You are on page 1of 26

P1: OTA/XYZ P2: ABC

JWBT335-c120033 JWBT335/Comprehensive Physiology June 8, 2013 8:4 Printer Name: Yet to Come

Muscle as a Secretory Organ


Bente K. Pedersen*1

ABSTRACT
Skeletal muscle is the largest organ in the body. Skeletal muscles are primarily characterized
by their mechanical activity required for posture, movement, and breathing, which depends on
muscle fiber contractions. However, skeletal muscle is not just a component in our locomotor
system. Recent evidence has identified skeletal muscle as a secretory organ. We have suggested
that cytokines and other peptides that are produced, expressed, and released by muscle fibers
and exert either autocrine, paracrine, or endocrine effects should be classified as “myokines.”
The muscle secretome consists of several hundred secreted peptides. This finding provides a
conceptual basis and a whole new paradigm for understanding how muscles communicate
with other organs such as adipose tissue, liver, pancreas, bones, and brain. In addition, several
myokines exert their effects within the muscle itself. Many proteins produced by skeletal muscle are
dependent upon contraction. Therefore, it is likely that myokines may contribute in the mediation
of the health benefits of exercise. 
C 2013 American Physiological Society. Compr Physiol 3:1337-

1362, 2013.

Introduction physical activity. Such factors might directly or indirectly in-


fluence the function of other organs such as the adipose tissue,
Skeletal muscle can be identified as an organ that produces and the liver, the cardiovascular system, and the brain.
releases cytokines and other peptides, which we have named In our view, the pluralis form “exercise factors” would be
“myokines” (170). Given that skeletal muscle is the largest or- more applicable given the fact that multiple metabolic and
gan in the human body, the discovery that contracting skeletal physiologic changes are induced by exercise. This view has
muscle secretes proteins sets a novel paradigm: skeletal mus- been substantiated by the identification of skeletal muscle as
cle is a secretory organ producing and releasing myokines in a secretory organ, which produces several secreted factors.
response to contraction, which can influence metabolism and It has been obvious that several effects of exercise might be
function of both muscle tissue and other tissues and organs, explained by the release of one or more humoral factors from
Figure 1. the muscle that either directly or indirectly could influence
metabolism and function in other organs.
An example is the fact that the dramatically increased glu-
History—Myokines in the Context of cose uptake by contracting skeletal muscle corresponds to an
Exercise Physiology increased glucose production by the liver, whereby glucose
homeostasis is maintained. It is not fully understood how con-
For nearly half a century, researchers hypothesized that skele- tracting muscles modulate metabolism in the liver, but it is ob-
tal muscle cells possessed a “humoral” factor that was released vious that a muscle-derived humoral factor could be involved.
in response to increased glucose demand during contraction Another example is given when, in response to exercise, adi-
(69). Due to lack of more precise knowledge, the unidentified pose tissue increases the release of free fatty acids (FFAs) into
contraction-induced factor was named “the work stimulus,” the circulation. Again, it is not known how contracting skeletal
“the work factor,” or the “exercise factor” (178). muscle and adipose tissue communicate. Yet another example
The early view on the exercise factor concept was pred- is found in the notion that many individuals claim a stronger
icated on the fact that contracting skeletal muscle mediates feeling of pleasantness after exercise. This may be ascribed
metabolic and physiologic responses in other organs, which
are not mediated via the nervous system.
* Correspondence to bkp@rh.dk
This view has been supported by the fact that electrical
1 The Centre of Inflammation and Metabolism at Department of
stimulation of paralyzed muscles in spinal cord injured pa-
Infectious Diseases, and Copenhagen Muscle Research Centre,
tients with no afferent or efferent impulses induces many of The Faculty of Health Sciences, University of Copenhagen,
the same physiological changes as in intact human beings Copenhagen, Denmark
(110, 136). Therefore, it was clear that contracting skeletal Published online, July 2013 (comprehensivephysiology.com)
muscles were able to communicate to other organs via hu- DOI: 10.1002/cphy.c120033
moral factors, which are released into the circulation during Copyright 
C American Physiological Society

Volume 3, July 2013 1337


P1: OTA/XYZ P2: ABC
JWBT335-c120033 JWBT335/Comprehensive Physiology June 8, 2013 8:4 Printer Name: Yet to Come

Muscle as a Secretory Organ Comprehensive Physiology

Browning
Lipolysis

Irisin Follistatin
LIF Hepatic glucose
IL-6 IL-4 production
IL-6
during exercise
IL-7
Unknown
IL-15 Myostatin Hepatic CXCL-1
IL-6 exercise
IL-6 production
stimulus
BDNF Hypertrophy
IL-6 Lipolysis IL-6
AMPK
Glucose
uptake
Fat oxidation
Angiogenesis

IGF-1 FGF-21 FSTL-1


FGF-2 IL-8?
IL-6 CXCL-1?
Promotes endothellal
function and
revascularization
IL-6 increases
insulin secretion via
GLP-1

Figure 1 Skeletal muscle is a secretory organ. Leukemia inhibitory factor (LIF), interleukin (IL)-
4, IL-6, IL-7, and IL-15 promote muscle hypertrophy. Myostatin inhibits muscle hypertrophy and
exercise provokes the release of a myostatin inhibitor, follistatin, from the liver. Brain-derived
neurotropic factor (BDNF) and IL-6 are involved in AMP-activated protein kinase (AMPK)-mediated
fat oxidation and IL-6 enhances insulin-stimulated glucose uptake. IL-6 appears to have systemic
effects on the liver and adipose tissue and increases insulin secretion via upregulation of GLP-1.
Insulin-like growth factor-1 (IGF-1) and FGF-2 are involved in bone formation, and follistatin-
related protein 1 improves endothelial function and revascularization of ischemic vessels. Irisin
has a role in “browning” of white adipose tissue. Adapted, with permission, from (172).

Myokines

Myokines Proinflammatory
adipokines
Type 2 diabetes mellitus,
cardiovascular disease, cancer,
osteoporosis

Figure 2 The interplay between adipokines and myokines represents a Yin-Yang balance.
Especially under conditions of obesity, adipose tissue secretes adipokines that contribute to
establish a chronic inflammatory environment, promoting pathological processes such as
atherosclerosis and insulin resistance. Skeletal muscles are capable of producing myokines
that confer some of the health benefits of exercise. Such myokines might counteract the
harmful effects of proinflammatory adipokines. Adapted, with permission, from (172).

1338 Volume 3, July 2013


P1: OTA/XYZ P2: ABC
JWBT335-c120033 JWBT335/Comprehensive Physiology June 8, 2013 8:4 Printer Name: Yet to Come

Comprehensive Physiology Muscle as a Secretory Organ

Pro inflammatory Anti inflammatory 1000

IL-6

Plasma IL-6 (fold change from rest)


TNF 100
IL-1ra
TNF-R IL-10

10

Sepsis

Anti inflammatory
1
Knee-extensor Bicycling Running Eccentric
IL-6
Figure 4 Different modes of exercise and the corresponding in-
crease in plasma interleukin-6 (IL-6) levels. Graph is based on 73
exercise trials and represents ∼800 subjects. Each dot indicates one
exercise trial; the corresponding bars represent geometric means with
IL-1ra 95% confidence intervals. Although different modes of exercise are as-
IL-10
sociated with different levels of muscle damage, the increase in plasma
IL-6 levels postexercise is a consistent finding. Modified, with permis-
sion, from (58, 173).

itary function is abnormal, circulating thymic peptide levels


Excercise
undergo a premature decline (186). These pioneering stud-
ies led to the hypothesis that normal development of the
immune system is dependent on factors produced by the
Figure 3 Comparison of sepsis-induced verses exercise-induced in-
creases in circulating cytokines. During sepsis, there is a marked and hypothalamic-pituitary axis. It later appeared that a number of
rapid increase in circulating tumor necrosis factor-alpha (TNF-α), which pituitary hormones, for example, prolactin, growth hormone,
is followed by an increase in interleukin-6 (IL-6). In contrast, during ex- and adrenocorticotropin, may serve as immunomodulatory
ercise, the marked increase in IL-6 is not preceded by elevated TNF-α.
Adapted, with permission, from (175). factors (63, 99, 175). Initially, cytokines (glycoproteins with
molecular masses of 15,000-30,000) (45) were known for
their immunoregulatory roles. However, other studies demon-
to a handful of neurotrophic factors and hormones, includ- strated that cytokines were involved in complex networks of
ing brain-derived neurotropic factor (BDNF), betaendorphins, communication between the neuroendocrine and the immune
serotonin, dopamine, and noradrenaline (124). However, it system. It appeared that cytokines were involved in modulat-
is not known how contracting muscles communicate to the ing the secretion from the hypopituitary-hypothalamus axis
brain and facilitate increased production, action, or signal- and an important neuroendocrine-immune loop was identified
ing of these neurotransmitters. These examples all illustrate (40, 221) (175).
effects of exercise, which may be mediated by one or more During the past 20 to 30 years, it has become evident that
secreted products from the muscle. In the early days, it was exercise induces considerable changes in the immune sys-
anticipated that the exercise factor would be a simple metabo- tem, especially with regard to the innate immune system, also
lite or ion released from the active muscle like K+ ion, lactic known as nonspecific immune system and first line of defense.
acid, adenosine, etc. However, to date, metabolites have not Studies of the interactions between exercise and the immune
been identified to mediate muscle-organ cross-talk (7). system have provided unique opportunities to evaluate the
In the search for soluble factors that could mediate the role of the underlying endocrine and cytokine mechanisms
communication between muscle and other organs, we found (175, 176).
it obvious to focus on candidates, previously known from the As with many paradigm-shifting studies, our initial find-
immune system. Research that dates back to 1930 demon- ings that led to the identification of interleukin-6 (IL-6) as a
strated that if the pituitary gland was removed in rats, the myokine were somewhat serendipitous. In 1988, we had ini-
thymus would undergo atrophy (175, 220). In 1976, Pelletier tiated systematic studies on the effects of exercise on the im-
et al. showed that in the dwarf mouse, in which the pitu- mune system (183). This research soon opened a new research

Volume 3, July 2013 1339


P1: OTA/XYZ P2: ABC
JWBT335-c120033 JWBT335/Comprehensive Physiology June 8, 2013 8:4 Printer Name: Yet to Come

Muscle as a Secretory Organ Comprehensive Physiology

field called “exercise immunology,” leading in 1993 to the binding protein-4 have been added to the growing list of
foundation of the International Society of Exercise and Im- adipokines [for review, see (213)]. Notwithstanding the role
munology. It was while looking for a mechanistic explana- of adiponectin (218), most of the factors that are produced by
tion to understand exercise-induced changes in the distribu- adipocytes are, however, considered to be proinflammatory,
tion and concentrations of lymphocyte subpopulations that we for example, tumor necrosis factor-alpha (TNF-α), mono-
and others decided to focus on cytokines and their possible cyte chemoattractant protein-1, and plasminogen activator
roles as a link between muscle contractions and cellular im- inhibitor type 1, and potentially harmful with regard to the
mune changes (176). This research soon led to the discovery development of obesity-induced metabolic and cardiovascu-
that exercise provokes an increase in a number of cytokines lar diseases (CVDs) (172). To neutralize the effect of the
(52, 53, 163, 174, 178, 179, 182). proinflammatory adipokines, it is obvious that another organ
In year 2000, we were able to demonstrate that contract- or tissue might offer protection and contribute to produce
ing human skeletal muscle releases significant amounts of anti-inflammatory components that could provide a counter-
IL-6 into the circulation during prolonged exercise (232). balance to the proinflammatory factors that are produced by
An accompanying editorial to this publication suggested adipocytes. Given that exercise offers multiple health bene-
that muscle-derived IL-6 could have metabolic roles (66), fits, it was reasonable to suggest that skeletal muscle might
a fact soon supported by experimental studies (106, 228). secrete proteins that could counteract the harmful effects of
In a historical context, the identification of muscle as a the proinflammatory adipokines secreted by adipose tissue in
cytokine-producing endocrine organ may be regarded as the obese state (Fig. 6).
a breakthrough. The finding that IL-6 was produced by The word “myokine” is derived from the Greek words
contracting muscles and released into the blood (232) soon for “muscle” and “motion” and in 2003, we suggested this
led to the discovery that muscle-derived cytokines may play term should be used as a classification for cytokines or other
a role in mediating some of the exercise-associated metabolic peptides that are produced, expressed, and released by muscle
changes, as well as the metabolic changes following training fibers and exert endocrine effects (178).
adaptation. While the word adipokine refers to factors secreted from
In continuation, we suggested that cytokines or other pep- adipose tissue, the term “myokine” refers to a protein that
tides that are produced, expressed, and released by muscle is secreted from myocytes. Characterization of a number of
fibers and exert either paracrine or endocrine effects should myokines reveal that skeletal muscles are capable of produc-
be classified as “myokines” (170). Thus, although the idea ing and releasing proteins that can both communicate with
of an “exercise factor” can be traced back many years, the cells locally within the muscles (autocrine/paracrine) or to
identification of skeletal muscle as a myokine-producing or- other distant tissues (endocrine).
gan opens for a whole new field of research. During the past This review provides an update on some of the muscle-
decade, myocytes have been identified as cells with a high derived cytokines that have been identified so far.
secretory capacity in parallel with the concept of adipocytes
being major endocrine cells. It appears that muscle cells, here
defined as myoblasts or myocytes, have the capacity to pro- Myokines
duce several hundred secreted factors (20, 82, 254).
The fact that skeletal muscle was identified as a cytokine- Characteristics of a myokine
producing organ led to studies, showing that muscle-derived
r Myokines are cytokines or other peptides that are produced,
cytokines played a role in mediating not only some of the expressed, and released by muscle fibers.
exercise-induced immune changes, but also the exercise- r Myokines may exert autocrine, paracrine, or endocrine
associated metabolic changes (172).
effects.
r Myokines may balance and counteract the effects of
Myokines and Adipokines in a adipokines.
Yin-Yang Concept r The muscle-cell secretome consists of several hundred se-
Adipose tissue was initially considered an inert storage com- creted products.
partment for triglycerides, not the least due to pioneering
work from the Spiegelman and Flier (38) laboratories in the r Identified myokines include myostatin, leukemia inhibitory
mid-1980s, who demonstrated that adipocytes are capable of factor (LIF), IL-6, IL-7, BDNF, insulin-like growth factors
releasing a specific secretory protein, called adipsin or com- (IGF)-1, fibroblast growth factor 2 (FGF-2), FSTL-1, and
plement factor D. Friedman and colleagues later identified irisin.
leptin as a fat cell-specific secretory factor, deficient in the
ob/ob mouse and responsible for mediating a hormonal sig-
r Myokines may mediate protective effects of muscular ex-
nal between fat and the brain (258). Since then, adiponectin, ercise, with regard to diseases associated with a physically
resistin, acylation-stimulating protein, visfatin, and retinol- inactive lifestyle.

1340 Volume 3, July 2013


P1: OTA/XYZ P2: ABC
JWBT335-c120033 JWBT335/Comprehensive Physiology June 8, 2013 8:4 Printer Name: Yet to Come

Comprehensive Physiology Muscle as a Secretory Organ

Myostatin centration of IL-6 increases during muscular exercise (58).


In 1997, Se-Jin Lee’s laboratory identified myostatin, which This increase is followed by the appearance of IL-1ra and the
appears to be the first discovery of a secreted muscle factor that anti-inflammatory cytokine IL-10. In general, the cytokine
fulfills the criteria for a myokine. Myostatin is produced by response to exercise and sepsis differs with regard to TNF-α.
skeletal muscle and secreted into the circulation. It is a highly Thus, the cytokine response to exercise is not preceded by an
conserved member of the TGF-β superfamily and myostatin increase in plasma-TNF-α.
knockout results in extensive skeletal muscle hypertrophy in Following exercise, the basal plasma IL-6 concentration
mice (130), cattle, and humans (202). may increase up to 100-fold, but less dramatic increases are
Myostatin has effects on muscle growth and is also more frequent (58, 173).
involved in the modulation of adipose tissue function and The exercise-induced increase of plasma IL-6 occurs in
mass (2, 55, 74, 119, 131, 259). Both aerobic exercise and an exponential manner (59, 157, 232) and the peak IL-6 level
resistance training in humans and animals attenuate myostatin is reached at the end of the exercise or shortly thereafter
expression and myostatin inactivation seems to potentiate the (59, 157, 162). It is the combination of mode, intensity, and
beneficial effects of endurance exercise on metabolism (3). duration of the exercise that determines the magnitude of the
Myostatin secretion is enhanced from myotubes derived from exercise-induced increase of plasma IL-6 (58). IL-6 has pre-
myoblasts isolated from muscle biopsies of obese compared viously been classified as a proinflammatory cytokine. There-
with nonobese women (88). In addition, patients with type 2 fore, it was first thought that the exercise-induced IL-6 re-
diabetes have higher levels of muscle myostatin mRNA sponse was related to muscle damage (29). However, it has
content than matched control subjects and muscle myostatin become evident that eccentric exercise is not associated with a
mRNA is correlated with indices of impaired glucose larger increase in plasma IL-6 than exercise involving concen-
metabolism and poor fitness (23). tric “nondamaging” muscle contractions. This finding clearly
Follistatin represents another member of the TGF-β su- demonstrates that muscle damage is not required to provoke
perfamily. Follistatin is a naturally occurring inhibitor of myo- an increase in plasma IL-6 during exercise. As a matter of fact,
statin with regard to its regulatory role in skeletal muscle. Its eccentric exercise may result in a delayed peak and a much
presence in plasma is increased with acute exercise. Interest- slower decrease of plasma IL-6 during recovery (81,125,249).
ingly, experimental studies show a marked increase in fol- The amount of IL-6 produced is correlated to the amount
listatin levels in the liver in response to exercise, suggesting of muscle mass engaged in the exercise. Muscles of the upper
the existence of a possible muscle-liver cross-talk during and extremities may be insufficient to increase plasma IL-6 above
following exercise (78). preexercise level (18, 86, 154). In contrast, running—which
involves several large muscle groups—is the mode of exercise
during which the most dramatic plasma IL-6 increases have
Conclusion been observed (Fig. 4).
Myostatin has effects on muscle growth and its expression is In a review by C. Fischer (100), he shows that exercise
regulated by both aerobic exercise and resistance. High levels duration is the single most important factor determining the
of myostatin inhibit muscle growth and vice versa. Myostatin postexercise plasma IL-6 amplitude and that 50% of the vari-
is negatively regulated by follistatin, which appears to be ation in plasma IL-6 following exercise can be explained by
released from the liver during acute exercise. exercise duration alone (P < 10−12 ) (58).
Based on the log-log linear relationship between time and
fold increase of plasma IL-6, a tenfold increase of plasma IL-
Interleukin-6 6 requires exercise for 1.9 h (CI 1.6-2.9 h, P < 0.0001), while
While myostatin was the first muscle-derived peptide to fulfill a 100-fold increase of plasma IL-6 requires exercise lasting
the criteria for a myokine, the gp130 receptor cytokine IL-6 6.0 h (CI 4.5-8.1 h, P < 0.0001) (Fig. 5).
was the first myokine that was found to be secreted into the IL-6 has been shown to be synthesized and released from
blood stream in response to muscle contractions (175). contracting muscles alone and not from resting muscles ex-
posed to the same hormonal changes (100, 232), demonstrat-
ing that circulating systemic factors cannot explain why con-
Exercise and systemic levels of IL-6
tracting muscles synthesize and release IL-6. Of note, the
Aerobic exercise provokes a systemic cytokine response, in- increase of IL-6 in the circulation occurs during exercise with-
cluding, for example, IL-6, IL-1 receptor antagonist (IL-1ra), out any sign of muscle damage (58). Until the beginning of
and IL-10. For reviews, see (8, 32, 49, 67, 68, 143, 162, 164, this millennium, it was commonly thought that the increase
171, 175, 181, 184, 240, 245, 255) (Fig. 3). in IL-6 during exercise was a consequence of an immune re-
IL-6 was serendipitously discovered as a myokine be- sponse due to local damage in the working muscles (151) and
cause of the observation that it increased in an exponential it was hypothesized that macrophages were responsible for
fashion proportional to the length of exercise and the amount this increase (140). However, an early study (239) demon-
of muscle mass engaged in the exercise [for review, see (58)]. strated that IL-6 mRNA in monocytes did not increase as a
It has been consistently demonstrated that the plasma con- result of exercise. Further work confirmed this finding at the

