Sie sind auf Seite 1von 15

Ageing Research Reviews 26 (2016) 2236

Contents lists available at ScienceDirect

Ageing Research Reviews


journal homepage: www.elsevier.com/locate/arr

Review

Skeletal muscle regeneration and impact of aging and nutrition


Carla Domingues-Faria a,b , Marie-Paule Vasson c,e , Nicolas Goncalves-Mendes c ,
Yves Boirie a,d , Stephane Walrand a,b,
a
Clermont Universit, Universit dAuvergne, Unit de Nutrition Humaine, Equipe NuTriM, CRNH Auvergne; INRA, UMR 1019, UNH, CRNH Auvergne, 63000
Clermont-Ferrand, France
b
INRA, UMR1019, UNH, CRNH Auvergne, 63000 Clermont-Ferrand, France
c
Clermont Universit, Universit dAuvergne, Unit de Nutrition Humaine, Equipe ECREIN, CLARA, CRNH Auvergne; INRA, UMR 1019, UNH, CRNH Auvergne,
63000 Clermont-Ferrand, France
d
CHU Clermont-Ferrand, Service de Nutrition Clinique, 63003 Clermont-Ferrand, France
e
Centre Jean Perrin, Unit de Nutrition, 63000 Clermont-Ferrand, France

a r t i c l e i n f o a b s t r a c t

Article history: After skeletal muscle injury a regeneration process takes place to repair muscle. Skeletal muscle recovery
Received 4 June 2015 is a highly coordinated process involving cross-talk between immune and muscle cells. It is well known
Received in revised form 1 December 2015 that the physiological activities of both immune cells and muscle stem cells decline with advancing age,
Accepted 7 December 2015
thereby blunting the capacity of skeletal muscle to regenerate. The age-related reduction in muscle repair
Available online 9 December 2015
efciency contributes to the development of sarcopenia, one of the most important factors of disability
in elderly people. Preserving muscle regeneration capacity may slow the development of this syndrome.
Keywords:
In this context, nutrition has drawn much attention: studies have demonstrated that nutrients such as
Immunity
Aging
amino acids, n-3 polyunsaturated fatty acids, polyphenols and vitamin D can improve skeletal muscle
Muscle regeneration by targeting key functions of immune cells, muscle cells or both. Here we review the process
Regeneration of skeletal muscle regeneration with a special focus on the cross-talk between immune and muscle cells.
Nutrition We address the effect of aging on immune and skeletal muscle cells involved in muscle regeneration.
Finally, the mechanisms of nutrient action on muscle regeneration are described, showing that quality
of nutrition may help to preserve the capacity for skeletal muscle regeneration with age.
2015 Elsevier B.V. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 23
2. Immune and muscle cell cross-talk during muscle regeneration and impact of aging . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 23
2.1. Immune cell inltration in muscle lesion and pro-inammatory response development. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .23
2.2. Pro-inammatory macrophages stimulate satellite cell proliferation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 24
2.3. Pro-inammatory macrophages are converted to anti-inammatory macrophages for further progression of the muscle regeneration
process . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 25
2.4. Anti-inammatory macrophages stimulate satellite cell differentiation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 26
2.5. Other immune cells involved in muscle regeneration . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 27
2.6. Myokines in skeletal muscle regeneration . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 28
3. Effect of aging on muscle cell behavior and function . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 29
4. Effect of aging on immune and muscle cell cross-talk and consequences for skeletal muscle repair . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 30
5. Nutrition and muscle regeneration . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 31

Corresponding author.
E-mail address: swalrand@clermont.inra.fr (S. Walrand).

http://dx.doi.org/10.1016/j.arr.2015.12.004
1568-1637/ 2015 Elsevier B.V. All rights reserved.
C. Domingues-Faria et al. / Ageing Research Reviews 26 (2016) 2236 23

6. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 33
Conicts of interest . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 33
Acknowledgment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 33
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 33

1. Introduction response against an antigen. It has been shown that the com-
plement system is rapidly activated in injured muscle, causing
Skeletal muscle is a post-mitotic tissue with a very low turnover immune cell inltration in the lesion site (Frenette et al., 2000).
rate (Spalding et al., 2005). Even so, muscle has a strong ability The involvement of mast cells in the initiation of the pro-
to regenerate after injury. Muscle regeneration is a tightly coordi- inammatory response following muscle damage has also been
nated process composed of four consecutive, interlinked phases: demonstrated. Mast cells secrete pro-inammatory cytokines such
(i) necrosis, (ii) inammation, (iii) activation and differentiation of as tumor necrosis factor- (TNF-) allowing the recruitment of
satellite cells, and (iv) maturation of newly formed muscle bers immune cells at the injury site (Radley and Grounds, 2006).
and remodeling of the regenerated muscle (Barberi et al., 2013). To our knowledge, very little is known about the changes in
After activation and proliferation, satellite cells, i.e., muscle stem mast cell function with advancing age. It has been demonstrated
cells, return to their quiescent state to maintain a pool of satellite in an ischemia-reperfusion model that the number and volume of
cells ready for the next regeneration process. Alternatively, satellite mast cells are increased in aged compared with young rats (Harris
cells differentiate and merge to form new mature multinucleated and Rumbaut, 2001). Mast cells are known to be a source of TNF-
muscle cells, i.e., myobers, or fuse with damaged bers (Hawke , thereby contributing to the low-grade inammation observed
and Garry, 2001). with aging (Payne, 2006). If the number of mast cells is increased
The regulation of proliferation and differentiation processes during skeletal muscle regeneration with aging, we hypothesize
in satellite cells depends in part on immune cell function. Dur- that the recruitment of immune cells at the lesion site could be
ing muscle regeneration, immune cells that have inltrated in altered, impacting the whole process of regeneration.
the lesion develop a pro-inammatory phenotype. The biological Besides mast cells, polynuclear neutrophils (also called neu-
function of the rst immune response is to regulate the immune trophils) are among the immune cells that rstly inltrate muscle
cells themselves, and promote migration of satellite cells to the lesions. They express lymphocyte antigen 6C (Ly6C) and G (Ly6C)
site of injury and proliferation. In turn, muscle cells, both dam- cell surface markers. Neutrophils are also involved in the initia-
aged and intact, modulate the immune response. Neutrophils and tion of pro-inammatory response following muscle injury (Tidball
pro-inammatory macrophages are involved in the lysis of dam- and Villalta, 2010). The inltration of these cells is dependent
aged muscle cells and the phagocytosis and destruction of cell on integrin 2. In rodents, blocking integrin 2 function by tar-
debris. Pro-inammatory macrophages are known to stimulate the geting one of its subunits (CD11b or CD18) before muscle lesion
proliferation of satellite cells. The pro-inammatory phenotype of prevents neutrophil inltration at the site of injury (Pizza et al.,
immune cells then turns into an anti-inammatory one. This pro- 2005; Zerria et al., 2006). Neutrophils phagocyte cellular debris
motes the differentiation of satellite cells, leading to the formation and secrete enzymes such as myeloperoxidase (MPO), cytokines or
or the repair of myobers. The whole process is thus highly regu- reactive oxygen species, which contribute to the lysis of damaged
lated, and requires close cross-talk between immune and muscle muscle bers and the aggravation of muscle injury (Kharraz et al.,
cells during regeneration (Tidball and Villalta, 2010). 2013; Nguyen and Tidball, 2003b). The inhibition of neutrophil
Immunosenescence is characterized by an overall alteration of function results in the preservation of muscle bers close to the
immune cell functions (Shaw et al., 2013; Vasson et al., 2013). injury site (Nguyen and Tidball, 2003b; Zerria et al., 2006). Although
The secretion prole of immune cells is modied, resulting in neutrophils are involved in the lysis of muscle bers and in the
an increase in circulating pro-inammatory cytokines such as phagocytosis of cell debris, their presence is required for an optimal
TNF-, IL-6 or IL-1, and leading to the development of chronic muscle regeneration process. Hence injection of a myotoxin in the
low-grade inammation, called inammaging (Franceschi and muscle of neutrophil-depleted mice results in an incomplete mus-
Campisi, 2014). Knowing that immune cells are in closed cross-talk cle recovery and the persistence of necrotic bers (Teixeira et al.,
with muscle cells during muscle regeneration, immunosenescence 2003).
may impact the process of muscle repair. Immunosenescence have With aging, a clinical study has reported that the circulating
been extensively and recently reviewed elsewhere (Agarwal and neutrophil count remains unchanged (Chatta et al., 1994), while it
Busse, 2010; Fulop et al., 2011; Montoya-Ortiz, 2013; Panda et al., has been demonstrated more recently that a small decrease in neu-
2009; Shaw et al., 2013). In this section, we review the process of trophil number occurs with aging (De Martinis et al., 2004). These
skeletal muscle regeneration with a special focus on the cross-talk conicting results can be explained by the difference in the age
between immune cells and muscle cells. In addition, we point out ranges of the subjects included in the two studies: 7080 versus
the main characteristics of age-related change in immune function 80100 years old (Chatta et al., 1994; De Martinis et al., 2004).
that may impact skeletal muscle regeneration. Among the main functions of this cell type, chemotactism, bac-
tericidy and phagocytosis are blunted with age (Niwa et al., 1989;
2. Immune and muscle cell cross-talk during muscle Wenisch et al., 2000). Thus, if the number of circulating neutrophils
regeneration and impact of aging is modied with aging, jointly with phagocytosis and chemo-
tactism, we suppose that the initiation of the pro-inammatory
2.1. Immune cell inltration in muscle lesion and response following muscle injury could be reduced, affecting skele-
pro-inammatory response development tal muscle repair.
It should be noted that the implication of complement system,
Activation of the complement system following acute muscle mast cells and polynuclear neutrophils in the initiation of immune
injury is involved in the initiation of the inammatory response response were demonstrated in rodent models. Effect of aging on
during muscle regeneration. The complement system is composed mast cell functions implicated in muscle repair was also highlighted
of circulating innate immunity proteins that allow a rapid immune in animal models. Thus investigations are needed to conrm that
24 C. Domingues-Faria et al. / Ageing Research Reviews 26 (2016) 2236

