O Papel Imunomodulador Do Exercício Na Doença Metabólica

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 8

TREIMM-1081; No.

of Pages 8

Review

The immunomodulating role of


exercise in metabolic disease
Graeme I. Lancaster and Mark A. Febbraio
Cellular and Molecular Metabolism Laboratory, Baker IDI Heart and Diabetes Institute, Melbourne, Australia

A lack of physical activity is linked to the development of The past two decades have identified inflammation as a
many chronic diseases. It is now well established that key nexus between obesity and the development of chronic
the immune system and inflammation play a central role metabolic diseases [1]. A plethora of studies have described
in the development of numerous chronic metabolic dis- how inappropriate activation of components of the immune
eases including insulin resistance, type 2 diabetes, ath- system and induction of local inflammation in key cells and
erosclerosis, nonalcoholic fatty liver disease, and tissues underpins the development of chronic metabolic
specific types of cancer. Physical exercise elicits potent diseases. For example, in obesity the immune cell profile of
anti-inflammatory effects that are likely to account for the adipose tissue is substantially altered such that proin-
many of the salutary actions of regular exercise on flammatory macrophages, neutrophils, and CD8+ T lym-
chronic metabolic diseases. Here we review the anti- phocytes accumulate, while regulatory T cells and
inflammatory and immunomodulatory mechanisms by eosinophils decrease, and initiate a state of local inflam-
which the beneficial effects of exercise on chronic meta- mation that contributes to the development of systemic
bolic diseases may be mediated. insulin resistance. In cardiovascular disease, immune cells
accumulate within the arterial vasculature and contribute
Inflammation at the nexus of metabolic disease to the formation of atherosclerotic plaques [6–8].
The World Health Organization has estimated that in Exercise elicits potent and wide-ranging effects on the
excess of 1 billion people worldwide are overweight, with immune system [9,10], principle amongst these being its
300 million defined as clinically obese. Obesity is associat- anti-inflammatory effects. Although exercise can be proin-
ed with the development of a cluster of chronic metabolic flammatory and has, in some instances, been associated
diseases such as insulin resistance, type 2 diabetes, ath- with increased production of proinflammatory mediators
erosclerosis, nonalcoholic liver disease, hypertension, and such as tumor necrosis factor-a (TNF-a), interleukin-1b
some forms of cancer [1]. Physical inactivity is major (IL-1b), and C-reactive protein, this is typically only the
contributor to the development of chronic metabolic dis- case after extreme forms of exercise such as marathon
eases [2]. Furthermore, regular exercise prevents, or at running or Ironman triathlon, which are associated with
least delays, the progression of a host of metabolic diseases muscle damage and even systemic endotoxemia [11,12].
including cardiovascular disease, insulin resistance, type 2 However, in most instances exercise is anti-inflammatory
diabetes, and hypertension [2]. Indeed, it has been ob- in nature. The cytokine profile induced by exercise is
served that exercise as a therapeutic intervention can be classically anti-inflammatory, comprising marked
as effective as the medications that are prescribed for increases in the levels of several potent anti-inflammatory
chronic metabolic diseases [3,4]. Although some of the cytokines such as IL-10, IL-1 receptor antagonist (IL-1ra),
beneficial effects of exercise in chronic metabolic diseases and IL-6 [12]. An elegant and powerful demonstration of
are likely to be attributable to an increase in energy the anti-inflammatory effects of exercise is the observation
expenditure and consequently a reduction in the accumu- that prolonged cycling exercise reduced endotoxin-induced
lation of fat mass in individuals who consume calories in TNF-a production in vivo in humans [13]. Preventing or
excess of their daily requirements, numerous studies have attenuating inflammation is likely to be an important
demonstrated that the salutary actions of exercise are mechanism by which physical activity and exercise protect
independent of its effect on weight loss [5]. Exercise pro- against the development of chronic metabolic diseases
motes numerous molecular and cellular changes in several [3,14].
tissues and these adaptations are likely to underpin many Here we discuss some of the latest research on the anti-
of the salutary actions of exercise. inflammatory effects of exercise in the context of chronic
metabolic diseases (Figure 1), outline several novel hypoth-
Corresponding authors: Lancaster, G.I. (graeme.lancaster@bakeridi.edu.au);
Febbraio, M.A. (mark.febbraio@bakeridi.edu.au). eses, and highlight areas for further research. Specifically,
Keywords: exercise; muscle; immune cells; inflammation; metabolic disease; we discuss how exercise can impact on several tissues that
anti-inflammatory.
are deleteriously affected in metabolic disease, such as
1471-4906/$ – see front matter adipose tissue – a key site in the control of systemic
ß 2014 Elsevier Ltd. All rights reserved. http://dx.doi.org/10.1016/j.it.2014.02.008
metabolism – and liver and skeletal muscle. We also
discuss the role of exercise as a modulator of Toll-like
receptor (TLR) function and how this may counteract some
of the negative effects of obesity. Finally, we discuss the
Trends in Immunology xx (2014) 1–8 1
TREIMM-1081; No. of Pages 8

Review Trends in Immunology xxx xxxx, Vol. xxx, No. x

Adipose ssue size


Recruitment of M1 macrophages
and CD8+ T lymphocytes
TLR4 expression and acvaon

TLR4

MyD88
MyD88

IL-10

Monocyte/ IL-1ra
IL-6 macrophage

TLR4 Lipid accumulaon


MyD88
TLR4 expression and acvaon

TLR4
MyD88
MyD88
Lipid accumulaon
TLR4 expression and acvaon
TRENDS in Immunology

