-mediated signaling during exercise and recovery (as
illustrated in the hypothetical schema in Fig. 1). For example, After contractile activity, skeletal muscle cells increase the
lactate is produced and released into blood circulation, production and release of several molecules such as cytokines
reaching multiple different tissues, then altering the metab- or other small proteins (*5–20 kDa) and proteoglycan pep-
olism, and finally promoting an ROS-mediated signaling. tides that show autocrine, paracrine, and/or endocrine effects.
Simultaneously, intense ATP hydrolyses, mostly found dur- Several well-known cytokines (e.g., IL-6, IL-8, IL-10, IL-15,
ing intense exercise, promote the release of inositol and etc.) have been shown to be secreted by skeletal muscle
hypoxanthine, which mostly reaches its peak after exercise. during physical exercise and, as a result, they are referred to
Once in the liver, urate is produced from hypoxanthine and as myokines (123). Myokines are involved in exercise-
released into circulating blood, delivering a potent antioxi- associated metabolic changes and not only in muscle but also
dant and probably counteracting the peaks of ROS produc- in other tissues such as heart, adipose tissue, liver, pancreas
tion. This hypothetical possibility elaborates the fine-tuning bone, and brain. Myokines play important roles in metabolic
process required for the exchange of purines and lactates that changes that are induced by physical exercise. They also
are derived from contracting muscle and is dictated by the participate in tissue regeneration and repair, immunomodula-
intensity of exercise. When this fascinating and natural tion. and cell differentiation (124). The interconnections be-
communication is perturbed, a disaccord in redox-mediated tween contracting muscle and practically all cells are complex
REDOX SIGNALING IN WIDESPREAD EFFECTS OF EXERCISE 7

and not a completely deciphered system. Currently, several preventing oxidative stress and tissue damage. Nrf2 activates
myokines have been described to be stimulated by physical a well-known antioxidant pathway that protects against var-
exercise, including IL-6 (121), Irisin (19), IL-15 (146), FGF21 ious pathological conditions (43, 97, 99), and its activation
(156), LIF (23), and MCP1 (28). Interestingly, the PGC-1a and seems to correlate with the antifibrotic and antioxidative ef-
Akt/mTOR pathways seem to be involved in myokine secre- fects of irisin (33, 45, 171). For instance, irisin treatment
tion, and both of them are redox-sensitive intracellular path- protects hepatocytes in vitro from overproduction of ROS
ways (Fig. 4). The most well-studied myokines are IL-6 and that is induced by H2O2 (13). Moreover, irisin mediates part
Irisin (121). of the protective effect of dexmedetomidine in livers under
Skeletal muscle IL-6 secretion increases after a single bout intestinal ischemia/reperfusion damages via decreasing in-
of physical exercise (124), which induces liver glucose out- flammasome markers and ROS production (51). In addition,
put (52). IL-6 secretion during physical exercise is not related irisin is known to decrease several pro-inflammatory cyto-
to tissue damage or pro-inflammatory processes (124, 142, kines and increase antioxidant defenses (e.g., SOD and GPX)
150), and it seems to be regulated by intracellular calcium in hepatocytes through Nrf2 upregulation (45). In endothelial
levels (167). It was demonstrated that the administration of a cells, irisin stimulates angiogenesis via AKT/mTOR/S6K1/
Downloaded by UNIV OF MIAMI MILLER SCH MED Package NERL from www.liebertpub.com at 04/27/20. For personal use only.

cocktail of antioxidants containing Vitamins A, C, and E Nrf2 signaling, which reduces the higher ROS content in-
(161) or Vitamin C and E (54) before a single bout of physical duced by oxidized LDL (Fig. 5) (171).
exercise attenuated IL-6 secretion in humans. L-NAME, a A major role of oxidative stress in fibrosis development is
nitric oxide synthase (NOS) inhibitor, also attenuated the caused by over-activated Smad-3 signaling (18) that leads to
increase in IL-6 mRNA levels in response to exercise (151). exacerbated ROS production (14). Mechanistically, irisin-
In addition, it has been suggested that NOX enzymes are induced Nrf2 activation in cardiac cells suppresses the ROS
involved in IL-6 secretion by skeletal muscle during physical generation that is induced by angiotensin II, which is triggered
exercise, since apocynin, an NOX inhibitor, attenuated this by TGF-b1 in fibroblasts (33). Moreover, irisin was found to
phenomenon, which also occurred in a gp91phox knockout prevent heart remodeling (98) and reduce blood pressure of
animal (75). spontaneously hypertensive rats through endothelial cells
IL-6, secreted by contracting skeletal muscle during re- oxidative stress reduction via AMPK and NO signaling (56).
sistance training, seems to activate satellite cells through In a type 2 diabetes mice model, irisin ameliorated endothelial
the activation of STAT3, which is crucial for hypertrophy function by reducing the overproduction of superoxide and
(144). It has not been investigated whether this action is peroxynitrite (172). However, in endothelial cells, irisin also
dependent on ROS-mediated signaling; however, ROS can reverted the oxidative stress induced by advanced glycation
stimulate STAT3 via a modification on its Tyr 7055 and end products by decreasing endothelial NOS activity and
Cys 253 (147). Further, IL-6 stimulates ROS production in therefore, preventing the endothelial damage that is secondary
plenty of different cell types (163), so it is possible that to inflammasome overactivation (40).
ROS can act as an intermediate in the effect of IL-6 in In macrophages, irisin was found to increase proliferation
remote tissues during exercise. For instance, increased in- and phagocytosis ability (105) and protected these cells
sulin sensitivity and fatty acid oxidation that is stimulated against oxidative stress damage via upregulation of antioxi-
by IL-6 (26) might, in part, be dependent on the activation dant defenses, even under LPS stimulation (106). Given the
of AMP-activated protein kinase (AMPK) by ROS (157). undeniable implication of macrophages in the development
Also, the stimulus of muscle glucose uptake after exercise and progression of many diseases, a direct action of exercise-
might be through a similar mechanism (61). The interplay induced irisin on macrophages could contribute to the
between IL-6 and AMPK has also been confirmed in dif- widespread beneficial effects of exercise. Some of these ef-
ferent tissues (88). fects are illustrated in Figure 5.
Irisin was first described as a myokine with the ability to Previous studies have suggested that myokines mediate the
induce a browning phenotype in white adipose tissue, raising beneficial effects of exercise in brain function (Fig. 5). Under
basal metabolic rates (Fig. 5) (19). Several studies showed an ischemia/reperfusion stroke, brain cells experiment a
that irisin promotes widespread effects in remote tissues, glucose/oxygen deprivation that triggers an intense ROS

FIG. 4. Myokines and ROS dependence. The most studied myokines are illustrated and their ROS dependence is
highlighted. IL-6, IL-8, and IL-15 are regulated by ROS. The control of several myokines relies on the activation of human
PGC-1a and AKT/mTOR signaling, but their dependence on redox signaling has not yet been completely investigated.
AKT/mTOR, protein kinase B/mechanistic target of rapamycin; IL, interleukin; PGC-1a, peroxisome proliferator-activated
receptor gamma coactivator 1-alpha. Color images are available online.
8 LOUZADA ET AL.