these results are relevant to humans. Of note, the effect of aging on also essential for the attraction of satellite cells to the lesion site
polynuclear neutrophil functions was evidenced in humans. and for their proliferation.
Following neutrophil inltration, circulating monocytes
migrate from blood to the site of the lesion, where they differ- 2.2. Pro-inammatory macrophages stimulate satellite cell
entiate into macrophages, gradually replacing neutrophils as the proliferation
main population of immune cells (Tidball et al., 1999; Zerria et al.,
2006). Monocyte migration starts in response to chemokines (such During muscle regeneration (Fig. 1), M1 macrophages attract
as interleukin-1 or IL-1, and interleukin-8 or IL-8) secreted by satellite cells to the injured site (Robertson et al., 1993), and stimu-
neutrophils present at the injury site (Pizza et al., 2005). Monocyte late their proliferation (Saclier et al., 2013a). Several in vitro studies
migration also depends on chemokine (CC motif) ligand 2 (CCL2), have also reported that macrophages migrate towards satellite
also called monocyte chemoattractant protein-1 (MCP1), which cells, and thus potentiate satellite cell proliferation (Arnold et al.,
is secreted by injured muscle bers, and resident or inltrated 2007; Merly et al., 1999; Saclier et al., 2013b). Muscle cell pro-
macrophages in muscle (Lu et al., 2011a). The expression of liferation is in part dependent on IL-6 secreted by macrophages
monocyte CC chemokine receptor type 2 (CCR2), the receptor (Zhang et al., 2013), and does not necessarily require any direct con-
of CCL2, seems important for monocyte migration to the lesion tact between macrophages and muscle cells (Cantini et al., 2002).
site, since macrophage inltration is reduced in CCR2/ mice These ndings have been conrmed in in vivo studies demonstrat-
following injection of cardiotoxin (Martinez et al., 2010). ing the importance of macrophage/muscle cell cross-talk during
Chemokines, in particular the CC class partially produced by muscle regeneration. In a mouse model of muscle lesion induced
macrophages and neutrophils, play an important role in mus- by notexin infusion, the depletion of macrophages in muscle tissue
cle repair. Following muscle cryolesion in mice, the expression results in persistence of necrotic bers, and impairs tissue regen-
of the three chemokines CCL2, CCL3 and CCL4, jointly with their eration (Arnold et al., 2007). Researchers have also demonstrated
main receptors CCR5 and CCR2 is increased (Warren et al., 2004). a greater participation of muscle cells in skeletal muscle regenera-
Recently, a study has demonstrated that iNOS expression in tion in the presence of macrophages: proliferation, migration and
macrophages regulates chemokine expression, and its invalidation engraftment of myoblasts (activated satellite cells) transplanted in
leads to increased inltration and accumulation of immune cells muscle of dystrophic mice are improved when myoblasts are co-
at the lesion site, leading to perturbation of the muscle repair pro- injected with macrophages, especially M1 macrophages (Bencze
cess (Rigamonti et al., 2013). The CCL2/CCR2 axis is likely involved et al., 2012; Lesault et al., 2012). Moreover, macrophages limit
in muscle regeneration, since the neutralization of CCL2 by anti- myotube and satellite cell apoptosis during muscle regeneration
body infusion or CCR2 invalidation is associated with a reduction (Chazaud et al., 2003; Sonnet et al., 2006). Of note, macrophages
in macrophage inltration in injured muscle, leading to an incom- interact with satellite cells to further augment monocyte chemo-
plete process of muscle repair (Ochoa et al., 2007; Warren et al., tactism (Chazaud et al., 2003).
2005, 2004). Soon after muscle injury, inltrated neutrophils and
Macrophages potentiate their own inltration at the injured site. macrophages produce the pro-inammatory cytokine TNF-,
Zhang et al. (2013) have demonstrated in an IL6/ mouse model which contributes to the pro-inammatory response (Collins and
with a muscle lesion that IL-6 produced by macrophages induces Grounds, 2001; Zador et al., 2001). Several roles are assigned to
CCL2 expression through the signal transducer and activator of the TNF- during muscle regeneration. TNF- production stimulates
transcription 3 (STAT3) pathway. In this way, macrophages are able iNOS expression in macrophages, resulting in nitric oxide (NO )
to self-amplify their inltration into a muscle lesion (Zhang et al., production, which promotes muscle damage (Tidball and Villalta,
2013). Macrophages inltrated into injured muscle express specic 2010). In vitro, treatment of rodent myoblasts with TNF- increases
markers such as CD86 or iNOS (inducible nitric oxide synthase), and their proliferation rate, and in vivo, an intraperitoneal injection
secrete cytokines/chemokines such as TNF-, IL-1 (interleukin- of this cytokine in mice stimulates satellite cell proliferation
1), MCP-1, IFN- (interferon ) and IL-6 (interleukin-6). Due to (Li, 2003). It has been found that the pro-proliferative effect of
this phenotype, these cells are considered pro-inammatory or TNF- is dependent on the action of NF-B (nuclear factor-kappa
M1 cells. M1 macrophages lyze muscle cells and phagocyte cel- B). TNF- activates the NF-B pathway in muscle cells, which
lular debris (Rigamonti et al., 2014; Tidball and Villalta, 2010). positively controls cell proliferation through increased cyclin D1
Macrophage phagocytosis can be activated by oxidized low den- expression (Guttridge et al., 1999; Langen et al., 2001). Addition-
sity lipoprotein (LDL) binding to the membrane CD68 (Ottnad ally, TNF- inhibits muscle differentiation, causing destabilization
et al., 1995; Ramprasad et al., 1995). Interestingly, the subset of the mRNA of the myogenic regulatory factor MyoD and the
of neutrophils is able to secrete MPO known to generate reac- degradation of the related protein, which is known to regulate the
tive species oxidizing LDL (Mertens and Holvoet, 2001; Nguyen expression of muscle-specic genes (Langen et al., 2001, 2004).
et al., 2005; Nguyen and Tidball, 2003b). In addition, the co-culture Interestingly, pro-inammatory cytokines, such as TNF- and IL-1,
of myobers, neutrophils and macrophages has demonstrated but also NO stimulate NF-B activation, which in turns induces
that neutrophils potentiate macrophage cytotoxicity (Nguyen and TNF-, IL-1 and iNOS expression. Thus the NF-B pathway activa-
Tidball, 2003a). These data suggest that lysis and phagocytosis func- tion is self-amplied. These substances also increase expression
tions are regulated by neutrophil and macrophage cross-talk. of CCL2 and IL-6, two entities involved in monocyte/macrophage
Of note, the migration and the inltration of monocytes to the inltration in muscle lesions (Ghosh et al., 1998; Mourkioti and
lesion site, their differentiation into macrophages, the potentiation Rosenthal, 2008). Not only does TNF- secreted by neutrophils and
of their own inltration, their rst pro-inammatory phenotype M1 macrophages exert a central action in immune cell attraction;
and the implication of chemokines in their recruitment have this cytokine also plays a chemoattractant role on satellite cells
been essentially demonstrated in animal models. Their lysis and at the damaged site of the muscle (Lolmede et al., 2009; Torrente
phagocytosis functions were essentially evidenced in in vitro mod- et al., 2003).
els. Thus, attention have to be paid concerning the rst steps To sum up, TNF- stimulates muscle proliferation and inhibits
of macrophage response during muscle regeneration in human muscle differentiation through the NF-B pathway. Nevertheless,
beings. it seems that TNF- also stimulates muscle differentiation at later
Lysis and phagocytosis are not the only roles played by M1 stages in the regeneration process, independently of the NF-kB
macrophages during skeletal muscle regeneration. These cells are pathway. In a rodent model of muscle regeneration, the expression
C. Domingues-Faria et al. / Ageing Research Reviews 26 (2016) 2236 25

Fig. 1. Cross-talk between macrophages, neutrophils and satellite cells during skeletal muscle regeneration. (For interpretation of the references to color in this gure legend,
the reader is referred to the web version of this article.)
Following muscle lesion, immune cells such as neutrophils and macrophages inltrate the injury site. They rstly produce a pro-inammatory response, notably characterized
by the secretion of pro-inammatory cytokines such as TNF-, chemokines such as CCL2, and reactive oxygen species such as NO (blue characters). This pro-inammatory
reaction leads to further recruitment of immune cells at the lesion site, and also attracts and stimulates the proliferation of muscle satellite cells. Of note, the Notch signaling
pathway is activated in proliferating satellite cells to control this process. The pro-inammatory response then turns into an anti-inammatory one. Immune cells secrete
anti-inammatory cytokines (yellow characters). Anti-inammatory macrophages can inhibit the pro-inammatory reaction, and they promote the differentiation of satellite
cells. Wnt signaling is activated in satellite cells during their differentiation, as this pathway regulates the expression of genes related to this process.

of TNF- mRNA is induced following injury, and again one week / mice or in mice receiving IFN- receptor antagonist (Cheng
later when the muscle differentiation process is already started et al., 2008).
(Zador et al., 2001). Thus this cytokine likely has a regulating action The stimulation of satellite cell proliferation by pro-
even in the late steps of muscle regeneration, when satellite cells inammatory macrophages have been demonstrated in in vitro
differentiate. TNF- activates p38 kinase and stimulates the differ- and animal models. Thus investigations are needed to conrm the
entiation of C2C12 muscle cells. Inversely, blocking TNF- action relevance on these results in humans.
using anti-TNF- or inhibiting p38 kinase activity decreases the
expression of muscle differentiation markers such as MyoD, myo-
genin or myosin (Chen et al., 2007; Wu et al., 2000). 2.3. Pro-inammatory macrophages are converted to
Like TNF-, IL-6 stimulates monocyte/macrophage migration to anti-inammatory macrophages for further progression of the
the muscle lesion site through the activation of the STAT3 pathway. muscle regeneration process
This process activates satellite cell proliferation. In IL6/ mice, the
migration and inltration of monocyte/macrophages to the site of Macrophage conversion from a pro-inammatory (M1) to an
muscle injury is reduced, and as a result satellite cell proliferation is anti-inammatory (M2) phenotype is a central process in mus-
decreased (Zhang et al., 2013). Of note, IL-6 is produced not only by cle regeneration (Fig. 1). The M1 population stimulates muscle
macrophages but also by satellite cells and myobers. Interestingly, proliferation, whereas M2 cells play a key role in the process of dif-
the proliferation and migration of myoblasts isolated from IL6/ ferentiation, i.e. from satellite cells to myobers (Arnold et al., 2007;
mice are reduced compared with myoblasts isolated from wild type Saclier et al., 2013b). Several molecular and cellular processes are
mice (Serrano et al., 2008). involved in macrophage phenotype transition. Macrophage phago-
IFN- is also involved in muscle regeneration. Satellite cell cytosis is one of the mechanisms leading to M1/M2 conversion.
proliferation and regenerating ber numbers are diminished in IFN- The expression of the anti-inammatory cytokine transforming
growth factor- (TGF-) is enhanced in macrophages cultivated
with lyzed satellite cells (Arnold et al., 2007). If phagocytosis is
26 C. Domingues-Faria et al. / Ageing Research Reviews 26 (2016) 2236