Figure 1. The anti-inflammatory effects of exercise in the context of obesity. As discussed in the text, exercise may mediate potent salutary actions in the context of
metabolic disease via its anti-inflammatory effects. Given the proinflammatory effects of several lipids, limiting lipid accumulation, either by limiting the expansion of
adipose tissue or by decreasing the accumulation of lipid in the skeletal muscle and liver, is likely to be a key anti-inflammatory effect of exercise. Furthermore, by inhibiting
the expansion of adipose tissue, exercise limits the recruitment of proinflammatory M1 macrophages and CD8+ T lymphocytes, cells known to promote insulin resistance. It
is well established that exercising muscle releases myokines. The release of interleukin-6 (IL-6) from skeletal muscle and subsequent production of IL-1 receptor antagonist
(IL-1ra) by monocytes and macrophages may represent an important anti-inflammatory action of exercise. Finally, exercise can decrease the expression and activation of
Toll-like receptor 4 (TLR4) in several tissue and cell types and this may contribute to exercise’s ability to protect against the deleterious effects of obesity.

role of IL-1b and the inflammasome in metabolic diseases profile [21]; however, in each instance adipose tissue
and suggest that a primary anti-inflammatory effect of expansion due to adipocyte hypertrophy is central. An
exercise may be via the inhibition of IL-1 signaling increase in adipose tissue mass is a hallmark of human
(Figure 1). obesity. A key beneficial effect of exercise is via its effects
on weight loss and/or the prevention of weight gain. Ac-
Obesity and adipose tissue-resident immune cells cordingly, by limiting the expansion of adipose tissue,
A hallmark of obesity is a shift in the immune cell profile of exercise would be predicted to ameliorate obesity-induced
the adipose tissue [8]. Thus, the proportion of resident M2 changes in the adipose tissue immune cell profile and,
anti-inflammatory, alternatively activated macrophages importantly, contribute to the maintenance of whole-body
[15], eosinophils [16], and CD4+ T lymphocytes [17] insulin sensitivity.
decreases and the proportions of M1 proinflammatory, clas- Several studies have shown that chronic exercise train-
sically activated macrophages [15], neutrophils [18], B lym- ing in mice fed a high-fat diet, which is associated with
phocytes [19] and CD8+ T lymphocytes [17] are increased. reductions in adipose tissue mass, results in a marked
Mechanistically, this change in the adipose tissue immune reduction in the levels of proinflammatory cytokines in
cell profile initiates a state of local inflammation within the adipose tissue compared with non-exercising controls [22–
adipose tissue that induces both local and systemic insulin 25]. Importantly, this reduction in adipose tissue inflam-
resistance. Remarkably, the targeted deletion of even a mation following exercise training is associated with a
single immune cell type through genetic or antibody-medi- substantial reduction in macrophage adipose tissue con-
ated approaches can protect against the development of tent [22–25], an effect that is probably attributable to a
insulin resistance in mice fed a high-fat diet [17–20]. It is decrease in the recruitment of M1 proinflammatory macro-
likely that the obesity-associated transition of the adipose phages [23,24]. Chronic exercise training was also associ-
tissue immune cell profile from principally anti-inflamma- ated with an increase in the expression of CD163, a marker
tory cell types toward proinflammatory cell types is a key of M2 macrophages [24]. Studies examining adipose tissue
event initiating the development of local and systemic inflammation in humans following exercise are limited, but
insulin resistance [8]. Several hypotheses have been pro- there is experimental support that a combination of diet
posed to explain the key events that lead to the shift in the and exercise reduces adipose tissue inflammation and
adipose tissue immune cell profile in obesity [8]. The death macrophage recruitment [26], although others report little
of adipocytes within the adipose tissue, adipose tissue effect of endurance training alone on adipose tissue inflam-
hypoxia, production of chemotactic factors, and increased mation [27]. Of note, however, in this latter study, endur-
free fatty acid (FFA) fluxes have all been proposed to ance exercise training increased the number of CD163+
regulate the obesity-associated shift in the immune cell cells within the adipose tissue, indicative of increased
2
TREIMM-1081; No. of Pages 8