Volume 3, July 2013 1341


P1: OTA/XYZ P2: ABC
JWBT335-c120033 JWBT335/Comprehensive Physiology June 8, 2013 8:4 Printer Name: Yet to Come

Muscle as a Secretory Organ Comprehensive Physiology

Linear regression
1000 Plasma IL-6 (fold change) = antilog10[1.030 · log10[duration (h)] + 0.695]
n = 74 exercise trials (~800 subjects)

Plasma IL-6 (fold increase from rest)


2
R = 0.51
-12
P < 10

100
Knee-extensor
Bicycling
10 Running
Eccentric
Regression

0.1
0.1 1 10 100
Exercise duration (h)

Figure 5 The overall log10 -log10 linear relation (straight solid line) between
exercise duration and increase in plasma interleukin-6 (IL-6) (fold change from
preexercise level) indicates that 51% of the variation in fold plasma IL-6 increase
may be explained by the duration of exercise. Modified, with permission, from
(58).

protein level (225, 227). In addition, the liver clears, rather adaptation attenuate the exercise-sensitive increase in IL-6
than secretes, IL-6 during exercise (51). plasma concentration (60, 169).
The finding that the nuclear transcription rate for IL-6 and It has been shown that skeletal muscle cells are capable
the IL-6mRNA levels were rapidly and markedly increased of producing IL-6 in response to various stimuli such as incu-
after the onset of exercise (106, 231) suggested that a factor bation with lipopolysaccharide, reactive oxygen species, and
associated with contraction was responsible for the increase inflammatory cytokines. In response to the latter stimuli, the
in IL-6 transcriptional rate within the nuclei from myocytes. upstream signaling events that lead to the induction of IL-6
Febbraio’s group found further evidence that contracting mus- have been well categorized. As mentioned above, human
cle fibers themselves were a source of IL-6 mRNA and pro- skeletal muscle is unique as it can produce IL-6 in response to
tein. Such data were achieved by analysis of biopsies from contraction in the absence of inflammation (52) and notewor-
the human vastus lateralis using in situ hybridization and im- thy in a TNF-independent fashion (104). Thereby, muscle-
munohistochemistry techniques (87). derived IL-6 is being linked to metabolism rather than in-
The microdialysis technique indicated that the concentra- flammation. Muscle-glycogen level is a determining factor as
tion of IL-6 within the contracting skeletal muscle could be 5- both intramuscular IL-6 mRNA expression (106) and protein
to 100-fold higher than the levels found in the circulation and release (228) are exacerbated when intramuscular glycogen
that IL-6 appears to accumulate within the contracting muscle is compromised, suggesting that IL-6 is somehow regulated
fibers as well as in the interstitium during exercise (203). by glycogen content.
Measurement of arteriovenous IL-6 concentrations and Several studies have shown that glucose ingestion dur-
blood flow across the leg has demonstrated that IL-6 is re- ing exercise attenuates the exercise-induced increase in
leased in relatively high quantities into the circulation from plasma-IL-6 (83, 114, 116-118, 140, 146-148, 151), but not
the exercising leg (232). IL-6 mRNA expression within the contracting muscle itself
A number of studies have confirmed that IL-6 is indeed (54, 140, 151, 226). Contraction may lead to IL-6 gene tran-
produced by muscle cells. Thus, IL-6 has been shown to be scription via calcium (Ca2+ ) being released from the sar-
expressed by human myoblasts (15,41) and by human cultured coplasmic reticulum to activate IL-6 through activation of
myotubes (104). In addition, IL-6 is produced by growing nuclear factor of activated T (NFAT) cells (93). In human
murine myofibers and associated muscle stem cells (satellite skeletal muscle cell cultures, IL-6 mRNA increases time and
cells) (217) and released from human primary muscle cell dose dependently with ionomycin stimulation, an effect that
cultures from healthy individuals (72, 79) and from patients is blunted by the presence of the calcineurin-inhibitor Cy-
with type 2 diabetes (72, 211). closporin A. In contrast, TNF-α gene expression is decreased
in response to ionomycin treatment, demonstrating that IL-
6 and TNF-α are regulated differentially in skeletal muscle
IL-6 is an energy sensor cells in response to a Ca2+ stimulus (104).
Muscle-derived IL-6 works as an exercise sensor (89, 169, Regular exercise leads to a number of physiological, adap-
179, 206). Thus, enhanced glucose availability and training tive responses. These include increased levels of basal skeletal

1342 Volume 3, July 2013


P1: OTA/XYZ P2: ABC
JWBT335-c120033 JWBT335/Comprehensive Physiology June 8, 2013 8:4 Printer Name: Yet to Come

Comprehensive Physiology Muscle as a Secretory Organ

Muscular expression of IL-6R

IL-6

Concentration muscle glycogen

Concentration IL-6
IL-6
Gly
cog
en

Gl
yc
og
en IL-6
G
ly
co
ge
n

Exercise Exercise Exercise

Time

Figure 6 The figure presents a model on how interleukin-6 (IL-6) is regulated


in response to training adaptation. Regular exercise leads to an enhancement
of glycogen synthase and a trained muscle will consequently store more mus-
cle glycogen. During acute exercise, the untrained muscle is highly dependent
on glycogen as substrate, whereas training leads to an enhancement of beta-
oxidating enzymes and an enhanced capability to oxidize fat and hence to use
fat as substrate during exercise. This means that the trained muscle uses less
glycogen during work. The activation of muscle-IL-6 is glycogen dependent.
At conditions with low muscle glycogen, the transcription rate of IL-6 is faster
and relatively more IL-6 is produced at the same relative work compared to
conditions with high muscle glycogen. Thus, the acute plasma IL-6 response is
lower in a trained versus an untrained subject. The mechanisms whereby basal
plasma-IL-6 is decreased by training and whereby the muscular expression of
IL-6 receptors (IL-6R) is enhanced are not fully understood. However, it appears
that a trained muscle may be more sensitive to IL-6. Adapted, with permission,
from (175).

muscle glycogen content, enhanced activity of key enzymes compared to untrained conditions (105), suggesting that the
involved in the β-oxidation, increased sensitivity of adipose sensitivity to IL-6 is increased (Fig. 6).
tissue to epinephrine-stimulated lipolysis, and increased ca- We recently reported that insulin-resistant individuals
pacity to oxidize fat. Consequently, the trained skeletal muscle demonstrated IL-6 resistance (211). Accordingly, we suspect
is less dependent on plasma glucose and muscle glycogen as that muscle disuse may lead to IL-6 resistance. The elevated
substrates during exercise (175, 190). circulating levels of IL-6 that accompany obesity and physi-
Several observational studies have reported a negative as- cal inactivity may represent a compensatory mechanism. This
sociation between the amount of regular physical activity and would be in line with the well-known facts that insulin resis-
the resting plasma IL-6 levels: the more physically active, the tance is accompanied by hyperinsulinemia and that chronic
lower basal plasma IL-6 (58). On the other hand, high plasma high circulating levels of leptin may reflect leptin resistance.
levels of IL-6 are closely associated with physical inactivity
and the metabolic syndrome. Intervention studies show that
IL-6—a role in glucose and lipid metabolism
basal levels of IL-6 are reduced after training (58). It also
appears that the exercise-induced increase in plasma IL-6 and IL-6 has been shown to enhance glucose production both in
muscular IL-6 mRNA is less pronounced in trained versus un- vitro and in vivo (172) and cytokine signaling through AMP-
trained individuals (60). However, while plasma-IL-6 appears activated protein kinase (AMPK) appears to play a major role
to be down regulated by training, the muscular expression of (234). Several studies have shown that IL-6 enhances glu-
the IL-6 receptor (IL-6R) is unregulated. Moreover, the basal cose uptake and increases intramyocellular (28, 33, 189) or
IL-6R mRNA content in trained skeletal muscle is increased whole body (241) fatty acid oxidation via an effect on AMPK

Volume 3, July 2013 1343


P1: OTA/XYZ P2: ABC
JWBT335-c120033 JWBT335/Comprehensive Physiology June 8, 2013 8:4 Printer Name: Yet to Come

Muscle as a Secretory Organ Comprehensive Physiology

(33, 101). It appears that IL-6 activates AMPK in skeletal study, Ellingsgaard et al. demonstrated the existence of an
muscle by increasing the concentration of cAMP, and sec- adipose tissue and skeletal muscle enteroendocrine-islet axis.
ondarily, the AMP:ATP ratio (107). IL-6 mediates signaling This cross-talk between insulin-sensitive tissues and insulin-
through the gp130 receptor and exhibits many “leptin-like” producing cells was shown to be mediated through IL-6 acting
actions (133, 233, 234, 246). In healthy skeletal muscle, and on L cells and alpha cells to promote Glucagon-like peptide-
not least in humans, the IL-6-induced activation of AMPK 1 (GLP-1) secretion and production, thereby allowing for an
appears to override the IL-6-induced activation of suppressor adaptation to increased insulin demand during obesity and im-
of cytokine signaling (SOCS)-3. Accordingly, IL-6 knockout proved beta cell function in response to physical training (48).
mice develop mature onset obesity and glucose intolerance
(243), supporting the notion that IL-6 may exert beneficial
Conclusion
effects on metabolism. IL-6 increases insulin-stimulated glu-
cose uptake in vitro in healthy individuals, while infusion IL-6 is the myokine prototype (Fig. 7). The systemic level
of recombinant human (rh) IL-6 into healthy humans during of IL-6 increases markedly with exercise and skeletal muscle
a hyperinsulinemic, euglycemic clamp increases the glucose is the main source of origin. Muscle contractions lead to the
infusion rate without affecting the total suppression of en- production and release into the circulation of IL-6, which
dogenous glucose production (EGP) (33). appears to have numerous biological effects, including effects
IL-6 can be classified as a myokine with an endocrinolog- on glucose and fat metabolism. In addition, IL-6 has a role
ical activity since it contributes to hepatic glucose production in myogenesis and mediates anti-inflammatory effects, see
during exercise (50). However, it is not clear how the tightly below.
controlled production and clearance of glucose during mus-
cular work are regulated. It is possible that an unidentified
“work factor” exists that influences the contraction-induced BDNF
increase in EGP. We infused rhIL-6 at physiological concen- Neurotrophins are a family of structurally related growth
trations into resting human subjects. Acute administration of factors, including BDNF, which exert many of their effects
rhIL-6 had no effect on whole-body glucose disposal, glu- on neurons primarily through Trk receptor tyrosine kinases.
cose uptake or EGP (123, 189, 230). In contrast, we found Among these, BDNF and its receptor TrkB are the ones most
that IL-6 contributed to the contraction-induced increase in widely and abundantly expressed in the brain (97). However,
EGP. Healthy men performed 2 h of bicycle exercise on three several studies verify that skeletal muscle is also capable of
separate occasions: (i) at a relatively high intensity (HI) and expressing BDNF (165, 166, 177).
(ii) at a low intensity with (LO + IL-6) or without (LO) an Studies in rodents demonstrate that both exercise and elec-
infusion of rhIL-6 that mimicked the circulating concentra- trical stimulation (and contraction) of skeletal muscle lead to
tion of IL-6 observed during HI exercise. This study revealed an induction of BNDF expression in muscle (39,70,128,216).
a direct muscle-liver “cross-talk” (50). Several studies have also reported that exercise induces
Infusion of rhIL-6 into healthy humans caused an increase an expression of BDNF in skeletal muscle. For example, Co-
in lipolysis in the absence of hypertriglyceridaemia or changes pray et al. (39) found that intense contraction of the soleus
in catecholamines, glucagon, insulin, or any adverse effects in muscle in both normal and diabetic rats caused an increase
healthy individuals (123, 189, 241). These findings combined in the expression of BDNF. In addition, ultrastructural stud-
with cell culture experiments showed that IL-6 had direct ies from these same authors found that BDNF expression was
effects on both lipolysis and fat oxidation and identify IL-6 localized within muscle fibers and activated satellite cells. Im-
as a lipolytic factor (189). In a recent study, we were able portantly, no expression of BDNF was observed in Schwann
to distinguish between lipolysis in muscle and adipose tissue cells or fibroblasts, suggesting that the localization of BDNF
and found that IL-6 primarily stimulated lipolysis in skeletal was defined within the muscle fibers.
muscle, whereas abdominal adipose tissue was unaffected In other studies, Gomez-Pinilla et al. (70) found that
(251). BDNF mRNA and protein levels in rodents increased in the
Ellingsgaard et al. found that the pancreatic alpha cell is a soleus muscle after 3 and 7 days of exercise. Interestingly, fol-
primary target of IL-6 action (47) and that IL-6 promotes al- lowing paralysis of the soleus muscle, BDNF mRNA levels
pha cell proliferation and inhibits apoptosis. They showed that were reduced, demonstrating that active muscle contraction
in response to a high-fat diet, alpha cell mass expands in an is important in modulating BDNF levels in muscle. In addi-
IL-6-dependent manner and that whole-body IL-6 knockout tion, BDNF appears to play a role in the development and
mice with no alpha cell expansion showed increased glycemia differentiation of myoblasts and muscle fibers (134, 138).
after feeding caused by impaired insulin secretion. These find- It is well known that BDNF increases in brain tissue in re-
ings show that alpha cell expansion in response to a high-fat sponse to acute exercise and exercise training and may account
diet may be required for functional beta cell compensation for the effect of exercise in the protection against neurodegen-
and that systemically increased IL-6 abundance induced by erative diseases such as dementia (128). We studied whether
a high-fat diet is an adaptive response necessary to maintain human skeletal muscle would produce BDNF in response
proper insulin secretion and glucose homeostasis. In another to exercise (128) and found that BDNF mRNA and protein

1344 Volume 3, July 2013


P1: OTA/XYZ P2: ABC
JWBT335-c120033 JWBT335/Comprehensive Physiology June 8, 2013 8:4 Printer Name: Yet to Come

Comprehensive Physiology Muscle as a Secretory Organ

IL-6

IL-6Rα/gp130Rβ

Pl3-K p-STAT3

p-Akt p-AMPK Blood vessel

Glucose uptake Fat oxidation IL-6


Liver

IL-6
Increased hepatic glucose
Contraction production during exercise

IL-6 IL-6

IL-6 IL-6
IL-6
IL-6
Adipose tissue

IL-6

Increased lipolysis

Figure 7 Biological role of contraction-induced interleukin-6 (IL-6). Skeletal muscle expresses and releases myokines
into the circulation. In response to muscle contractions, both type I and type II muscle fibers express the myokine IL-
6, which subsequently exerts its effects both locally within the muscle (e.g., through activation of AMPK) and—when
released into the circulation—peripherally in several organs in a hormone-like fashion. Specifically in skeletal muscle,
IL-6 acts in an autocrine or paracrine manner to signal through a gp130Rβ/IL-6Rα homodimer resulting in activation
of AMP-kinase and/or PI3-kinase to increase glucose uptake and fat oxidation. IL-6 is also known to increase hepatic
glucose production during exercise or lipolysis in adipose tissue. Modified, with permission, from (173).

expression were modestly increased in human skeletal muscle tal muscle. In addition, muscle-derived BDNF plays a role
after exercise. However, muscle-derived BDNF appeared not in muscle repair, regeneration, and differentiation. Thus, in
to be released into the circulation. BDNF mRNA and protein addition to its well-known role in neurobiology, BDNF can
expression were clearly increased in muscle cells that were be identified as a myokine that plays a role in peripheral
electrically stimulated. Interestingly, BDNF increased phos- metabolism, myogenesis, and muscle regeneration.
phorylation of AMPK and Acetyl-CoA carboxylase (ACC)
and enhanced fat oxidation both in vitro and ex vivo. Thus,
we were able to identify BDNF as a novel contraction- Interleukin-7
induced muscle cell-derived protein that may increase fat Interleukin-7 (IL-7) is a cytokine that is required for T and
oxidation in skeletal muscle in an AMPK-dependent fash- B cell development, whereas possible biological functions of
ion (165, 166, 177). Other studies consistently demonstrate IL-7 in nonimmune cells have not been explored. Recently,
that muscle-derived BDNF and other neurotrophins serve as Haugen et al. identified IL-7 as a myokine (79).
important regulators of the maintenance, function, and regen- IL-7 mRNA and IL-7 protein were detected in conditioned
eration of skeletal muscle fibers. Thus, BDNF is an injury- media from primary cultures of human myotubes as well
related factor that is involved in the survival and function of as inside the myotubes. The amount of IL-7 in the medium
innervating motorneurons [reviewed in (210)]. increased with incubation time (79).
Incubation with recombinant IL-7 during differentia-
tion induced a reduction in mRNA for the terminal myo-
Conclusion
genic markers myosin heavy chain (MHC) 2 and myogenin
Taken together, BDNF is a protein produced in skeletal mus- (MYOG). This finding suggests that IL-7 may act on satellite
cle cells, which is increased by contraction to enhance fat cells and may be involved in myogenesis. The authors also
oxidation in an AMPK-dependent fashion, most probably by demonstrated that the muscular expression of IL-7 mRNA
acting in an autocrine and/or paracrine manner within skele- was increased several fold in resting musculus vastus lateralis

Volume 3, July 2013 1345


P1: OTA/XYZ P2: ABC
JWBT335-c120033 JWBT335/Comprehensive Physiology June 8, 2013 8:4 Printer Name: Yet to Come