blunted, macrophages retain their M1 phenotype (Arnold et al., 2.4. Anti-inammatory macrophages stimulate satellite cell
2007). Interestingly, the type of the element to be phagocyted differentiation
inuences M1/M2 conversion. The phagocytosis of apoptotic neu-
trophils inhibits the production of pro-inammatory cytokines, The main characteristics of M2 macrophages are that these
whereas the production of an anti-inammatory cytokine such as cells express different markers such as CD206, CD163 or arginase-
TGF- is augmented (Fadok et al., 1998). Also, TGF- production 1, and produce anti-inammatory cytokines such as IL-10 or
by macrophages following phagocytosis is greater when apop- TGF-. CD206 (or mannose receptor) is involved in the pro-
totic neutrophils are phagocyted compared with lyzed neutrophils inammatory response blockage. CD206 binds to MPO and then
(Fadok et al., 2001). internalizes this enzyme in a lysosome, which contributes to
Macrophage-secreted cytokines also orchestrate M1/M2 con- the reduction in both M1 response and muscle lysis (Shepherd
version. The pro-inammatory cytokine IFN- stimulates the M1 and Hoidal, 1990). CD163 also plays a central role in the inhi-
and inhibits the M2 phenotype. A reduction in muscle damage bition of pro-inammatory response. This cluster is known to
and an increase in M2 macrophage activation have been observed bind and internalize the hemoglobin-haptoglobin complex and to
in IFN-/ dystrophic mice (Villalta et al., 2011a). The anti- counteract oxidative damage caused by free radicals produced by
inammatory cytokine TGF- participates in the M1/M2 phenotype hemoglobin. Also, the internalization of hemoglobin-haptoglobin
transition, as the treatment of macrophages with lipopolysac- complex induces the secretion of the anti-inammatory cytokine
charide (LPS) and TGF- reduces pro-inammatory cytokine IL-10 (Philippidis et al., 2004). Thus the action of CD206 and CD163
production compared with treatment with LPS alone (Fadok seems to contribute to the promotion of the anti-inammatory
et al., 1998). The anti-inammatory cytokine IL-10 reduces M1 response.
macrophage activation and promotes M1/M2 conversion in vitro M2 macrophages stimulate muscle cell differentiation, leading
and in vivo (Deng et al., 2012; Villalta et al., 2011b) to myober formation or repair (Fig. 1) (Chazaud et al., 2009).
At the cellular level, the M1/M2 transition is coordinated A decrease in the percentage of centrally nucleated myobers,
by mitogen-activated protein kinase phosphatase-1 (MKP-1), which is a marker of regeneration, was observed in a murine
which regulates the activation of mitogen-activated protein kinase model of muscle regeneration characterized by a depletion of
(MAPK) p38 and the activity of Akt. This pathway allows a con- M2 macrophages (Tidball and Wehling-Henricks, 2007). In vitro,
trolled conversion from a pro-inammatory phenotype to an M2 macrophages promote the expression of myogenin, a marker
anti-inammatory phenotype in macrophages. The transition of of muscle differentiation, and myotube formation (Arnold et al.,
M1 macrophages to an anti-inammatory phenotype occurs too 2007). A study using human quadriceps biopsies, withdrawn after
early in injured skeletal muscle of MPK-1/ mice, and the expres- exercise, has revealed that M2 macrophages are preferentially
sion of pro-inammatory cytokines persists. Consequently, skeletal found in areas containing differentiating satellite cells (Saclier et al.,
muscle regeneration is incomplete. In addition, p38 induces the 2013b).
expression of the pro-inammatory cytokine IL-1, which stimu- The central importance of M2 macrophages during muscle
lates the expression of TGF-, showing that cytokines themselves repair has been demonstrated by numerous studies. In CCR2/
play a role in the M1/M2 transition (Perdiguero et al., 2011). mice, macrophages do not inltrate muscle lesions, and conse-
cAMP response element binding protein (CREB) binding on the quently the efciency of muscle regeneration is blunted. However,
promoter of CCAAT/enhancer binding protein (C/EBP) induces the the infusion of IGF-1 (insulin-like growth factor-1), a hormone pro-
expression of C/EBP, leading to the expression of M2 phenotype- duced by M2 macrophages, stimulates muscle repair in CCR2/
related genes such as IL-10. In mutant mice for the promoter mice (Lu et al., 2011b). Furthermore, the lack of M2 macrophages
of C/EBP, leading to the loss of function of this promoter, during the regeneration process leads to delayed angiogenesis, con-
macrophage invasion in the muscle lesion site is not modied, tributing to inefcient muscle repair (Ochoa et al., 2007).
but the expression of M2 specic genes is limited, and so muscle The anti-inammatory response has to cease at the end of
regeneration is incomplete (Ruffell et al., 2009). the muscle repair process. A study has demonstrated that when
The involvement of adenosine 5 -monophosphate-activated regeneration is completed, M2 macrophages become inactive,
protein kinase alpha (AMPK) activation in the conversion of and consequently anti-inammatory cytokine production is inter-
the macrophage phenotype has also been demonstrated. Thus rupted (Perdiguero et al., 2011). Macrophages then revert to a
the inhibition of AMPK in macrophages results in an increase quiescent state or are eliminated through apoptosis. Like the
in LPS-induced pro-inammatory cytokine (TNF-, IL-6) expres- M1/M2 transition, inammatory reaction is stopped by a coordi-
sions. Conversely, a signicant decrease in the expression of these nated action of p38/MKP-1 and AKT (Perdiguero et al., 2011).
cytokines is observed when macrophages express a constitutively With aging, monocyte/macrophage phagocytosis and cytokine
activated form of AMPK (Sag et al., 2008). More recently, a study secretions are altered but discrepancies among conclusions still
has shown in vitro and in vivo that the deletion of the AMPK exist. According to the species, i.e., humans or rodents, the tissue
gene in macrophages is a limiting factor for their conversion to origin or the nature of the body to be phagocyted, phagocytosis has
M2 phenotype and for muscle regeneration (Mounier et al., 2013). been described to be stimulated, inhibited, or unchanged (Gomez
Interestingly, the activation of AMPK induces CREB activation and et al., 2008; Shaw et al., 2013). Studies also conict concerning the
thus stimulates the expression of M2-related genes, and therefore capacity of monocytes/macrophages to produce cytokines. Some
M1/M2 conversion (Ruffell et al., 2009; Sag et al., 2008). have shown that the production of pro-inammatory cytokines by
These different studies show that the conversion of the M1/M2 human monocytes is increased with aging, whereas others have
macrophage phenotype is nely regulated, and that different reported a diminution (Hearps et al., 2012; Nyugen et al., 2010;
molecular targets and pathways are involved. It remains unclear Qian et al., 2012; van Duin et al., 2007). Of note, the method used
how all these elements are coordinated and whether other players to stimulate the cells differed among these studies. Also, they were
are involved for a successful M1/M2 transition. all conducted in vitro, which meant the cell environment formatted
Of note, the conversion of M1M2 phenotype in macrophages, by aging, which may inuence the function of immune cells, was
the role of cytokines and signaling pathways in this process have missing. Knowing the central role of macrophages in the regula-
been highlighted in in vitro and animal models. tion of the proliferation and the differentiation of satellite cells, we
presume that any changes that occur during aging in macrophage
C. Domingues-Faria et al. / Ageing Research Reviews 26 (2016) 2236 27

functions could reduce the capacity of skeletal muscle to regener- inammation known as Inammaging (Boren and Gershwin, 2004;
ate. Castelo-Branco and Soveral, 2014).
The stimulation of satellite cell differentiation by anti- Concerning NK cells, two populations exist in humans: cytotoxic
inammatory macrophages have been well described in in vitro NK and NK producing cytokines. During aging it has been demon-
and rodent models. Investigation carried out in humans seems to strated that the cytotoxicity of NK cells is decreased, while their
be consistent with in vitro and animal models as human quadri- number increases (the overall cytotoxicity is therefore preserved).
ceps biopsies have revealed that M2 macrophages are preferentially However, the number of NK cells able to produce cytokines, and
found in areas containing differentiating satellite cells (Saclier et al., their cytokine secretion, especially of IFN-, decrease (Almeida-
2013b). Interestingly, the effect of aging on macrophage functions Oliveira et al., 2011; Camous et al., 2012).
has been essentially demonstrated in humans. Future scientic Thus we suppose that alterations with aging of the number of
works will highlighted the impact of aging on the stimulation of T and NK cells together with changes in their ability to secrete
satellite cell proliferation, on the M1/M2 conversion and on the cytokines could modify the process of muscle regeneration, likely
promotion of satellite cell differentiation in order to prevent the through a reduction of satellite cell proliferation.
effect of aging on macrophage/muscle cell cross-talk. Of note, the total number of B cells diminish with aging. Hence,
As seen previously, pro-inammatory M1 macrophages are the number of naive B cells declines, while the number of mem-
converted into anti-inammatory M2 macrophages during skele- ory B cells increases with age (Montoya-Ortiz, 2013). These B cells
tal muscle regeneration. However, fully polarized M1 and M2 present an increased resistance to apoptosis. Antibody produc-
cells in their various versions are extremes of a continuum tion is also modied with aging. This production is lowered with
(Mantovani et al., 2004). There are actually several populations lower afnities and decreased opsonizing abilities, and the anti-
of M2 macrophages involved in muscle regeneration: M2a, M2b body response to new antigens is delayed (Castelo-Branco and
and M2c macrophages for which functions were already well Soveral, 2014). To our knowledge, B cell involvement in muscle
described in reviews (Mantovani et al., 2004; Tidball and Villalta, repair has not yet been demonstrated
2010). Generally speaking, M2a are activated by IL-4 and IL-13 and A particular population of lymphocytes, named Treg cells, has
these cells reduce the lysis rate of muscle cells induced by M1 been identied as key modulators of pro- to anti-inammatory
macrophages (Villalta et al., 2009). M2b macrophages are activated response transition. Following acute injury, Treg cells inltrate the
by the toll-like receptors (TLR) or immune complexes (antigen- lesion site, and this inltration coincides with M1/M2 response con-
antibody complex) and are involved in the anti-inammatory version (Burzyn et al., 2013). This transition is not possible when
response through cytokine secretion. Finally, M2c are activated by acutely injured mice are depleted in Treg cells (Burzyn et al., 2013).
IL-4 and IL-10, and promote lesion resorption (Tidball and Villalta, Additionally, M1 macrophage activation and the production of pro-
2010). Here we chose to focus on the function of M2 macrophage inammatory cytokines are increased in mdx mice depleted in Treg
population in its entirety. To our knowledge M2a, M2b and M2c cells (Villalta et al., 2014).
macrophage functions were not yet investigated during skeletal With aging, the number of Treg cells rises whereas their function
muscle regeneration and aging. is altered, thereby affecting the function of the other immune cells
(Garg et al., 2014). The fact that Treg are unable to modulate pro- to
anti-inammatory response transition may result in an incomplete
2.5. Other immune cells involved in muscle regeneration regeneration process as we age.
The presence of monocyte-derived dendritic cells has been iden-
Neutrophil and macrophage functions during muscle repair tied in a mouse model of acute injury. This immune cell type may
have been thoroughly investigated. However, other types of provide longstanding immune surveillance until full restoration of
immune cells also contribute to this process (Table 1). myober integrity (Brigitte et al., 2010).
In mdx mice, a model of Duchennes muscular dystrophy, or It has been demonstrated that cytokine production by dendritic
in muscle-injured mice, CD4+ T cells, CD8+ T cells and natural cells in response to stimulation is reduced in older people (Panda
killer (NK) cells inltrate the injured site, jointly with macrophages. et al., 2010; Qian et al., 2011). In addition, phagocytosis and migra-
These cells are sources of IFN-, a cytokine known to stimulate tion are reduced (Agrawal et al., 2007). Thus we presume that
satellite cell proliferation. Furthermore, the depletion of CD8+ T the alteration of dendritic cell function with aging could reduce
cells reduces the concentration of apoptotic myonuclei, as well their capacity to provide immune surveillance during skeletal mus-
as muscle ber necrosis in mdx mice, demonstrating that these cle regeneration. Consequently, immune response during muscle
immune cells are involved in the severity of Duchenne muscular repair could be disorganized and not adapted to the muscle regen-
dystrophy (Cheng et al., 2008; Spencer et al., 1997). It has been eration context.
demonstrated in mice decient in CD8+ T cells that these cells are Polynuclear eosinophil cells have also been observed in mus-
needed for normal muscle regeneration (Zhang et al., 2014). CD8+ T cle necrotic areas of mdx mice, where they promote myober lysis.
cells regulate MCP-1 secretion to recruit M1 macrophages at muscle However, this immune cell type has seldom been identied in mus-
injury sites, facilitating myoblast proliferation (Zhang et al., 2014). cle after acute injury (Wehling-Henricks et al., 2008).
With aging, investigations of adaptive immunity have demon- Little is known about the effect of age on polynuclear eosinophil
strated that the reserve pool of naive CD4+ T cells is reduced, and the function. However, Mathur et al. (2008) have highlighted that
CD4+ T cell/CD8+ T cell ratio decreased. Furthermore, the reserve degranulation of these cells (as measured by IL-5induced degran-
pool of memory T cells (CD4 + or CD8 +) increases. T cell prolif- ulation of eosinophil-derived neurotoxin) and their free radical
eration requires the presence of IL-2 and IL-2 receptors, both of production are reduced in asthmatic older people compared with
which decline with aging (Boren and Gershwin, 2004). In addition, younger ones. Polynuclear eosinophil are very rarely observed dur-
stimulation of the T cell receptor leads to a decrease in transmem- ing skeletal muscle regeneration and his role is not known.
brane signaling. This decline leads to a decrease in transcription Taken together, these ndings demonstrate the importance of
factor in T cell and consequently the reduction in gene expres- immune cells in the orchestration of muscle regeneration. This
sion involved in the induction of the immune response (Boren and being the case, modications of the phenotype, and therefore
Gershwin, 2004). Finally, the modications of the whole immune the function of these cells, may impact immune and muscle cell
response contributes to the increased levels of circulating pro- cross-talk, leading to inefcient muscle regeneration. With aging,
inammatory cytokines leading to a chronic state of low-grade
28 C. Domingues-Faria et al. / Ageing Research Reviews 26 (2016) 2236

Table 1
Role of immune cells in muscle regeneration and their known cross-talk with other cells.