Review Trends in Immunology xxx xxxx, Vol. xxx, No. x

numbers of M2 macrophages. CD8+ T lymphocytes accu- cise. Exercise training in mice fed a high-fat–high-fructose
mulate in the adipose tissue of high-fat diet-fed mice diet, which induces hepatosteatosis and nonalcoholic fatty
progressively over the course of the dietary period and liver disease, reduced hepatic inflammation as evidenced
CD8+ T lymphocytes contribute to obesity-induced inflam- by reduced macrophage infiltration and TNF-a expression
mation and insulin resistance [17]. In mice fed a high-fat [36].
diet for 16 weeks, exercise training (60 min/day, 5 days/ Skeletal muscle lipid accumulation is a central feature
week) reduced high feeding-induced increases in adipose of obesity, insulin resistance, and type 2 diabetes. Al-
tissue CD8+ T lymphocyte recruitment compared with non- though immune cells, most notably macrophages, have a
exercise trained control mice [23]. Collectively, these data well established role in obesity-induced inflammation in
argue that exercise training prevents the deleterious adipose tissue and liver [8], the contribution of macro-
changes in the adipose tissue immune cell profile that phages to skeletal muscle insulin resistance, or indeed
occur in obesity, attenuating the recruitment of M1 pro- whether obesity promotes macrophage accumulation in
inflammatory macrophages and CD8+ T lymphocytes and skeletal muscle, is unclear. However, two recent studies
increasing numbers of M2 macrophages. These effects are have examined the effect of obesity and type 2 diabetes on
most parsimoniously explained by the effects of exercise in the recruitment of macrophages into skeletal muscles.
preventing the expansion of adipose tissue. First, it was shown that overweight, type 2 diabetic
humans had significantly greater expression of CD11b
Exercise and lipid accumulation and inflammation in and CD11c (macrophage markers) in skeletal muscle biop-
liver and skeletal muscle sies compared with overweight, non-type 2 diabetics [37].
Hepatic lipid accumulation is a central feature of several Interestingly, obesity per se did not appear to promote
metabolic diseases. For example, excess hepatic lipid ac- macrophage recruitment because no differences were ob-
cumulation induces hepatic insulin resistance leading to served between overweight non-type 2 diabetics and lean
elevated hepatic glucose production and hyperglycemia non-type 2 diabetics [37]. However, in a second study the
and plays a key role in the development of nonalcoholic same group showed that mice fed a high-fat diet for 1 week
fatty liver disease. Although excess hepatic lipid accumu- had elevated F4/80 and CD11c expression in quadriceps
lation is likely to exert deleterious actions via multiple muscle, indicative of increased macrophage recruitment
mechanisms, increased liver inflammation is a hallmark of [38].
hepatosteatosis and has been mechanistically linked to the Although the role of immune cells in skeletal muscle
development of obesity-induced hepatocellular carcinoma insulin resistance is poorly defined, obesity induces the
[28], alcoholic liver disease [29], and obesity-induced insu- activation of c-Jun N-terminal kinase (JNK) in skeletal
lin resistance [30]. Specifically, obesity-induced hepatocel- muscle [39,40], which contributes to the development of
lular carcinoma is associated with marked macrophage skeletal muscle insulin resistance [40]. Given the impor-
and neutrophil recruitment into the liver and increased tant role of adipose tissue hypertrophy and hepatostea-
production of proinflammatory cytokines, such as TNF-a tosis in promoting adipose tissue and liver inflammation,
and IL-6, which are critical in promoting obesity-induced respectively, we hypothesize that the accumulation of
tumorigenesis [28]. Inflammation in alcoholic liver disease lipids is critical in mediating obesity-induced inflamma-
has been shown to be dependent on inflammasome activa- tion, and potentially macrophage recruitment, in skeletal
tion and IL-1b production in Kupffer cells (liver-resident muscle. Importantly, exercise training in obese patients
macrophages) [29]. Finally, in obesity-associated liver in- with type 2 diabetes increases mitochondrial content and
flammation, numerous proinflammatory cytokines are el- the capacity of skeletal muscle to oxidize fatty acids [41].
evated in the liver of mice fed a high-fat diet [31]. Furthermore, 8 weeks of aerobic exercise training (cycling
Activation of Kupffer cells is likely to be important in and running 5 days/week) was recently shown to increase
the pathogenesis of obesity-associated insulin resistance the expression of lipolytic proteins concomitant with a
because their depletion improves insulin sensitivity [32]. decrease in intramuscular lipid content in obese humans
However, the activation of inflammatory pathways in liver [42]. Based on these findings, we hypothesize that a likely
parenchymal cells is also likely to contribute to obesity- anti-inflammatory effect of exercise training is via an
induced liver inflammation because hepatocyte-specific increased capacity of the skeletal muscle to utilize fatty
deletion of IkB kinase beta (IKKb), a critical regulator of acids, resulting in decreased lipid content. This would be
the nuclear factor kB (NF-kB) family of transcription predicted to decrease recruitment of macrophages, and
factors, reduces liver inflammation and improves insulin potentially other immune cells, into skeletal muscle and
sensitivity [30]. Several human studies have demonstrated limit the activation of known proinflammatory signaling
that aerobic exercise training such as brisk walking (5 pathways (e.g., JNK). Although there is some evidence
days/week for 30–60 min) or cycling (3 days/week for that obesity and a high-fat diet can promote macrophage
30–45 min) for 4–16 weeks reduces hepatosteatosis recruitment into skeletal muscle, further work is required
[33,34] and, furthermore, can improve hepatic lipid com- to substantiate these findings. Of particular interest
position by altering the profile of liver lipids toward an would be a time-course analysis to determine when macro-
increase in polyunsaturated lipids [35]. Given the crucial phages initially start to accumulate in the skeletal muscle
role of hepatosteatosis in promoting liver inflammation and to what degree they increase on prolonged high-fat
and of liver inflammation in the development of numerous feeding. Furthermore, given the important roles of numer-
diseases, reductions in hepatic lipid content are likely to ous other immune cells in addition to macrophages in
represent an important anti-inflammatory effect of exer- adipose tissue, determining whether other immune cell
3
TREIMM-1081; No. of Pages 8