Muscle as a Secretory Organ Comprehensive Physiology

and musculus trapezius biopsies taken from male individuals Conclusion


undergoing a strength training program.
IL-8 and CXCL-1 increase in contracting skeletal muscle and
IL-8 is released from human muscle cells in vitro. The phys-
Conclusion iological function of IL-8 within the muscle is, however, still
IL-7 has recently been identified as a myokine involved in unknown. The main part of the systemic increase in IL-8 as
myogenesis. Its expression in resting skeletal muscle is in- seen during exercise with an eccentric component is most
creased with training adaptation. likely due to an inflammatory response. However, muscle-
derived IL-8 is likely to occur locally and exert its effect in
an autocrine or paracrine fashion. A more likely function of
Interleukin-8 muscle-derived IL-8 is to stimulate angiogenesis in skele-
tal muscle. IL-8 associates with the CXC receptor 1 and 2
Interleukin-8 (IL-8) belongs to the CXC family of
(CXCR1 and CXCR2) and IL-8 has been proven to play a
chemokines. The CXC nomenclature relates to the presence
direct role in endochelial cell survival, proliferation, and an-
of two conserved cysteine residues at the amino terminus sep-
giogenesis (108, 115).
arated by one amino acid. IL-8 belongs to a subdivision of
CXC-chemokines, which has an amino acid sequence Glu-
Leu-Arg (ELR) preceding the first conserved cysteine amino Interleukin-15
acid residue in the primary structure of these proteins (10). IL-15 is a 14 kDa cytokine. It belongs to the IL-2 superfamily
Murine CXCL-1 shares the highest sequence homology with and was originally isolated on the basis of its ability to
human CXCL-1, but it is often mentioned as the functional support natural killer T-lymphocyte proliferation. IL-15 is
homolog to human IL-8 (205). expressed at the mRNA level in a variety of nonlymphoid
CXCL-1 and IL-8 possess neutrophil chemoattractant ac- tissues, with particularly high expressions in skeletal muscle
tivity. In addition, they are involved in the processes of an- and placenta. IL-15 is also expressed abundantly in cardiac
giogenesis (122). The ability of IL-8 to induce angiogenesis muscle, lung, liver, kidney, brain, and pancreas (71). The
is distinct from its ability to induce inflammation (103). IL- regulatory role of muscle contraction with regard to IL-15 re-
8 associates with the CXC receptors 1 and 2 (CXCR1 and mains unclear. Nieman et al. found that muscle IL-15mRNA
CXCR2) (17). CXCR2 is expressed by human microvascular levels were unchanged immediately after a 3-h run (148),
endothelial cells and is the receptor responsible for IL-8- and Ostrowski et al. found that plasma IL-15 (measured up
induced angiogenesis (1). to 6 h into recovery) did not change in response to 2.5 h of
The production of different chemokines in the ELR+ CXC treadmill running (157). Skeletal muscle IL-15 mRNA levels,
family has been shown to be induced by IL-6 (237). There- measured immediately after a 2-h weight training bout, did
fore, we studied the role of exercise and IL-6 in the regulation not differ from baseline (146), whereas in one study plasma
of murine CXCL-1 (185). Following a single bout of exercise, IL-15 protein increased immediately after acute resistance
CXCL-1 increased in serum, muscle, and liver. The exercise- exercise (201). We demonstrated that IL-15 mRNA levels
induced regulation of liver CXCL-1 mRNA expression was are unregulated in human skeletal muscle following a bout
completely blunted in IL-6 knockout mice. When IL-6 was of strength training (142).
overexpressed in murine muscles, we found a marked in- IL-15 has been identified as an anabolic factor that is
crease in serum CXCL-1 and liver CXCL-1 mRNA expres- highly expressed in skeletal muscle (71). Furthermore, IL-15
sion. These data demonstrate a robust muscle-liver cross-talk has been suggested to play a role in muscle-adipose tissue
during exercise in which exercise-induced IL-6 production interaction (6). In human skeletal myogenic cultures, IL-15
stimulates the liver to produce CXCL-1. The study found a induces an increase in the accumulation of the protein MHC
higher expression of CXCL-1 in liver compared to muscle. in differentiated muscle cells. This suggests that IL-15 is an
However, it has clearly been demonstrated that muscular IL-8 anabolic factor in muscle growth (61). Moreover, IL-15 stim-
mRNA levels are enhanced by exercise (147) and that IL-8 is ulates myogenic differentiation independently of IGFs (198)
released by human primary cultured myotubes (79). and in contrast to IGF-1, IL-15 has effects on fully differen-
The plasma concentration of IL-8 increases in response to tiated myoblasts (196). The ability of IL-15 to antagonize the
exhaustive exercise such as running, which involves eccentric enhanced muscle protein breakdown as demonstrated in an in
muscle contractions (148-150, 158, 236). In addition, a slight vivo cancer cachexia model points to the potential therapeutic
increase in IL-8 plasma concentration has been reported dur- effect of IL-15 (31, 57).
ing intense cycle ergometry (139). Interestingly, while IL-15 has been reliably demonstrated
The possibility of contracting skeletal muscle express- to have anabolic effects on skeletal muscle in vitro and in vivo,
ing IL-8 has received some attention. In a pioneering study IL-15 seems also to play a role in reducing adipose tissue
by Nieman and co-workers, a several-fold increase in IL-8 mass, as IL-15 decreases lipid deposition in preadipocytes and
mRNA was found in skeletal muscle biopsies from subjects decreases the mass of white adipose tissue (WAT) (30, 199).
having completed a 3-h-treadmill-run concomitantly with in- When IL-15 was administered to adult rats for 7 days,
creased plasma levels of IL-8 (148). it resulted in a 33% decrease in WAT mass (30). The tissue

1346 Volume 3, July 2013


P1: OTA/XYZ P2: ABC
JWBT335-c120033 JWBT335/Comprehensive Physiology June 8, 2013 8:4 Printer Name: Yet to Come

Comprehensive Physiology Muscle as a Secretory Organ

response to IL-15 was related to the amount of IL-15/IL-15 IL-15 injection inhibits fat deposition in both wild-type
receptor complex expression, suggesting a direct action of IL- and ob/ob mice (5). Physical inactivity leads to loss of mus-
15 on adipose tissue (5). IL-15 mRNA expression has been cle mass and accumulation of visceral fat (155) and there are
examined in both 3T3-L1 adipogenic cells and C2C12 murine some pieces of evidence pointing at IL-15 somehow being
skeletal myogenic cells. Quantitative real-time PCR indicated involved in the regulation of abdominal adiposity. In humans,
that IL-15 mRNA was expressed by C2C12 skeletal myogenic we found a negative association between plasma IL-15 con-
cells and was unregulated more than tenfold in differentiated centration and trunk fat mass, but not limb fat mass. In support,
skeletal myotubes compared to undifferentiated myoblasts. we demonstrated a decrease in visceral fat mass, but not sub-
In contrast, 3T3-L1 cells expressed little or no IL-15 mRNA cutaneous fat mass, when IL-15 was overexpressed in murine
on either the undifferentiated preadipocyte or differentiated muscle (141).
adipocyte stages (199). These findings provide support for Both humans and laboratory mice exhibit detectable lev-
the hypothesis that IL-15 may function in a muscle-to-fat els of IL-15 in the circulation (195). The possibility, therefore,
endocrine axis that modulates fat; lean body composition and exists that IL-15 may exert endocrine (as well as paracrine)
insulin sensitivity. effects on cell types that do not themselves express IL-15.
The effect of IL-15 on adipocyte differentiation was ana- Still, at present it is unknown from which tissues the circu-
lyzed using the 3T3-L1 preadipose cell line. The data showed lating IL-15 originates. Skeletal muscles express high levels
that IL-15 tends to reduce the rate of adipocyte prolifera- of IL-15 and it has been suggested that IL-15 functions as a
tion, induces apoptosis, and partially stops differentiation. myokine that exerts positive effects on body composition via
The signaling molecules behind these actions of the cytokine an endocrine mechanism (30, 141, 165, 199). Although IL-15
on adipose cells are: p42/p44 MAPK (which seem to be as- appears to play a role in muscle-fat cross-talk, controversy
sociated with the reduced rate of proliferation induced by the exists with regard to whether IL-15 should be classified as a
cytokine), signal transducer and activator of transcription 5 true myokine (195).
(STAT5) (which is related to the actions of IL-15 on differ-
entiation), and stress-activated protein kinase (SAPK)/c-Jun
Conclusion
N-terminal kinase (JNK) (which are related to the increased
apoptosis induced by IL-15). Altogether, results from cell IL-15 has been identified as an anabolic factor that is constitu-
culture studies suggest that IL-15 is involved in the regula- tively expressed by skeletal muscle and regulated by strength
tion of adipose tissue size (62). In support of these in vitro training. While IL-15 has solid anabolic effects, it also seems
studies, a negative association was found in humans between to play a role in reducing adipose tissue mass and it is therefore
plasma IL-15 on one hand and total fat mass, trunk fat mass, suggested that IL-15 may play a role in muscle-fat cross-talk.
and percent fat mass, on the other (141). A similar finding We suggest that muscle-derived IL-15 should be classified as
was reported by Barra et al. who observed that obese human a potential myokine.
subjects exhibited lower circulating IL-15 levels than lean
subjects (14). This could indicate that IL-15 was involved in
exerting an antiobesity effect. LIF
However, Christiansen et al. reported decreased circulat- LIF is a newly discovered myokine (26). Nevertheless, LIF
ing IL-15 concentrations following a diet-induced weight loss was identified already in 1988 as a protein secreted from as-
in obese human subjects (37). cites tumor cells (84). The initial observed function of LIF
IL-15KO mice have higher amounts of body fat than con- was its ability to induce terminal differentiation of myeloid
trol mice (14) whereas transgenic mice with elevated circulat- leukemic cells (hence its name LIF). Today it is known that
ing levels of IL-15 due to a skeletal muscle-specific promoter LIF has a wide array of actions, including acting as a stim-
have lower levels of body fat than controls and are resistant ulus for platelet formation, proliferation of hematopoietic
to diet-induced obesity (197). In the latter study by Quinn cells, bone formation, neural survival and formation, mus-
et al., it was shown that mice that expressed high intramus- cle satellite cell proliferation, and acute phase production by
cular levels of IL-15 without concomitant increased levels of hepatocytes (132). LIF is a long chain four α-helix bundle
serum IL-15 levels showed no differences in adiposity com- cytokine, which is highly glycosylated and may be present
pared to controls. The data suggest that IL-15 have to be with a weight of 38 to 67 kDa that can be deglycosylated to
secreted into the circulation to exert its effects on adipose ∼20 kDa (80, 85, 215). The effects of LIF are initiated when
tissue. LIF binds the specific LIF receptor and gp130 (64), which
IL-15 has also been introduced into rodents by injection leads to phosphorylation and thereby activation of janus ki-
of recombinant IL-15 protein, by adenoviral expression vec- nase (JAK) and the STAT (44, 235). This further results in
tors (14, 30) and by DNA electrotransfer into skeletal muscle expression of SOCS-3, which negatively regulates LIF sig-
(141). The findings from these studies were that IL-15 ad- naling at the receptor level (44). Several tissues, including
ministration reduces fat mass by as much as 30% in normal skeletal muscle, express LIF. Hence, LIF is constitutively ex-
rodents and by 10% in obese rodents without an effect on pressed at a low level in type 1 muscle fibers (102, 208) and
food consumption. is implicated in conditions affecting skeletal muscle growth

Volume 3, July 2013 1347


P1: OTA/XYZ P2: ABC
JWBT335-c120033 JWBT335/Comprehensive Physiology June 8, 2013 8:4 Printer Name: Yet to Come

Muscle as a Secretory Organ Comprehensive Physiology

and regeneration (73, 102, 208, 223). LIF protein expression Although muscular LIF mRNA levels appear responsive
is augmented in mechanically overloaded rat plantaris mus- to different types of exercise, LIF protein levels remain un-
cle and in denervated rat muscle (208, 209), thus endogenous altered (26), suggesting that repetitive bouts of exercise are
LIF production is modulated by factors influencing muscle necessary to induce accumulation of LIF protein in skele-
activity. Furthermore, LIF restores the hypertrophic response tal muscle, although the latter suggestion needs to be ad-
to increased loading in LIF (-/-) mice, and in that respect LIF dressed in long-term endurance training studies. In addition,
has been denoted as an important factor in skeletal muscle muscle-derived LIF seems to be muscle specific as LIF was
hypertrophy (223). undetectable in plasma in human subjects following bicycle
Another, but perhaps related function of LIF is the potency exercise (26). Besides detection limitations, it is possible that
to induce myoblast proliferation and inhibit differentiation LIF is secreted to the interstitial space between muscle fibers
of myoblasts into multinucleated myotubes (9, 44, 222, 235). and never reaches the circulation. This suggests that LIF does
Consequently, LIF seems to affect intact skeletal muscle in not function as a systemic myokine, as does for example IL-
vivo as well as isolated muscle cell cultures in vitro. Seeing 6, but is more likely to affect skeletal muscle in an autocrine
that LIF is produced by skeletal muscle and affects intact mus- and/or a paracrine fashion.
cle as well as isolated muscle cells, we hypothesized that LIF Austin and co-workers demonstrated that LIF stimulates
would be a myokine (175). Although the LIF peptide contains myoblast proliferation in culture (9) thereby showing that LIF
a secretory amino acid sequence specifying that LIF be di- functions as a mitogenic growth factor when added experi-
rected out of the cell in which the protein is synthesized (85) mentally to muscle precursor cells in vitro. To date, differ-
no studies had investigated whether LIF is actually secreted ent groups have confirmed this finding and shown that LIF
from muscle cells or from intact skeletal muscle. We, there- induces satellite cell and myoblast proliferation, while pre-
fore, undertook a study to determine the potential of LIF as a venting premature differentiation, by activating a signaling
secreted myokine. First, we isolated and propagated satellite cascade involving JAK1, STAT1, and STAT3 (4,44,222,235).
cells from muscle biopsies obtained from healthy men, as pre- In line with this, the specific LIF receptor is primarily ex-
viously described (26), and examined whether the cells could pressed by satellite cells and not by mature muscle fibers
produce and secrete LIF into the cell media. We observed an (102). Muscle satellite cells start to form at the late stage of
accumulation of LIF in the cell media, indicating that LIF vertebrate embryo development (42). In the adult muscle, the
was produced by cultured muscle cells and secreted sponta- satellite cells are quiescent and located beneath the basal lam-
neously. Thus, LIF was not stored within the cells. Secondly, ina and the plasma membrane (242). However, in response
we used the electrotransfer technique described previously to muscle injury or exercise the normally quiescent cells be-
(141) to overexpress LIF in m. tibialis of mice and assessed come activated, re-enter the cell cycle and start to proliferate.
the abundance of LIF in serum. Whereas LIF was undetectable Later in the process, the cells irreversibly withdraw from the
in the control mice, which had saline injected into musculus cell cycle and fuse with preexisting myofibers (42). There is
tibialis (m. tibialis), the mice electrotransferred with LIF in increasing evidence that muscle adaptation and hypertrophy
m. tibialis demonstrated high LIF plasma levels 48 h after the depend on the addition of new myonuclei by way of prolifer-
electrotransfer, indicating that LIF was effectively secreted ation and further fusion of satellite cells to the adult muscle
from the intact mouse muscle. Hence, we concluded that LIF fibers (42). Hence, LIF may be involved in muscle adaptation
is a muscle-expressed protein released from cultured muscle to exercise through its potent effect on muscle satellite cells.
cells in vitro and intact mouse muscle in vivo. Indeed, Spangenburg and co-workers showed that LIF (-/-)
In a study by Broholm et al., it was found that aerobic mice were unable to enlarge their muscle size in response
exercise induces expression of LIF in human skeletal muscle to increased muscle load. However, the hypertrophic muscle
(26). The study showed that aerobic exercise and concentric response was restored when the mice were given systemic
muscle contractions regulate muscular LIF mRNA expres- treatments with LIF. Accordingly, the authors suggested that
sion in humans. With regard to the molecular mechanism LIF was an important factor in muscle hypertrophy (223).
responsible for the increase in LIF in relation to exercise, it Muscle regeneration is another process relying on activation
was shown that human muscle cells that are stimulated with and proliferation of satellite cells (42), and in this regard LIF
the Ca2+ ionophore, ionomycin, increase their expression of also demonstrates in vivo effects. LIF stimulates muscle re-
both LIF mRNA and protein (26). Thus, oscillations in Ca2+ generation in mice suffering from muscle dystrophy (112),
concentration may be the signal conveying neuromuscular ac- and LIF (-/-) mice show reduced muscle regeneration follow-
tivity into changes in the transcription of the LIF gene during ing muscle injury (109), thereby demonstrating that LIF is
muscle contractions. Since the human LIF promoter contains directly involved in regeneration of skeletal muscle. Thus, the
three putative (NFAT) cells binding sites (11) the calcineurin- possibility exists that the proliferative effects of LIF on satel-
NFAT pathway could represent a possible mechanism for LIF lite cells are closely linked to the role of LIF in muscle hyper-
gene activation by Ca2+ in myocytes. IL-6 is also regulated trophy and regeneration. Depending on the type and duration
by Ca2+ , possibly through calcineurin (12) thereby suggesting of exercise, muscle adaptation may involve processes such
that Ca2+ oscillations constitute a common signal to increase as muscle growth and muscle regeneration. LIF is produced
transcription of myokines during exercise. during exercise and might contribute to muscle adaptation

1348 Volume 3, July 2013


P1: OTA/XYZ P2: ABC
JWBT335-c120033 JWBT335/Comprehensive Physiology June 8, 2013 8:4 Printer Name: Yet to Come