Immune cells Role in muscle regeneration and known cross-talk References


with other cells

Mast cells Secretion of TNF- for immune cell recruitment at Radley and Grounds (2006)
lesion site

Polynuclear Promotion of myobre lysis in mdx model Wehling-Henricks et al.


eosinophils Very scarce in muscle acute injury (2008)

Polynuclear Muscle lysis and phagocytosis of cellular debris Kharraz et al. (2013), Nguyen and Tidball (2003b) and
neutrophils Teixeira et al. (2003)
Initiation of pro-inammatory response Tidball and Villalta (2010), Zerria et al. (2006) and Pizza
et al. (2005)
Recruitment of immune cells such as macrophages Pizza et al. (2005)

Macrophages Muscle lysis (M1) and phagocytosis of cellular debris Tidball and Villalta (2010), Rigamonti et al. (2014), Ottnad
(M1) et al. (1995) and Ramprasad et al. (1995)
Secretion of chemokines Warren et al. (2004), Ochoa et al. (2007), Rigamonti et al.
(2013) and Warren et al. (2005)
Recruitment of immune cells such as Lu et al. (2011a), Martinez et al. (2010) and Zhang et al.
monocytes/macrophages (2013)
Recruitment of satellite cells Robertson et al.(1993)
Secretion of pro-inammatory cytokines (M1) Tidball and Villalta (2010)
Stimulation of the proliferation of satellite cells (M1) Saclier et al. (2013a,b), Arnold et al. (2007), Bencze et al.
(2012) and Lesault et al. (2012)
Control their own conversion from M1 to M2 Arnold et al. (2007), Fadok et al. (1998, 2001), Villalta et al.
phenotype (2011a,b), Deng et al. (2012), Perdiguero et al. (2011),
Ruffell et al. (2009) and Mounier et al. (2013)
Stimulation of the differentiation of satellite cells (M2) Saclier et al. (2013b), Arnold et al. (2007), Chazaud et al.
(2009), Tidball and Wehling-Henricks (2007) and Lu et al.
(2011b)
Secretion of anti-inammatory cytokines (M2) Tidball and Villalta (2010)
Limitation of myotube and satellite cell apoptosis Chazaud et al. (2003) and Sonnet et al. (2006)
Arrest of pro-inammatory response (M2) Arnold et al. (2007), Shepherd and Hoidal (1990) and
Philippidis et al. (2004)

NK cells Source of IFN-, which stimulates satellite cell Cheng et al. (2008)
proliferation

Monocyte-derived dendritic cells Immune surveillance Brigitte et al. (2010)

CD4+ T cells Source of IFN-, which stimulates satellite cell Cheng et al. (2008)
proliferation

CD8+ T cells Source of IFN-, which stimulates satellite cell Spencer et al. (1997)
proliferation
Regulate MCP-1 secretion for M1 macrophage Zhang et al. (2014)
recruitment, facilitating satellite cell proliferation

Treg cells Promotion of the conversion of M1 to M2 macrophages Burzyn et al. (2013) and Villalta et al. (2014)

immune function is altered, and this jointly with muscle cell aging most of myokines have been studied in the context of physical
likely contributes to an overall change in muscle repair capacity. activity, some of them have been identied during skeletal muscle
The implication of CD4+ and CD8+ T cells, Treg cells, NK cells, regeneration and/or likely have a key role in this process.
dendritic cells and polynuclear eosinophils has been evidenced in In vitro, human myoblasts express and secrete IL-6 in response
animal models of muscle regeneration. Inversely, the effect of aging to inammatory stimuli mimics by IL-1, TNF- and lipopolysac-
has been studied in humans. In this context, the role of CD4+ and charide (Gallucci et al., 1998). In addition, muscle cell-derived IL-6
CD8+ T cells, Treg cells, NK cells, dendritic cells and polynuclear stimulates migration and proliferation of muscle cells as these
eosinophils in human skeletal muscle repair has to be conrmed. processes are decreased in myoblasts derived from IL-6/ mice
Furthermore, there is a gap concerning the consequences of the (Serrano et al., 2008). Of note, immune cells inltrating the mus-
aging of theses immune cells on muscle regeneration. Additional cle following an injury secrete pro-inammatory cytokines. Thus
studies are needed to bridge this gap. immune cell-derived cytokines may further stimulate IL-6 pro-
duction by muscle cells. IL-6 is also implicated in the myogenic
differentiation program. In C2C12 muscle cells, interference of IL-6
2.6. Myokines in skeletal muscle regeneration
mRNA reduces C2C12 differentiation whereas IL-6 overexpression
increased muscle differentiation (Baeza-Raja and Munoz-Canoves,
During skeletal muscle regeneration, immune cells secrete
2004; Hoene et al., 2013). Similarly, myotube formation was
cytokines to improve satellite cell proliferation or differentiation.
impaired in IL-6/ skeletal muscle cells (Hoene et al., 2013). Taken
However, skeletal muscle also produces factors that may possibly
together, this results demonstrate the key role of IL-6 myokine in
inuenced muscle repair process. Cytokines and other peptides that
myoblast migration, proliferation and differentiation. As it is pro-
are produced, expressed and released by muscle bers and exert
duced by both immune and muscle cells during skeletal muscle
autocrine or endocrine effect are classied as myokines (Pedersen,
regeneration (Kurek et al., 1996; Zhang et al., 2013), IL-6 is prepon-
2011; Pedersen and Febbraio, 2008). According to Pedersen (2011),
derant in this process.
contracting skeletal muscle release myokines, which work in
Leukaemia inhibitory factor (LIF) is produced and released from
hormone-like fashion, exerting specic endocrine effects on other
muscle cells in vitro and from skeletal muscle in vivo, partic-
organs or which work locally via paracrine mechanisms. Even if
C. Domingues-Faria et al. / Ageing Research Reviews 26 (2016) 2236 29

ularly during exercise (Broholm and Pedersen, 2010). However, 1997; Hawke and Garry, 2001; Renault et al., 2000). The increased
LIF myokine is also produced by muscle cells during regeneration expression of cell cycle inhibitors seems to contribute to the reduc-
(Barnard et al., 1994; Kurek et al., 1996). Furthermore, Barnard et al. tion in satellite cell proliferation with age (Barberi et al., 2013). The
(1994) have demonstrated in a rodent model that administration of aged niche disrupts muscle stem cell quiescence and self-renewal
LIF to the site of muscle injury results in an increase rate of regen- capacity through a constant production of FGF-2 (broblast growth
eration. This effect is likely due to the modulation of muscle cell factor-2), even without muscle lesion (Chakkalakal et al., 2012).
proliferation and differentiation. In vitro, human myoblast prolif- Interestingly, this phenomenon leads to a non-polarized overacti-
eration is increased by recombinant LIF and decreased by the siRNA vation of the p38 pathway during satellite cell division (Bentzinger
knockdown of the endogenous LIF receptor (Broholm et al., 2011). and Rudnicki, 2014).
In addition, LIF stimulates C2C12 differentiation trough JAK2/STAT2 The differentiation capacity of satellite cells is also reduced with
pathway (Yang et al., 2009). Thus, LIF myokine is implicated in aging. In vitro, it has been demonstrated that satellite cell entry
skeletal muscle regeneration, probably through its role on muscle into the differentiation program is delayed in aged compared with
cell proliferation and differentiation. young mice, as a result of reduced capacity to proliferate (Shefer
IL-8 is a myokine and CXC ligand 1 (CXCL-1) is often mentioned et al., 2006). Furthermore, the number of satellite cells engaged
as the functional homologue of IL-8 (Pedersen and Febbraio, 2012). in the differentiation program (expressing differentiation markers
It has been highlighted that IL-8 and CXCL-1 have a neutrophil such as desmin and myogenin) is decreased in aged mice (Charge
chemoattractant activity (Lira et al., 1994; Pedersen and Febbraio, et al., 2002; Collins et al., 2007).
2012). To our knowledge, the implication of IL-8 and CXCL-1 in Studies have found that the expression of myogenic regulatory
skeletal muscle regeneration has to be determined. Due to their factors (MRF) known to control the differentiation program, such as
chemoattractiveness, the secretion of these compounds by muscle Myf-5, MyoD, myogenin, and MRF4/Myf6, is reduced in senescent
cells may contribute to the attraction of neutrophils at the injury human satellite cells and in aged rat muscle (Alway et al., 2002;
site at the beginning of regeneration process. Bigot et al., 2008; Degens, 2007). The decrease in MRF expression
Human skeletal muscle cells also express and secrete IL-7 is likely due to the increased expression of inhibitors of the differ-
(Haugen et al., 2010). In vitro, IL-7 expression increases during dif- entiation (Id) process (Alway et al., 2002). Finally, the regenerating
ferentiation of satellite cells into human myotubes, and inversely myober numbers and the expression of the terminal differentia-
the expression of IL-7 receptor decreases (Haugen et al., 2010). IL-7 tion marker, i.e. myosin, are reduced in aged mice following a lesion
treatment of satellite cells increases cell migration but inhibits cell (Conboy et al., 2003, 2005). Similarly, the differentiation of satellite
differentiation (Haugen et al., 2010). In a context of muscle regen- cells into myotubes, and the expression of myosin are decreased
eration, IL-7 may be implicated in the attraction of satellite cells to in aged rat muscle (Lees et al., 2006). Additionally, the culture of
the lesion site and may prevent precocious differentiation of these satellite cells isolated from aged and young subjects has revealed
cells. the altered muscle cell differentiation with aging, illustrated by the
It has been demonstrated in vitro that following myotube for- decrease in fusion index and by the reduction in the number of
mation, nascent myotube secretes IL-4 to further recruit myoblasts desmin-positive satellite cells (Beccaco et al., 2007).
in order to merge with nascent myotube, leading to muscle growth Interestingly, a signaling pathway has been identied as respon-
(Horsley et al., 2003). Therefore, during muscle repair, IL-4 may sible, at least in part, for the decrease in muscle regeneration
be secreted by newly formed myober to further enhance their capacity, namely the Notch signaling pathway (Conboy et al., 2003).
growth. Following muscle lesion, the Notch pathway is activated, promoting
With aging, muscle functions decline and it is likely that satellite cell proliferation and self-renewal (Fig. 2). In addition, the
myokine production is altered. Consequently, part of skeletal mus- inhibition of this pathway allows the cells to enter the differentia-
cle regeneration process that could be dependent of myokines may tion program (Conboy and Rando, 2002). Several in vitro and in vivo
be impaired, contributing to the age-related muscle repair inef- studies have clearly established that the activity of the Notch sig-
ciency. naling pathway decreases with aging (Carlson and Faulkner, 1989;
Carlson et al., 2009; Conboy et al., 2003, 2005). Another pathway,
the Wnt signaling pathway, is also involved in the reduction in mus-
3. Effect of aging on muscle cell behavior and function cle repair capacity with aging (Arthur and Cooley, 2012). Following
muscle injury, when Notch activity is inhibited to allow muscle
With aging, muscle regeneration capacity declines (Ehrhardt cell differentiation, the Wnt pathway is activated (Fig. 2) (Jang
and Morgan, 2005). The impoverishment of the reserve pool of et al., 2011). It then promotes the entry and progression of satellite
satellite cells contributes to this process (Welle, 2002). It has been cells through the differentiation program (Brack et al., 2008; Yin
demonstrated that the number of satellite cells per myober in et al., 2013). However, the required balanced activation between
mice is reduced with aging (Shefer et al., 2006). In humans, this the Notch and Wnt pathways is altered with aging. Furthermore,
decrease seems to be more evident for satellite cells associated with the hyperactivity of Wnt signaling leads to exit of satellite cells from
type II myobers, corresponding to the specicity of ber loss with the myogenic differentiation program and induces a brogenic pro-
advancing age, i.e., a specic loss of type II bers (Verdijk et al., gram (Brack et al., 2007).
2014). The reduction in satellite cell number is partly explained by It should be noted that the reduction of the number of satel-
changes in (i) proliferation, (ii) response to stimuli inducing prolif- lite cells with aging have been observed in rodents models and in
eration, and (iii) capacity to replenish the reserve pool of satellite humans. In addition, the effect of aging of Notch signaling pathway
cells (Conboy et al., 2005; Day et al., 2010). Barani et al. have demon- and its consequence on satellite cells were observed in both rodents
strated that entry into the proliferation phase is delayed for muscle and humans. Similarly, the decrease in satellite cell differentiation
cells derived from aged compared with young rats. Specically, observed in animals was conrmed in humans. Thus future inves-
the doubling time of these cells is increased, and the expression of tigations have to develop strategies to prevent the reduction of
the proliferation marker PCNA in response to proliferating stimuli satellite cell proliferation and differentiation associated with aging.
is decreased (Barani et al., 2003). Telomere reduction throughout All of these data show that muscle regeneration declines with
the proliferation cycles likely contributes to the decrease in the aging not only through the intrinsic modications of satellite cells,
proliferation capacity of satellite cells, as demonstrated in satel- but also through the changes in their environment, which has a
lite cells isolated from aged and young individuals (Decary et al., direct impact on cell function. Of note, the loss of muscle regenera-
30 C. Domingues-Faria et al. / Ageing Research Reviews 26 (2016) 2236