Review Trends in Immunology xxx xxxx, Vol. xxx, No. x

types are also recruited to skeletal muscle in obesity is Observations that both acute exercise (90 min of cycling
of interest. Finally, determining whether the macro- in humans) [59] and exercise training (resistance training
phages, and potentially other immune cells, that are in elderly women) [60,61] decrease the expression of TLR4
recruited to the skeletal muscle are causally involved on the surface of monocytes have led to the proposal that a
in skeletal muscle insulin resistance would be important. major anti-inflammatory effect of exercise may be via
In vivo this may be a difficult question to address, but in downregulation of TLR4 expression [3,14]. Many [62–
vitro findings support the idea that activated macro- 65], although not all [66], subsequent studies have con-
phages can promote insulin resistance in skeletal muscle firmed these initial findings. Circulating monocytes are the
[43]. precursors to tissue macrophages and given the important
role for proinflammatory macrophages in the development
Exercise ameliorates TLR-dependent inflammation of several metabolic diseases, including insulin resistance,
The TLRs are a family of transmembrane receptors that type 2 diabetes, and atherosclerosis [8], exercise-induced
are central to innate immunity [44]. However, in addition decreases in monocyte TLR4 expression may be an impor-
to their central role in regulating responses to pathogens, tant mechanism by which the anti-inflammatory effects of
the activation of specific TLRs, most notably TLR4, has exercise are mediated. In support of this, it was recently
been suggested to play a role in the development of obesity- shown that exercise-trained apolipoprotein E (ApoE)-defi-
induced insulin resistance, type 2 diabetes, and atheroscle- cient mice fed a high-fat diet – a model of atherosclerosis –
rosis [45,46]. Lipopolysaccharide (LPS), a component of the had significantly reduced vascular TLR4 expression [67].
outer cell wall of Gram-negative bacteria, is the canonical Importantly, however, the effects of exercise on TLR4
ligand for TLR4. However, in addition to recognizing clas- expression are not limited to monocytes. Feeding rats a
sical pathogen-associated molecular patterns such as LPS, high-fat diet caused a dramatic increase in not only the
it is now well established that TLR4 is a receptor for expression of TLR4 but also the activation of TLR4 signal-
numerous host molecules [47]. These damage-associated ing in skeletal muscle, liver, and adipose tissue [68], effects
molecular patterns are typically molecules that, under that were significantly attenuated by either chronic exer-
homeostatic conditions, are not exposed to TLR4, but on cise training (5 days/week for 8 weeks) or acute exercise
cell damage are released and can subsequently induce (two 3-h acute exercise bouts separated by a 45-min rest
inflammatory responses [47]. In the context of metabolic period) [68]. This exercise-induced decrease in the expres-
disease, it has been proposed that specific TLRs can be sion and activation of the TLR4 signaling pathway in high-
activated by numerous ligands (e.g., saturated FFAs [48– fat diet-fed rats was closely associated with reduced acti-
50], oxidized low-density lipoproteins [LDLs] [51]) that are vation of JNK, a serine kinase that is strongly implicated
elevated in metabolic diseases. Deletion of TLR4 has been in the development of obesity-induced insulin resistance,
shown to ameliorate obesity-induced inflammation and and decreased serine phosphorylation of insulin receptor
insulin resistance [52,53] and atherosclerosis in mouse substrate-1 (IRS1), a marker of inactivation of the insulin
models of disease [53]. signaling pathway [68].
There are several features of acute exercise and chron- A hallmark of acute exercise is an increase in lipolysis
ic exercise training that suggest that a principal anti- and consequently an increase in the circulating levels of
inflammatory effect of exercise may be mediated via FFAs [69]. The primary function of the exercise-induced
effects on TLR pathway activation. First, chronic exercise increase in lipolysis of stored lipid is to provide the con-
training can decrease the circulating levels of TLR tracting skeletal muscles with a fuel source to maintain
ligands that are known to be elevated in metabolic dis- continued contraction [69]. However, the finding that cer-
ease. Specifically, given the proposed roles for saturated tain FFAs, primarily saturated FFAs, are ligands for TLR4
FFAs and oxidized LDLs as mediators of TLR activation [48,49] raises the intriguing idea that saturated FFAs
in metabolic disease, the ability of chronic exercise train- released from adipose tissue stores during exercise may
ing to reduce the levels of these ligands may reduce TLR- initiate proinflammatory signaling. It was recently dem-
dependent inflammation in numerous metabolic diseases onstrated that mice deficient in either TLR2 or TLR4 had
[33,54]. Furthermore, it was recently shown that the greatly reduced activation of the mitogen-activated protein
liver-secretory protein fetuin-A may serve as an adaptor kinases p38 and JNK in skeletal muscles following acute
protein, facilitating the presentation of FFAs to TLR4 and exercise [70]. Furthermore, administration of heparin to
thereby contributing to the development of obesity-in- elevate plasma FFA level to that seen during exercise
duced inflammation and insulin resistance [50]. Elevated mimicked the effects of exercise, arguing that exercise-
fetuin-A levels have been linked to numerous metabolic induced increases in plasma FFA levels activate mitogen-
diseases [55,56]. Interestingly, exercise training has been activated protein kinase (MAPK) signaling in skeletal
shown to reduce circulating fetuin-A levels [57,58] and muscles via the activation of TLR2 or TLR4 [70]. We
fetuin-A levels are inversely correlated with maximal hypothesize that it is unlikely that the exercise-induced
oxygen uptake during an exercise test, a measure of increase in circulating FFA levels constitutes a major
cardiorespiratory fitness [58]. These data suggest that proinflammatory action of exercise. Indeed, following pro-
reductions in circulating fetuin-A levels and, therefore, a longed exercise, where circulating FFA levels are greatly
decrease in the capacity of FFAs to be presented to TLR4, increased, it has been repeatedly demonstrated that proin-
in addition to a reduction in the levels of specific TLR flammatory cytokine production by circulating monocytes
ligands, may constitute an important anti-inflammatory is unaltered, arguing that exercise-induced increased
effect of exercise training. plasma FFA levels are not proinflammatory [59,71]. The
4
TREIMM-1081; No. of Pages 8