Comprehensive Physiology Muscle as a Secretory Organ

following exercise by stimulating muscle satellite cell pro- comitantly led to an increase in total body energy expenditure
liferation, a process important for muscle hypertrophy and and modest improvements in glucose intolerance.
regeneration. In consequence, we hypothesize that the pri-
mary function of LIF, as a contraction-induced myokine, is Conclusion
that of a mitogenic growth factor affecting nearby satellite
cells in a paracrine fashion (27). Irisin was recently identified as a myokine, which is regulated
by exercise and plays a role in driwing white fat cells into
“brite” cells with a brown-fat-like phenotype (168).
Conclusion
LIF is a newly discovered myokine, which is induced in skele- Erythropoietin
tal muscle following exercise and affects satellite cells, muscle
Erythropoietin (EPO) is well known as a kidney-produced
growth, and regeneration.
hormone with distinct effects on erythropoiesis. However,
EPO may also be classified as a nonimmunologic cytokine,
Irisin which belongs to the IL-2 subfamily and recent studies sug-
gest that EPO should also be classified as a myokine. We
Irisin was recently identified as a new myokine, which is re-
overexpressed EPO in murine skeletal muscles by gene elec-
leased into the circulation during exercise. Irisin drives white
trotransfer. This resulted in a 100-fold increase in serum
fat cells into “brite” cells; white fat cells with a brown-fat-like
EPO and concomitant increases in hemoglobin levels. After
phenotype (21) (Fig. 8).
12 weeks, EPO expression resulted in a 23% weight reduc-
Brown fat generates heat via the mitochondrial uncoupling
tion in EPO transfected obese mice and a reduction in adipose
protein UCP1 and it has been suggested that there are two
tissue mass. EPO expression also induced a 14% increase in
distinct types of brown fat: classical brown fat derived from a
muscle volume and a 25% increase in vascularization of the
myf-5 cellular lineage and UCP1-positive cells that emerge in
EPO transfected muscle. EPO overexpression was accompa-
white fat from a non-myf-5 lineage. It was recently reported
nied by an improvement in fasting insulin levels and glucose
that so-called “beige” cells could be isolated from murine
tolerance in the high-fat fed mice. In addition, muscular fat
white fat depots. Beige cells resemble white fat cells in having
oxidation was increased 1.8-fold in both the EPO transfected
extremely low basal expression of UCP1, but, like classical
and the contralateral muscle. Thus, it appeared that supra-
brown fat, they respond to cyclic AMP stimulation with high
physiological levels of EPO have substantial metabolic effects
UCP1 expression and respiration rates. The gene expression
(90). In the latter model study, EPO mimiced a myokine in
pattern of beige cells is distinct from either white or brown
that it is produced and secreted from skeletal muscles and ex-
fat. Beige cells are, however, preferentially sensitive to the
hibits paracrine and endocrine effects on other muscles. The
polypeptide hormone irisin, which turns beige cells into so-
DNA electrotransfer method represents, however, an artificial
called brite cells (252).
model to overexpress and secrete EPO from muscles and our
Peroxisome-proliferator-activated receptor-gamma
findings do not prove that EPO is indeed secreted natively
coactivator-1 alpha (PGC-1α) plays a critical role in the
from muscles. Of interest, however, Rundqvist et al. reported
maintenance of glucose, lipid, and energy homeostasis and
that EPO is released from the exercising leg to the circula-
is involved in the pathology associated with obesity-related
tion, possibly corresponding to an increased bioavailability
disorders such as diabetes, CVD and neurodegeneration
of EPO. This finding suggests that EPO may represent a true
(120). Moreover, muscle specific PGC-1α overexpression
myokine (207).
renders mice resistant to age-related obesity and diabetes and
increases lifespan (247) suggesting that PGC-1α may con-
tribute to regulate metabolism of other tissues, notably WAT. Conclusion
By comparing muscle gene-expression profiles of trans- EPO may represent a true myokine with various metabolic
genic and wild-type mice, it was recently demonstrated that effects on muscle and adipose tissue.
PGC-1α induced the expression of several genes with secreted
protein products, including FNDC5 (21). Irisin is a proteolytic
cleavage product of the membrane protein FNDC5 and its ex- Other myokines
pression within skeletal muscle was found to be increased by By building the so-called “Myo-mouse” that is able to in-
exercise in both mice and humans. Irisin is regulated by exer- duce growth of functional type II muscle by stimulating Akt-
cise. Thus, a twofold increase in basal plasma levels of irisin 1signaling, Kenneth Walsh, Boston has identified a couple of
was observed after 10 weeks of regular exercise in humans, muscle-secreted factors (98, 244). Follistatin-like 1 (FSTL-1)
suggesting that the FNDC5/irisin complex plays a role in is a myokine that activates Akt-Endothelial nitric oxide syn-
training adaptation to exercise. When mice were injected with thase (eNOS) signaling in endothelial cells and appears to
FNDC5 expressing adenoviral particles, the systemic levels of have cardioprotective effects (156, 159) (219).
irisin increased by three- to fourfold. Overexpression of irisin Overexpression of FSTL-1 stimulates ischemia-induced
induced a brown-fat cell-like development of WAT and con- revascularization in mice through activation of eNOS (159).

Volume 3, July 2013 1349


P1: OTA/XYZ P2: ABC
JWBT335-c120033 JWBT335/Comprehensive Physiology June 8, 2013 8:4 Printer Name: Yet to Come

Muscle as a Secretory Organ Comprehensive Physiology

Skeletal
muscle
Exercise PGC-1α

FNDC5

Irisin

White Brite

UCP1

Figure 8 Exercise increases the intramuscular expression of peroxisome prolif-


erator activated receptor γ coactivator 1α (PGC-1α). Boström and colleagues re-
cently reported that PGC-1α, a transcriptional coactivator, stimulates the expres-
sion of the membrane protein fibronectin type III domain containing 5 (FNDC5),
which is proteolytically cleaved to form irisin, a myokine. Irisin drives the transfor-
mation of white fat cells into brite cells—white fat cells with a phenotype similar
to that of brown fat cells, as indicated by a marked increase in the expression
of uncoupling protein 1 (UCP1) in white adipose tissue. The investigators also
showed that an elevated level of plasma irisin, achieved through gene replace-
ment, is followed by a reduction in body weight and an improvement in metabolic
homeostasis in obese mice. Adapted, with permission, from (168).

FGF21 induces hepatic expression of PGC-1α, which is cells, eosinophils, and neutrophils (35). Horsley et al. showed
a key transcriptional regulator of energy homeostasis. More- that myogenic cells could be a target for IL-4 (94). They
over, FGF21 causes corresponding increases in fatty acid ox- demonstrated in a mouse model that IL-4 is a crucial factor
idation, tricarboxylic acid cycle flux, and gluconeogenesis in muscle growth.
without increasing glycogenesis (46, 194). Studies in humans Lafreniere et al. (113) showed that IL-4 was secreted dur-
support that FGF-21 may be an insulin-regulated myokine ing human myoblast differentiation and is required for my-
(92). otube maturation. In addition, more recent studies suggest the
Insulin-like 6 (Insl6) was identified as a myokine that is existence of secreted factors from muscle cells, which may
upregulated in skeletal muscle downstream of Akt signaling influence cancer cell growth (91) and distant organs such as
and in regenerating muscle in response to injury. Skeletal pancreas (22).
muscle-specific Insl6 transgenic mice exhibited normal mus- A role for myokines in muscle-bone interactions has been
cle mass under basal conditions but elevated satellite cell suggested (76). Two well-known osteogenic factors, IGF-1
activation and enhanced muscle regeneration in response to and FGF-2, are localized to the muscle-bone interface in vivo,
injury. In addition, overexpression of Insl6-stimulated pro- they are abundant in homogenized muscle tissue, and they are
liferation and reduced apoptosis in cultured myogenic cells, secreted from cultured myotubes in vitro (172).
whereas knockdown of Insl6 reduced proliferation and in-
creased apoptosis. These data indicate that Insl6 is an injury-
regulated myokine that functions as a myogenic regenerative Myokine screening
factor. Interleukin-4 (IL-4) is a complex glycoprotein pro- As many as 10% of encoded human genes appear to have the
duced mostly by mast cells, basophils, a subset of activated T capacity to express proteins that potentially can be secreted

1350 Volume 3, July 2013


P1: OTA/XYZ P2: ABC
JWBT335-c120033 JWBT335/Comprehensive Physiology June 8, 2013 8:4 Printer Name: Yet to Come

Comprehensive Physiology Muscle as a Secretory Organ

from cells (244). A couple of research groups have con-


tributed to the identification of the muscle cell secretome
Diseasome of physical inactivity
using various forms of proteomic analyses. In a study led by Type 2 diabetes
Kratchmarowa, a quantitative proteomics platform was used Cardiovascular
diseases
to investigate the factors secreted during the differentiation Breast cancer
of murine C2C12 skeletal muscle cells. This study identified
and quantitatively analyzed 635 secreted proteins, including
35 growth factors, 40 cytokines, and 36 metallopeptidases
(82). Yoon et al. treated differentiated L6 rat skeletal mus-
Depression
cle cells with or without insulin and comparatively analyzed Colon cancer
the proteins secreted into the media. They identified a total
of 254 proteins, among which 153 were classified as secre-
tory proteins. They reported that 14 proteins were secreted
Dementia
at higher levels under insulin stimulation, including several
proteins known to be highly secreted in metabolic diseases Figure 9 Type 2 diabetes, cardiovascular diseases, colon cancer,
(254). A study led by Schiaffino and his group reported that postmenopausal breast cancer, dementia, and depression constitute
the resulting putative skeletal muscle secretome consisted of a cluster of diseases, which can be identified as “the diseasome of
physical inactivity”. Adapted, with permission, from (165).
319 proteins, including 78 still uncharacterized proteins (20).
Norheim and coinvestigators detected a total of 236 proteins
by proteome analysis in medium conditioned by cultured hu-
in terms of phenotypical presentation and required treatment
man myotubes. Interestingly, they also showed that 15 of the
(165).
secreted muscle proteins had markedly enhanced mRNA ex-
pression in the vastus lateralis and/or trapezius muscles after
11 weeks of strength training among healthy volunteers (153).
So far, the biological effects have been mapped for only some The diseasome of physical inactivity
of the identified myokines. However, it is obvious that several The so-called “diseasome of physical inactivity” includes
myokines may serve as potential pharmaceutical targets and type 2 diabetes, CVDs, colon cancer, postmenopausal breast
provide a basis to explain the mechanisms whereby muscles cancer, dementia, and depression, which constitute a cluster
communicate with other organs. of diseases (Fig. 9).
The above diagnoses are not meant to represent an exclu-
sive list of diseases or disorders related to physical inactiv-
ity. In fact, Booth et al. listed 35 chronic diseases in which
Myokines in a Clinical Perspective
physical inactivity was thought to be a primary cause (19).
The identification of myokines provides a conceptual basis However, the diseases within the “diseasome of physical in-
for understanding how skeletal muscles communicate to other activity” are all frequent chronic diseases, associated with an
organs and why several diseases occur in networks. According enhanced risk of premature morbidity.
to our theory, muscle disuse will lead to impaired myokine Physical inactivity increases the risk of type 2 diabetes
production, which may cause simultaneous malfunction of (238), CVDs (152), colon cancer (250), postmenopausal
several organs and the occurrence of several diseases. breast cancer (137), dementia (204), and depression (161).
It is well known that many human diseases are dependent It has been documented that patients with type 2 diabetes
on each other. They may occur in, for example, genetic have a markedly increased risk of CVD (43). Furthermore,
networks (13) or so-called social networks, which include all type 2 diabetes is associated with both Alzheimer’s disease
human-to-human interactions (e.g., familial, friendship, sex- and vascular dementia, and individuals with type 2 diabetes
ual, and proximity-based contacts) that play a role not only in also have a high prevalence of depression [reviewed in (111)].
the spread of pathogens (13), but also in the spread of obesity. In addition, it has been shown that patients with type 2 diabetes
It appears that friends have an even more important effect have an elevated risk of cancer, for example, colon and breast
on a person’s risk of obesity than genes do (36). The latter cancer (200). Thus, without doubt there is a major overlap
study showed that if one friend became obese during a given between several diagnoses within the diseasome of physical
time interval, the other friend’s chances of following suit inactivity, which points at the possibility that although these
increased by 171%. In contrast, if one sibling became obese, disorders have very different phenotypical presentations, they
the chance that the other would become obese increased share some underlying pathogenetic mechanisms.
by 40%. The identification of muscle as a secretory organ provides
A couple of years ago, we suggested the existence of a the basis for a new understanding of how muscles communi-
“lifestyle network” or more specifically a “physical inactivity cate with other organs such as adipose tissue, liver, pancreas,
network” of key importance in understanding why several bones, and brain. According to this new paradigm, exercise
diseases appear in clusters, although they are highly different leads to the production and release of a variety of myokines

Volume 3, July 2013 1351


P1: OTA/XYZ P2: ABC
JWBT335-c120033 JWBT335/Comprehensive Physiology June 8, 2013 8:4 Printer Name: Yet to Come

Muscle as a Secretory Organ Comprehensive Physiology

Physical inactivity

Abdominal adiposity

Macrophage infiltration of visceral fat

Chronic systemic inflammation

Insulin resistance, atherosclerosis, neurodegeneration, tumor growth

Diseasome of physical inactivity

Type 2 diabetes

Cardiovascular
Breast cancer diseases

Depression
Colon cancer

Dementia

Figure 10 Hypothesis: physical inactivity leads to accumulation of visceral fat and conse-
quently to the activation of a network of inflammatory pathways, which promotes develop-
ment of insulin resistance, atherosclerosis, neurodegeneration, and tumor growth, leading
to the development of “the diseasome of physical inactivity.” Adapted, with permission, from
(165).

that contribute to protect against a whole network of diseases, quences of abdominal adiposity and physical inactivity are
including CVDs, type 2 diabetes, cancer, and cognitive very similar.
diseases. It is well known that both physical inactivity (175) and
abdominal adiposity (256) are associated with persistent sys-
temic low-grade inflammation (56, 77). Models of lipodys-
The link between physical inactivity, abdominal trophy suggest that if the subcutaneous fat becomes inflamed
adiposity, and systemic inflammation and adipocytes undergo apoptosis/necrosis, the fat storing ca-
It is well supported that physical inactivity is an indepen- pacity is impaired and fat will consequently be deposited as
dent and strong risk factor for accumulation of visceral fat ectopic fat (34). One obvious explanation to the differential
and consequently the activation of a network of inflamma- outcome of accumulating fat subcutaneously or as ectopic
tory pathways that promote development of insulin resis- fat could be that when fat is stored in “the wrong places,” it
tance, atherosclerosis, neurodegenation, and tumor growth will stimulate an inflammatory response. Evidence exists that
and thereby also the development of the diseases belonging visceral fat is more inflamed than subcutaneous fat and con-
to the “diseasome of physical inactivity” (165, 167) (Fig. 10). stitutes an important source of systemic inflammation (256).
Even a large amount of subcutaneous adipose tissue may We recently conducted a real-life model of physical inac-
have only little or no damaging effect and may even offer tivity that clearly demonstrated a direct link between physical
protection against chronic diseases, whereas strong evidence inactivity and accumulation of visceral fat. We included a
exists for the detrimental effects of visceral fat and fat in the group of young healthy men, who did not train on a reg-
liver and in muscle (165, 191). ular basis, and asked them to decrease their daily stepping
Abdominal adiposity is associated with CVD (75), type 2 for 2 weeks to 1500 steps from the range recommended
diabetes (16), dementia (248), colon cancer (65), and breast for adults of around 10,000. During the 14 days of reduced
cancer (253) as well as all-cause mortality independently stepping, they developed a markedly impaired glucose toler-
of body mass index, that is, also in people with a normal ance as well as attenuation of postprandial lipid metabolism.
body weight (191). Thus, it appears that the health conse- They experienced a 7% increase in intra-abdominal fat mass,

1352 Volume 3, July 2013


P1: OTA/XYZ P2: ABC
JWBT335-c120033 JWBT335/Comprehensive Physiology June 8, 2013 8:4 Printer Name: Yet to Come

Comprehensive Physiology Muscle as a Secretory Organ

Before After

Figure 11 MR-scanning demonstrating visceral fat mass before and after 14 days of reduced daily
stepping as described in (155).

measured by Magnetic Resonance (MR)-scanning, without a addition, evidence suggests that TNF-α plays a direct role
change in total fat mass while total fat-free mass and body in linking insulin resistance to vascular disease (193, 257).
mass index decreased (155) (Fig. 11). Moreover, in CVDs, activated immune cells also play ma-
This was accompanied by a marked decline in periph- jor roles, particularly in the etiology of atherosclerosis (127).
eral insulin sensitivity without an effect on hepatic EGP. The Importantly, also tumor initiation, promotion, and progres-
2-week period also induced a 7% decline in VO2 max sion is stimulated by systemic elevation of proinflammatory
(mL/min; cardiovascular fitness). cytokines (77, 121).
Of note, 2 weeks of inactivity did not provoke an increase Several downstream mediators and signaling pathways
in circulating levels of TNF. However, evidence exists of an seem to provide the cross talk between inflammatory and
association between physical inactivity and low-grade sys- metabolic signaling (165). These include the discovery of
temic inflammation in healthy, young individuals (187). These JNK and I kappa beta kinase (IκβK) as critical regulators
findings are compatible with the notion that accumulation of of insulin action activated by TNF-α (96). In human TNF-
visceral fat actually precedes chronic systemic inflammation α infusion studies, TNF-α increases phosphorylation of p70
and may represent the source of origin with regard to systemic S6 kinase, extracellular signal-regulated kinase-1/2, and c-
inflammation. Jun NH(2)-terminal kinase, concomitant with increased ser-
Although insulin resistance clearly can occur without ine, and reduced tyrosine phosphorylation of insulin receptor
inflammation, it is constantly found that chronic low-grade substrate-1. These signaling effects are associated with im-
inflammation promotes or aggravates development of in- paired phosphorylation of Akt substrate 160, the most proxi-
sulin resistance, atherosclerosis, neurodegenation, and tumor mal step identified in the insulin signaling cascade regulating
growth (77) and concomitantly the development of the dis- GLUT4 translocation and glucose uptake (193).
eases belonging to the “diseasome of physical inactivity.” The role of IL-6 in insulin resistance is highly controver-
Accumulating data suggest that although TNF-α is not the sial [as reviewed in (165, 175)]. In short, infusion of rhIL-6
pathogenetic factor, it plays a direct role in the metabolic syn- into resting healthy humans has no effect on glucose home-
drome [recently reviewed in (165,175)]. In short, patients with ostasis, although IL-6 contributes to the contraction-induced
diabetes have a high protein expression of TNF-α in skele- increase in EGP.
tal muscle and increased TNF-α levels in plasma, and it is A number of studies indicate that IL-6 enhances lipol-
likely that adipose tissue, which produces TNF-α, is the main ysis, as well as fat oxidation, via an activation of AMPK
source of the circulating TNF-α. In vitro studies demonstrate [reviewed in (175)]. Consistent with this idea, Wallenius
that TNF-α has direct inhibitory effects on insulin signaling. et al. (243) demonstrated that IL-6-deficient mice developed
In addition, TNF-α infusion in healthy humans induces in- mature-onset obesity and insulin resistance. Of note, both
sulin resistance in skeletal muscle, without an effect on EGP. TNF-α and IL-6 induce lipolysis, whereas IL-6 only appears
It has also been proposed that TNF-α causes insulin resistance to induce fat oxidation (192,241). Given the different biologi-
indirectly in vivo by increasing the release of FFAs from adi- cal profiles of TNF-α and IL-6 and given that TNF-α may trig-
pose tissue. TNF-α increases lipolysis in human and 3T3-L1 ger an IL-6 release, one theory holds that it is TNF-α derived
adipocytes. However, TNF-α has no effect on muscle fatty from adipose tissue that is actually the major “driver” behind
acid oxidation, but increases fatty acid incorporation into di- inflammation-induced insulin resistance and atherosclerosis.
acylglycerol, which may be involved in the development of The beneficial effect of exercise in the protection against
the TNF-α-induced insulin resistance in skeletal muscle. In diseases associated with chronic inflammation may to some

Volume 3, July 2013 1353


P1: OTA/XYZ P2: ABC
JWBT335-c120033 JWBT335/Comprehensive Physiology June 8, 2013 8:4 Printer Name: Yet to Come