Fig. 2. Notch and canonical Wnt signaling pathways activated in satellite cells during skeletal muscle differentiation.
After injury the expression of the transmembrane ligand Delta-like is stimulated in satellite cells and myobers. Delta-like binds to Notch receptor, which activates the Notch
pathway. Two proteolytic cleavage events then occur. The rst involves an ADAM protease (a disintegrin and a metalloprotease) cleaving the Notch receptor and generating a
transmembrane fragment of Notch: TM Notch. TM Notch is then cleaved by a -secretase complex, the second cleavage event, leading to the release of the intracellular domain
of Notch: Nicd Notch. Nicd Notch migrates to the nucleus where it acts as a transcription factor. It interacts with RBP-J associated with corepressors allowing the recruitment
of co-activators, and the stimulation of the expression of target genes. These genes are involved in the inhibition of differentiation and the maintenance of cell renewal. The
activation of the Notch pathway promotes the proliferation and renewal of satellite cells following injury, and the inhibition of this pathway by Numb protein allows the
satellite cells to enter the differentiation program. During the differentiation phase, the canonical Wnt pathway is activated. Wnt proteins bind to the Frizzeld (Fzd) receptor
and to the co-receptor low-density lipoprotein receptor-related protein (LRP). Heterotrimeric G proteins and disheveled (Dsh) are then activated, leading to the release of
-catenin from its complex of degradation made of axin, casein kinase 1 (CK1), adenomatous polyposis coli (APC) and serine-threonine kinase glycogen synthase kinase-3
(GSK-3). Thus -catenin accumulates in the cytoplasm and translocates into the nucleus, where it acts as a transcriptional co-activator. -catenin binds to the TCF/LEF
family of transcription factors. In this context, canonical Wnt signaling leads to the expression of genes involved in the regulation of the muscle cell differentiation. During
skeletal muscle regeneration, a switch from Notch to canonical Wnt signaling is necessary for the transition of the proliferation phase to the differentiation phase in satellite
cells.

tion capacity with age appears to be reversible. This observation cle cells, likely contributing to the decrease in muscle regeneration
opens up the possibility of devising strategies for slowing the with age. The dialog between these two cell types has been widely
decline of the process. studied in in vitro muscle regeneration models or using young ani-
mals (for review (Pillon et al., 2013; Tidball and Villalta, 2010)),
but infrequently in a context of aging. During muscle regeneration,
4. Effect of aging on immune and muscle cell cross-talk and macrophages protect satellite cells from apoptosis (Sonnet et al.,
consequences for skeletal muscle repair 2006). However, this protection is blunted during aging (Krajnak
et al., 2006). It has been shown that numbers of activated satellite
With aging, the efcacy of muscle regeneration is signicantly cells (i.e. those expressing MyoD) increase in both aged (30 month)
decreased. Studies have demonstrated that the reduction in the and young (3 month) rats after a muscle lesion (Krajnak et al., 2006).
potential of muscle to regenerate is due to a decrease in the activa- Nevertheless, the number of satellite cells expressing MyoD and the
tion, proliferation and renewal of the satellite cells, as well as in the pro-apoptotic marker Bax were higher in aged rats, contributing to
differentiation into myobers, although some of the observations the reduced muscle regeneration in these animals (Krajnak et al.,
reported in the literature are controversial (Ciciliot and Schiafno, 2006).
2010; Garcia-Prat et al., 2013; Shefer et al., 2006). The alteration In vitro, the percentage of satellite cells expressing activated
of the immune response with aging is also a muscle regeneration caspases following TNF- treatment was augmented in muscle cell
disturbing factor due to its role in the stimulation of satellite cell population isolated from aged rats (31 months) compared with
proliferation and differentiation (Shaw et al., 2013). Immunose- adult (9 months) or young rats (3 months) (Jejurikar et al., 2006).
nescence contributes to the development of a chronic low-grade Of note, the expression of the anti-apoptotic marker Bcl-2 (B-cell
inammation state. We hypothesize that this phenomenon, in addi- lymphoma 2) was reduced in aged compared with adult and young
tion to the age-related changes of muscle cell metabolism and rats (Jejurikar et al., 2006). These results demonstrate that muscle
function, could disrupt the cross-talk between immune and mus-
C. Domingues-Faria et al. / Ageing Research Reviews 26 (2016) 2236 31

satellite cells are likely more sensitive to apoptosis process with The profound changes in the immune and muscle cell functions
aging. disrupt the metabolic and functional cross-talk between these two
All these ndings show that environmental and intrinsic cell types, and so modify muscle regeneration. Additional studies
changes in satellite cells with aging make them more suscepti- are needed to understand the consequences of aging on muscle
ble to apoptosis, rendering muscle regeneration less efcient. In and immune cell function, and thereby on muscle regeneration.
addition, the increased sensitivity of satellite cells to TNF- implies The specic mechanisms and molecular pathways that are modi-
that functional and secretory modications of immune cells, jointly ed with aging remain to be elucidated. Such work will help us to
with age-related changes in muscle cells, leads to the perturbation devise strategies to preserve or improve the capacity of muscle to
of immune/muscle cell cross-talk, and thus impairs muscle repair regenerate as we age.
capacity.
The involvement of functional modications of immune cells
during muscle regeneration and with aging has been demon- 5. Nutrition and muscle regeneration
strated in humans. In the basal state, muscles of older men (71
years) contained fewer macrophages than those of young men Dietary habits directly impact the capacities of muscle to regen-
(32 years) (Przybyla et al., 2006). Furthermore, pro-inammatory erate. In recent years, some studies have demonstrated that diets
macrophages were less abundant, whereas the number of anti- and/or nutrients can modulate muscle repair capacity, even in early
inammatory macrophages remained unchanged compared with life. In mice exposed to a prenatal undernutrition and/or to a post-
young volunteers (Przybyla et al., 2006). After an acute resistance natal high-fat diet, frequencies (% of live cells) of skeletal muscle
exercise, the number of pro- and anti-inammatory macrophages precursors and muscle regeneration were reduced following freeze
rose in the muscles of young men, but not in older men (Przybyla injury (Woo et al., 2011).
et al., 2006). Interestingly, at the basal state, gene expressions of Obesity is associated with a decrease in the potential of skeletal
pro-inammatory cytokine IL-1 and anti-inammatory cytokines muscle to regenerate (for review (Akhmedov and Berdeaux, 2013)).
IL-1RA and IL-10 were increased in muscles of elderly subjects com- In obese individuals, lipids accumulate excessively in adipose tis-
pared with younger ones (Przybyla et al., 2006). After exercise, the sues, and ectopically in non-adipose tissues like skeletal muscles.
expression of these cytokines was unchanged in older subjects, but In this tissue, accumulated lipids are likely able to impair the repair
was stimulated in the younger group (Przybyla et al., 2006). It has process following injury. Moreover, obesity is recognized as a state
also been demonstrated that 2 h after acute exercise, the expres- of chronic inammation (Akhmedov and Berdeaux, 2013). Acute
sions of pro-inammatory cytokines MCP-1, IL-6 and IL-8 were inammation is one of the main steps in the process of muscle
greater in elderly than in young subjects, but a 12-week period of regeneration (Tidball and Villalta, 2010), and so the tight cross-talk
training attenuated this difference (Della Gatta et al., 2014). These between muscle and immune cells is likely to be modied in such
ndings evidence a failure in the regulation of macrophage function a situation. Interestingly, loss of muscle mass with no loss of body
with aging, leading to altered immune response following muscle weight is systematically reported with aging, as signicant accu-
injury, and consequently to the modication of immune and mus- mulation of body fat occurs. Accumulation of intramuscular fat was
cle cell cross-talk. It is noteworthy that exercise is not equal to observed in this context (Walrand et al., 2011). Thus aging together
muscle injury as the mechanism of skeletal muscle repair is differ- with tissue lipotoxicity likely impairs skeletal muscle regeneration.
ent (Paulsen et al., 2012). To elucidate skeletal muscle regeneration Beside the effect of lipid inltration, nutritional strategies to
process in animals, protocols specically designed to induce exten- stimulate muscle regeneration have been attempted. In young
sive muscle damages (infusion of cardiotoxin, infusion of myotoxin, healthy subjects, whey protein supplementation seems to accel-
crush injury. . .) have been used. Obviously, such muscle injuries erate the proliferation of satellite cells following high-intensity
are ethically non-feasible in humans. Thus, protocols of exercises eccentric exercise (Farup et al., 2014). Dietary whey protein is rich
have been used to induce muscle lesions in order to study skeletal in leucine, an essential amino acid. Pereira et al. (2014a) have
muscle regeneration in humans (Przybyla et al., 2006). demonstrated that leucine supplementation increases myober
An in vitro study clearly demonstrated that aged muscle cells size, and limits the decrease in muscle function and macrophage
were less sensitive to immune secretions (Dumke and Lees, 2011). A inltration following the cryolesion of young rat muscles. How-
T cell-conditioned medium stimulated the proliferation and migra- ever, the overall macrophage population was investigated without
tion of satellite cells isolated from muscles of young rats, but failed distinction of phenotypes. Author concluded from this result
to produce the same effects on satellite cells isolated from mus- that the regenerating muscles supplemented with leucine might
cles of old rats. Finally, this environment, i.e., a T cell-conditioned have an acceleration of inammatory process installation and a
medium, did not modify the differentiation of satellite cells from more efcient function of immune cells stimulated by increased
young animals, but inhibited the differentiation of satellite cells amino acid substrates (Pereira et al., 2014a). This modication
sampled in old rats (Dumke and Lees, 2011). of immune response following leucine supplementation likely
These results demonstrate that the satellite cell response to improves muscle regeneration capacity. Finally, leucine supple-
immune cells changes with aging, as well as the immune response mentation increased the expression of type II myosin heavy chain,
to muscle cells. This alteration likely contributes to the age-related limited the development of brosis, and prevented the decrease
change in the muscle regeneration process. in muscle function in regenerating skeletal muscles (Pereira et al.,
Of note in vitro and animal studies have demonstrated that aged 2014b).
muscle cells are less sensitive to immune cell effects, i.e. protec- Beside whey protein, benecial effects of codsh protein on
tion from apoptosis and stimulation of proliferation and migration. skeletal muscle recovery have been reported (Dort et al., 2012). In
In addition, it has been demonstrated in humans that inltration rats fed with casein or codsh proteins, codsh protein supplemen-
of immune cells following muscle injury is modied during aging. tation decreased M1 macrophage inltration in the muscle lesion,
This results predicts a possible alteration of the cross-talk between whereas it increased M2 macrophage inltration and myober
immune and muscle cells that can possibly alter skeletal muscle cross-sectional area compared with casein group (Dort et al.,
repair. In this context investigations are needed to understand the 2012). Authors concluded that effects of codsh protein might
consequences of aging on immune and muscle cell cross-talk to result in a reduced necrotic zone and an accelerated switch of
boost muscle repair efciency during aging. pro-inammatory M1 macrophages toward the anti-inammatory
phenotype (M2), leading to the enhancement of the muscle repair
32 C. Domingues-Faria et al. / Ageing Research Reviews 26 (2016) 2236