Review Trends in Immunology xxx xxxx, Vol. xxx, No. x

functional consequences of FFA-induced activation of TLR note, the levels of IL-1ra observed following exercise are
signaling in skeletal muscle is at present unknown and considerably higher than that seen for other cytokines.
awaits further research. Although the precise source of the exercise-induced
These data provide evidence that some of the anti- increase in IL-1ra has not been definitely established,
inflammatory and beneficial health effects of exercise peripheral blood mononuclear cells are a likely source.
may be mediated through multiple effects on TLR4. Spe- Given the critical role of the proinflammatory cytokines
cifically, exercise may reduce the availability of endoge- IL-1a and IL-1b in a range of metabolic disorders and the
nous TLR4 ligands, decrease the expression of TLR4, and potential therapeutic utility of targeting IL-1b and IL-1a,
decrease the activation of TLR4 signaling. it is an intriguing idea that during exercise the contracting
skeletal muscle releases IL-6 that subsequently induces
Exercise tempers inflammatory signals upstream and the release of IL-1ra from circulating immune cells and
downstream of IL-1 potentially tissue macrophages. We hypothesize that exer-
Proinflammatory cytokines are elevated in numerous met- cise-induced increases in IL-1ra may contribute to the anti-
abolic diseases [1]. Although mechanistic roles have been inflammatory effects of exercise by inhibiting the deleteri-
described for some of these, the cytokine that is perhaps ous actions of IL-1a and IL-1b. This could be tested in
best characterized in terms of its involvement in disease several ways. First, given our suggestion that skeletal
pathogenesis is IL-1b [72]. IL-1b is a master cytokine, muscle IL-6 production during exercise stimulates IL-
controlling local and systemic inflammation [72]. IL-1b 1ra production, deletion of IL-6 specifically from skeletal
levels are increased in and have been shown to contribute muscle using IL-6 ‘floxed’ mice would provide an interest-
to numerous metabolic diseases, including atherosclerosis, ing model to test this hypothesis. A complementary ap-
obesity-induced insulin resistance [73], type 2 diabetes proach would be to study mice in which IL-1ra was
[74], and alcoholic liver disease [29]. The production of specifically deleted from immune cells. To date, IL-1ra
IL-1b is regulated by the NLRP3 inflammasome [47]. ‘floxed’ mice have not been reported in the literature,
Recent evidence also proposes an important role for IL- but the deletion of IL-1ra from the immune system specifi-
1a in vascular inflammation in atherosclerosis, although cally could be achieved by transplanting bone marrow from
IL-1a release is inflammasome independent [75]. To date, donor global IL-1ra knockout mice into recipient wild type
three agents that target the IL-1 pathway have been mice. In both models, mice would be high-fat diet fed with
approved for therapeutic use in various inflammatory dis- or without the addition of regular exercise training and
eases [72]. Preliminary studies support the therapeutic metabolic analysis conducted after a set period of high-fat
potential of targeting IL-1 in type 2 diabetes and clinical diet.
trials evaluating the targeting of IL-1b in myocardial As discussed in the preceding section, TLRs belong to a
infarction, stroke, and cardiovascular deaths are ongoing large collection of receptors known as pattern-recognition
[72,76]. Collectively, basic and clinical research supports receptors [47]. Another class of pattern-recognition recep-
an important role for IL-1 in the development and progres- tor that has been implicated in the development of numer-
sion of several metabolic diseases. ous metabolic diseases is the NLRP3 inflammasome [81],
The release of specific cytokines from contracting skele- which controls the release of IL-1b. Similar to the case with
tal muscle was first demonstrated over a decade ago [77]. TLRs, ligands for the NLRP3 inflammasome (e.g., FFAs,
These discoveries led to the concept that muscle is a oxidized LDL) are elevated in chronic metabolic diseases.
secretory organ, releasing myokines during exercise, and Therefore, as described above for TLRs, exercise-induced
potentially in disease states, that influence metabolism decreases in the levels of these ligands may also reduce
within other tissues [78]. Although numerous cytokines activation of the NLRP3 inflammasome and, therefore,
have been identified as being released from exercising reduce the levels of IL-1b.
skeletal muscle [e.g., leukemia inhibitory factor (LIF),
IL-7, myostatin, insulin-like growth factor 1 (IGF-1), fibro- Concluding remarks and outlook
blast growth factor 2 (FGF-2)], the prototypical cytokine As discussed above, the adipose tissue immune cell profile
released from contracting muscle is IL-6 [78]. However, in changes dramatically in the transition from the lean to the
addition to the release of myokines, exercise also results in obese state. Numerous immune cell types have been shown
an increase in the circulating concentrations of numerous to contribute to adipose tissue inflammation and systemic
other cytokines not released from the contracting skeletal insulin resistance in the obese state or, by contrast, con-
muscle. Most prominent among these is IL-1ra [12,79]. tribute to the maintenance of insulin sensitivity in the
Both IL-1b and IL-1a induce signaling through the IL-1 lean, healthy state [8]. This raises several important ques-
receptor (IL-1R). IL-1ra binds to the IL-1R but exerts no tions for further research. First, does exercise prevent the
biological activity and therefore functions as a competitive accumulation of other immune cell types that contribute to
inhibitor of IL-1 signaling. In contrast to IL-6, which tends adipose tissue inflammation and insulin resistance, such
to peak at the cessation of exercise, circulating IL-1ra as neutrophils [18]? Second, does exercise prevent the
levels are modestly elevated at the end of exercise, but high-fat diet-mediated loss of adipose tissue immune cells
increase considerably in the hours following exercise. This that are important in preventing inflammation in adipose
observation can be most parsimoniously explained by the tissue and regulate insulin sensitivity, such as eosinophils
fact that IL-6 is a potent inducer of IL-1ra, and, indeed, [16] and CD4+CD25+ T-regulatory lymphocytes [82]? Given
infusion of recombinant IL-6 into humans at levels seen that the effects of exercise on the adipose tissue immune
during exercise increases circulating IL-1ra levels [80]. Of cell profile are likely to be mediated in the main by
5
TREIMM-1081; No. of Pages 8