FIG. 5. Crosstalk between


skeletal muscle and other
tissues. The regular practice
of physical exercise promotes
widespread health benefits
Downloaded by UNIV OF MIAMI MILLER SCH MED Package NERL from www.liebertpub.com at 04/27/20. For personal use only.

that can prevent and treat


many diseases. Most of the
desirable effects of physical
exercise are mediated by
myokines that can facilitate
communication of contract-
ing muscles with remote tis-
sues. The mechanism of
action of many myokines re-
lies directly or indirectly on
redox signaling. Color ima-
ges are available online.

production that can lead to an irreversible injury. Using activity and cancer prevention, most of them have an in-
in vitro model, irisin protected neuron cells from oxidative complete description of physical activity (such as type, in-
stress via decreasing the higher ROS production and in- tensity, duration, and metabolic responses).
flammation through increased SOD expression, consequently Mechanistically, an antiproliferative effect of serum col-
protecting neurons from stroke-induced apoptosis (127). lected from mice after moderate exercise intensity (50% of
In 1944, one of the first reports suggested that exercise maximal exercise capacity), but not from mice submitted to
reduced tumor growth and progression in mice (136). Since high-intensity swimming exercise (50% and 80% of maximal
then, there are several evidences showing that a healthy capacity, respectively), confirms the idea that a mix of
lifestyle hinders the development of several types of cancer myokines are secreted that are able to affect tumor cells,
and regular exercise can protect from a large range of dis- reducing tumor growth (2). However, another study showed
eases (49, 119, 134). However, the precise mechanisms are that high-intensity training (corresponding to 85% of VO2
still unclear. Regarding the effect of exercise on tumor max) was able to promote a reduction in tumor mass (10). By
growth, there are many conflicting results that have been screening 28 potential candidate molecules upregulated in
obtained from rodent experiments. Recent studies have serum after swimming exercise, Oncostatin M has emerged
shown that exercise may decrease tumor growth if tumor- as a candidate for reducing mammary cancer cells growth
bearing mice were trained 4 weeks before inoculation with (78). Another candidate able to prevent the initiation of a
B16 melanoma cells (125), whereas another study suggested tumor was the ‘‘Secreted Protein Acidic and Rich in Cy-
avoiding free access to exercise because they observed in- steine’’ (SPARC). The level of circulating SPARC increased
creased tumor growth and decreased lifespan in a model of in mice and humans after a single bout of exercise. Low-
intramuscular tumor inoculation (8). Despite the gap of intensity exercise induces SPARC expression and secretion
knowledge about the mechanisms of actions involved in this that reduces the formation of aberrant crypt foci and prevents
process, regular exercise has been recommended as a sup- the onset of colon cancer (6). SPARC expression seems to be
plementary therapy for cancer patients (83). Based on exist- induced by low and moderate exercise programs (149) and is
ing evidence, some public health organizations have issued mainly secreted by slow-twitch fibers instead of fast-twitch
physical activity guidelines also for cancer prevention. Re- fibers after resistance exercise (Fig. 5) (148). Apart from its
garding the large number of studies conducted on physical possible direct action as an antitumor molecule, SPARC also
REDOX SIGNALING IN WIDESPREAD EFFECTS OF EXERCISE 9

seems to mimic some of the beneficial effects of exercise on is mediated by ATP-binding cassette transporter (ABCA1)
glucose metabolism via AMPK activation (5). transmembrane transporters (4). Further, three other mecha-
Therefore, myokines secreted by the muscles may mediate nisms can also be related to Hsp70 release, such as: (i) cell
the beneficial effects of exercise on tumor development. lysis with consequent release of cytoplasmic content; (ii) cell
Although it is a promising and innovative theme, the effects surface blebbing and Hsp70 release in microvesicles to the
of contracting muscle myokines (e.g., IL-6, Irisin, FGF21, extracellular fluid; and (iii) through endolysosomes (58, 103,
Oncostatin M, SPARC, etc.) on tumor growth and progres- 104). HSPs are molecular chaperones that facilitate protein
sion using xenograph and inoculated rodent models are still folding and maintain protein structure and function during
relatively unknown. cellular stress. HSPs are involved in a wide range of cellular
processes that are perturbed by oxidative stress, such as
protein folding, apoptosis, and inflammation (85, 86). As
Extracellular Vesicles Released After Exercise
recently discussed (108), the intercellular communication via
and Physiological Perspectives
exosomes might compensate for the imbalance in HSP levels
Other important products secreted by skeletal muscle in almost every cell (155). For example, differentiated neu-
Downloaded by UNIV OF MIAMI MILLER SCH MED Package NERL from www.liebertpub.com at 04/27/20. For personal use only.

during contraction are extracellular vesicles that include rons seem to be dependent on a transfer of HSP from other
exosomes and microvesicles (also known as shedding vesi- cells to maintain their cytoprotection against stress (101). It
cles, ectosome). Exosomes are complex 20–100-nm vesicles has been demonstrated that intercellular HSP transfer occurs
formed by the inward budding of endosomal membranes to by exosomes after exercise (93, 164), raising the idea that this
form large multivesicular bodies. These vesicles are released intercellular communication is critical to ensure an integrated
out of the cell when the multivesicular body fuses with the stress response across different tissues (160a). The exosomes
plasma membrane. The exosome is characterized by soluble secreted after acute exercise have higher levels of HSPs (55,
and membrane-bound proteins, lipids, metabolites, DNA, 168). On the contrary, muscle denervation alters the content
and RNA (mRNA, miRNAs, and other small regulatory of exosomes (38), suggesting a crucial role for contractile
RNAs) that are involved in a protective lipid bilayer that can activity on exosome content and secretion.
modify signaling pathways and protein expression in remote Alzheimer’s is a neurodegenerative disease in which there
tissues (160). is a prevalent protein folding disorder in the brain, and the
Physical exercise considerably increases the metabolic HSP response in some neurons becomes reduced (25). It has
demands of the body. and the vesicle trafficking of metabolic been extensively demonstrated that HSP is involved in pro-
mediators might be a tool through which tissues can share tein homeostasis in the brain, and an accumulation of mis-
resources during this physiological situation. Using quanti- folded proteins (b-amyloid peptides) contributes to the
tative proteomic techniques and intravital imaging experi- progression of this neurodegenerative disease (7). Interest-
ments, Whitham et al. demonstrated that skeletal muscle ingly, an increase in HSP70 expression improves cognition
contraction stimulates extracellular vesicle release and pro- function and reduces amyloid-b peptide levels in Alzhei-
vides the means for tissue crosstalk during exercise (168). mer’s model mice (152). Based on the mechanisms men-
Although the temporal aspects of exosome and small vesicle tioned earlier, it is tempting to suggest that HSP transfer
biogenesis and transport in exercise are unknown, it has been through exosomes, which follows exercise, could retard or
demonstrated that the release of exosomes is associated with even prevent the development of brain diseases; and this
increases in the levels of intracellular calcium (141). Because hypothesis is illustrated in Figure 5.
motor neuron stimulation of skeletal muscle fibers leads to a
rapid release of Ca2+ from the sarcoplasmic reticulum (109),
Conclusions
it is plausible that a transient Ca2+ increase during skeletal
muscle contraction is likely involved in exosome release. The widespread beneficial effects of physical exercise are
ROS seems to be involved in the secretion of extracellular mediated by multiple soluble factors that interfere with a
vesicles since pro-oxidant conditions seem to induce their variety of signaling pathways, and the decoding of these
release (15). In addition, NADPH oxidase and NOS-2 in- pathways is a challenge for basic and translational research.
hibitors reduced the production and release of neutrophil Fascinatingly, optimal ROS-mediated signaling is necessary
microvesicles (158). Further, tumor and senescent cells have to achieve the beneficial effects of exercise. In a common
altered redox balances with elevated ROS levels, which is way, exercise-secreted mediators seem to operate via PGC-
related to the higher number of microvesicles secreted by 1a and Nrf2, two redox sensible pathways that promote
these cells (29, 80). From an exercise perspective, more re- healthier conditions globally.
search is necessary to elucidate the relationship between in- Recently, attention has focused on the mechanisms of
creases in ROS production during skeletal muscle contraction action of new myokines. Many exercise responses and ad-
and microvesicle release (Fig. 1). However, some evidence aptations may be dictated by the (un)balance of ‘‘cytokines
suggests that disrupted ROS-mediated signaling caused by cocktail,’’ defined as a net balance between deleterious
NAD(P)H oxidase inhibition (75), or aging, through exac- hampering molecules and benefit enhancing molecules. Un-
erbated ROS generation (107), can affect the nature of exo- derstanding how aging and obesity blunt some of these ex-
somes (e.g., heat shock proteins [HSPs] expression), ercise adaptations and proposing a multiple and combined
delivering and potentially hampering the widespread bene- therapeutic strategy that will optimize the beneficial effect of
ficial effect of exercise. exercise in many diseases is a critical issue in exercise bi-
HSPs, for example, can be transferred between two cells ology. In addition, the ROS-rich environment that is created
(170) and participate in the maintenance of protein homeo- after exercise is not exclusively restricted to contracting
stasis (155). Previous studies suggested that Hsp70 secretion muscle. Understanding the processes by which the beneficial
10 LOUZADA ET AL.