Muscle as a Secretory Organ Comprehensive Physiology

extent be ascribed to an anti-inflammatory effect of regu- increase primarily in IL-6, followed by an increase in IL-1ra
lar exercise. However, we have suggested that the long-term and IL-10.
anti-inflammatory effects of exercise may be mediated via It has been clearly demonstrated that both the upstream
effects of exercise leading to a reduction in visceral fat mass and downstream signaling pathways for IL-6 differ markedly
(165). IL-6 may also work in an endocrine fashion to increase between myocytes and macrophages (Fig. 12).
hepatic glucose production during exercise or lipolysis in adi- Unlike IL-6 signaling in macrophages, which is dependent
pose tissue [reviewed in (175)]. However, although it has not upon activation of the NFκB signaling pathway, it appears
been demonstrated that IL-6 has specific effects on visceral fat that intramuscular IL-6 expression is regulated by a network
mass, it appears to play an important role in mediating AMPK- of signaling cascades that among other pathways is likely
induced fat oxidation in skeletal muscle. As described above, to involve cross-talk between the Ca2+/NFAT and glyco-
IL-15 is a potential myokine, which appears to be involved gen/p38 MAPK pathways. Therefore, when IL-6 is signaling
in the regulation of visceral fat as overexpression of IL-15 in monocytes or macrophages, it creates a proinflammatory
protected mice from obesity, especially with regard to accu- response, whereas IL-6 activation and signaling in muscle is
mulation of visceral fat (141,143). In addition, BDNF appears totally independent upon a preceding TNF-response or NFκB
to work in an autocrine or paracrine fashion with strong ef- activation (165, 175).
fects on peripheral metabolism, including fat oxidation with a Without doubt, an acute bout of exercise elicits an anti-
subsequent effect on the size of adipose tissue (177). Finally, inflammatory response and IL-6 appears to mediate some of
EPO may play a role in muscle-fat cross-talk and contribute the anti-inflammatory effects of exercise (144, 187). IL-6 in-
to minimize the abdominal adiposity (90). hibits lipo polysaccharide (LPS)-induced TNF production in
cultured human monocytes (214). Other studies show that
levels of TNF-α are elevated in anti-IL-6-treated mice and in
The anti-inflammatory effects of an acute bout IL-6 deficient knockout mice (135). In healthy humans, both
of exercise exercise and rhIL-6 infusion inhibit the endotoxin-induced
Regular exercise appears to induce direct anti-inflammatory increase in circulating levels of TNF-α (224). Moreover, IL-6
effects, suggesting that physical activity per se may suppress contributes to mediate anti-inflammatory effects by stimulat-
systemic low-grade inflammation (24, 68, 126, 144, 187, 188, ing the production of the classic anti-inflammatory cytokines
212). IL-1ra and IL-10 (229).
A substantial amount of studies have demonstrated that The possibility exists that with regular exercise, the anti-
markers of inflammation are reduced following longer-term inflammatory effects of an acute bout of exercise will protect
behavioral changes involving both reduced energy intake and against chronic systemic low-grade inflammation, but such a
increased physical activity [reviewed in (165, 187)]. direct link between the acute effects of exercise and the long-
A number of mechanisms may be responsible for this term benefits has yet to be established. Another possibility
effect. Exercise increases the release of epinephrine, cortisol, is that regular exercise could protect against accumulation of
growth hormone, prolactin, and other factors that have visceral fat and ectopic fat as such and thereby also protect
immunomodulatory effects (77, 145, 165). Acute exercise against chronic systemic inflammation.
has been shown to induce a true anti-inflammatory response. The myokine concept provides a new platform for un-
A model of “low grade inflammation” was established derstanding some of the molecular mechanisms underlying
in our laboratory. A very low dose of Escherichia. Coli muscle organ, including muscle-fat cross-talk and for under-
endotoxin was administered to healthy volunteers, who were standing the anti-inflammatory effects of exercise and thereby
randomized to either rest or exercise prior to endotoxin how exercise may contribute to protect against a whole net-
administration. In resting subjects, endotoxin induced a 2- to work of chronic diseases with very different phenotypic
threefold increase in circulating levels of TNF-α. In contrast, presentations.
when the subjects performed 3 h of ergometer cycling and
received the endotoxin bolus at 2.5 h, the TNF-α response
was totally blunted (224), suggesting that acute exercise may Myokines in myogenesis
inhibit TNF production. Although some myokines exert their actions on other organs
The cytokine response to exercise differs markedly from in a hormone-like fashion, many of them operate locally on
that elicited by severe infections in the fact that the clas- skeletal muscle itself. Skeletal muscles may adjust to increas-
sical proinflammatory cytokines, TNF-α and IL-1β, do not ing demands by enlarging fiber size, and it is perhaps not
increase with exercise [as reviewed in (175)]. surprising that many of the factors secreted from the working
In relation to exercise, IL-6 is the first cytokine released muscle act to regulate muscle hypertrophy and repair. In this
into the blood. The level of circulating IL-6 increases in an sense, myokines provide a feedback loop for the muscle to reg-
exponential fashion (up to 100-fold) during acute exercise and ulate its own growth and regeneration allowing, for example,
declines in the postexercise period. The circulating levels of for adaptation to exercise training. Myokines may regulate
the well-known anti-inflammatory cytokines, IL-1ra and IL- skeletal myogenesis in numerous ways, including a process
10, also increase after exercise. Thus, exercise provokes an that involves sequential satellite cell activation and migration,

1354 Volume 3, July 2013


P1: OTA/XYZ P2: ABC
JWBT335-c120033 JWBT335/Comprehensive Physiology June 8, 2013 8:4 Printer Name: Yet to Come

Comprehensive Physiology Muscle as a Secretory Organ

Macrophage
LPS
CD14
Skeletal muscle

TLR4
MyD88 IRAKs

Ca2+
TRAF-6
Calcineurin
p38 MAPK
IKK-α IKK-β
IKK-γ
CREBP NFAT AP-1
p300 CBP
NF κ B

IL-1β IL-6
IL-6
TNF-α

Figure 12 The proposed cytokine signaling pathways for macrophages and contracting skeletal muscle. While it is well known that transcription
of interleukin-6 (IL-6) and other proinflammatory cytokines such as tumor necrosis factor-alpha (TNF-α) and IL-β is principally regulated by the
Toll like receptor (##TLR) receptor signaling cascade that results in nuclear translocation and activation of NFκB, evidence in contracting skeletal
muscle suggests that contraction leads to increased cytosolic Ca2+ and activation of p38 MAPK and/or calcineurin, which leads to activation of
transcription factors depending upon these upstream events. Adapted, with permission, from (175).

with subsequent proliferation and differentiation of myoblasts to be regulated by IL-4 in mouse skeletal muscle. Muscle
into mature myotubes. In fact, each of these steps has been cells lacking IL-4 have been shown to be smaller in size
shown to be regulated by one or more myokines. and to have less myonuclei (95). Of note, IL-4 is secreted
In short, LIF increases myoblast proliferation and upreg- from differentiating human muscle cell cultures and causes
ulates expression of c-Myc and Jun-b, while knockdown of increased migration but not proliferation of myoblasts (113).
the LIF receptor leads to decreased myoblast proliferation, Myostatin is a member of the TGF-β superfamily. In hu-
suggesting a role for LIF as a positive regulator of satellite man skeletal myoblasts, myostatin negatively regulates pro-
cell proliferation (25). liferation by an upregulation of the cyclin-dependent kinase
IL-6 is expressed by human myoblasts (15, 41) and hu- (Cdk) inhibitor p21, causing an inhibition of Cdk2 and the
man cultured myotubes (104). Moreover, IL-6 is locally and Cdk2 downstream target retinoblastoma (129).
transiently produced by growing murine myofibers and asso- When we consider the numerous functions of myokines in
ciated satellite cells (217). In addition, IL-6 is released from the regulation of skeletal muscle growth and maintenance, it
human primary muscle cell cultures from healthy individuals is possible that myokines may provide a potential therapeutic
(72, 79) and from patients with type 2 diabetes (72). target for the treatment of muscle growth and regeneration
IL-7 has been identified as a myokine secreted from differ- disorders and explain why regular exercise retards aging pro-
entiating human myotubes (79). Although IL-7 expression has cesses. By targeting these myogenic myokines, it might be
been shown to increase throughout differentiation of cultured possible to ameliorate the symptoms of muscle wasting dis-
muscle cells, the expression of the IL-7 receptor was shown to orders and age-related sarcopenia.
be highest in undifferentiated cells. Haugen et al. showed that
the addition of recombinant IL-7 caused a decrease in mRNA Conclusion
expression of both MYOG and MHC-2 and an upregulation
of the satellite cell marker Pax7, and furthermore increased
migration of satellite cells (79).
A central step of myocyte differentiation is the fusion of
Final Conclusion
myoblasts with existing fibers to form mature, multinucleated Two millennia ago, Hippocrates observed that “walking is
myocytes. This process was demonstrated by Horsley et al. man’s best medicine.” Already then, the benefits of physical

Volume 3, July 2013 1355


P1: OTA/XYZ P2: ABC
JWBT335-c120033 JWBT335/Comprehensive Physiology June 8, 2013 8:4 Printer Name: Yet to Come

Muscle as a Secretory Organ Comprehensive Physiology

Physical activity reveal how myokine production are regulated on the basal
level; whether myokine resistance and abnormal myokine sig-
naling are dominant in pathophysiology, whether myokines .
Myokines may be proven to play a role as biomarkers of phys-
ical activity/inactivity. They may also be useful in monitoring
exercise effects both with regard to disease and performance.
Considering the numerous functions of myokines in
the regulation of skeletal muscle growth and maintenance,
myokines provide a potential therapeutic target for the treat-
Myokines
ment of muscle growth and regeneration disorders. Targeting
these myogenic myokines could thus provide means to ame-
liorate the symptoms of muscle wasting disorders, muscular
Muscle hypertrophy (myostatin, LIF, IL-4, IL-6, IL-7, IL-15) dystrophies, and age-related sarcopenia. It is possible that
Adipose tissue oxidation (IL-6, BDNF) physical inactivity or muscle disuse may lead to an altered or
Insulin sensitivity (IL-6)
Osteogenesis (IGF-1, FGF-2) impaired myokine response and/or resistance to the effects of
Anti-inflammation (IL-6) myokines, explaining why lack of physical activity increases
Antitumor defence [unidentified secreted factor(s)]
Pancreas function [unidentified secreted factor(s)] the risk of a whole network of diseases, such as cancer, CVDs,
type 2 diabetes, dementia, and osteoporosis.

Decreased risk of chronic diseases and premature mortality


Acknowledgements
Figure 13 The finding that muscle produces and releases myokines I wish to gratefully acknowledge my collaborators, postdoc-
provides a conceptual basis for understanding some of the molecular
mechanisms that link physical activity to protection against premature toral fellows, students, and technicians who have contributed
mortality (172). much of the work reported in this review. I want to thank
Inge Holm for critical review of the article. The Centre of
Inflammation and Metabolism (CIM) is supported by a grant
activity to health were recognized. Since then, the benefits of from the Danish National Research Foundation (02-512-55).
physical activity in lowering the risk of death from any cause This study was further supported by the Danish Council for
and improving longevity have been well documented (160). In Independent Research—Medical Sciences, the Commission
the past few years, exercise research has contributed tremen- of the European Communities (Grant Agreement no. 223576-
dously to our understanding of the benefits of exercise on a MYOAGE). CIM is part of the UNIK Project: Food, Fitness
molecular level and recently also to the concept of considering & Pharma for Health and Disease, supported by the Danish
skeletal muscle as a secretory organ. The identification of the Ministry of Science, Technology, and Innovation. CIM is a
muscle secretome provides a new platform for understanding member of DD2—the Danish Center for Strategic Research in
how muscles communicate with other organs and to explain Type 2 Diabetes (the Danish Council for Strategic Research,
how a healthy muscle tissue is developed and maintained grant nos. 09-067009 and 09-075724). The Copenhagen Mus-
(Fig. 13). Some myokines may be involved in communicat- cle Research Centre is supported by a grant from the Capital
ing to other organs, such as adipose tissue, liver, pancreas, Region of Denmark.
bones, and brain. However, some myokines exert their effects
within the muscle itself. Thus, myostatin, LIF, IL-4, IL-6, and
IL-7 are involved in muscle hypertrophy and myogenesis,
whereas BDNF and IL-6 are involved in AMPK-mediated fat References
oxidation. BDNF and Insl6 are involved in regeneration fol-
1. Addison CL, Daniel TO, Burdick MD, Liu H, Ehlert JE, Xue YY,
lowing injury. IL-6 also appears to have systemic effects on Buechi L, Walz A, Richmond A, Strieter RM. The CXC chemokine
the liver, adipose tissue, and the immune system, and mediates receptor 2, CXCR2, is the putative receptor for ELR+ CXC chemokine-
induced angiogenic activity. J Immunol 165: 5269-5277, 2000.
cross-talk between the intestine and pancreatic islets. Other 2. Allen DL, Cleary AS, Speaker KJ, Lindsay SF, Uyenishi J, Reed JM,
myokines include the osteogenic factors IGF-1 and FGF-2; Madden MC, Mehan RS. Myostatin, activin receptor IIb, and follistatin-
like-3 gene expression are altered in adipose tissue and skeletal muscle
FSTL-1, which improve the endothelial function of the vascu- of obese mice. Am J Physiol Endocrinol Metab 294: E918-E927, 2008.
lar system; and the PGC-1α-dependent myokine irisin, which 3. Allen DL, Hittel DS, McPherron AC. Expression and function of myo-
statin in obesity, diabetes, and exercise adaptation. Med Sci Sports Exerc
drives brown-fat-like development. Furthermore, studies in 43: 1828-1835, 2011.
the past few years suggest the existence of yet unidentified 4. Alter J, Rozentzweig D, Bengal E. Inhibition of myoblast differentiation
by tumor necrosis factor alpha is mediated by c-Jun N-terminal kinase
factors, secreted from muscle cells, which may influence can- 1 and leukemia inhibitory factor. J Biol Chem 283: 23224-23234, 2008.
cer cell growth and pancreas function. 5. Alvarez B, Carbo N, Lopez-Soriano J, Drivdahl RH, Busquets S, Lopez-
The identification of the muscle secretome could set a new Soriano FJ, Argiles JM, Quinn LS. Effects of interleukin-15 (IL-15) on
adipose tissue mass in rodent obesity models: Evidence for direct IL-15
agenda for the scientific community. Future studies should action on adipose tissue. Biochim Biophys Acta 1570: 33-37, 2002.

1356 Volume 3, July 2013


P1: OTA/XYZ P2: ABC
JWBT335-c120033 JWBT335/Comprehensive Physiology June 8, 2013 8:4 Printer Name: Yet to Come