Review Trends in Immunology xxx xxxx, Vol. xxx, No. x

preventing adipose tissue expansion, we hypothesize that 3 Handschin, C. and Spiegelman, B.M. (2008) The role of exercise and
PGC1alpha in inflammation and chronic disease. Nature 454, 463–469
exercise training may prevent the accumulation of delete-
4 Knowler, W.C. et al. (2002) Reduction in the incidence of type 2 diabetes
rious immune cells that promote inflammation and insulin with lifestyle intervention or metformin. N. Engl. J. Med. 346, 393–403
resistance and prevent the loss of adipose tissue immune 5 Krogh-Madsen, R. et al. (2013) Normal physical activity obliterates the
cells that are important in the maintenance of insulin deleterious effects of a high-caloric intake. J. Appl. Physiol. (1985) 116,
sensitivity. Third, what is the effect of both acute exercise 231–239
6 Mathis, D. (2013) Immunological goings-on in visceral adipose tissue.
and exercise training on the adipose tissue immune cell
Cell Metab. 17, 851–859
profile in the non-obese state? Because exercise training 7 Libby, P. et al. (2013) Immune effector mechanisms implicated in
markedly increases systemic insulin sensitivity, and the atherosclerosis: from mice to humans. Immunity 38, 1092–1104
immune cell profile of the adipose tissue has a major 8 Osborn, O. and Olefsky, J.M. (2012) The cellular and signaling
impact on systemic insulin sensitivity, it is an intriguing networks linking the immune system and metabolism in disease.
Nat. Med. 18, 363–374
hypothesis that effects on the adipose tissue immune cell 9 Gleeson, M. (2007) Immune function in sport and exercise. J. Appl.
profile may contribute to the beneficial effects of exercise Physiol. (1985) 103, 693–699
training on whole-body insulin sensitivity. For example, 10 Walsh, N.P. et al. (2011) Position statement. Part one: immune
does exercise training affect the numbers of eosinophils, function and exercise. Exerc. Immunol. Rev. 17, 6–63
M2 alternatively activated macrophages, or CD4+CD25+ T- 11 Jeukendrup, A.E. et al. (2000) Relationship between gastro-intestinal
complaints and endotoxaemia, cytokine release and the acute-phase
regulatory lymphocytes – cells that are critical to the reaction during and after a long-distance triathlon in highly trained
regulation of systemic insulin sensitivity in the lean and men. Clin. Sci. (Lond.) 98, 47–55
healthy state – within the adipose tissue? Finally, does 12 Ostrowski, K. et al. (1999) Pro- and anti-inflammatory cytokine balance
acute exercise (i.e., a single bout of exercise) influence the in strenuous exercise in humans. J. Physiol. 515, 287–291
13 Starkie, R. et al. (2003) Exercise and IL-6 infusion inhibit endotoxin-
adipose tissue immune cell profile and might this contrib-
induced TNF-alpha production in humans. FASEB J. 17, 884–886
ute to post-exercise insulin sensitivity? In this regard, it is 14 Gleeson, M. et al. (2011) The anti-inflammatory effects of exercise:
intriguing to note that fasting-induced lipolysis and the mechanisms and implications for the prevention and treatment of
subsequent release of FFAs from adipose tissue is a potent disease. Nat. Rev. Immunol. 11, 607–615
stimulus that promotes the recruitment of M2 alternative- 15 Lumeng, C.N. et al. (2007) Obesity induces a phenotypic switch in
adipose tissue macrophage polarization. J. Clin. Invest. 117, 175–184
ly activated macrophages into adipose tissue [83]. Exercise
16 Wu, D. et al. (2011) Eosinophils sustain adipose alternatively activated
is also a potent inducer of adipose tissue lipolysis and, macrophages associated with glucose homeostasis. Science 332, 243–
accordingly, we hypothesize that acute exercise may dy- 247
namically regulate the adipose tissue immune cell profile, 17 Nishimura, S. et al. (2009) CD8+ effector T cells contribute to
promoting the accumulation of M2 alternatively activated macrophage recruitment and adipose tissue inflammation in obesity.
Nat. Med. 15, 914–920
macrophages. This may represent an important, weight 18 Talukdar, S. et al. (2012) Neutrophils mediate insulin resistance in
loss-independent mechanism by which exercise promotes a mice fed a high-fat diet through secreted elastase. Nat. Med. 18, 1407–
favorable adipose tissue immune cell profile. 1412
Given the multitude of mechanisms by which exercise 19 Winer, D.A. et al. (2011) B cells promote insulin resistance through
promotes an anti-inflammatory state, it seems likely that modulation of T cells and production of pathogenic IgG antibodies. Nat.
Med. 17, 610–617
the anti-inflammatory effects of exercise are central to its 20 Patsouris, D. et al. (2008) Ablation of CD11c-positive cells normalizes
salutary actions in chronic metabolic diseases. Given these insulin sensitivity in obese insulin resistant animals. Cell Metab. 8,
diverse beneficial effects of exercise in chronic metabolic 301–309
disease, it is little wonder that the concept that ‘exercise is 21 Sun, K. et al. (2011) Adipose tissue remodeling and obesity. J. Clin.
Invest. 121, 2094–2101
medicine’ is gaining considerable support [14]. Future
22 Jung, D.Y. et al. (2013) Short-term weight loss attenuates local tissue
studies will be required to delineate the molecular mecha- inflammation and improves insulin sensitivity without affecting
nisms by which the anti-inflammatory actions of exercise adipose inflammation in obese mice. Am. J. Physiol. Endocrinol.
prevent the development of chronic metabolic diseases. Metab. 304, E964–E976
However, perhaps the greatest challenge will be to get 23 Kawanishi, N. et al. (2013) Exercise attenuates M1 macrophages and
CD8+ T cells in the adipose tissue of obese mice. Med. Sci. Sports Exerc.
more people exercising and being more physically active.
45, 1684–1693
Given the myriad benefits of exercise, encouraging more 24 Kawanishi, N. et al. (2010) Exercise training inhibits inflammation in
people to do more exercise and be more physically active adipose tissue via both suppression of macrophage infiltration and
will be a key goal for the future. acceleration of phenotypic switching from M1 to M2 macrophages in
high-fat-diet-induced obese mice. Exerc. Immunol. Rev. 16, 105–118
25 Vieira, V.J. et al. (2009) Effects of exercise and low-fat diet on adipose
tissue inflammation and metabolic complications in obese mice. Am. J.
Acknowledgments
Physiol. Endocrinol. Metab. 296, E1164–E1171
The authors thank all of their current and past staff and collaborators
26 Bruun, J.M. et al. (2006) Diet and exercise reduce low-grade
who have contributed to this body of work. They gratefully acknowledge
inflammation and macrophage infiltration in adipose tissue but not
the Australian Research Council and the National Health and Medical
in skeletal muscle in severely obese subjects. Am. J. Physiol.
Research Council (NHMRC) for continued funding support. M.A.F. is a
Endocrinol. Metab. 290, E961–E967
Senior Principal Research Fellow of the NHMRC.
27 Auerbach, P. et al. (2013) Differential effects of endurance training and
weight loss on plasma adiponectin multimers and adipose tissue
References macrophages in younger, moderately overweight men. Am. J.
1 Hotamisligil, G.S. (2006) Inflammation and metabolic disorders. Physiol. Regul. Integr. Comp. Physiol. 305, R490–R498
Nature 444, 860–867 28 Park, E.J. et al. (2010) Dietary and genetic obesity promote liver
2 Booth, F.W. et al. (2012) Lack of exercise is a major cause of chronic inflammation and tumorigenesis by enhancing IL-6 and TNF
diseases. Comp. Physiol. 2, 1143–1211 expression. Cell 140, 197–208