effects of exercise occurs is, therefore, a prerequisite to de- 8. Assi M, Derbré F, Lefeuvre-Orfila L, Saligaut D, Stock N,
fining how lack of exercise contributes to a poor quality of Ropars M, and Rébillard A. Maintaining a regular phys-
life and high incidence of disease. Therefore, further studies ical activity aggravates intramuscular tumor growth in an
designed to decipher the mechanism of action of myokines orthotopic liposarcoma model. Am J Cancer Res 7: 1037–
are indispensable to understand the redox code that underlies 1053, 2017.
the beneficial effects of exercise and, thus, will expand the 9. Ayala A, Muñoz MF, and Argüelles S. Lipid peroxidation:
therapeutic strategies for many diseases. production, metabolism, and signaling mechanisms of
malondialdehyde and 4-hydroxy-2-nonenal. Oxid Med
Cell Longev 2014: 360438, 2014.
Authors’ Contributions
10. Bacurau AVN, Belmonte MA, Navarro F, Moraes MR,
All authors listed have made a substantial, direct, and in- Pontes FL, Pesquero JL, Araújo RC, and Bacurau RFP.
tellectual contribution to the work, and approved it for pub- Effect of a high-intensity exercise training on the metab-
lication. R.A.L., J.B., and L.P.M. wrote the article; D.P.C., olism and function of macrophages and lymphocytes of
C.D., and J.P.W-.d.-C. revised the article; and R.A.L. and walker 256 tumor-bearing rats. Exp Biol Med 232: 1289–
1299, 2007.
Downloaded by UNIV OF MIAMI MILLER SCH MED Package NERL from www.liebertpub.com at 04/27/20. For personal use only.

R.S.F. designed the outline of the article and wrote the article.
11. Bailey DM, Young IS, McEneny J, Lawrenson L, Kim J,
Barden J, and Richardson RS. Regulation of free radical
Acknowledgment
outflow from an isolated muscle bed in exercising hu-
The authors would like to thank Sapiens scientific illus- mans. Am J Physiol Heart Circ Physiol 287: H1689–
trations for the design of the figures. H1699, 2004.
12. Baker SK, Mccullagh KJA, and Bonen A. Training
intensity-dependent and tissue-specific increases in lactate
Funding Information
uptake and MCT-1 in heart and muscle. J Appl Physiol 84:
The study was supported by research grants from CNPq, 987–994, 1998.
Fundaçao Carlos Chagas Filho de Amparo a Pesquisa do 13. Batirel S, Bozaykut P, Mutlu Altundag E, Kartal Ozer N,
Estado do Rio de Janeiro (FAPERJ), and Coordenação de and Mantzoros CS. The effect of Irisin on antioxidant
Aperfeiçoamento de Pessoal de Nı́vel Superior (CAPES). system in liver. Free Radic Biol Med 75: S16, 2014.
14. Bendall JK, Cave AC, Heymes C, Gall N, and Shah AM.
Pivotal role of a gp91phox-containing NADPH oxidase in
References
angiotensin II-induced cardiac hypertrophy in mice. Cir-
1. Allen D and Westerblad H. Lactic acid—the latest culation 105: 293–296, 2002.
performance-enhancing drug. Science 305: 1112–1113, 15. Benedikter BJ, Weseler AR, Wouters EFM, Savelkoul
2004. PHM, Rohde GGU, and Stassen FRM. Redox-dependent
2. Almeida PWM, Gomes-Filho A, Ferreira AJ, Rodrigues thiol modifications: implications for the release of extra-
CEM, Dias-Peixoto MF, Russo RC, Teixeira MM, Cassali cellular vesicles. Cell Mol Life Sci 75: 2321–2337, 2018.
GD, Ferreira E, Santos IC, Garcia AMC, Silami-Garcia E, 16. Bergman BC, Butterfield GE, Wolfel EE, Lopaschuk GD,
Wisløff U, and Pussieldi GA. Swim training suppresses Casazza GA, Horning MA, and Brooks GA. Muscle net
tumor growth in mice. J Appl Physiol 107: 261–265, 2009. glucose uptake and glucose kinetics after endurance
3. Amin MN, Hussain MS, Sarwar MS, Rahman Moghal training in men. Am J Physiol Endocrinol Metab 277:
MM, Das A, Hossain MZ, Chowdhury JA, Millat MS, and E81–E92, 1999.
Islam MS. How the association between obesity and in- 17. Bjørnsen T, Salvesen S, Berntsen S, Hetlelid KJ, Stea TH,
flammation may lead to insulin resistance and cancer. Lohne-Seiler H, Rohde G, Haraldstad K, Raastad T, Køpp
Diabetes Metab Syndr 13: 1213–1224, 2019. U, Haugeberg G, Mansoor MA, Bastani NE, Blomhoff R,
4. Andrei C, Dazzi C, Lotti L, Torrisi MR, Chimini G, and Stølevik SB, Seynnes OR, and Paulsen G. Vitamin C and
Rubartelli A. The secretory route of the leaderless protein E supplementation blunts increases in total lean body
interleukin 1beta involves exocytosis of endolysosome- mass in elderly men after strength training. Scand J Med
related vesicles. Mol Biol Cell 10: 1463–1475, 1999. Sci Sports 26: 755–763, 2016.
5. Aoi W, Hirano N, Lassiter DG, Björnholm M, Chibalin 18. Black D, Lyman S, Qian T, Lemasters JJ, Rippe RA, Nitta
AV, Sakuma K, Tanimura Y, Mizushima K, Takagi T, T, Kim JS, and Behrns KE. Transforming growth factor
Naito Y, Zierath JR, and Krook A. Secreted protein acidic beta mediates hepatocyte apoptosis through Smad3 gen-
and rich in cysteine (SPARC) improves glucose tolerance eration of reactive oxygen species. Biochimie 89: 1464–
via AMP-activated protein kinase activation. FASEB J 33: 1473, 2007.
10551–10562, 2019. 19. Boström P, Wu J, Jedrychowski MP, Korde A, Ye L, Lo
6. Aoi W, Naito Y, Takagi T, Tanimura Y, Takanami Y, JC, Rasbach KA, Boström EA, Choi JH, Long JZ, Kaji-
Kawai Y, Sakuma K, Hang LP, Mizushima K, Hirai Y, mura S, Zingaretti MC, Vind BF, Tu H, Cinti S, Højlund
Koyama R, Wada S, Higashi A, Kokura S, Ichikawa H, K, Gygi SP, and Spiegelman BM. A PGC1-a-dependent
and Yoshikawa T. A novel myokine, secreted protein myokine that drives brown-fat-like development of white
acidic and rich in cysteine (SPARC), suppresses colon fat and thermogenesis. Nature 481: 463–468, 2012.
tumorigenesis via regular exercise. Gut 62: 882–889, 20. Bourdeau Julien I, Sephton CF, and Dutchak PA. Meta-
2013. bolic networks influencing skeletal muscle fiber compo-
7. Arispe N and De Maio A. Memory loss and the onset of sition. Front Cell Dev Biol 6: 125, 2018.
Alzheimer’s disease could be under the control of extra- 21. Braakhuis AJ, Hopkins WG, and Lowe TE. Effects of
cellular heat shock proteins. J Alzheimers Dis 63: 927– dietary antioxidants on training and performance in fe-
934, 2018. male runners. Eur J Sport Sci 14: 160–168, 2014.
REDOX SIGNALING IN WIDESPREAD EFFECTS OF EXERCISE 11

22. Brady PS, Brady LJ, and Ullrey DE. Selenium, vitamin E 38. De Gasperi R, Hamidi S, Harlow LM, Ksiezak-Reding H,
and the response to swimming stress in the rat. J Nutr 109: Bauman WA, and Cardozo CP. Denervation-related al-
1103–1109, 1979. terations and biological activity of miRNAs contained in
23. Broholm C, Mortensen OH, Nielsen S, Akerstrom T, exosomes released by skeletal muscle fibers. Sci Rep 7:
Zankari A, Dahl B, and Pedersen BK. Exercise induces 12888, 2017.
expression of leukaemia inhibitory factor in human skel- 39. Deldicque L, Cani PD, Delzenne NM, Baar K, and
etal muscle. J Physiol 586: 2195–2201, 2008. Francaux M. Endurance training in mice increases the
24. Brooks GA. The science and translation of lactate shuttle unfolded protein response induced by a high-fat diet. J
theory. Cell Metab 27: 757–785, 2018. Physiol Biochem 69: 215–225, 2013.
25. Calderwood SK and Murshid A. Molecular chaperone 40. Deng X, Huang W, Peng J, Zhu TT, Sun XL, Zhou XY,
accumulation in cancer and decrease in Alzheimer’s dis- Yang H, Xiong JF, He HQ, Xu YH, and He YZ. Irisin
ease: the potential roles of HSF1. Front Neurosci 11: 1–8, alleviates advanced glycation end products-induced in-
2017. flammation and endothelial dysfunction via inhibiting
26. Carey AL, Steinberg GR, Macaulay SL, Thomas WG, ROS-NLRP3 inflammasome signaling. Inflammation 41:
Holmes AG, Ramm G, Prelovsek O, Hohnen-Behrens C, 260–275, 2018.
Downloaded by UNIV OF MIAMI MILLER SCH MED Package NERL from www.liebertpub.com at 04/27/20. For personal use only.