Comprehensive Physiology Muscle as a Secretory Organ

6. Argiles JM, Lopez-Soriano J, Almendro V, Busquets S, Lopez-Soriano 33. Carey AL, Steinberg GR, Macaulay SL, Thomas WG, Holmes AG,
FJ. Cross-talk between skeletal muscle and adipose tissue: A link with Ramm G, Prelovsek O, Hohnen-Behrens C, Watt MJ, James DE,
obesity? Med Res Rev 25: 49-65, 2005. Kemp BE, Pedersen BK, Febbraio MA. Interleukin-6 increases insulin-
7. Asmussen E. Ventilation at transition from rest to exercise. Acta Physiol stimulated glucose disposal in humans and glucose uptake and fatty
Scand 89: 68-78, 1973. acid oxidation in vitro via AMP-activated protein kinase. Diabetes 55:
8. Astrom MB, Feigh M, Pedersen BK. Persistent low-grade inflammation 2688-2697, 2006.
and regular exercise. Front Biosci (Schol Ed) 2: 96-105, 2010. 34. Caron-Debarle M, Lagathu C, Boccara F, Vigouroux C, Capeau J. HIV-
9. Austin L, Burgess AW. Stimulation of myoblast proliferation in culture associated lipodystrophy: From fat injury to premature aging. Trends
by leukaemia inhibitory factor and other cytokines. J Neurol Sci 101: Mol Med 16: 218-229, 2010.
193-197, 1991. 35. Chomarat P, Banchereau J. An update on interleukin-4 and its receptor.
10. Baggiolini M. Chemokines in pathology and medicine. J Intern Med Eur Cytokine Netw 8: 333-344, 1997.
250: 91-104, 2001. 36. Christakis NA, Fowler JH. The spread of obesity in a large social
11. Bamberger AM, Jenatschke S, Schulte HM, Ellebrecht I, Beil FU, network over 32 years. N Engl J Med 357: 370-379, 2007.
Bamberger CM. Regulation of the human leukemia inhibitory factor 37. Christiansen T, Paulsen SK, Bruun JM, Pedersen SB, Richelsen B. Ex-
gene by ETS transcription factors. Neuroimmunomodulation 11: 10- ercise training versus diet-induced weight-loss on metabolic risk factors
19, 2004. and inflammatory markers in obese subjects: A 12-week randomized
12. Banzet S, Koulmann N, Simler N, Birot O, Sanchez H, Chapot R, intervention study. Am J Physiol Endocrinol Metab 298: E824-E831,
Peinnequin A, Bigard X. Fibre-type specificity of interleukin-6 gene 2010.
transcription during muscle contraction in rat: Association with cal- 38. Cook KS, Min HY, Johnson D, Chaplinsky RJ, Flier JS, Hunt CR,
cineurin activity. J Physiol 566: 839-847, 2005. Spiegelman BM. Adipsin: A circulating serine protease homolog se-
13. Barabasi AL. Network medicine–from obesity to the “diseasome”. N creted by adipose tissue and sciatic nerve. Science 237: 402-405, 1987.
Engl J Med 357: 404-407, 2007. 39. Copray S, Liem R, Brouwer N, Greenhaff P, Habens F, Fernyhough
14. Barra NG, Reid S, MacKenzie R, Werstuck G, Trigatti BL, Richards C, P. Contraction-induced muscle fiber damage is increased in soleus
Holloway AC, Ashkar AA. Interleukin-15 contributes to the regulation muscle of streptozotocin-diabetic rats and is associated with elevated
of murine adipose tissue and human adipocytes. Obesity (Silver Spring) expression of brain-derived neurotrophic factor mRNA in muscle fibers
18: 1601-1607, 2010. and activated satellite cells. Exp Neurol 161: 597-608, 2000.
15. Bartoccioni E, Michaelis D, Hohlfeld R. Constitutive and cytokine- 40. Crofford LJ. The hypothalamic-pituitary-adrenal axis in the pathogen-
induced production of interleukin-6 by human myoblasts. Immunol esis of rheumatic diseases. Endocrinol Metab Clin North Am 31: 1-13,
Lett 42: 135-138, 1994. 2002.
16. Bays HE. “ Sick fat,” metabolic disease, and atherosclerosis. Am J Med 41. De Rossi M, Bernasconi P, Baggi F, de Waal MR, Mantegazza R.
122: S26-S37, 2009. Cytokines and chemokines are both expressed by human myoblasts:
17. Belperio JA, Keane MP, Arenberg DA, Addison CL, Ehlert JE, Burdick Possible relevance for the immune pathogenesis of muscle inflamma-
MD, Strieter RM. CXC chemokines in angiogenesis. J Leukoc Biol 68: tion. Int Immunol 12: 1329-1335, 2000.
1-8, 2000. 42. Dhawan J, Rando TA. Stem cells in postnatal myogenesis: Molecular
18. Bergfors M, Barnekow-Bergkvist M, Kalezic N, Lyskov E, Eriksson mechanisms of satellite cell quiescence, activation and replenishment.
JW. Short-term effects of repetitive arm work and dynamic exercise on Trends Cell Biol 15: 666-673, 2005.
glucose metabolism and insulin sensitivity. Acta Physiol Scand 183: 43. Diamant M, Tushuizen ME. The metabolic syndrome and endothelial
345-356, 2005. dysfunction: Common highway to type 2 diabetes and CVD. Curr Diab
19. Booth FW, Roberts CK, Laye MJ. Lack of exercise is a major cause of Rep 6: 279-286, 2006.
chronic diseases. Comprehensive Physiology, 2(2): 1143-1211, 2012. 44. Diao Y, Wang X, Wu Z. SOCS1, SOCS3, and PIAS1 promote myogenic
20. Bortoluzzi S, Scannapieco P, Cestaro A, Danieli GA, Schiaffino S. differentiation by inhibiting the leukemia inhibitory factor-induced
Computational reconstruction of the human skeletal muscle secretome. JAK1/STAT1/STAT3 pathway. Mol Cell Biol 29: 5084-5093, 2009.
Proteins 62: 776-792, 2006. 45. Dinarello CA, Mier JW. Interleukins. Annu Rev Med 37: 173-178, 1986.
21. Bostrom P, Wu J, Jedrychowski MP, Korde A, Ye L, Lo JC, Rasbach 46. Domouzoglou EM, Maratos-Flier E. Fibroblast growth factor 21 is a
KA, Bostrom EA, Choi JH, Long JZ, Kajimura S, Zingaretti MC, Vind metabolic regulator that plays a role in the adaptation to ketosis. Am J
BF, Tu H, Cinti S, Hojlund K, Gygi SP, Spiegelman BM. A PGC1- Clin Nutr 93: 901S-905, 2011.
alpha-dependent myokine that drives brown-fat-like development of 47. Ellingsgaard H, Ehses JA, Hammar EB, Van LL, Quintens R, Martens
white fat and thermogenesis. Nature 481: 463-468, 2012. G, Kerr-Conte J, Pattou F, Berney T, Pipeleers D, Halban PA, Schuit
22. Bouzakri K, Plomgaard P, Berney T, Donath MY, Pedersen BK, Halban FC, Donath MY. Interleukin-6 regulates pancreatic alpha-cell mass
PA. Bimodal effect on pancreatic â-cells of secretory products from expansion. Proc Natl Acad Sci U S A 105: 13163-13168, 2008.
normal or insulin-resistant human skeletal muscle. Diabetes 60: 1111- 48. Ellingsgaard H, Hauselmann I, Schuler B, Eppler E, Meier D, Bouzakri
1121, 2011. K, Wueest S, Muller Y, Reinecke M, Konrad D, Gassmann M, Halban P,
23. Brandt C, Nielsen AR, Fischer CP, Hansen J, Pedersen BK, Plomgaard Gromada J, Ehses J, Donath M. Interleukin-6 enhances insulin secretion
P. Plasma and muscle myostatin in relation to type 2 diabetes. PLoS by increasing glucagon-like peptide-1 secretion from L cells and alpha
One 7: e37236, 2012. cells. Nat Med 17: 1481-1489, 2011.
24. Brandt C, Pedersen BK. The role of exercise-induced myokines in 49. Febbraio MA. Signaling pathways for IL-6 within skeletal muscle.
muscle homeostasis and the defense against chronic diseases. J Biomed Exerc Immunol Rev 9: 34-9, 2003.
Biotechnol 2010: 520258, 2010. 50. Febbraio MA, Hiscock N, Sacchetti M, Fischer CP, Pedersen BK.
25. Broholm C, Laye MJ, Brandt C, Vadalasetty R, Pilegaard H, Peder- Interleukin-6 is a novel factor mediating glucose homeostasis during
sen BK, Scheele C. LIF is a contraction-induced myokine stimulating skeletal muscle contraction. Diabetes 53: 1643-1648, 2004.
human myocyte proliferation. J Appl Physiol 111: 251-259, 2011. 51. Febbraio MA, Ott P, Nielsen HB, Steensberg A, Keller C, Krustrup P,
26. Broholm C, Mortensen OH, Nielsen S, Akerstrom T, Zankari A, Dahl Secher NH, Pedersen BK. Hepatosplanchnic clearance of interleukin-
B, Pedersen BK. Exercise induces expression of leukaemia inhibitory 6 in humans during exercise. Am J Physiol Endocrinol Metab 285:
factor in human skeletal muscle. J Physiol 586: 2195-2201, 2008. E397-E402, 2003.
27. Broholm C, Pedersen BK. Leukaemia inhibitory factor–an exercise- 52. Febbraio MA, Pedersen BK. Muscle-derived interleukin-6: Mecha-
induced myokine. Exerc Immunol Rev 16: 77-85, 2010. nisms for activation and possible biological roles. FASEB J 16: 1335-
28. Bruce CR, Dyck DJ. Cytokine regulation of skeletal muscle fatty acid 1347, 2002.
metabolism: Effect of interleukin-6 and tumor necrosis factor-alpha. 53. Febbraio MA, Pedersen BK. Contraction-induced myokine production
Am J Physiol Endocrinol Metab 287: E616-E621, 2004. and release: Is skeletal muscle an endocrine organ? Exerc Sport Sci Rev
29. Bruunsgaard H, Galbo H, Halkjaer-Kristensen J, Johansen TL, 33: 114-119, 2005.
MacLean DA, Pedersen BK. Exercise-induced increase in interleukin-6 54. Febbraio MA, Steensberg A, Keller C, Starkie RL, Krustrup P, Ott P,
is related to muscle damage. J Physiol Lond 499: 833-841, 1997. Secher NH, Pedersen BK. Glucose ingestion attenuates interleukin-6
30. Carbo N, Lopez-Soriano J, Costelli P, Alvarez B, Busquets S, Bac- release from contracting skeletal muscle in humans. J Physiol (London)
cino FM, Quinn LS, Lopez-Soriano FJ, Argiles JM. Interleukin-15 549: 607-612, 2003.
mediates reciprocal regulation of adipose and muscle mass: A poten- 55. Feldman BJ, Streeper RS, Farese RV, Jr., Yamamoto KR. Myo-
tial role in body weight control. Biochim Biophys Acta 1526: 17-24, statin modulates adipogenesis to generate adipocytes with favorable
2001. metabolic effects. Proc Natl Acad Sci U S A 103: 15675-15680, 2006.
31. Carbo N, Lopez-Soriano J, Costelli P, Busquets S, Alvarez B, Baccino 56. Festa A, D’Agostino R, Jr., Tracy RP, Haffner SM. Elevated levels of
FM, Quinn LS, Lopez-Soriano FJ, Argiles JM. Interleukin-15 antag- acute-phase proteins and plasminogen activator inhibitor-1 predict the
onizes muscle protein waste in tumour-bearing rats. Br J Cancer 83: development of type 2 diabetes: The insulin resistance atherosclerosis
526-531, 2000. study. Diabetes 51: 1131-1137, 2002.
32. Carey AL, Febbraio MA. Interleukin-6 and insulin sensitivity: Friend 57. Figueras M, Busquets S, Carbo N, Barreiro E, Almendro V, Argiles
or foe? Diabetologia 47: 1135-1142, 2004. JM, Lopez-Soriano FJ. Interleukin-15 is able to suppress the increased

Volume 3, July 2013 1357


P1: OTA/XYZ P2: ABC
JWBT335-c120033 JWBT335/Comprehensive Physiology June 8, 2013 8:4 Printer Name: Yet to Come

Muscle as a Secretory Organ Comprehensive Physiology

DNA fragmentation associated with muscle wasting in tumour-bearing 85. Hinds MG, Maurer T, Zhang JG, Nicola NA, Norton RS. Resonance
rats. FEBS Lett 569: 201-206, 2004. assignments, secondary structure and topology of leukaemia inhibitory
58. Fischer CP. Interleukin-6 in acute exercise and training: What is the factor in solution. J Biomol NMR 9: 113-126, 1997.
biological relevance? Exerc Immunol Rev 12: 6-33, 2006. 86. Hirose L, Nosaka K, Newton M, Laveder A, Kano M, Peake J, Suzuki
59. Fischer CP, Hiscock N, Basu S, Vessby B, Kallner A, Sjöberg LB, K. Changes in inflammatory mediators following eccentric exercise of
Febbraio MA, Pedersen BK. Supplementation with vitamins C and E the elbow flexors. Exerc Immunol Rev 10: 75-90.: 75-90, 2004.
inhibits the release of interleukin-6 from contracting human skeletal 87. Hiscock N, Chan MH, Bisucci T, Darby IA, Febbraio MA. Skeletal
muscle. J Physiol 558: 633-645, 2004. myocytes are a source of interleukin-6 mRNA expression and protein
60. Fischer CP, Plomgaard P, Hansen AK, Pilegaard H, Saltin B, Pedersen release during contraction: Evidence of fiber type specificity. FASEB J
BK. Endurance training reduces the contraction-induced interleukin-6 18: 992-994, 2004.
mRNA expression in human skeletal muscle. Am J Physiol Endocrinol 88. Hittel DS, Berggren JR, Shearer J, Boyle K, Houmard JA. Increased
Metab 287: E1189-E1194, 2004. secretion and expression of myostatin in skeletal muscle from extremely
61. Furmanczyk PS, Quinn LS. Interleukin-15 increases myosin accretion obese women. Diabetes 58: 30-38, 2009.
in human skeletal myogenic cultures. Cell Biol Int 27: 845-851, 2003. 89. Hoene M, Weigert C. The role of interleukin-6 in insulin resistance,
62. Fuster G, Almendro V, Fontes-Oliveira CC, Toledo M, Costelli P, body fat distribution and energy balance. Obes Rev 9: 20-29, 2008.
Busquets S, Lopez-Soriano FJ, Argiles JM. Interleukin-15 affects dif- 90. Hojman P, Brolin C, Gissel H, Brandt C, Zerahn B, Pedersen BK, Gehl
ferentiation and apoptosis in adipocytes: Implications in obesity. Lipids J. Erythropoietin over-expression protects against diet-induced obesity
46: 1033-1042, 2011. in mice through increased fat oxidation in muscles. PLoS One 4: e5894,
63. Gala RR. Prolactin and growth hormone in the regulation of the immune 2009.
system. Proc Soc Exp Biol Med 198: 513-527, 1991. 91. Hojman P, Dethlefsen C, Brandt C, Hansen J, Pedersen L, Pedersen BK.
64. Giese B, Roderburg C, Sommerauer M, Wortmann SB, Metz S, Hein- Exercise-induced muscle-derived cytokines inhibit mammary cancer
rich PC, Muller-Newen G. Dimerization of the cytokine receptors cell growth. Am J Physiol Endocrinol Metab 301: E504-E510, 2011.
gp130 and LIFR analysed in single cells. J Cell Sci 118: 5129-5140, 92. Hojman P, Pedersen M, Nielsen AR, Krogh-Madsen R, Yfanti C, Ak-
2005. erstrom T, Nielsen S, Pedersen BK. Fibroblast growth factor-21 is
65. Giovannucci E. Metabolic syndrome, hyperinsulinemia, and colon can- induced in human skeletal muscles by hyperinsulinemia. Diabetes 58:
cer: A review. Am J Clin Nutr 86: s836-s842, 2007. 2797-2801, 2009.
66. Gleeson M. Interleukins and exercise. J Physiol 529: 1, 2000. 93. Holmes AG, Watt MJ, Carey AL, Febbraio MA. Ionomycin, but
67. Gleeson M. Immune function in sport and exercise. J Appl Physiol 103: not physiologic doses of epinephrine, stimulates skeletal muscle
693-699, 2007. interleukin-6 mRNA expression and protein release. Metabolism 53:
68. Gleeson M, Bishop NC, Stensel DJ, Lindley MR, Mastana SS, Nimmo 1492-1495, 2004.
MA. The anti-inflammatory effects of exercise: Mechanisms and impli- 94. Horsley V, Jansen KM, Mills ST, Pavlath GK. IL-4 acts as a myoblast
cations for the prevention and treatment of disease. Nat Rev Immunol recruitment factor during mammalian muscle growth. Cell 113: 483-
11: 607-615, 2011. 494, 2003.
69. Goldstein MS. Humoral nature of the hypoglycemic factor of muscular 95. Horsley V, Pavlath GK. Forming a multinucleated cell: Molecules that
work. Diabetes 10: 232-234, 1961. regulate myoblast fusion. Cells Tissues Organs 176: 67-78, 2004.
70. Gomez-Pinilla F, Ying Z, Roy RR, Molteni R, Edgerton VR. Voluntary 96. Hotamisligil GS. Inflammatory pathways and insulin action. Int J Obes
exercise induces a BDNF-mediated mechanism that promotes neuro- Relat Metab Disord 27(Suppl 3): S53-S55, 2003.
plasticity. J Neurophysiol 88: 2187-2195, 2002. 97. Huang EJ, Reichardt LF. Neurotrophins: Roles in neuronal develop-
71. Grabstein KH, Eisenman J, Shanebeck K, Rauch C, Srinivasan S, Fung ment and function. Annu Rev Neurosci 24: 677-736, 2001.
V, Beers C, Richardson J, Schoenborn MA, Ahdieh M. Cloning of a T 98. Izumiya Y, Hopkins T, Morris C, Sato K, Zeng L, Viereck J, Hamilton
cell growth factor that interacts with the beta chain of the interleukin-2 JA, Ouchi N, LeBrasseur NK, Walsh K. Fast/Glycolytic muscle fiber
receptor. Science 264: 965-968, 1994. growth reduces fat mass and improves metabolic parameters in obese
72. Green CJ, Pedersen M, Pedersen BK, Scheele C. Elevated NF- mice. Cell Metab 7: 159-172, 2008.
{kappa}B activation is conserved in human myocytes cultured from 99. Johnson HM, Smith EM, Torres BA, Blalock JE. Regulation of the in
obese type 2 diabetic patients and attenuated by AMP-activated protein vitro antibody response by neuroendocrine hormones. Proc Natl Acad
kinase. Diabetes 60: 2810-2819, 2011. Sci U S A 79: 4171-4174, 1982.
73. Gregorevic P, Williams DA, Lynch GS. Effects of leukemia inhibitory 100. Jonsdottir IH, Schjerling P, Ostrowski K, Asp S, Richter EA, Pedersen
factor on rat skeletal muscles are modulated by clenbuterol. Muscle BK. Muscle contractions induce interleukin-6 mRNA production in rat
Nerve 25: 194-201, 2002. skeletal muscles. J Physiol (London) 528: 157-163, 2000.
74. Guo T, Jou W, Chanturiya T, Portas J, Gavrilova O, McPherron AC. 101. Kahn BB, Alquier T, Carling D, Hardie DG. AMP-activated protein
Myostatin inhibition in muscle, but not adipose tissue, decreases fat kinase: Ancient energy gauge provides clues to modern understanding
mass and improves insulin sensitivity. PLoS One 4: e4937, 2009. of metabolism. Cell Metab 1: 15-25, 2005.
75. Haffner SM. Abdominal adiposity and cardiometabolic risk: Do we 102. Kami K, Senba E. Localization of leukemia inhibitory factor and
have all the answers? Am J Med 120: S10-S16, 2007. interleukin-6 messenger ribonucleic acids in regenerating rat skeletal
76. Hamrick MW. A role for myokines in muscle-bone interactions. Exerc muscle. Muscle Nerve 21: 819-822, 1998.
Sport Sci Rev 39: 43-47, 2011. 103. Keane MP, Arenberg DA, Lynch JP, III, Whyte RI, Iannettoni MD, Bur-
77. Handschin C, Spiegelman BM. The role of exercise and PGC1[alpha] dick MD, Wilke CA, Morris SB, Glass MC, DiGiovine B, Kunkel SL,
in inflammation and chronic disease. Nature 454: 463-469, 2008. Strieter RM. The CXC chemokines, IL-8 and IP-10, regulate angiogenic
78. Hansen J, Brandt C, Nielsen AR, Hojman P, Whitham M, Febbraio activity in idiopathic pulmonary fibrosis. J Immunol 159: 1437-1443,
MA, Pedersen BK, Plomgaard P. Exercise induces a marked increase 1997.
in plasma follistatin: Evidence that follistatin is a contraction-induced 104. Keller C, Hellsten Y, Steensberg A, Pedersen BK. Differential regula-
hepatokine. Endocrinology 152: 164-171, 2011. tion of IL-6 and TNF-alpha via calcineurin in human skeletal muscle
79. Haugen F, Norheim F, Lian H, Wensaas AJ, Dueland S, Berg O, Fun- cells. Cytokine 36: 141-147, 2006.
derud A, Skalhegg BS, Raastad T, Drevon CA. IL-7 is expressed and 105. Keller C, Steensberg A, Hansen AK, Fischer CP, Plomgaard P, Pedersen
secreted by human skeletal muscle cells. Am J Physiol Cell Physiol BK. The effect of exercise, training, and glycogen availability on IL-
298: C807-C816, 2010. 6 receptor expression in human skeletal muscle. J Appl Physiol 99:
80. Heinrich PC, Behrmann I, Muller-Newen G, Schaper F, Graeve L. 2075-2079, 2005.
Interleukin-6-type cytokine signalling through the gp130/Jak/STAT 106. Keller C, Steensberg A, Pilegaard H, Osada T, Saltin B, Pedersen BK,
pathway. Biochem J 334: 297-314, 1998. Neufer PD. Transcriptional activation of the IL-6 gene in human con-
81. Hellsten Y, Frandsen U, Orthenblad N, Sjodin N, Richter EA. Xanthine tracting skeletal muscle: Influence of muscle glycogen content. FASEB
oxidase in human skeletal muscle following eccentric exercise: A role J 15: 2748-2750, 2001.
of inflammation. J Physiol (London) 498: 239-248, 1997. 107. Kelly M, Gauthier MS, Saha AK, Ruderman NB. Activation of AMP-
82. Henningsen J, Rigbolt KT, Blagoev B, Pedersen BK, Kratchmarova I. activated protein kinase (AMPK) by Interleukin-6 in rat skeletal muscle:
Dynamics of the skeletal muscle secretome during myoblast differen- Association with changes in cAMP, energy state, and endogenous fuel
tiation. Mol Cell Proteomics 9: 2482-96, 2010. mobilization. Diabetes 58: 1953-1960, 2009.
83. Henson DA, Nieman DC, Nehlsen-Cannarella SL, Fagoaga OR, Shan- 108. Kim GY, Lee JW, Ryu HC, Wei JD, Seong CM, Kim JH. Proinflam-
non M, Bolton MR, Davis JM, Gaffney CT, Kelln WJ, Austin MD, matory cytokine IL-1beta stimulates IL-8 synthesis in mast cells via a
Hjertman JM, Schilling BK. Influence of carbohydrate on cytokine leukotriene B4 receptor 2-linked pathway, contributing to angiogenesis.
and phagocytic responses to 2 h of rowing. Med Sci Sports Exerc 32: J Immunol 184: 3946-3954, 2010.
1384-1389, 2000. 109. Kishimoto T. Signal transduction through homo- or heterodimers of
84. Hilton DJ, Nicola NA, Metcalf D. Purification of a murine leukemia gp130. Stem Cells Dayt 12(Suppl 1): 37-44, 1994.
inhibitory factor from Krebs ascites cells. Anal Biochem 173: 359-367, 110. Kjaer M, Secher NH, Bangsbo J, Perko G, Horn A, Mohr T, Galbo
1988. H. Hormonal and metabolic responses to electrically induced cycling