efcacy (Dort et al., 2012). The anti-inammatory action of codsh results demonstrate the direct effect of vitamin D on muscle cells,
protein seems to be driven by its high levels of arginine, glycine, leading to the improvement of skeletal muscle recovery.
taurine and lysine (Dort et al., 2013). Taken together, these ndings Knowing that vitamin D has effects not only on muscle cells
show that the quality of dietary proteins is important for improving (Ceglia and Harris, 2013), but also on immune cell functions
skeletal muscle regeneration through the modulation of immune (Hewison, 2011), it seems important to investigate the action of
response. vitamin D on specic immune response during muscle regenera-
Long chain n-3 polyunsaturated fatty acids (n-3 PUFAs) such as tion.
docosahexaenoic acid (DHA) or eicosapentaenoic acid (EPA) have In an animal model of skeletal muscle regeneration, the num-
been described as presenting a strong anti-inammatory action ber of inltrated immune cells on injury site was not modied
(Calder, 2006). In this context, n-3 PUFAs are likely able to modulate by vitamin infusion (Stratos et al., 2013). Of note, in this study,
immune and muscle cell cross-talk. the overall immune cell population was investigated without dis-
It has been demonstrated that EPA treatment prevents TNF- tinction of immune cell types. Interestingly in another rodent
-mediated loss of myosin heavy chain expression in C2C12 model, vitamin D infusion interfered with the increase in pro-
myotubes (Magee et al., 2008). Furthermore, EPA protected C2C12 inammatory cytokine expressions in muscle following intensive
from TNF--mediated cell death and increased myogenic fusion physical exercise in rats (Choi et al., 2013). In addition, it has
(Magee et al., 2008). been demonstrated that vitamin D-decient subjects displayed a
Treatment with DHA of human myotubes cocultured with decrease of blood concentration of anti-inammatory cytokines
inammatory macrophages reduced the expression of the muscle- IL-10 and IL-13 following physical training, compared to healthy
specic ubiquitin ligase muscle RING-nger protein-1 (MuRF1) volunteers. Furthermore, muscle strength was more rapidly recov-
involved in proteolysis (Murton et al., 2008). In this context, DHA ered in non-decient men (Barker et al., 2013, 2014). Taken
may favor protein anabolism by inhibiting proteolysis, which pro- together, this results highlight that vitamin D is able to modulate
motes recovery of regenerating skeletal muscles as previously circulating and muscle-specic cytokine expression, and possibly
described for leucine supplementation (Pereira et al., 2014a). These immune and muscle cell cross-talk during muscle repair. These
in vitro studies demonstrate that long chain n-3 PUFAs may have a studies conrm that vitamin D likely modulates immune response
benecial effect on skeletal muscle regeneration. following muscle injury.
Polyphenols also belong to the family of nutrients displaying Finally, our research group have demonstrated in old rats that
anti-inammatory properties. The intake of these compounds may vitamin D deciency down-regulates the Notch pathway, known to
therefore modify the cross-talk between immune and muscle cells, play a leading role in muscle regeneration (Domingues-Faria et al.,
and consequently the time course of muscle regeneration. In rats, 2014). In this context, vitamin D deciency could aggravate the
the administration of proanthocyanidolic oligomer (PCO) resulted age-related decrease in muscle regeneration capacity.
in earlier activation of satellite cells following muscle contusion Taken together, these results show that the effect of nutrition on
injury (Myburgh et al., 2012). Neutrophil number was also blunted skeletal muscle regeneration efcacy is an emerging topic of inter-
and macrophage response was normal but earlier in the muscle est. Unfortunately, investigations on nutrition and muscle repair in
of PCO group. Muscle ber regeneration was more precocious in the context of aging are very scarce. More research are needed in
the muscle of PCO group (Myburgh et al., 2012). These ndings this scientic area to dene strategies able to prevent ineffective
demonstrate that the modulation of inammatory response can and/or incomplete muscle repair. This would be of great interest
improve skeletal muscle regeneration. particularly in older people in which ineffective muscle repair con-
The consumption of a diet enriched with green tea extract rich tributes to muscle mass loss and sarcopenia (Walrand et al., 2011).
in polyphenols decreased serum creatine kinase, an indicator of Nutritional strategies need to be considered not only during aging
muscle damage, in mdx mice (Evans et al., 2010). In addition, the but in all situations where muscle repair may be committed: in
area of muscle ber was increased, whereas regenerating ber area pathologies associated with muscle damage, or among athletes,
was reduced. Lastly, NF-B activity was diminished in regenerat- who regularly undergo muscle regeneration processes.
ing muscle bers of mdx mice supplemented with green tea extract Note that the effect of n-3 PUFAs on muscle cells have only
(Evans et al., 2010). In dystrophic muscle pathology, muscle degen- been demonstrated in an in vitro model and further conrma-
eration is a characteristic of the disease and skeletal muscle repair is tion in rodents and/or humans have to be undertaken. However,
unable to occur successfully (Tidball and Wehling-Henricks, 2004). the possible benecial effects of nutrition and nutrients, i.e. pro-
These results demonstrate that green tea extract decreases dys- teins, polyphenols and vitamin D, in muscle regeneration have been
trophic muscle pathology potentially by regulating NF-kB activity highlighted in animal models and human. Nutritional strategies
in regenerating muscle bers and by limiting degenerative areas. have to be taken into consideration to improve skeletal muscle
Vitamin D is likely a good candidate to stimulate muscle recov- regeneration, particularly during aging, but more investigations
ery, as immune and muscle cells are preferred targets of this are needed in this area. To this day, vitamin D supplementation
nutrient (Ceglia and Harris, 2013; Hewison, 2011). Srikuea et al. seems to be a meaningful nutritional strategy to improve skeletal
(2012) have demonstrated that expressions of vitamin D recep- muscle regeneration in young and old people. Numerous studies
tor (VDR) and 25-hydroxyvitamin D3 1--hydroxylase (also called have demonstrated that immune and muscle cells are vitamin D
CYP27B1) were up-regulated following acute lesions in adult mice. targets (Ceglia and Harris, 2013; Hewison, 2011), these two types
In another animal model, the infusion of vitamin D3 led to an of cells being implicated in muscle repair (Tidball and Villalta,
increase in proliferation and a decrease in apoptosis in injured mus- 2010). During skeletal muscle regeneration, markers of vitamin D
cle (Stratos et al., 2013). Owens et al. (2015) have demonstrated metabolism are up-regulated showing that vitamin D is implicated
that muscle cell migration dynamics, myotube fusion, differentia- in this process (Srikuea et al., 2012). Clinical trials have demon-
tion and hyperthophy of myoblasts derived from biopsies of young strated that vitamin D insufciency impairs functional recovery
volunteers presenting vitamin D insufciency were improved fol- of skeletal muscle after damaging physical exercise (Barker et al.,
lowing vitamin D treatment. In a clinical trial, the same research 2013; Owens et al., 2015). Furthermore, vitamin D supplementa-
team highlighted that the supplementation in vitamin D3 of young tion leads to the improvement of skeletal muscle recovery (Owens
men presenting vitamin D insufciency leads to the improvement et al., 2015). Thus, vitamin D seems to be a promising nutritional
of peak torque recovery following damaging eccentric contractions strategy to stimulate skeletal muscle repair. Interestingly, skele-
of the knee extensors (Owens et al., 2015). Taken together, this tal muscle regeneration capacity is impaired with aging and it is
C. Domingues-Faria et al. / Ageing Research Reviews 26 (2016) 2236 33