Watt MJ, James DE, Kemp BE, Pedersen BK, and Feb- 41. Di Meo S, Napolitano G, and Venditti P. Mediators of
braio MA. Interleukin-6 increases insulin-stimulated glu- physical activity protection against ROS-linked skeletal
cose disposal in humans and glucose uptake and fatty acid muscle damage. Int J Mol Sci 20: 1–38, 2019.
oxidation in vitro via AMP-activated protein kinase. 42. Dillard CJ, Litov RE, Savin WM, Dumelin EE, and
Diabetes 55: 2688–2697, 2006. Tappel AL. Effects of exercise, vitamin E, and ozone on
27. Catalá A and Dı́az M. Editorial: impact of lipid perox- pulmonary function and lipid peroxidation. J Appl Physiol
idation on the physiology and pathophysiology of cell 45: 927–932, 1978.
membranes. Front Physiol 7: 423, 2016. 43. Divya T, Dineshbabu V, Soumyakrishnan S, Sureshkumar
28. Catoire M, Mensink M, Kalkhoven E, Schrauwen P, and A, and Sudhandiran G. Celastrol enhances Nrf2 mediated
Kersten S. Identification of human exercise-induced antioxidant enzymes and exhibits anti-fibrotic effect
myokines using secretome analysis. Physiol Genomics 46: through regulation of collagen production against
256–267, 2014. bleomycin-induced pulmonary fibrosis. Chem Biol Inter-
29. Chandrasekaran A, Idelchik MDPS, and Melendez JA. act 246: 52–62, 2016.
Redox control of senescence and age-related disease. 44. Done AJ and Traustadóttir T. Nrf2 mediates redox adap-
Redox Biol 11: 91–102, 2017. tations to exercise. Redox Biol 10: 191–199, 2016.
30. Chang S, Linderholm A, Franzi L, Kenyon N, Grasberger 45. Du J, Fan X, Yang B, Chen Y, Liu KX, and Zhou J. Irisin
H, and Harper R. Dual oxidase regulates neutrophil re- pretreatment ameliorates intestinal ischemia/reperfusion
cruitment in allergic airways. Free Radic Biol Med 65: injury in mice through activation of the Nrf2 pathway. Int
38–46, 2013. Immunopharmacol 73: 225–235, 2019.
31. Chapados NA and Lavoie J-M. Exercise training increases 46. E L, Lu J, Selfridge JE, Burns JM, and Swerdlow RH.
hepatic endoplasmic reticulum (er) stress protein expres- Lactate administration reproduces specific brain and liver
sion in MTP-inhibited high-fat fed rats. Cell Biochem exercise-related changes. J Neurochem 127: 91–100,
Funct 28: 202–210, 2010. 2013.
32. Chazaud B. Inflammation during skeletal muscle regen- 47. Egan B, Carson BP, Garcia-Roves PM, Chibalin AV,
eration and tissue remodeling: application to exercise- Sarsfield FM, Barron N, McCaffrey N, Moyna NM,
induced muscle damage management. Immunol Cell Biol Zierath JR, and O’Gorman DJ. Exercise intensity-
94: 140–145, 2016. dependent regulation of peroxisome proliferator-activated
33. Chen RR, Fan XH, Chen G, Zeng GW, Xue YG, Liu XT, receptor c coactivator-1a mRNA abundance is associated
and Wang CY. Irisin attenuates angiotensin II-induced with differential activation of upstream signalling kinases
cardiac fibrosis via Nrf2 mediated inhibition of ROS/ in human skeletal muscle. J Physiol 588: 1779–1790,
TGFb1/Smad2/3 signaling axis. Chem Biol Interact 302: 2010.
11–21, 2019. 48. Emhoff CAW, Messonnier LA, Horning MA, Fattor JA,
34. Close GL, Kayani AC, Ashton T, McArdle A, and Jackson Carlson TJ, and Brooks GA. Gluconeogenesis and hepatic
MJ. Release of superoxide from skeletal muscle of adult glycogenolysis during exercise at the lactate threshold. J
and old mice: an experimental test of the reductive hotspot Appl Physiol 114: 297–306, 2013.
hypothesis. Aging Cell 6: 189–195, 2007. 49. Eschke RCKR, Lampit A, Schenk A, Javelle F, Steindorf
35. Cuevas MJ, Almar M, Garcı́a-Glez JC, Garcı́a-López D, K, Diel P, Bloch W, and Zimmer P. Impact of physical
De Paz JA, Alvear-Ordenes I, and González-Gallego exercise on growth and progression of cancer in rodents-a
J. Changes in oxidative stress markers and NF-kappaB systematic review and meta-analysis. Front Oncol 9: 35,
activation induced by sprint exercise. Free Radic Res 39: 2019.
431–439, 2005. 50. Estébanez B, De Paz JA, Cuevas MJ, and González-
36. Davies KJA, Quintanilha AT, Brooks GA, and Packer Gallego J. Endoplasmic reticulum unfolded protein re-
L. Free radicals and tissue damage produced by exercise. sponse, aging and exercise: an update. Front Physiol 9:
Biochem Biophys Res Commun 107: 1198–1205, 1982. 1744, 2018.
37. Davies KJA, Sevanian A, Muakkassah-Kelly SF, and 51. Fan X, Du J, Wang MH, Li JM, Yang B, Chen Y, Dai JC,
Hochstein P. Uric acid-iron ion complexes. A new aspect Zhang C, and Zhou J. Irisin contributes to the hepato-
of the antioxidant functions of uric acid. Biochem J 235: protection of dexmedetomidine during intestinal ischemia/
747–754, 1986. reperfusion. Oxid Med Cell Longev 2019: 7857082, 2019.
12 LOUZADA ET AL.

52. Febbraio MA, Hiscock N, Sacchetti M, Fischer CP, and 67. Habibovic A, Hristova M, Heppner DE, Danyal K, Ather
Pedersen BK. Interleukin-6 is a novel factor mediating JL, Janssen-Heininger YMW, Irvin CG, Poynter ME,
glucose homeostasis during skeletal muscle contraction. Lundblad LK, Dixon AE, Geiszt M, and van der Vliet
Diabetes 53: 1643–1648, 2004. A. DUOX1 mediates persistent epithelial EGFR activa-
53. Feng H, Kang C, Dickman JR, Koenig R, Awoyinka I, tion, mucous cell metaplasia, and airway remodeling
Zhang Y, and Ji LL. Training-induced mitochondrial ad- during allergic asthma. JCI Insight 1: e88811, 2016.
aptation: role of peroxisome proliferator-activated recep- 68. Handschin C and Spiegelman B. The role of exercise and
tor c coactivator-1a, nuclear factor-jB and b-blockade. PGC1alpha in inflammation and chronic disease. Nature
Exp Physiol 98: 784–795, 2013. 454: 463–469, 2008.
54. Fischer CP, Hiscock NJ, Penkowa M, Basu S, Vessby B, 69. Hashimoto T and Brooks GA. Mitochondrial lactate oxi-
Kallner A, Sjöberg LB, and Pedersen BK. Supplementa- dation complex and an adaptive role for lactate produc-
tion with vitamins C and E inhibits the release of tion. Med Sci Sports Exerc 40: 486–494, 2008.
interleukin-6 from contracting human skeletal muscle. J 70. Hawley JA, Hargreaves M, Joyner MJ, and Zierath JR.
Physiol 558: 633–645, 2004. Integrative biology of exercise. Cell 159: 738–749, 2014.
55. Frühbeis C, Helmig S, Tug S, Simon P, and Krämer- 71. Hellsten Y, Richter EA, Kiens B, and Bangsbo J. AMP
Downloaded by UNIV OF MIAMI MILLER SCH MED Package NERL from www.liebertpub.com at 04/27/20. For personal use only.