6
TREIMM-1081; No. of Pages 8

Review Trends in Immunology xxx xxxx, Vol. xxx, No. x

29 Petrasek, J. et al. (2012) IL-1 receptor antagonist ameliorates 55 Mori, K. et al. (2012) Fetuin-A and the cardiovascular system. Adv.
inflammasome-dependent alcoholic steatohepatitis in mice. J. Clin. Clin. Chem. 56, 175–195
Invest. 122, 3476–3489 56 Stefan, N. and Haring, H.U. (2013) The role of hepatokines in
30 Arkan, M.C. et al. (2005) IKK-beta links inflammation to obesity- metabolism. Nat. Rev. Endocrinol. 9, 144–152
induced insulin resistance. Nat. Med. 11, 191–198 57 Jenkins, N.T. et al. (2011) Plasma fetuin-A concentrations in young and
31 Cai, D. et al. (2005) Local and systemic insulin resistance resulting older high- and low-active men. Metabolism 60, 265–271
from hepatic activation of IKK-beta and NF-kappaB. Nat. Med. 11, 58 Malin, S.K. et al. (2013) Fetuin-A is linked to improved glucose
183–190 tolerance after short-term exercise training in nonalcoholic fatty
32 Huang, W. et al. (2010) Depletion of liver Kupffer cells prevents the liver disease. J. Appl. Physiol. (1985) 115, 988–994
development of diet-induced hepatic steatosis and insulin resistance. 59 Lancaster, G.I. et al. (2005) The physiological regulation of Toll-
Diabetes 59, 347–357 like receptor expression and function in humans. J. Physiol. 563,
33 Johnson, N.A. et al. (2009) Aerobic exercise training reduces hepatic 945–955
and visceral lipids in obese individuals without weight loss. Hepatology 60 Flynn, M.G. et al. (2003) Toll-like receptor 4 and CD14 mRNA
50, 1105–1112 expression are lower in resistive exercise-trained elderly women. J.
34 Sullivan, S. et al. (2012) Randomized trial of exercise effect on Appl. Physiol. (1985) 95, 1833–1842
intrahepatic triglyceride content and lipid kinetics in nonalcoholic 61 McFarlin, B.K. et al. (2004) TLR4 is lower in resistance-trained older
fatty liver disease. Hepatology 55, 1738–1745 women and related to inflammatory cytokines. Med. Sci. Sports Exerc.
35 Haus, J.M. et al. (2013) Improved hepatic lipid composition following 36, 1876–1883
short-term exercise in nonalcoholic fatty liver disease. J. Clin. 62 Oliveira, M. and Gleeson, M. (2010) The influence of prolonged cycling
Endocrinol. Metab. 98, E1181–E1188 on monocyte Toll-like receptor 2 and 4 expression in healthy men. Eur.
36 Kawanishi, N. et al. (2012) Exercise training attenuates hepatic J. Appl. Physiol. 109, 251–257
inflammation, fibrosis and macrophage infiltration during diet 63 Simpson, R.J. et al. (2009) Toll-like receptor expression on classic and
induced-obesity in mice. Brain Behav. Immun. 26, 931–941 pro-inflammatory blood monocytes after acute exercise in humans.
37 Fink, L.N. et al. (2013) Expression of anti-inflammatory Brain Behav. Immun. 23, 232–239
macrophage genes within skeletal muscle correlates with insulin 64 Stewart, L.K. et al. (2005) Influence of exercise training and age on
sensitivity in human obesity and type 2 diabetes. Diabetologia 56, CD14+ cell-surface expression of Toll-like receptor 2 and 4. Brain
1623–1628 Behav. Immun. 19, 389–397
38 Fink, L.N. et al. (2014) Pro-inflammatory macrophages increase in 65 Timmerman, K.L. et al. (2008) Exercise training-induced lowering of
skeletal muscle of high fat-fed mice and correlate with metabolic risk inflammatory (CD14+CD16+) monocytes: a role in the anti-
markers in humans. Obesity (Silver Spring) 22, 747–757 inflammatory influence of exercise? J. Leukoc. Biol. 84, 1271–1278
39 Hirosumi, J. et al. (2002) A central role for JNK in obesity and insulin 66 Reyna, S.M. et al. (2013) Short-term exercise training improves insulin
resistance. Nature 420, 333–336 sensitivity but does not inhibit inflammatory pathways in immune
40 Sabio, G. et al. (2010) Role of muscle c-Jun NH2-terminal kinase 1 in cells from insulin-resistant subjects. J. Diabetes Res. http://dx.doi.org/
obesity-induced insulin resistance. Mol. Cell Biol. 30, 106–115 10.1155/2013/107805
41 Sparks, L.M. et al. (2013) Nine months of combined training improves 67 Wu, X.D. et al. (2013) Effect of aerobic exercise on miRNA–TLR4
ex vivo skeletal muscle metabolism in individuals with type 2 diabetes. signaling in atherosclerosis. Int. J. Sports Med. http://dx.doi.org/
J. Clin. Endocrinol. Metab. 98, 1694–1702 10.1055/s-0033-1349075
42 Louche, K. et al. (2013) Endurance exercise training up-regulates 68 Oliveira, A.G. et al. (2011) Physical exercise reduces circulating
lipolytic proteins and reduces triglyceride content in skeletal muscle lipopolysaccharide and TLR4 activation and improves insulin