considered as a key factor of sarcopenia (Decary et al., 1997; Jang skeletal muscle injury switch into antiinammatory macrophages to support
et al., 2011; Lenk et al., 2010). In parallel, vitamin D deciency is myogenesis. J. Exp. Med. 204, 10571069.
Arthur, S.T., Cooley, I.D., 2012. The effect of physiological stimuli on sarcopenia;
very common in elderly population and has been associated with impact of Notch and Wnt signaling on impaired aged skeletal muscle repair.
muscle weakness (Holick, 2007). In this context, vitamin D sup- Int. J. Biol. Sci. 8, 731760.
plementation is likely the meaningful strategy to enhance skeletal Baeza-Raja, B., Munoz-Canoves, P., 2004. p38 MAPK-induced nuclear
factor-kappaB activity is required for skeletal muscle differentiation: role of
muscle regeneration in old subjects in order to limit sarcopenia interleukin-6. Mol. Biol. Cell 15, 20132026.
development. Barani, A.E., Durieux, A.C., Sabido, O., Freyssenet, D., 2003. Age-related changes in
the mitotic and metabolic characteristics of muscle-derived cells. J. Appl.
Physiol. (1985) 95, 20892098.
6. Conclusion Barberi, L., Scicchitano, B.M., De Rossi, M., Bigot, A., Duguez, S., Wielgosik, A.,
Stewart, C., McPhee, J., Conte, M., Narici, M., Franceschi, C., Mouly, V.,
Butler-Browne, G., Musaro, A., 2013. Age-dependent alteration in muscle
Muscle regeneration is a highly controlled and complex pro- regeneration: the critical role of tissue niche. Biogerontology 14, 273292.
cess, with the intervention of many cell types. Any change in the Barker, T., Henriksen, V.T., Martins, T.B., Hill, H.R., Kjeldsberg, C.R., Schneider, E.D.,
functional capacities of one of these cells may compromise skeletal Dixon, B.M., Weaver, L.K., 2013. Higher serum 25-hydroxyvitamin D
concentrations associate with a faster recovery of skeletal muscle strength
muscle repair. During aging, the alteration of satellite cell activa-
after muscular injury. Nutrients 5, 12531275.
tion, proliferation and differentiation, jointly with the changes in Barker, T., Martins, T.B., Hill, H.R., Kjeldsberg, C.R., Dixon, B.M., Schneider, E.D.,
immune cell functions and ability to secrete key controllers, impair Henriksen, V.T., Weaver, L.K., 2014. Vitamin D sufciency associates with an
increase in anti-inammatory cytokines after intense exercise in humans.
the capacity of skeletal muscle to regenerate. The decreased ef-
Cytokine 65, 134137.
ciency of skeletal muscle recovery is one of the factors contributing Barnard, W., Bower, J., Brown, M.A., Murphy, M., Austin, L., 1994. Leukemia
to sarcopenia (Ali and Garcia, 2014; Beyer et al., 2012), dened as inhibitory factor (LIF) infusion stimulates skeletal muscle regeneration after
the loss of skeletal muscle mass and strength as we age. Sarcopenia injury: injured muscle expresses lif mRNA. J. Neurol. Sci. 123, 108113.
Beccaco, S., Puglielli, C., Pietrangelo, T., Bellomo, R., Fano, G., Fulle, S., 2007.
is one of the most important factors of disability in older people Age-dependent effects on functional aspects in human satellite cells. Ann. N. Y.
(Walrand et al., 2011). Thus strategies need to be developed to Acad. Sci. 1100, 345352.
counteract or at least to limit the inefciency of muscle repair with Bencze, M., Negroni, E., Vallese, D., Yacoub-Youssef, H., Chaouch, S., Wolff, A.,
Aamiri, A., Di Santo, J.P., Chazaud, B., Butler-Browne, G., Savino, W., Mouly, V.,
aging. Nutritional strategies seem to be a good and a safe way to Riederer, I., 2012. Proinammatory macrophages enhance the regenerative
improve muscle regeneration with aging, as some nutrients are able capacity of human myoblasts by modifying their kinetics of proliferation and
to modulate immune or muscle cell functions, or both. In this con- differentiation. Mol. Ther. 20, 21682179.
Bentzinger, C.F., Rudnicki, M.A., 2014. Rejuvenating aged muscle stem cells. Nat.
text, the quality of both diet and nutrient supplementation has to Med. 20, 234235.
be oriented to improve and maintain muscle regeneration capacity. Beyer, I., Mets, T., Bautmans, I., 2012. Chronic low-grade inammation and
Developing these nutritional strategies will be of great interest not age-related sarcopenia. Curr. Opin. Clin. Nutr. Metab. Care 15, 1222.
Bigot, A., Jacquemin, V., Debacq-Chainiaux, F., Butler-Browne, G.S., Toussaint, O.,
only in the context of aging, but also in all situations leading to mus-
Furling, D., Mouly, V., 2008. Replicative aging down-regulates the myogenic
cle damage and consequent muscle regeneration. This can be the regulatory factors in human myoblasts. Biol. Cell/Under Auspices Eur. Cell Biol.
case for chronic pathologies associated with muscle damage such as Org. 100, 189199.
Boren, E., Gershwin, M.E., 2004. Inamm-aging: autoimmunity, and the
cachexia. Finally, some non-pathological situations are associated
immune-risk phenotype. Autoimmun. Rev. 3, 401406.
with strong and frequent periods of skeletal muscle regeneration, Brack, A.S., Conboy, I.M., Conboy, M.J., Shen, J., Rando, T.A., 2008. A temporal switch
e.g., among athletes. In this population, dietary prescription has to from notch to Wnt signaling in muscle stem cells is necessary for normal adult
consider improvement of both physical performance and muscle myogenesis. Cell Stem Cell 2, 5059.
Brack, A.S., Conboy, M.J., Roy, S., Lee, M., Kuo, C.J., Keller, C., Rando, T.A., 2007.
regeneration capacities. A suitable diet could help lower recovery Increased Wnt signaling during aging alters muscle stem cell fate and
time, and improve the quality of muscle repair between successive increases brosis. Science 317, 807810.
physical training sessions. Brigitte, M., Schilte, C., Plonquet, A., Baba-Amer, Y., Henri, A., Charlier, C.,
Tajbakhsh, S., Albert, M., Gherardi, R.K., Chretien, F., 2010. Muscle resident
macrophages control the immune cell reaction in a mouse model of
Conicts of interest notexin-induced myoinjury. Arthritis Rheum. 62, 268279.
Broholm, C., Laye, M.J., Brandt, C., Vadalasetty, R., Pilegaard, H., Pedersen, B.K.,
Scheele, C., 2011. LIF is a contraction-induced myokine stimulating human
All the authors declare that there are no conicts of interest. myocyte proliferation. J. Appl. Physiol. (1985) 111, 251259.
Broholm, C., Pedersen, B.K., 2010. Leukaemia inhibitory factoran
exercise-induced myokine. Exerc. Immunol. Rev. 16, 7785.
Acknowledgment Burzyn, D., Kuswanto, W., Kolodin, D., Shadrach, J.L., Cerletti, M., Jang, Y., Sek, E.,
Tan, T.G., Wagers, A.J., Benoist, C., Mathis, D., 2013. A special population of
regulatory T cells potentiates muscle repair. Cell 155, 12821295.
Figs. 1 and 2 were produced with the assistance of Servier Med- Calder, P.C., 2006. n-3 polyunsaturated fatty acids, inammation, and
ical Art. inammatory diseases. Am. J. Clin. Nutr. 83, 1505S1519S.
Camous, X., Pera, A., Solana, R., Larbi, A., 2012. NK cells in healthy aging and
age-associated diseases. J. Biomed. Biotechnol. 2012, 195956.
References Cantini, M., Giurisato, E., Radu, C., Tiozzo, S., Pampinella, F., Senigaglia, D., Zaniolo,
G., Mazzoleni, F., Vitiello, L., 2002. Macrophage-secreted myogenic factors: a
Agarwal, S., Busse, P.J., 2010. Innate and adaptive immunosenescence. Ann. Allergy promising tool for greatly enhancing the proliferative capacity of myoblasts
Asthma Immunol. 104, 183190, quiz 190-182, 210. in vitro and in vivo. Neurol. Sci. 23, 189194.
Agrawal, A., Agrawal, S., Cao, J.N., Su, H., Osann, K., Gupta, S., 2007. Altered innate Carlson, B.M., Faulkner, J.A., 1989. Muscle transplantation between young and old
immune functioning of dendritic cells in elderly humans: a role of rats: age of host determines recovery. Am. J. Physiol. 256, C12621266.
phosphoinositide 3-kinase-signaling pathway. J. Immunol. 178, 69126922. Carlson, M.E., Suetta, C., Conboy, M.J., Aagaard, P., Mackey, A., Kjaer, M., Conboy, I.,
Akhmedov, D., Berdeaux, R., 2013. The effects of obesity on skeletal muscle 2009. Molecular aging and rejuvenation of human muscle stem cells. EMBO
regeneration. Front. Physiol. 4, 371. Mol. Med. 1, 381391.
Ali, S., Garcia, J.M., 2014. Sarcopenia, cachexia and aging: diagnosis, mechanisms Castelo-Branco, C., Soveral, I., 2014. The immune system and aging: a review.
and therapeutic optionsa mini-review. Gerontology 60 (4), 294305. Gynecol. Endocrinol. 30, 1622.
Almeida-Oliveira, A., Smith-Carvalho, M., Porto, L.C., Cardoso-Oliveira, J., Ribeiro Ceglia, L., Harris, S.S., 2013. Vitamin D and its role in skeletal muscle. Calcif. Tissue
Ados, S., Falcao, R.R., Abdelhay, E., Bouzas, L.F., Thuler, L.C., Ornellas, M.H., Int. 92, 151162.
Diamond, H.R., 2011. Age-related changes in natural killer cell receptors from Chakkalakal, J.V., Jones, K.M., Basson, M.A., Brack, A.S., 2012. The aged niche
childhood through old age. Hum. Immunol. 72, 319329. disrupts muscle stem cell quiescence. Nature 490, 355360.
Alway, S.E., Degens, H., Lowe, D.A., Krishnamurthy, G., 2002. Increased myogenic Charge, S.B., Brack, A.S., Hughes, S.M., 2002. Aging-related satellite cell
repressor Id mRNA and protein levels in hindlimb muscles of aged rats. Am. J. differentiation defect occurs prematurely after Ski-induced muscle
Physiol. Regul. Integr. Comp. Physiol. 282, R411R422. hypertrophy. Am. J. Physiol. Cell Physiol. 283, C1228C1241.
Arnold, L., Henry, A., Poron, F., Baba-Amer, Y., van Rooijen, N., Plonquet, A., Chatta, G.S., Price, T.H., Stratton, J.R., Dale, D.C., 1994. Aging and marrow neutrophil
Gherardi, R.K., Chazaud, B., 2007. Inammatory monocytes recruited after reserves. J. Am. Geriatr. Soc. 42, 7781.
34 C. Domingues-Faria et al. / Ageing Research Reviews 26 (2016) 2236