1358 Volume 3, July 2013


P1: OTA/XYZ P2: ABC
JWBT335-c120033 JWBT335/Comprehensive Physiology June 8, 2013 8:4 Printer Name: Yet to Come

Comprehensive Physiology Muscle as a Secretory Organ

during epidural anesthesia in humans. J Appl Physiol 80: 2156-2162, 135. Mizuhara H, O’Neill E, Seki N, Ogawa T, Kusunoki C, Otsuka K,
1996. Satoh S, Niwa M, Senoh H, Fujiwara H. T cell activation-associated
111. Komulainen P, Pedersen M, Hanninen T, Bruunsgaard H, Lakka TA, hepatic injury: Mediation by tumor necrosis factors and protection by
Kivipelto M, Hassinen M, Rauramaa TH, Pedersen BK, Rauramaa R. interleukin 6. J Exp Med 179: 1529-1537, 1994.
BDNF is a novel marker of cognitive function in ageing women: The 136. Mohr T, Andersen JL, Biering-Sorensen F, Galbo H, Bangsbo J, Wag-
DR’s EXTRA Study. Neurobiol Learn Mem 90: 596-603, 2008. ner A, Kjaer M. Long-term adaptation to electrically induced cycle
112. Kurek JB, Bower J, Romanella M, Austin L. Leukaemia inhibitory training in severe spinal cord injured individuals. Spinal Cord 35: 1-16,
factor treatment stimulates muscle regeneration in the mdx mouse. 1997.
Neurosci Lett 212: 167-170, 1996. 137. Monninkhof EM, Elias SG, Vlems FA, van dT, I, Schuit AJ, Voskuil
113. Lafreniere JF, Mills P, Bouchentouf M, Tremblay JP. Interleukin-4 DW, van Leeuwen FE. Physical activity and breast cancer: A systematic
improves the migration of human myogenic precursor cells in vitro and review. Epidemiology 18: 137-157, 2007.
in vivo. Exp Cell Res 312: 1127-1141, 2006. 138. Mousavi K, Jasmin BJ. BDNF is expressed in skeletal muscle satellite
114. Lancaster GI, Jentjens RL, Moseley L, Jeukendrup AE, Gleeson M. cells and inhibits myogenic differentiation. J Neurosci 26: 5739-5749,
Effect of pre-exercise carbohydrate ingestion on plasma cytokine, stress 2006.
hormone, and neutrophil degranulation responses to continuous, high- 139. Mucci P, Durand F, Lebel B, Bousquet J, Prefaut C. Interleukins 1-beta,
intensity exercise. Int J Sport Nutr Exerc Metab 13: 436-453, 2003. -8, and histamine increases in highly trained, exercising athletes. Med
115. Li A, Dubey S, Varney ML, Dave BJ, Singh RK. IL-8 directly enhanced Sci Sports Exerc 32: 1094-1100, 2000.
endothelial cell survival, proliferation, and matrix metalloproteinases 140. Nehlsen Cannarella SL, Fagoaga OR, Nieman DC, Henson DA, But-
production and regulated angiogenesis. J Immunol 170: 3369-3376, terworth DE, Schmitt RL, Bailey EM, Warren BJ, Utter A, Davis JM.
2003. Carbohydrate and the cytokine response to 2.5 h of running. J Appl
116. Li TL, Gleeson M. The effect of single and repeated bouts of prolonged Physiol 82: 1662-1667, 1997.
cycling on leukocyte redistribution, neutrophil degranulation, IL-6, and 141. Nielsen AR, Hojman P, Erikstrup C, Fischer CP, Plomgaard P, Mounier
plasma stress hormone responses. Int J Sport Nutr Exerc Metab 14: R, Mortensen OH, Broholm C, Taudorf S, Krogh-Madsen R, Linde-
501-516, 2004. gaard B, Petersen AM, Gehl J, Pedersen BK. Association between
117. Li TL, Gleeson M. The effects of carbohydrate supplementation dur- IL-15 and obesity: IL-15 as a potential regulator of fat mass. J Clin
ing the second of two prolonged cycling bouts on immunoendocrine Endocrinol Metab 93: 4486-93, 2008.
responses. Eur J Appl Physiol 95: 391-399, 2005. 142. Nielsen AR, Mounier R, Plomgaard P, Mortensen OH, Penkowa M,
118. Li TL, Wu CL, Gleeson M, Williams C. The effects of pre-exercise Speerschneider T, Pilegaard H, Pedersen BK. Expression of interleukin-
high carbohydrate meals with different glycemic indices on blood 15 in human skeletal muscle effect of exercise and muscle fibre type
leukocyte redistribution, IL-6, and hormonal responses during a subse- composition. J Physiol 584: 305-312, 2007.
quent prolonged exercise. Int J Sport Nutr Exerc Metab 14: 647-656, 143. Nielsen AR, Pedersen BK. The biological roles of exercise-induced
2004. cytokines: IL-6, IL-8, and IL-15. Appl Physiol Nutr Metab 32: 833-
119. Lin J, Arnold HB, la-Fera MA, Azain MJ, Hartzell DL, Baile CA. 839, 2007.
Myostatin knockout in mice increases myogenesis and decreases adi- 144. Nielsen S, Pedersen BK. Skeletal muscle as an immunogenic organ.
pogenesis. Biochem Biophys Res Commun 291: 701-706, 2002. Curr Opin Pharmacol 8: 346-351, 2008.
120. Lin J, Handschin C, Spiegelman BM. Metabolic control through the 145. Nieman DC. Current perspective on exercise immunology. Curr Sports
PGC-1 family of transcription coactivators. Cell Metabolism 1: 361- Med Rep 2: 239-242, 2003.
370, 2005. 146. Nieman DC, Davis JM, Brown VA, Henson DA, Dumke CL, Utter AC,
121. Lin WW, Karin M. A cytokine-mediated link between innate immunity, Vinci DM, Downs MF, Smith JC, Carson J, Brown A, McAnulty SR,
inflammation, and cancer. J Clin Invest 117: 1175-1183, 2007. McAnulty LS. Influence of carbohydrate ingestion on immune changes
122. Lira SA, Zalamea P, Heinrich JN, Fuentes ME, Carrasco D, Lewin after 2 h of intensive resistance training. J Appl Physiol 96: 1292-1298,
AC, Barton DS, Durham S, Bravo R. Expression of the chemokine 2004.
N51/KC in the thymus and epidermis of transgenic mice results in 147. Nieman DC, Davis JM, Henson DA, Gross SJ, Dumke CL, Ut-
marked infiltration of a single class of inflammatory cells. J Exp Med ter AC, Vinci DM, Carson JA, Brown A, McAnulty SR, McAnulty
180: 2039-2048, 1994. LS, Triplett NT. Muscle cytokine mRNA changes after 2.5 h of cy-
123. Lyngso D, Simonsen L, Bulow J. Interleukin-6 production in human cling: Influence of carbohydrate. Med Sci Sports Exerc 37: 1283-1290,
subcutaneous abdominal adipose tissue: The effect of exercise. J Phys- 2005.
iol 543: 373-378, 2002. 148. Nieman DC, Davis JM, Henson DA, Walberg-Rankin J, Shute M,
124. Ma Q. Beneficial effects of moderate voluntary physical exercise and Dumke CL, Utter AC, Vinci DM, Carson JA, Brown A, Lee WJ, McAn-
its biological mechanisms on brain health. Neurosci Bull 24: 265-270, ulty SR, McAnulty LS. Carbohydrate ingestion influences skeletal mus-
2008. cle cytokine mRNA and plasma cytokine levels after a 3-h run. J Appl
125. MacIntyre DL, Sorichter S, Mair J, Berg A, McKenzie DC. Markers of Physiol 94: 1917-1925, 2003.
inflammation and myofibrillar proteins following eccentric exercise in 149. Nieman DC, Henson DA, McAnulty SR, McAnulty L, Swick NS, Utter
humans. Eur J Appl Physiol 84: 180-186, 2001. AC, Vinci DM, Opiela SJ, Morrow JD. Influence of vitamin C supple-
126. Mathur N, Pedersen BK. Exercise as a mean to control low-grade mentation on oxidative and immune changes after an ultramarathon.
systemic inflammation. Mediators Inflamm 2008: 109502, 2008. J Appl Physiol 92: 1970-1977, 2002.
127. Matter CM, Handschin C. RANTES (regulated on activation, normal T 150. Nieman DC, Henson DA, Smith LL, Utter AC, Vinci DM, Davis JM,
cell expressed and secreted), inflammation, obesity, and the metabolic Kaminsky DE, Shute M. Cytokine changes after a marathon race.
syndrome. Circulation 115: 946-948, 2007. J Appl Physiol 91: 109-114, 2001.
128. Matthews VB, Astrom MB, Chan MH, Bruce CR, Krabbe KS, 151. Nieman DC, Nehlsen-Canarella SL, Fagoaga OR, Henson DA, Utter
Prelovsek O, Akerstrom T, Yfanti C, Broholm C, Mortensen OH, A, Davis JM, Williams F, Butterworth DE. Influence of mode and
Penkowa M, Hojman P, Zankari A, Watt MJ, Bruunsgaard H, Ped- carbohydrate on the cytokine response to heavy exertion. Med Sci Sports
ersen BK, Febbraio MA. Brain-derived neurotrophic factor is produced Exerc 30: 671-678, 1998.
by skeletal muscle cells in response to contraction and enhances fat 152. Nocon M, Hiemann T, Muller-Riemenschneider F, Thalau F, Roll S,
oxidation via activation of AMP-activated protein kinase. Diabetologia Willich SN. Association of physical activity with all-cause and car-
52: 1409-1418, 2009. diovascular mortality: A systematic review and meta-analysis. Eur J
129. McFarlane C, Hui GZ, Amanda WZ, Lau HY, Lokireddy S, Xiaojia Cardiovasc Prev Rehabil 15: 239-246, 2008.
G, Mouly V, Butler-Browne G, Gluckman PD, Sharma M, Kambadur 153. Norheim F, Raastad T, Thiede B, Rustan AC, Drevon CA, Haugen
R. Human myostatin negatively regulates human myoblast growth and F. Proteomic identification of secreted proteins from human skeletal
differentiation. Am J Physiol Cell Physiol 301: C195-C203, 2011. muscle cells and expression in response to strength training. Am J
130. McPherron AC, Lawler AM, Lee SJ. Regulation of skeletal muscle Physiol Endocrinol Metab 301: E1013-E1021, 2011.
mass in mice by a new TGF-beta superfamily member. Nature 387: 154. Nosaka K, Clarkson PM. Changes in indicators of inflammation after
83-90, 1997. eccentric exercise of the elbow flexors. Med Sci Sports Exerc 28: 953-
131. McPherron AC, Lee SJ. Suppression of body fat accumulation in 961, 1996.
myostatin-deficient mice. J Clin Invest 109: 595-601, 2002. 155. Olsen RH, Krogh-Madsen R, Thomsen C, Booth FW, Pedersen BK.
132. Metcalf D. The unsolved enigmas of leukemia inhibitory factor. Stem Metabolic responses to reduced daily steps in healthy nonexercising
Cells 21: 5-14, 2003. men. JAMA 299: 1261-1263, 2008.
133. Minokoshi Y, Kim YB, Peroni OD, Fryer LG, Muller C, Carling D, 156. Oshima Y, Ouchi N, Sato K, Izumiya Y, Pimentel DR, Walsh K.
Kahn BB. Leptin stimulates fatty-acid oxidation by activating AMP- Follistatin-like 1 is an Akt-regulated cardioprotective factor that is se-
activated protein kinase. Nature 415: 339-343, 2002. creted by the heart. Circulation 117: 3099-3108, 2008.
134. Miura P, Amirouche A, Clow C, Belanger G, Jasmin BJ. Brain-derived 157. Ostrowski K, Hermann C, Bangash A, Schjerling P, Nielsen JN, Ped-
neurotrophic factor expression is repressed during myogenic differen- ersen BK. A trauma-like elevation of plasma cytokines in humans in
tiation by miR-206. J Neurochem 120: 230-238, 2012. response to treadmill running. J Physiol 513: 889-894, 1998.

Volume 3, July 2013 1359


P1: OTA/XYZ P2: ABC
JWBT335-c120033 JWBT335/Comprehensive Physiology June 8, 2013 8:4 Printer Name: Yet to Come