Albers E-M. Physical exercise induces rapid release of deamination and purine exchange in human skeletal
small extracellular vesicles into the circulation. J Extra- muscle during and after intense exercise. J Physiol 520:
cell Vesicles 4: 28239, 2015. 909–920, 1999.
56. Fu J, Han Y, Wang J, Liu Y, Zheng S, Zhou L, Jose PA, 72. Hellsten Y, Sjödin B, Richter EA, and Bangsbo J. Urate
and Zeng C. Irisin lowers blood pressure by improvement uptake and lowered ATP levels in human muscle after
of endothelial dysfunction via AMPK-Akt-eNOS-NO high-intensity intermittent exercise. Am J Physiol En-
pathway in the spontaneously hypertensive rat. J Am docrinol Metab 274: E600–E606, 1998.
Heart Assoc 5, 2016. 73. Hellsten Y, Tullson PC, Richter EA, and Bangsbo
57. Furrer R and Handschin C. Optimized engagement of J. Oxidation of urate in human skeletal muscle during
macrophages and satellite cells in the repair and regen- exercise. Free Radic Biol Med 22: 169–174, 1997.
eration of exercised muscle. In: Hormones, Metabolism 74. Henrı́quez-Olguı́n C, Boronat S, Cabello-Verrugio C,
and the Benefits of Exercise, edited by Spiegelman Jaimovich E, Hidalgo E, and Jensen TE. The emerging
B. Chamcham: Springer, 2017. roles of nicotinamide adenine dinucleotide phosphate
58. Gastpar R, Gehrmann M, Bausero MA, Asea A, Gross C, oxidase 2 in skeletal muscle redox signaling and metab-
Schroeder JA, and Multhoff G. Heat shock protein 70 olism. Antioxid Redox Signal 31: 1371–1410, 2019.
surface-positive tumor exosomes stimulate migratory and 75. Henrı́quez-Olguı́n C, Dı́az-Vegas A, Utreras-Mendoza Y,
cytolytic activity of natural killer cells. Cancer Res 65: Campos C, Arias-Calderón M, Llanos P, Contreras-Ferrat
5238–5247, 2005. A, Espinosa A, Altamirano F, Jaimovich E, and Valla-
59. Gertz EW, Wisneski JA, Stanley WC, and Neese RA. dares DM. NOX2 inhibition impairs early muscle gene
Myocardial substrate utilization during exercise in hu- expression induced by a single exercise bout. Front Phy-
mans. Dual carbon-labeled carbohydrate isotope experi- siol 7: 282, 2016.
ments. J Clin Invest 82: 2017–2025, 1988. 76. This reference has been deleted.
60. Gladden LB. Muscle as a consumer of lactate. Med Sci 77. Henrı́quez-Olguin C, Knudsen JR, Raun SH, Li Z, Dal-
Sports Exerc 32: 764–771, 2000. bram E, Treebak JT, Sylow L, Holmdahl R, Richter EA,
61. Glund S, Deshmukh A, Yun CL, Moller T, Koistinen HA, Jaimovich E, and Jensen TE. Cytosolic ROS production
Caidahl K, Zierath JR, and Krook A. Interleukin-6 directly by NADPH oxidase 2 regulates muscle glucose uptake
increases glucose metabolism in resting human skeletal during exercise. Nat Commun 10: 4623, 2019.
muscle. Diabetes 56: 1630–1637, 2007. 78. Hojman P, Dethlefsen C, Brandt C, Hansen J, Pedersen L,
62. Goldstein MS. Humoral nature of the hypoglycemic factor and Pedersen BK. Exercise-induced muscle-derived cy-
of muscular work. Diabetes 10: 232–234, 1961. tokines inhibit mammary cancer cell growth. Am J Physiol
63. Gomes EC, Silva AN, and Oliveira MRD. Oxidants, an- Endocrinol Metab 301: 504–510, 2011.
tioxidants, and the beneficial roles of exercise-induced 79. Huang H, Liu H, Liu C, Fan L, Zhang X, Gao A, Hu X,
production of reactive species. Oxid Med Cell Longev Zhang K, Cao X, Jiang K, Zhou Y, Hou J, Nan F, and Li
2012: 756132, 2012. J. Disruption of the unfolded protein response (UPR) by
64. Gomez-Cabrera MC, Domenech E, Romagnoli M, Ardu- lead compound selectively suppresses cancer cell growth.
ini A, Borras C, Pallardo FV, Sastre J, and Viña J. Oral Cancer Lett 360: 257–268, 2015.
administration of vitamin C decreases muscle mitochon- 80. Huber V, Fais S, Iero M, Lugini L, Canese P, Squarcina P,
drial biogenesis and hampers training-induced adaptations Zaccheddu A, Colone M, Arancia G, Gentile M, Seregni
in endurance performance. Am J Clin Nutr 87: 142–149, E, Valenti R, Ballabio G, Belli F, Leo E, Parmiani G, and
2008. Rivoltini L. Human colorectal cancer cells induce T-cell
65. Groussard C, Morel I, Chevanne M, Monnier M, Cillard J, death through release of proapoptotic microvesicles: role
and Delamarche A. Free radical scavenging and antioxi- in immune escape. Gastroenterology 128: 1796–1804,
dant effects of lactate ion: an in vitro study. J Appl Physiol 2005.
89: 169–175, 2000. 81. Hulmi JJ, Hentilä J, DeRuisseau KC, Oliveira BM, Pa-
66. Groussard C, Rannou-Bekono F, Machefer G, Chevanne paioannou KG, Autio R, Kujala UM, Ritvos O, Kainu-
M, Vincent S, Sergent O, Cillard J, and Gratas- lainen H, Korkmaz A, and Atalay M. Effects of muscular
Delamarche A. Changes in blood lipid peroxidation dystrophy, exercise and blocking activin receptor IIB li-
markers and antioxidants after a single sprint anaerobic gands on the unfolded protein response and oxidative
exercise. Eur J Appl Physiol 89: 14–20, 2003. stress. Free Radic Biol Med 99: 308–322, 2016.
REDOX SIGNALING IN WIDESPREAD EFFECTS OF EXERCISE 13

82. Iatsenko I, Boquete JP, and Lemaitre B. Microbiota- 97. Li J, Ichikawa T, Villacorta L, Janicki JS, Brower GL,
derived lactate activates production of reactive oxygen Yamamoto M, and Cui T. Nrf2 protects against mala-
species by the intestinal NADPH oxidase Nox and short- daptive cardiac responses to hemodynamic stress. Arter-
ens Drosophila lifespan. Immunity 49: 929–942.e5, 2018. ioscler Thromb Vasc Biol 29: 1843–1850, 2009.
83. Irwin ML, Smith AW, McTiernan A, Ballard-Barbash R, 98. Li RL, Wu SS, Wu Y, Wang XX, Chen HY, juan Xin J, Li
Cronin K, Gilliland FD, Baumgartner RN, Baumgartner H, Lan J, Xue KY, Li X, Zhuo CL, Cai YY, He JH, Zhang
KB, and Bernstein L. Influence of pre- and postdiagnosis HY, Tang CS, Wang W, and Jiang W. Irisin alleviates
physical activity on mortality in breast cancer survivors: pressure overload-induced cardiac hypertrophy by induc-
the health, eating, activity, and lifestyle study. J Clin ing protective autophagy via mTOR-independent activa-
Oncol 26: 3958–3964, 2008. tion of the AMPK-ULK1 pathway. J Mol Cell Cardiol
84. Jackson MJ. Control of reactive oxygen species produc- 121: 242–255, 2018.
tion in contracting skeletal muscle. Antioxid Redox Signal 99. Li X, Han D, Tian Z, Gao B, Fan M, Li C, Li X, Wang Y,
15: 2477–2486, 2011. Ma S, and Cao F. Activation of cannabinoid receptor type
85. Kalmar B and Greensmith L. Activation of the heat shock II by AM1241 ameliorates myocardial fibrosis via Nrf2-
response in a primary cellular model of motoneuron mediated inhibition of TGF-b1/Smad3 pathway in myo-
Downloaded by UNIV OF MIAMI MILLER SCH MED Package NERL from www.liebertpub.com at 04/27/20. For personal use only.