of obese subjects. J. Clin. Endocrinol. Metab. 98, 4863–4871 signaling in tissues of DIO rats. Diabetes 60, 784–796
43 Samokhvalov, V. et al. (2009) Palmitate- and lipopolysaccharide- 69 Jeppesen, J. and Kiens, B. (2012) Regulation and limitations to fatty
activated macrophages evoke contrasting insulin responses in acid oxidation during exercise. J. Physiol. 590, 1059–1068
muscle cells. Am. J. Physiol. Endocrinol. Metab. 296, E37–E46 70 Zbinden-Foncea, H. et al. (2012) TLR2 and TLR4 activate p38 MAPK
44 Kawai, T. and Akira, S. (2010) The role of pattern-recognition receptors and JNK during endurance exercise in skeletal muscle. Med. Sci.
in innate immunity: update on Toll-like receptors. Nat. Immunol. 11, Sports Exerc. 44, 1463–1472
373–384 71 Starkie, R.L. et al. (2001) Circulating monocytes are not the source of
45 Konner, A.C. and Bruning, J.C. (2011) Toll-like receptors: linking elevations in plasma IL-6 and TNF-alpha levels after prolonged
inflammation to metabolism. Trends Endocrinol. Metab. 22, 16–23 running. Am. J. Physiol. Cell Physiol. 280, C769–C774
46 Seneviratne, A.N. et al. (2012) Toll-like receptors and macrophage 72 Dinarello, C.A. et al. (2012) Treating inflammation by blocking
activation in atherosclerosis. Clin. Chim. Acta 413, 3–14 interleukin-1 in a broad spectrum of diseases. Nat. Rev. Drug
47 Chen, G.Y. and Nunez, G. (2010) Sterile inflammation: sensing and Discov. 11, 633–652
reacting to damage. Nat. Rev. Immunol. 10, 826–837 73 Wen, H. et al. (2011) Fatty acid-induced NLRP3–ASC inflammasome
48 Nguyen, M.T. et al. (2007) A subpopulation of macrophages infiltrates activation interferes with insulin signaling. Nat. Immunol. 12, 408–
hypertrophic adipose tissue and is activated by free fatty acids via Toll- 415
like receptors 2 and 4 and JNK-dependent pathways. J. Biol. Chem. 74 Masters, S.L. et al. (2010) Activation of the NLRP3 inflammasome by
282, 35279–35292 islet amyloid polypeptide provides a mechanism for enhanced IL-1beta
49 Shi, H. et al. (2006) TLR4 links innate immunity and fatty acid-induced in type 2 diabetes. Nat. Immunol. 11, 897–904
insulin resistance. J. Clin. Invest. 116, 3015–3025 75 Freigang, S. et al. (2013) Fatty acid-induced mitochondrial uncoupling
50 Pal, D. et al. (2012) Fetuin-A acts as an endogenous ligand of TLR4 elicits inflammasome-independent IL-1alpha and sterile vascular
to promote lipid-induced insulin resistance. Nat. Med. 18, 1279– inflammation in atherosclerosis. Nat. Immunol. 14, 1045–1053
1285 76 Ridker, P.M. et al. (2012) Effects of interleukin-1beta inhibition with
51 Stewart, C.R. et al. (2010) CD36 ligands promote sterile inflammation canakinumab on hemoglobin A1c, lipids, C-reactive protein,
through assembly of a Toll-like receptor 4 and 6 heterodimer. Nat. interleukin-6, and fibrinogen: a phase IIb randomized, placebo-
Immunol. 11, 155–161 controlled trial. Circulation 126, 2739–2748
52 Higashimori, M. et al. (2011) Role of Toll-like receptor 4 in intimal foam 77 Steensberg, A. et al. (2000) Production of interleukin-6 in contracting
cell accumulation in apolipoprotein E-deficient mice. Arterioscler. human skeletal muscles can account for the exercise-induced increase
Thromb. Vasc. Biol. 31, 50–57 in plasma interleukin-6. J. Physiol. 529, 237–242
53 Michelsen, K.S. et al. (2004) Lack of Toll-like receptor 4 or myeloid 78 Pedersen, B.K. and Febbraio, M.A. (2012) Muscles, exercise and
differentiation factor 88 reduces atherosclerosis and alters plaque obesity: skeletal muscle as a secretory organ. Nat. Rev. Endocrinol.
phenotype in mice deficient in apolipoprotein E. Proc. Natl. Acad. 8, 457–465
Sci. U.S.A. 101, 10679–10684 79 Ostrowski, K. et al. (1998) A trauma-like elevation of plasma
54 Kraus, W.E. et al. (2002) Effects of the amount and intensity of exercise cytokines in humans in response to treadmill running. J. Physiol.
on plasma lipoproteins. N. Engl. J. Med. 347, 1483–1492 513, 889–894

7
TREIMM-1081; No. of Pages 8

Review Trends in Immunology xxx xxxx, Vol. xxx, No. x

80 Steensberg, A. et al. (2003) IL-6 enhances plasma IL-1ra, IL-10, and 82 Feuerer, M. et al. (2009) Lean, but not obese, fat is enriched for a unique
cortisol in humans. Am. J. Physiol. Endocrinol. Metab. 285, E433– population of regulatory T cells that affect metabolic parameters. Nat.
E437 Med. 15, 930–939
81 Grant, R.W. and Dixit, V.D. (2013) Mechanisms of disease: 83 Kosteli, A. et al. (2010) Weight loss and lipolysis promote a dynamic
inflammasome activation and the development of type 2 diabetes. immune response in murine adipose tissue. J. Clin. Invest. 120, 3466–
Front. Immunol. 4, 50 3479

You might also like