Muscle as a Secretory Organ Comprehensive Physiology

158. Ostrowski K, Rohde T, Asp S, Schjerling P, Pedersen BK. Chemokines expression is linked to muscle derived IL-6 expression. J Physiol 589:
are elevated in plasma after strenuous exercise in humans. Eur J Appl 1409-1420, 2011.
Physiol 84: 244-245, 2001. 186. Pelletier M, Montplaisir S, Dardenne M, Bach JF. Thymic hormone
159. Ouchi N, Oshima Y, Ohashi K, Higuchi A, Ikegami C, Izumiya Y, activity and spontaneous autoimmunity in dwarf mice and their litter-
Walsh K. Follistatin-like 1, a secreted muscle protein, promotes en- mates. Immunology 30: 783-788, 1976.
dothelial cell function and revascularization in ischemic tissue through 187. Petersen AM, Pedersen BK. The anti-inflammatory effect of exercise.
a nitric-oxide synthase-dependent mechanism. J Biol Chem 283: 32802- J Appl Physiol 98: 1154-1162, 2005.
32811, 2008. 188. Petersen AM, Pedersen BK. The role of IL-6 in mediating the anti-
160. Paffenbarger RS, Jr., Hyde RT, Wing AL, Hsieh CC. Physical activity, inflammatory effects of exercise. J Physiol Pharmacol 57(Suppl 10):
all-cause mortality, and longevity of college alumni. N Engl J Med 314: 43-51, 2006.
605-613, 1986. 189. Petersen EW, Carey AL, Sacchetti M., Steinberg GR, Macaulay SL,
161. Paffenbarger RS, Jr., Lee IM, Leung R. Physical activity and personal Febbraio M.A., Pedersen BK. Acute IL-6 treatment increases fatty
characteristics associated with depression and suicide in American col- acid turnover in elderly humans in vivo and in tissue culture in vitro:
lege men. Acta Psychiatr Scand Suppl 377: 16-22, 1994. Evidence that IL-6 acts independently of lipolytic hormones. Am J
162. Pedersen BK. Special feature for the Olympics: Effects of exercise on Physiol 288: E155-E162, 2005.
the immune system: Exercise and cytokines. Immunol Cell Biol 78: 190. Phillips SM, Green HJ, Tarnopolsky MA, Heigenhauser GF, Hill RE,
532-535, 2000. Grant SM. Effects of training duration on substrate turnover and oxi-
163. Pedersen BK. The anti-inflammatory effect of exercise: Its role in dia- dation during exercise. J Appl Physiol 81: 2182-2191, 1996.
betes and cardiovascular disease control. Essays Biochem 42: 105-117, 191. Pischon T, Boeing H, Hoffmann K, Bergmann M, Schulze MB, Over-
2006. vad K, van der Schouw YT, Spencer E, Moons KGM, Tjonneland A,
164. Pedersen BK. IL-6 signalling in exercise and disease. Biochem Soc Halkjaer J, Jensen MK, Stegger J, Clavel-Chapelon F, Boutron-Ruault
Trans 35: 1295-1297, 2007. MC, Chajes V, Linseisen J, Kaaks R, Trichopoulou A, Trichopoulos D,
165. Pedersen BK. The diseasome of physical inactivity and the role Bamia C, Sieri S, Palli D, Tumino R, Vineis P, Panico S, Peeters PHM,
of myokines in muscle-fat cross talk. J Physiol 587: 5559-5568, May AM, Bueno-De-Mesquita HB, van Duijnhoven FJB, Hallmans G,
2009. Weinehall L, Manjer J, Hedblad B, Lund E, Agudo A, Arriola L, Barri-
166. Pedersen BK. Exercise-induced myokines and their role in chronic carte A, Navarro C, Martinez C, Quiros JR, Key T, Bingham S, Khaw
diseases. Brain Behav Immun 25: 811-816, 2011. KT, Boffetta P, Jenab M, Ferrari P, Riboli E. General and abdominal
167. Pedersen BK. Muscles and their myokines. J Exp Biol 214: 337-346, adiposity and risk of death in Europe. New Engl J Med 359: 2105-2120,
2011. 2008.
168. Pedersen BK. A muscular twist on the fate of fat. N Engl J Med 366: 192. Plomgaard P, Fischer CP, Ibfelt T, Pedersen BK, van HG. TNF-alpha
1544-1545, 2012. modulates human in vivo lipolysis. J Clin Endocrinol Metab 93: 543-9,
169. Pedersen BK. Muscular IL-6 and Its Role as an Energy Sensor. Med 2007.
Sci Sports Exerc 44: 392-396, 2012. 193. Plomgaard P, Keller P, Keller C, Pedersen BK. TNF-alpha, but not
170. Pedersen BK, Akerstrom TC, Nielsen AR, Fischer CP. Role of IL-6, stimulates plasminogen activator inhibitor-1 expression in hu-
myokines in exercise and metabolism. J Appl Physiol 103(3): 1093- man subcutaneous adipose tissue. J Appl Physiol 98: 2019-2023,
1090, 2007. 2005.
171. Pedersen BK, Febbraio M. Muscle-derived interleukin-6: A possible 194. Potthoff MJ, Inagaki T, Satapati S, Ding X, He T, Goetz R, Mo-
link between skeletal muscle, adipose tissue, liver, and brain. Brain hammadi M, Finck BN, Mangelsdorf DJ, Kliewer SA, Burgess SC.
Behav Immun 19: 371-376, 2005. FGF21 induces PGC-1alpha and regulates carbohydrate and fatty acid
172. Pedersen BK, Febbraio MA. Muscle, exercise and obesity: Skeletal metabolism during the adaptive starvation response. Proc Natl Acad
muscle as a secretory organ. Nature Reviews Endocrinology 3: 457- Sci U S A 106: 10853-10858, 2009.
465, 2012. 195. Quinn LS, Anderson BG. Interleukin-15, IL-15 receptor-alpha, and
173. Pedersen BK, Fischer CP. Beneficial health effects of exercise—the obesity: Concordance of laboratory animal and human genetic studies.
role of IL-6 as a myokine. Trends Pharmacol Sci 28: 152-156, J Obes 2011: 456347, 2011.
2007. 196. Quinn LS, Anderson BG, Drivdahl RH, Alvarez B, Argiles JM. Over-
174. Pedersen BK, Fischer CP. Physiological roles of muscle-derived expression of interleukin-15 induces skeletal muscle hypertrophy in
interleukin-6 in response to exercise. Curr Opin Clin Nutr Metab Care vitro: Implications for treatment of muscle wasting disorders. Exp Cell
10: 265-271, 2007. Res 280: 55-63, 2002.
175. Pedersen BK, Febbraio MA. Muscle as an endocrine organ: Fo- 197. Quinn LS, Anderson BG, Strait-Bodey L, Stroud AM, Argiles JM.
cus on muscle-derived interleukin-6. Physiol Rev 88: 1379-1406, Oversecretion of interleukin-15 from skeletal muscle reduces adiposity.
2008. Am J Physiol Endocrinol Metab 296: E191-E202, 2009.
176. Pedersen BK, Hoffman-Goetz L. Exercise and the immune system: 198. Quinn LS, Haugk KL, Damon SE. Interleukin-15 stimulates C2 skeletal
Regulation, integration and adaptation. Physiol Rev 80: 1055-1081, myoblast differentiation. Biochem Biophys Res Commun 239: 6-10,
2000. 1997.
177. Pedersen BK, Pedersen M, Krabbe KS, Bruunsgaard H, Matthews VB, 199. Quinn LS, Strait-Bodey L, Anderson BG, Argiles JM, Havel PJ.
Febbraio MA. Role of exercise-induced brain-derived neurotrophic fac- Interleukin-15 stimulates adiponectin secretion by 3T3-L1 adipocytes:
tor production in the regulation of energy homeostasis in mammals. Exp Evidence for a skeletal muscle-to-fat signaling pathway. Cell Biol Int
Physiol 94: 1153-60, 2009. 29: 449-457, 2005.
178. Pedersen BK, Steensberg A, Fischer C, Keller C, Keller P, Plomgaard 200. Richardson LC, Pollack LA. Therapy insight: Influence of type 2 dia-
P, Febbraio M, Saltin B. Searching for the exercise factor: Is IL-6 a betes on the development, treatment and outcomes of cancer. Nat Clin
candidate? J Muscle Res Cell Motil 24: 113-119, 2003. Pract Oncol 2: 48-53, 2005.
179. Pedersen BK, Steensberg A, Fischer C, Keller C, Keller P, Plomgaard 201. Riechman SE, Balasekaran G, Roth SM, Ferrell RE. Association of
P, Wolsk-Petersen E, Febbraio M. The metabolic role of IL-6 produced interleukin-15 protein and interleukin-15 receptor genetic variation
during exercise: Is IL-6 an exercise factor? Proc Nutr Soc 63: 263-267, with resistance exercise training responses. J Appl Physiol 97: 2214-
2004. 2219, 2004.
180. Pedersen BK, Steensberg A, Keller P, Keller C, Fischer C, Hiscock 202. Rodgers BD, Garikipati DK. Clinical, agricultural, and evolutionary
N, Hall Gv, Plomgaard P, Febbraio MA. Muscle-derived interleukin-6: biology of myostatin: A comparative review. Endocr Rev 29: 513-534,
Lipolytic, anti-inflammatory and immune regulatory effects. Pflugers 2008.
Arch 446: 9-16, 2003. 203. Rosendal L, Sogaard K, Kjaer M, Sjogaard G, Langberg H, Kristiansen
181. Pedersen BK, Steensberg A, Schjerling P. Exercise and interleukin-6. J. Increase in interstitial interleukin-6 of human skeletal muscle with
Curr Opin Hematol 8: 137-141, 2001. repetitive low-force exercise. J Appl Physiol 98: 477-481, 2005.
182. Pedersen BK, Steensberg A, Schjerling P. Muscle-derived interleukin- 204. Rovio S, Kareholt I, Helkala EL, Viitanen M, Winblad B, Tuomilehto
6: Possible biological effects. J Physiol (London) 536: 329-337, J, Soininen H, Nissinen A, Kivipelto M. Leisure-time physical activity
2001. at midlife and the risk of dementia and Alzheimer’s disease. Lancet
183. Pedersen BK, Tvede N, Hansen FR, Andersen V, Bendix T, Bendixen Neurol 4: 705-711, 2005.
G, Bendtzen K, Galbo H, Haahr PM, Klarlund K and et al. Modulation 205. Rubio N, Sanz-Rodriguez F. Induction of the CXCL1 (KC) chemokine
of natural killer cell activity in peripheral blood by physical exercise. in mouse astrocytes by infection with the murine encephalomyelitis
Scand J Immunol 27: 673-678, 1988. virus of Theiler. Virology 358: 98-108, 2007.
184. Pedersen BK, Febbraio MA, Mooney RA. Interleukin-6 does/does not 206. Ruderman NB, Keller C, Richard AM, Saha AK, Luo Z, Xiang X,
have a beneficial role in insulin sensitivity and glucose homeostasis. Giralt M, Ritov VB, Menshikova EV, Kelley DE, Hidalgo J, Pedersen
J Appl Physiol 102: 814-816, 2007. BK, Kelly M. Interleukin-6 regulation of AMP-activated protein kinase:
185. Pedersen L, Pilegaard H, Hansen J, Brandt C, Adser H, Hidalgo J, Potential role in the systemic response to exercise and prevention of the
Olesen J, Pedersen BK, Hojman P. Exercise-induced liver CXCL-1 metabolic syndrome. Diabetes 55(Suppl 2): S48-S54, 2006.

1360 Volume 3, July 2013


P1: OTA/XYZ P2: ABC
JWBT335-c120033 JWBT335/Comprehensive Physiology June 8, 2013 8:4 Printer Name: Yet to Come

Comprehensive Physiology Muscle as a Secretory Organ

207. Rundqvist H, Rullman E, Sundberg CJ, Fischer H, Eisleitner K, istration does not impair muscle glucose uptake or whole body glucose
Stahlberg M, Sundblad P, Jansson E, Gustafsson T. Activation of the disposal in healthy humans. J Physiol 548: 631-638, 2003.
erythropoietin receptor in human skeletal muscle. Eur J Endocrinol 231. Steensberg A, Keller C, Starkie RL, Osada T, Febbraio MA, Pedersen
161: 427-434, 2009. BK. IL-6 and TNF-alpha expression in, and release from, contracting
208. Sakuma K, Watanabe K, Sano M, Uramoto I, Totsuka T. Differential human skeletal muscle. Am J Physiol Endocrinol Metab 283: E1272-
adaptation of growth and differentiation factor 8/myostatin, fibroblast E1278, 2002.
growth factor 6 and leukemia inhibitory factor in overloaded, regener- 232. Steensberg A, van HG, Osada T, Sacchetti M, Saltin B, Klarlund PB.
ating and denervated rat muscles. Biochim Biophys Acta 1497: 77-88, Production of interleukin-6 in contracting human skeletal muscles can
2000. account for the exercise-induced increase in plasma interleukin-6. J
209. Sakuma K, Watanabe K, Sano M, Uramoto I, Totsuka T. Postnatal Physiol 529(Pt 1): 237-242, 2000.
profiles of myogenic regulatory factors and the receptors of TGF-beta 233. Steinberg GR, Rush JW, Dyck DJ. AMPK expression and phosphory-
2, LIF and IGF-I in the gastrocnemius and rectus femoris muscles of lation are increased in rodent muscle after chronic leptin treatment. Am
dy mouse. Acta Neuropathol 99: 169-176, 2000. J Physiol Endocrinol Metab 284: E648-E654, 2003.
210. Sakuma K, Yamaguchi A. The recent understanding of the neu- 234. Steinberg GR, Watt MJ, Febbraio MA. Cytokine regulation of AMPK
rotrophin’s role in skeletal muscle adaptation. J Biomed Biotechnol signalling. Front Biosci 14: 1902-1916, 2009.
2011: 201696, 2011. 235. Sun L, Ma K, Wang H, Xiao F, Gao Y, Zhang W, Wang K, Gao X, Ip
211. Scheele C, Nielsen S, Kelly M, Broholm C, Nielsen AR, Taudorf S, N, Wu Z. JAK1-STAT1-STAT3, a key pathway promoting proliferation
Pedersen M, Fischer CP, Pedersen BK. Satellite cells derived from and preventing premature differentiation of myoblasts. J Cell Biol 179:
obese humans with type 2 diabetes and differentiated into myocytes 129-138, 2007.
in vitro exhibit abnormal response to IL-6. PLoS One 7: e39657, 236. Suzuki K, Nakaji S, Yamada M, Liu Q, Kurakake S, Okamura N, Kumae
2012. T, Umeda T, Sugawara K. Impact of a competitive marathon race on
212. Scheele C, Nielsen S, Pedersen BK. ROS and myokines promote systemic cytokine and neutrophil responses. Med Sci Sports Exerc 35:
muscle adaptation to exercise. Trends Endocrinol Metab 20: 95-99, 348-355, 2003.
2009. 237. Tseng YL, Wu MH, Yang HC, Wang CY, Lin CF. Autocrine IL-6
213. Scherer PE. Adipose tissue: From lipid storage compartment to en- regulates GRO-alpha production in thymic epithelial cells. Cytokine
docrine organ. Diabetes 55: 1537-1545, 2006. 51: 195-201, 2010.
214. Schindler R, Mancilla J, Endres S, Ghorbani R, Clark SC, Dinarello 238. Tuomilehto J, Lindstrom J, Eriksson JG, Valle TT, Hamalainen H,
CA. Correlations and interactions in the production of interleukin- Ilanne-Parikka P, Keinanen-Kiukaanniemi S, Laakso M, Louheranta
6 (IL-6), IL-1, and tumor necrosis factor (TNF) in human blood A, Rastas M, Salminen V, Uusitupa M. Prevention of type 2 diabetes
mononuclear cells: IL-6 suppresses IL-1 and TNF. Blood 75: 40-47, mellitus by changes in lifestyle among subjects with impaired glucose
1990. tolerance. N Engl J Med 344: 1343-1350, 2001.
215. Schmelzer CH, Burton LE, Tamony CM. Purification and partial char- 239. Ullum H, Haahr PM, Diamant M, Palmo J, Halkjaer Kristensen J,
acterization of recombinant human differentiation-stimulating factor. Pedersen BK. Bicycle exercise enhances plasma IL-6 but does not
Protein Expr Purif 1: 54-62, 1990. change IL-1alpha, IL-1beta, IL-6, or TNF-alpha pre-mRNA in BMNC.
216. Seidl K, Erck C, Buchberger A. Evidence for the participation of nerve J Appl Physiol 77: 93-7., 1994.
growth factor and its low-affinity receptor (p75NTR) in the regulation 240. van der, V, Janssen TW. The potential anti-inflammatory effect of
of the myogenic program. J Cell Physiol 176: 10-21, 1998. exercise in chronic obstructive pulmonary disease. Respiration 79: 160-
217. Serrano AL, Baeza-Raja B, Perdiguero E, Jardi M, Munoz-Canoves P. 174, 2010.
Interleukin-6 is an essential regulator of satellite cell-mediated skeletal 241. van Hall G, Steensberg A, Sacchetti M, Fischer C, Keller C, Schjer-
muscle hypertrophy. Cell Metab 7: 33-44, 2008. ling P, Hiscock N, Moller K, Saltin B, Febbraio MA, Pedersen BK.
218. Shetty S, Kusminski CM, Scherer PE. Adiponectin in health and dis- Interleukin-6 stimulates lipolysis and fat oxidation in humans. J Clin
ease: Evaluation of adiponectin-targeted drug development strategies. Endocrinol Metab 88: 3005-3010, 2003.
Trends Pharmacol Sci 30: 234-239, 2009. 242. Wagers AJ, Conboy IM. Cellular and molecular signatures of muscle
219. Shimano M, Ouchi N, Nakamura K, van WB, Ohashi K, Asaumi Y, regeneration: Current concepts and controversies in adult myogenesis.
Higuchi A, Pimentel DR, Sam F, Murohara T, van den Hoff MJ, Walsh Cell 122: 659-667, 2005.
K. Cardiac myocyte follistatin-like 1 functions to attenuate hypertrophy 243. Wallenius V, Wallenius K, Ahren B, Rudling M, Carlsten H, Dick-
following pressure overload. Proc Natl Acad Sci U S A 108: E899-E906, son SL, Ohlsson C, Jansson JO. Interleukin-6-deficient mice develop
2011. mature-onset obesity. Nat Med 8: 75-79, 2002.
220. Smith PE. Effect of hypophysectomy on the involution of the thymus 244. Walsh K. Adipokines, myokines and cardiovascular disease. Circ J 73:
in the rat. Anat Rec 47: 119-129, 1930. 13-18, 2009.
221. Spangelo BL, Gorospe WC. Role of the cytokines in the 245. Walsh NP, Gleeson M, Shephard RJ, Gleeson M, Woods JA,
neuroendocrine-immune system axis. Front Neuroendocrinol 16: 1-22, Bishop NC, Fleshner M, Green C, Pedersen BK, Hoffman-Goetz L,
1995. Rogers CJ, Northoff H, Abbasi A, Simon P. Position statement. Part
222. Spangenburg EE, Booth FW. Multiple signaling pathways mediate LIF- one: Immune function and exercise. Exerc Immunol Rev 17: 6-63,
induced skeletal muscle satellite cell proliferation. Am J Physiol Cell 2011.
Physiol 283: C204-C211, 2002. 246. Watt MJ, Dzamko N, Thomas WG, Rose-John S, Ernst M, Carling
223. Spangenburg EE, Booth FW. Leukemia inhibitory factor restores the D, Kemp BE, Febbraio MA, Steinberg GR. CNTF reverses obesity-
hypertrophic response to increased loading in the LIF(-/-) mouse. Cy- induced insulin resistance by activating skeletal muscle AMPK. Nat
tokine 34: 125-130, 2006. Med 12: 541-548, 2006.
224. Starkie R, Ostrowski SR, Jauffred S, Febbraio M, Pedersen BK. Exer- 247. Wenz T, Rossi SG, Rotundo RL, Spiegelman BM, Moraes CT. In-
cise and IL-6 infusion inhibit endotoxin-induced TNF-alpha production creased muscle PGC-1alpha expression protects from sarcopenia and
in humans. FASEB J 17: 884-886, 2003. metabolic disease during aging. Proc Natl Acad Sci U S A 106: 20405-
225. Starkie RL, Angus DJ, Rolland J, Hargreaves M, Febbraio M. Effect of 20410, 2009.
prolonged submaximal exercise and carbohydrate ingestion on mono- 248. Whitmer RA, Gustafson DR, Barrett-Connor E, Haan MN, Gunderson
cyte intracellular cytokine production in humans. J Physiol (London) EP, Yaffe K. Central obesity and increased risk of dementia more than
528: 647-655, 2000. three decades later. Neurology 71: 1057-1064, 2008.
226. Starkie RL, Arkinstall MJ, Koukoulas I, Hawley JA, Febbraio MA. 249. Willoughby DS, McFarlin B, Bois C. Interleukin-6 expression after
Carbohydrate ingestion attenuates the increase in plasma interleukin-6, repeated bouts of eccentric exercise. Int J Sports Med 24: 15-21,
but not skeletal muscle interleukin-6 mRNA, during exercise in humans. 2003.
J Physiol (London) 533: 585-591, 2001. 250. Wolin KY, Yan Y, Colditz GA, Lee IM. Physical activity and colon
227. Starkie RL, Rolland J, Angus DJ, Anderson MJ, Febbraio MA. Circu- cancer prevention: A meta-analysis. Br J Cancer 100: 611-616,
lating monocyes are not the source of elevations in plasma IL-6 and 2009.
TNF-alpha levels after prolonged running. Am J Physiol Cell Physiol 251. Wolsk E, Mygind H, Grondahl TS, Pedersen BK, van HG. IL-6 se-
280: C769-C774, 2001. lectively stimulates fat metabolism in human skeletal muscle. Am J
228. Steensberg A, Febbraio MA, Osada T, Schjerling P, van HG, Saltin B, Physiol Endocrinol Metab 299: E832-E840, 2010.
Pedersen BK. Interleukin-6 production in contracting human skeletal 252. Wu J, Bostrom P, Sparks LM, Ye L, Choi JH, Giang AH, Khandekar
muscle is influenced by pre-exercise muscle glycogen content. J Physiol M, Virtanen KA, Nuutila P, Schaart G, Huang K, Tu H, van Marken
537: 633-639, 2001. Lichtenbelt WD, Hoeks J, Enerback S, Schrauwen P, Spiegelman BM.
229. Steensberg A, Fischer CP, Keller C, Moller K, Pedersen BK. IL-6 Beige adipocytes are a distinct type of thermogenic fat cell in mouse
enhances plasma IL-1ra, IL-10, and cortisol in humans. Am J Physiol and human. Cell; 150: 366-376, 2012.
Endocrinol Metab 285: E433-E437, 2003. 253. Xue F, Michels KB. Diabetes, metabolic syndrome, and breast can-
230. Steensberg A, Fischer CP, Sacchetti M, Keller C, Osada T, Schjerling cer: A review of the current evidence. Am J Clin Nutr 86: s823-s835,
P, van Hall G, Febbraio MA, Pedersen BK. Acute interleukin-6 admin- 2007.

Volume 3, July 2013 1361


P1: OTA/XYZ P2: ABC
JWBT335-c120033 JWBT335/Comprehensive Physiology June 8, 2013 8:4 Printer Name: Yet to Come

Muscle as a Secretory Organ Comprehensive Physiology

254. Yoon JH, Yea K, Kim J, Choi YS, Park S, Lee H, Lee CS, Suh PG, 257. Yudkin JS, Eringa E, Stehouwer CD. “ Vasocrine” signalling from
Ryu SH. Comparative proteomic analysis of the insulin-induced L6 perivascular fat: A mechanism linking insulin resistance to vascular
myotube secretome. Proteomics 9: 51-60, 2009. disease. Lancet 365: 1817-1820, 2005.
255. You T, Nicklas BJ. Effects of exercise on adipokines and the metabolic 258. Zhang Y, Proenca R, Maffei M, Barone M, Leopold L, Friedman JM.
syndrome. Curr Diab Rep 8: 7-11, 2008. Positional cloning of the mouse obese gene and its human homologue.
256. Yudkin JS. Inflammation, obesity, and the metabolic syndrome. Horm Nature 372: 425-432, 1994.
Metab Res 39: 707-709, 2007. 259. Zhao B, Wall RJ, Yang J. Transgenic expression of myostatin propeptide
prevents diet-induced obesity and insulin resistance. Biochem Biophys
Res Commun 337: 248-255, 2005.

1362 Volume 3, July 2013

You might also like