neurodegeneration-evidence for neuroprotective and neu- cardial infarction mice. Cell Physiol Biochem 39: 1521–
rotoxic effects. Cell Mol Biol Lett 14: 319–335, 2009. 1536, 2016.
86. Kalmar B and Greensmith L. Induction of heat shock 100. MacNeil LG, Baker SK, Stevic I, and Tarnopolsky MA.
proteins for protection against oxidative stress. Adv Drug 17b-estradiol attenuates exercise-induced neutrophil in-
Deliv Rev 61: 310–318, 2009. filtration in men. Am J Physiol Regul Integr Comp Physiol
87. Kang C, O’Moore KM, Dickman JR, and Ji LL. Exercise 300: R1443–R1451, 2011.
activation of muscle peroxisome proliferator-activated 101. Maguire G. Amyotrophic lateral sclerosis as a protein
receptor-c coactivator-1a signaling is redox sensitive. level, non-genomic disease: therapy with S2RM exosome
Free Radic Biol Med 47: 1394–1400, 2009. released molecules. World J Stem Cells 9: 187–202, 2017.
88. Kelly M, Keller C, Avilucea PR, Keller P, Luo Z, Xiang 102. Mahoney DJ, Safdar A, Parise G, Melov S, Fu M, Mac-
X, Giralt M, Hidalgo J, Saha AK, Pedersen BK, and Neil L, Kaczor J, Payne ET, and Tarnopolsky MA. Gene
Ruderman NB. AMPK activity is diminished in tissues of expression profiling in human skeletal muscle during re-
IL-6 knockout mice: the effect of exercise. Biochem covery from eccentric exercise. Am J Physiol Regul Integr
Biophys Res Commun 320: 449–454, 2004. Comp Physiol 294: R1901–R1910, 2008.
89. Kim Y, Park M, Boghossian S, and York DA. Three 103. Mambula SS and Calderwood SK. Heat induced release of
weeks voluntary running wheel exercise increases endo- Hsp70 from prostate carcinoma cells involves both active
plasmic reticulum stress in the brain of mice. Brain Res secretion and passive release from necrotic cells. Int J
1317: 13–23, 2010. Hyperthermia 22: 575–585, 2006.
90. Kitaoka Y, Takeda K, Tamura Y, and Hatta H. Lactate 104. Mambula SS and Calderwood SK. Heat shock protein 70
administration increases mRNA expression of PGC-1a is secreted from tumor cells by a nonclassical pathway
and UCP3 in mouse skeletal muscle. Appl Physiol Nutr involving lysosomal endosomes. J Immunol 1950 177:
Metab 41: 695–698, 2016. 7849–7857, 2006.
91. Kong G, Jiang Y, Sun X, Cao Z, Zhang G, Zhao Z, Zhao 105. Mazur-Bialy AI. Irisin acts as a regulator of macrophages
Y, Yu Q, and Cheng G. Irisin reverses the IL-6 induced host defense. Life Sci 176: 21–25, 2017.
epithelial-mesenchymal transition in osteosarcoma cell 106. Mazur-Bialy AI, Kozlowska K, Pochec E, Bilski J, and
migration and invasion through the STAT3/Snail signal- Brzozowski T. Myokine irisin-induced protection against
ing pathway. Oncol Rep 38: 2647–2656, 2017. oxidative stress in vitro. Involvement of heme oxygenase-
92. Kristensen M, Albertsen J, Rentsch M, and Juel C. Lactate 1 and antioxidazing enzymes superoxide dismutase-2 and
and force production in skeletal muscle. J Physiol 562: glutathione peroxidase. J Physiol Pharmacol 69: 117–125,
521–526, 2005. 2018.
93. Lancaster GI and Febbraio MA. Mechanisms of stress- 107. McArdle A and Jackson MJ. The role of attenuated redox
induced cellular HSP72 release: implications for exercise- and heat shock protein responses in the age-related decline
induced increases in extracellular HSP72. Exerc Immunol in skeletal muscle mass and function. Essays Biochem 61:
Rev 11: 46–52, 2005. 339–348, 2017.
94. Le Moal E, Pialoux V, Juban G, Groussard C, Zouhal H, 108. McArdle A, Pollock N, Staunton CA, and Jackson MJ.
Chazaud B, and Mounier R. Redox control of skeletal Aberrant redox signalling and stress response in age-
muscle regeneration. Antioxid Redox Signal 27: 276–310, related muscle decline: role in inter- and intra-cellular
2017. signalling. Free Radic Biol Med 132: 50–57, 2019.
95. Levonen AL, Landar A, Ramachandran A, Ceaser EK, 109. Melzer W, Rios E, and Schneider MF. Time course of
Dickinson DA, Zanoni G, Morrow JD, and Darley-Usmar calcium release and removal in skeletal muscle fibers.
VM. Cellular mechanisms of redox cell signalling: role of Biophys J 45: 637–641, 1984.
cysteine modification in controlling antioxidant defences 110. Memme JM, Oliveira AN, and Hood DA. Chronology of
in response to electrophilic lipid oxidation products. UPR activation in skeletal muscle adaptations to chronic
Biochem J 378: 373–382, 2004. contractile activity. Am J Physiol Cell Physiol 310:
96. Li F, Liu BB, Cai M, Li JJ, and Lou S-J. Excessive en- C1024–C1036, 2016.
doplasmic reticulum stress and decreased neuroplasticity- 111. Merry TL and Ristow M. Nuclear factor erythroid-derived
associated proteins in prefrontal cortex of obese rats and 2-like 2 (NFE2L2, Nrf2) mediates exercise-induced mi-
the regulatory effects of aerobic exercise. Brain Res Bull tochondrial biogenesis and the anti-oxidant response in
140: 52–59, 2018. mice. J Physiol 594: 5195–5207, 2016.
14 LOUZADA ET AL.

112. Meyer C, Dostou JM, Welle SL, and Gerich JE. Role of mediators and metabolic implications. Am J Physiol En-
human liver, kidney, and skeletal muscle in postprandial docrinol Metab 304: E453–E463, 2013.
glucose homeostasis. Am J Physiol Endocrinol Metab 129. Powers SK and Jackson MJ. Exercise-induced oxidative
282: E419–E427, 2002. stress: cellular mechanisms and impact on muscle force
113. Molenaar RJ and van Noorden CJ. Type 2 diabetes and production. Physiol Rev 88: 1243–1276, 2008.
cancer as redox diseases? Lancet Lond Engl 384: 853, 130. Powers SK, Radak Z, and Ji LL. Exercise-induced oxi-
2014. dative stress: past, present and future. J Physiol 594:
114. Morales-Alamo D and Calbet JAL. Free radicals and 5081–5092, 2016.
sprint exercise in humans. Free Radic Res 48: 30–42, 131. Przybyla B, Gurley C, Harvey JF, Bearden E, Kortebein P,
2014. Evans WJ, Sullivan DH, Peterson CA, and Dennis RA.
115. Nalbandian M, Radak Z, and Takeda M. N-acetyl-L- Aging alters macrophage properties in human skeletal
cysteine prevents lactate-mediated PGC1-alpha expres- muscle both at rest and in response to acute resistance
sion in C2C12 myotubes. Biology 8: 44, 2019. exercise. Exp Gerontol 41: 320–327, 2006.
116. Nardai G, Stadler K, Papp E, Korcsmáros T, Jakus J, and 132. Quistorff B, Secher NH, and Van Lieshout JJ. Lactate
Csermely P. Diabetic changes in the redox status of the fuels the human brain during exercise. FASEB J 22: 3443–
Downloaded by UNIV OF MIAMI MILLER SCH MED Package NERL from www.liebertpub.com at 04/27/20. For personal use only.

microsomal protein folding machinery. Biochem Biophys 3449, 2008.


Res Commun 334: 787–795, 2005. 133. Ristow M, Zarse K, Oberbach A, Klöting N, Birringer M,
117. Nielsen OB, De Paoli F, and Overgaard K. Protective Kiehntopf M, Stumvoll M, Kahn CR, and Blüher
effects of lactic acid on force production in rat skeletal M. Antioxidants prevent health-promoting effects of
muslce. J Physiol 536: 161–166, 2001. physical exercise in humans. Proc Natl Acad Sci U S A
118. Niethammer P, Grabher C, Look AT, and Mitchison TJ. 106: 8665–8670, 2009.
A tissue-scale gradient of hydrogen peroxide mediates 134. Rogers CJ, Colbert LH, Greiner JW, Perkins SN, and
rapid wound detection in zebrafish. Nature 459: 996– Hursting SD. Physical activity and cancer prevention.
999, 2009. Sports Med 38: 271–296, 2008.
119. Nilsson MI, Bourgeois JM, Nederveen JP, Leite MR, 135. Rojo de la Vega M and Zhang DD. NRF2 induction for
Hettinga BP, Bujak AL, May L, Lin E, Crozier M, Ru- NASH treatment: a New Hope rises. Cell Mol Gastro-
siecki DR, Moffatt C, Azzopardi P, Young J, Yang Y, enterol Hepatol 5: 422–423, 2018.
Nguyen J, Adler E, Lan L, and Tarnopolsky MA. Lifelong 136. Rusch HP and Kline BE. The effect of exercise on the
aerobic exercise protects against inflammaging and can- growth of a mouse tumor. Cancer Res 4: 116–118, 1944.
cer. PLoS One 14: e0210863, 2019. 137. Saclier M, Cuvellier S, Magnan M, Mounier R, and
120. This reference has been deleted. Chazaud B. Monocyte/macrophage interactions with
121. Ostrowski K, Rohde T, Zacho M, Asp S, and Pedersen myogenic precursor cells during skeletal muscle regener-
BK. Evidence that interleukin-6 is produced in human ation. FEBS J 280: 4118–4130, 2013.
skeletal muscle during prolonged running. J Physiol 508: 138. Sakellariou GK, Vasilaki A, Palomero J, Kayani A, Zibrik
949–953, 1998. L, McArdle A, and Jackson MJ. Studies of mitochondrial
122. Pala R, Orhan C, Tuzcu M, Sahin N, Ali S, Cinar V, and nonmitochondrial sources implicate nicotinamide
Atalay M, and Sahin K. Coenzyme Q10 supplementation adenine dinucleotide phosphate oxidase(s) in the increased
modulates NFjB and Nrf2 pathways in exercise training. skeletal muscle superoxide generation that occurs during
J Sports Sci Med 15: 196–203, 2016. contractile activity. Antioxid Redox Signal 18: 603–621,
123. Pedersen BK, Akerström TCA, Nielsen AR, and Fischer 2013.
CP. Role of myokines in exercise and metabolism. J Appl 139. Sandiford SD, Kennedy KA, Xie X, Pickering JG, and Li
Physiol (1985) 103: 1093–1098, 2007. SS. Dual oxidase maturation factor 1 (DUOXA1) over-
124. Pedersen BK and Febbraio MA. Muscle as an endocrine expression increases reactive oxygen species production
organ: focus on muscle-derived interleukin-6. Physiol Rev and inhibits murine muscle satellite cell differentiation.
88: 1379–1406, 2008. Cell Commun Signal 12: 1–15, 2014.
125. Pedersen L, Idorn M, Olofsson GH, Lauenborg B, Noo- 140. San-Millán I and Brooks GA. Reexamining cancer me-
kaew I, Hansen RH, Johannesen HH, Becker JC, Pedersen tabolism: lactate production for carcinogenesis could be
KS, Dethlefsen C, Nielsen J, Gehl J, Pedersen BK, Thor the purpose and explanation of the Warburg effect. Car-
Straten P, and Hojman P. Voluntary running suppresses cinogenesis 38: 119–133, 2017.
tumor growth through epinephrine- and IL-6-dependent 141. Savina A, Furlán M, Vidal M, and Colombo MI. Exosome
NK cell mobilization and redistribution. Cell Metab 23: release is regulated by a calcium-dependent mechanism in
554–562, 2016. K562 cells. J Biol Chem 278: 20083–20090, 2003.
126. Pellerin L, Pellegri G, Bittar PG, Charnay Y, Bouras C, 142. Scheele C, Nielsen S, and Pedersen BK. ROS and myo-
Martin JL, Stella N, and Magistretti PJ. Evidence sup- kines promote muscle adaptation to exercise. Trends En-
porting the existence of an activity-dependent astrocyte- docrinol Metab 20: 95–99, 2009.
neuron lactate shuttle. Dev Neurosci 20: 291–299, 1998. 143. Scheiman J, Luber JM, Chavkin TA, MacDonald T, Tung
127. Peng J, Deng X, Huang W, Yu J-H, Wang J-X, Wang J-P, A, Pham LD, Wibowo MC, Wurth RC, Punthambaker S,
Yang S-B, Liu X, Wang L, Zhang Y, Zhou X-Y, Yang H, Tierney BT, Yang Z, Hattab MW, Avila-Pacheco J, Clish
He Y-Z, and Xu F-Y. Irisin protects against neuronal in- CB, Lessard S, Church GM, and Kostic AD. Meta-omics
jury induced by oxygen-glucose deprivation in part de- analysis of elite athletes identifies a performance-
pends on the inhibition of ROS-NLRP3 inflammatory enhancing microbe that functions via lactate metabolism.
signaling pathway. Mol Immunol 91: 185–194, 2017. Nat Med 25: 1104–1109, 2019.
128. Pillon NJ, Bilan PJ, Fink LN, and Klip A. Cross-talk be- 144. Serrano AL, Baeza-Raja B, Perdiguero E, Jardı́ M, and
tween skeletal muscle and immune cells: muscle-derived Muñoz-Cánoves P. Interleukin-6 is an essential regulator
REDOX SIGNALING IN WIDESPREAD EFFECTS OF EXERCISE 15

of satellite cell-mediated skeletal muscle hypertrophy. 158. Thom SR, Bhopale VM, and Yang M. Neutrophils gen-
Cell Metab 7: 33–44, 2008. erate microparticles during exposure to inert gases due to
145. Shanmugam G, Challa AK, Devarajan A, Athmanathan B, cytoskeletal oxidative stress. J Biol Chem 289: 18831–
Litovsky SH, Krishnamurthy P, Davidson CJ, and Raja- 18845, 2014.
sekaran NS. Exercise mediated Nrf2 signaling protects the 159. Um HS, Kang EB, Leem YH, Cho IH, Yang CH, Chae
myocardium from isoproterenol-induced pathological re- KR, Hwang DY, and Cho JY. Exercise training acts as a
modeling. Front Cardiovasc Med 6: 68, 2019. therapeutic strategy for reduction of the pathogenic phe-
146. Shin KO, Bae JY, Woo J, Jang KS, Kim KS, Park JS, Kim notypes for Alzheimer’s disease in an NSE/APPsw-
IK, and Kang S. The effect of exercise on expression of transgenic model. Int J Mol Med 22: 529–539, 2008.
myokine and angiogenesis mRNA in skeletal muscle of 160. Valadi H, Ekström K, Bossios A, Sjöstrand M, Lee JJ, and
high fat diet induced obese rat. J Exerc Nutr Biochem 6: Lötvall JO. Exosome-mediated transfer of mRNAs and
91–98, 2015. microRNAs is a novel mechanism of genetic exchange
147. Sobotta MC, Liou W, ouml Cker SS, Talwar D, Oehler M, between cells. Nat Cell Biol 9: 654–659, 2007.
Ruppert T, Scharf AND, and Dick TP. Peroxiredoxin-2 160a. van Oosten-Hawle P and Morimoto RI. Transcellular
and STAT3 form a redox relay for H2O2 signaling. Nat chaperone signaling: an organismal strategy for inte-
Downloaded by UNIV OF MIAMI MILLER SCH MED Package NERL from www.liebertpub.com at 04/27/20. For personal use only.

Chem Biol 11: 64–70, 2014. grated cell stress responses. J Exp Biol 217: 129–136,
148. Son JS, Kim JH, Kim HJ, Yoon DH, Kim JS, Song HS, 2014.
and Song W. Effect of resistance ladder training on 161. Vassilakopoulos T, Karatza MH, Katsaounou P, Kollintza
SPARC expression in skeletal muscle of hindlimb im- A, Zakynthinos S, and Roussos C. Antioxidants attenuate
mobilized rats. Muscle Nerve 53: 951–957, 2016. the plasma cytokine response to exercise in humans. J
149. Songsorn P, Ruffino J, and Vollaard NBJ. No effect of Appl Physiol 94: 1025–1032, 2003.
acute and chronic supramaximal exercise on circulating 162. Wafi AM, Yu L, Gao L, and Zucker IH. Exercise training
levels of the myokine SPARC. Eur J Sport Sci 17: 447– upregulates Nrf2 protein in the rostral ventrolateral me-
452, 2017. dulla of mice with heart failure. J Appl Physiol (1985)
150. Starkie RL, Rolland J, Angus DJ, Anderson MJ, and 127: 1349–1359, 2019.
Febbraio MA. Circulating monocytes are not the source of 163. Wajner SM, Goemann IM, Bueno AL, Larsen PR, and
elevations in plasma IL-6 and TNF-alpha levels after Maia AL. IL-6 promotes nonthyroidal illness syndrome by
prolonged running. Am J Physiol Cell Physiol 280: C769– blocking thyroxine activation while promoting thyroid
C774, 2001. hormone inactivation in human cells. J Clin Invest 121:
151. Steensberg A, Keller C, Hillig T, Frøsig C, Wojtaszewski 1834–1845, 2011.
JFP, Pedersen BK, Pilegaard H, and Sander M. Nitric 164. Walsh RC, Koukoulas I, Garnham A, Moseley PL, Har-
oxide production is a proximal signaling event controlling greaves M, and Febbraio MA. Exercise increases serum
exercise-induced mRNA expression in human skeletal Hsp72 in humans. Cell Stress Chaperones 6: 386–393,
muscle. FASEB J 21: 2683–2694, 2007. 2001.
152. Sun Y, Zhang J-R, and Chen S. Suppression of Alzhei- 165. Waring WS, Convery A, Mishra V, Shenkin A, Webb DJ,
mer’s disease-related phenotypes by the heat shock pro- and Maxwell SRJ. Uric acid reduces exercise-induced
tein 70 inducer, geranylgeranylacetone, in APP/PS1 oxidative stress in healthy adults. Clin Sci 105: 425–430,
transgenic mice via the ERK/p38 MAPK signaling path- 2003.
way. Exp Ther Med 14: 5267–5274, 2017. 166. Watson JD. Type 2 diabetes as a redox disease. Lancet
153. Sylow L, Kleinert M, Richter EA, and Jensen TE. 383: 841–843, 2014.
Exercise-stimulated glucose uptake—regulation and im- 167. Weigert C, Düfer M, Simon P, Debre E, Runge H,
plications for glycaemic control. Nat Rev Endocrinol 13: Brodbeck K, Häring HU, and Schleicher ED. Upregula-
133–148, 2017. tion of IL-6 mRNA by IL-6 in skeletal muscle cells: role
154. Takahashi H, Alves CRR, Stanford KI, Middelbeek RJW, of IL-6 mRNA stabilization and Ca2+-dependent mecha-
Pasquale Nigro null, Ryan RE, Xue R, Sakaguchi M, nisms. Am J Physiol Cell Physiol 293: C1139–C1147,
Lynes MD, So K, Mul JD, Lee M-Y, Balan E, Pan H, 2007.
Dreyfuss JM, Hirshman MF, Azhar M, Hannukainen JC, 168. Whitham M, Parker BL, Friedrichsen M, Hingst JR,
Nuutila P, Kalliokoski KK, Nielsen S, Pedersen BK, Kahn Hjorth M, Hughes WE, Egan CL, Cron L, Watt KI, Ku-
CR, Tseng Y-H, and Goodyear LJ. TGF-b2 is an exercise- chel RP, Jayasooriah N, Estevez E, Petzold T, Suter CM,
induced adipokine that regulates glucose and fatty acid Gregorevic P, Kiens B, Richter EA, James DE, Wojtas-
metabolism. Nat Metab 1: 291–303, 2019. zewski JFP, and Febbraio MA. Extracellular vesicles
155. Takeuchi T, Suzuki M, Fujikake N, Popiel HA, Kikuchi provide a means for tissue crosstalk during exercise. Cell
H, Futaki S, Wada K, and Nagai Y. Intercellular chaper- Metab 27: 237–251.e4, 2018.
one transmission via exosomes contributes to maintenance 169. Wu J, Ruas JL, Estall JL, Rasbach KA, Choi JH, Ye L,
of protein homeostasis at the organismal level. Proc Natl Boström P, Tyra HM, Crawford RW, Campbell KP,
Acad Sci U S A 112: E2497–E2506, 2015. Rutkowski DT, Kaufman RJ, and Spiegelman BM. The
156. Tanimura Y, Aoi W, Takanami Y, Kawai Y, Mizushima unfolded protein response mediates adaptation to exercise
K, Naito Y, and Yoshikawa T. Acute exercise increases in skeletal muscle through a PGC-1a/ATF6a complex.
fibroblast growth factor 21 in metabolic organs and cir- Cell Metab 13: 160–169, 2011.
culation. Physiol Rep 4: e12828, 2016. 170. Zhan R, Leng X, Liu X, Wang X, Gong J, Yan L, Wang L,
157. Thirupathi A and de Souza CT. Multi-regulatory network Wang Y, Wang X, and Qian L-J. Heat shock protein 70 is
of ROS: the interconnection of ROS, PGC-1 alpha, and secreted from endothelial cells by a non-classical pathway
AMPK-SIRT1 during exercise. J Physiol Biochem 73: involving exosomes. Biochem Biophys Res Commun 387:
487–494, 2017. 229–233, 2009.
16 LOUZADA ET AL.

171. Zhang M, Xu Y, and Jiang L. Irisin attenuates oxidized AMP ¼ adenosine monophosphate
low-density lipoprotein impaired angiogenesis through AMPK ¼ AMP-activated protein kinase
AKT/mTOR/S6K1/Nrf2 pathway. J Cell Physiol 234: ATP ¼ adenosine triphosphate
18951–18962, 2019. BiP ¼ binding immunoglobulin protein
172. Zhu D, Wang H, Zhang J, Zhang X, Xin C, Zhang F, Lee CAMK ¼ Ca2+/calmodulin-dependent protein kinases
Y, Zhang L, Lian K, Yan W, Ma X, Liu Y, and Tao DUOX ¼ dual oxidase
L. Irisin improves endothelial function in type 2 diabetes ER ¼ endoplasmic reticulum
through reducing oxidative/nitrative stresses. J Mol Cell ERS ¼ endoplasmic reticulum stress
Cardiol 87: 138–147, 2015. FGF21 ¼ fibroblast growth factor 21
GLUT4 ¼ glucose transporter 4
Address correspondence to: H2O2 ¼ hydrogen peroxide
Dr. Ruy A. Louzada HSPs ¼ heat shock proteins
Institut of Biophysics Carlos Chagas Filho IL ¼ interleukin
Federal University of Rio de Janeiro LIF ¼ leukemia inhibitory factor
Downloaded by UNIV OF MIAMI MILLER SCH MED Package NERL from www.liebertpub.com at 04/27/20. For personal use only.

LDH ¼ lactate dehydrogenase


Rio de Janeiro 21941-590
L-NAME ¼ N(G)-nitro-l-arginine methyl ester
Brazil
MAPK ¼ mitogen-activated protein kinase
MCP1 ¼ monocyte chemoattractant protein 1
E-mail: andraderuy@hotmail.com MCT ¼ monocarboxylate transporter
NADH ¼ nicotinamide adenine dinucleotides
Date of first submission to ARS Central, November 11, 2019; NOX ¼ NADPH oxidase
date of final revised submission, February 29, 2020; date of Nrf2 ¼ nuclear factor erythroid 2-related factor 2
acceptance, March 9, 2020. O2 ¼ oxygen
O2- ¼ superoxide
OH ¼ hydroxyl
PDK4 ¼ pyruvate dehydrogenase kinase 4
Abbreviations Used PGC-1a ¼ peroxisome proliferator-activated receptor
13-HODE ¼ 13-hydroxyoctadecadienoic acid gamma coactivator 1-alpha
4-HNE ¼ 4-hydroxynonenal ROS ¼ reactive oxygen species
AK ¼ adenylate kinase SPARC ¼ secreted protein acidic and rich
AKT/mTOR ¼ protein kinase B/mechanistic target in cysteine
of rapamycin TGF-b ¼ transforming growth factor beta

View publication stats

You might also like