Chazaud, B., Brigitte, M., Yacoub-Youssef, H., Arnold, L., Gherardi, R., Sonnet, C., Gallucci, S., Provenzano, C., Mazzarelli, P., Scuderi, F., Bartoccioni, E., 1998.
Lafuste, P., Chretien, F., 2009. Dual and benecial roles of macrophages during Myoblasts produce IL-6 in response to inammatory stimuli. Int. Immunol. 10,
skeletal muscle regeneration. Exerc. Sport Sci. Rev. 37, 1822. 267273.
Chazaud, B., Sonnet, C., Lafuste, P., Bassez, G., Rimaniol, A.C., Poron, F., Authier, F.J., Garcia-Prat, L., Sousa-Victor, P., Munoz-Canoves, P., 2013. Functional dysregulation
Dreyfus, P.A., Gherardi, R.K., 2003. Satellite cells attract monocytes and use of stem cells during aging: a focus on skeletal muscle stem cells. FEBS J. 280,
macrophages as a support to escape apoptosis and enhance muscle growth. J. 40514062.
Cell Biol. 163, 11331143. Garg, S.K., Delaney, C., Toubai, T., Ghosh, A., Reddy, P., Banerjee, R., Yung, R., 2014.
Chen, S.E., Jin, B., Li, Y.P., 2007. TNF-alpha regulates myogenesis and muscle Aging is associated with increased regulatory T-cell function. Aging Cell 13,
regeneration by activating p38 MAPK. Am. J. Physiol. Cell Physiol. 292, 441448.
C1660C1671. Ghosh, S., May, M.J., Kopp, E.B., 1998. NF-kappa B and Rel proteins: evolutionarily
Cheng, M., Nguyen, M.H., Fantuzzi, G., Koh, T.J., 2008. Endogenous conserved mediators of immune responses. Ann. Rev. Immunol. 16, 225260.
interferon-gamma is required for efcient skeletal muscle regeneration. Am. J. Gomez, C.R., Nomellini, V., Faunce, D.E., Kovacs, E.J., 2008. Innate immunity and
Physiol. Cell Physiol. 294, C1183C1191. aging. Exp. Gerontol. 43, 718728.
Choi, M., Park, H., Cho, S., Lee, M., 2013. Vitamin D3 supplementation modulates Guttridge, D.C., Albanese, C., Reuther, J.Y., Pestell, R.G., Baldwin Jr., A.S., 1999.
inammatory responses from the muscle damage induced by high-intensity NF-kappaB controls cell growth and differentiation through transcriptional
exercise in SD rats. Cytokine 63, 2735. regulation of cyclin D1. Mol. Cell. Biol. 19, 57855799.
Ciciliot, S., Schiafno, S., 2010. Regeneration of mammalian skeletal muscle. Basic Harris, N.R., Rumbaut, R.E., 2001. Age-related responses of the microcirculation to
Mech. Clin. Implic. Curr. Pharm. Des. 16, 906914. ischemia-reperfusion and inammation. Pathophysiology 8, 110.
Collins, C.A., Zammit, P.S., Ruiz, A.P., Morgan, J.E., Partridge, T.A., 2007. A population Haugen, F., Norheim, F., Lian, H., Wensaas, A.J., Dueland, S., Berg, O., Funderud, A.,
of myogenic stem cells that survives skeletal muscle aging. Stem Cells 25, Skalhegg, B.S., Raastad, T., Drevon, C.A., 2010. IL-7 is expressed and secreted by
885894. human skeletal muscle cells. Am. J. Physiol. Cell Physiol. 298, C807C816.
Collins, R.A., Grounds, M.D., 2001. The role of tumor necrosis factor-alpha Hawke, T.J., Garry, D.J., 2001. Myogenic satellite cells: physiology to molecular
(TNF-alpha) in skeletal muscle regeneration. Studies in TNF-alpha(-/-) and biology. J. Appl. Physiol. (1985) 91, 534551.
TNF-alpha(-/-)/LT-alpha(-/-) mice. J. Histochem. Cytochem. 49, 9891001. Hearps, A.C., Martin, G.E., Angelovich, T.A., Cheng, W.J., Maisa, A., Landay, A.L.,
Conboy, I.M., Conboy, M.J., Smythe, G.M., Rando, T.A., 2003. Notch-mediated Jaworowski, A., Crowe, S.M., 2012. Aging is associated with chronic innate
restoration of regenerative potential to aged muscle. Science 302, 15751577. immune activation and dysregulation of monocyte phenotype and function.
Conboy, I.M., Conboy, M.J., Wagers, A.J., Girma, E.R., Weissman, I.L., Rando, T.A., Aging Cell 11, 867875.
2005. Rejuvenation of aged progenitor cells by exposure to a young systemic Hewison, M., 2011. Vitamin D and innate and adaptive immunity. Vitam. Horm. 86,
environment. Nature 433, 760764. 2362.
Conboy, I.M., Rando, T.A., 2002. The regulation of Notch signaling controls satellite Hoene, M., Runge, H., Haring, H.U., Schleicher, E.D., Weigert, C., 2013. Interleukin-6
cell activation and cell fate determination in postnatal myogenesis. Dev. Cell 3, promotes myogenic differentiation of mouse skeletal muscle cells: role of the
397409. STAT3 pathway. Am. J. Physiol. Cell Physiol. 304, C128C136.
Day, K., Shefer, G., Shearer, A., Yablonka-Reuveni, Z., 2010. The depletion of skeletal Holick, M.F., 2007. Vitamin D deciency. N. Engl. J. Med. 357, 266281.
muscle satellite cells with age is concomitant with reduced capacity of single Horsley, V., Jansen, K.M., Mills, S.T., Pavlath, G.K., 2003. IL-4 acts as a myoblast
progenitors to produce reserve progeny. Dev. Biol. 340, 330343. recruitment factor during mammalian muscle growth. Cell 113, 483494.
De Martinis, M., Modesti, M., Ginaldi, L., 2004. Phenotypic and functional changes Jang, Y.C., Sinha, M., Cerletti, M., DallOsso, C., Wagers, A.J., 2011. Skeletal muscle
of circulating monocytes and polymorphonuclear leucocytes from elderly stem cells: effects of aging and metabolism on muscle regenerative function.
persons. Immunol. Cell Biol. 82, 415420. Cold Spring Harb. Symp. Quant. Biol. 76, 101111.
Decary, S., Mouly, V., Hamida, C.B., Sautet, A., Barbet, J.P., Butler-Browne, G.S., 1997. Jejurikar, S.S., Henkelman, E.A., Cederna, P.S., Marcelo, C.L., Urbanchek, M.G., Kuzon
Replicative potential and telomere length in human skeletal muscle: Jr., W.M., 2006. Aging increases the susceptibility of skeletal muscle derived
implications for satellite cell-mediated gene therapy. Hum. Gene Ther. 8, satellite cells to apoptosis. Exp. Gerontol. 41, 828836.
14291438. Kharraz, Y., Guerra, J., Mann, C.J., Serrano, A.L., Munoz-Canoves, P., 2013.
Degens, H., 2007. Age-related skeletal muscle dysfunction: causes and Macrophage plasticity and the role of inammation in skeletal muscle repair.
mechanisms. J. Musculoskelet. Neuronal Interact. 7, 246252. Mediators inamm. 2013, 491497.
Della Gatta, P.A., Garnham, A.P., Peake, J.M., Cameron-Smith, D., 2014. Effect of Krajnak, K., Waugh, S., Miller, R., Baker, B., Geronilla, K., Alway, S.E., Cutlip, R.G.,
exercise training on skeletal muscle cytokine expression in the elderly. Brain 2006. Proapoptotic factor Bax is increased in satellite cells in the tibialis
Behav. Immun. 39, 8086. anterior muscles of old rats. Muscle Nerve 34, 720730.
Deng, B., Wehling-Henricks, M., Villalta, S.A., Wang, Y., Tidball, J.G., 2012. IL-10 Kurek, J.B., Nouri, S., Kannourakis, G., Murphy, M., Austin, L., 1996. Leukemia
triggers changes in macrophage phenotype that promote muscle growth and inhibitory factor and interleukin-6 are produced by diseased and regenerating
regeneration. J. Immunol. 189, 36693680. skeletal muscle. Muscle Nerve 19, 12911301.
Domingues-Faria, C., Chanet, A., Salles, J., Berry, A., Giraudet, C., Patrac, V., Denis, P., Langen, R.C., Schols, A.M., Kelders, M.C., Wouters, E.F., Janssen-Heininger, Y.M.,
Bouton, K., Goncalves-Mendes, N., Vasson, M.P., Boirie, Y., Walrand, S., 2014. 2001. Inammatory cytokines inhibit myogenic differentiation through
Vitamin D deciency down-regulates Notch pathway contributing to skeletal activation of nuclear factor-kappaB. FASEB J. 15, 11691180.
muscle atrophy in old wistar rats. Nutr. Metab. 11, 47. Langen, R.C., Van Der Velden, J.L., Schols, A.M., Kelders, M.C., Wouters, E.F.,
Dort, J., Leblanc, N., Maltais-Giguere, J., Liaset, B., Cote, C.H., Jacques, H., 2013. Janssen-Heininger, Y.M., 2004. Tumor necrosis factor-alpha inhibits myogenic
Benecial effects of cod protein on inammatory cell accumulation in rat differentiation through MyoD protein destabilization. FASEB J. 18, 227237.
skeletal muscle after injury are driven by its high levels of arginine, glycine, Lees, S.J., Rathbone, C.R., Booth, F.W., 2006. Age-associated decrease in muscle
taurine and lysine. PloS One 8, e77274. precursor cell differentiation. Am. J. Physiol. Cell Physiol. 290, C609C615.
Dort, J., Sirois, A., Leblanc, N., Cote, C.H., Jacques, H., 2012. Benecial effects of cod Lenk, K., Schuler, G., Adams, V., 2010. Skeletal muscle wasting in cachexia and
protein on skeletal muscle repair following injury. Appl. Physiol. Nutr. Metab. sarcopenia: molecular pathophysiology and impact of exercise training. J.
37, 489498. Cachexia Sarcopenia Muscle 1, 921.
Dumke, B.R., Lees, S.J., 2011. Age-related impairment of T cell-induced skeletal Lesault, P.F., Theret, M., Magnan, M., Cuvellier, S., Niu, Y., Gherardi, R.K., Tremblay,
muscle precursor cell function. Am. J. Physiol. Cell Physiol. 300, C1226C1233. J.P., Hittinger, L., Chazaud, B., 2012. Macrophages improve survival,
Ehrhardt, J., Morgan, J., 2005. Regenerative capacity of skeletal muscle. Curr. Opin. proliferation and migration of engrafted myogenic precursor cells into MDX
Neurol. 18, 548553. skeletal muscle. PloS One 7, e46698.
Evans, N.P., Call, J.A., Bassaganya-Riera, J., Robertson, J.L., Grange, R.W., 2010. Green Li, Y.P., 2003. TNF-alpha is a mitogen in skeletal muscle. Am. J. Physiol. Cell Physiol.
tea extract decreases muscle pathology and NF-kappaB immunostaining in 285, C370C376.
regenerating muscle bers of mdx mice. Clin. Nutr. 29, 391398. Lira, S.A., Zalamea, P., Heinrich, J.N., Fuentes, M.E., Carrasco, D., Lewin, A.C., Barton,
Fadok, V.A., Bratton, D.L., Guthrie, L., Henson, P.M., 2001. Differential effects of D.S., Durham, S., Bravo, R., 1994. Expression of the chemokine N51/KC in the
apoptotic versus lysed cells on macrophage production of cytokines: role of thymus and epidermis of transgenic mice results in marked inltration of a
proteases. J. Immunol. 166, 68476854. single class of inammatory cells. J. Exp. Med. 180, 20392048.
Fadok, V.A., Bratton, D.L., Konowal, A., Freed, P.W., Westcott, J.Y., Henson, P.M., Lolmede, K., Campana, L., Vezzoli, M., Bosurgi, L., Tonlorenzi, R., Clementi, E.,
1998. Macrophages that have ingested apoptotic cells in vitro inhibit Bianchi, M.E., Cossu, G., Manfredi, A.A., Brunelli, S., Rovere-Querini, P., 2009.
proinammatory cytokine production through autocrine/paracrine Inammatory and alternatively activated human macrophages attract
mechanisms involving TGF-beta, PGE2, and PAF. J. Clin. Investig. 101, 890898. vessel-associated stem cells, relying on separate HMGB1- and
Farup, J., Rahbek, S.K., Knudsen, I.S., de Paoli, F., Mackey, A.L., Vissing, K., 2014. MMP-9-dependent pathways. J. Leukoc. Biol. 85, 779787.
Whey protein supplementation accelerates satellite cell proliferation during Lu, H., Huang, D., Ransohoff, R.M., Zhou, L., 2011a. Acute skeletal muscle injury:
recovery from eccentric exercise. Amino Acids 46, 25032516. CCL2 expression by both monocytes and injured muscle is required for repair.
Franceschi, C., Campisi, J., 2014. Chronic inammation (inammaging) and its FASEB J. 25, 33443355.
potential contribution to age-associated diseases. J. Gerontol. A Biol. Sci. Med. Lu, H., Huang, D., Saederup, N., Charo, I.F., Ransohoff, R.M., Zhou, L., 2011b.
Sci. 69 (Suppl. 1), S4S9. Macrophages recruited via CCR2 produce insulin-like growth factor-1 to repair
Frenette, J., Cai, B., Tidball, J.G., 2000. Complement activation promotes muscle acute skeletal muscle injury. FASEB J. 25, 358369.
inammation during modied muscle use. Am. J. Pathol. 156, 21032110. Magee, P., Pearson, S., Allen, J., 2008. The omega-3 fatty acid, eicosapentaenoic acid
Fulop, T., Larbi, A., Kotb, R., de Angelis, F., Pawelec, G., 2011. Aging, immunity, and (EPA), prevents the damaging effects of tumour necrosis factor (TNF)-alpha
cancer. Discov. Med. 11, 537550. during murine skeletal muscle cell differentiation. Lipids Health Dis. 7, 24.
C. Domingues-Faria et al. / Ageing Research Reviews 26 (2016) 2236 35

Mantovani, A., Sica, A., Sozzani, S., Allavena, P., Vecchi, A., Locati, M., 2004. The monocyte-macrophage responses in vitro, in resolving skin blisters in vivo,
chemokine system in diverse forms of macrophage activation and polarization. and after cardiopulmonary bypass surgery. Circ. Res. 94, 119126.
Trends Immunol. 25, 677686. Pillon, N.J., Bilan, P.J., Fink, L.N., Klip, A., 2013. Cross-talk between skeletal muscle
Martinez, C.O., McHale, M.J., Wells, J.T., Ochoa, O., Michalek, J.E., McManus, L.M., and immune cells: muscle-derived mediators and metabolic implications. Am.
Shireman, P.K., 2010. Regulation of skeletal muscle regeneration by J. Physiol. Endocrinol. Metab. 304, E453E465.
CCR2-activating chemokines is directly related to macrophage recruitment. Pizza, F.X., Peterson, J.M., Baas, J.H., Koh, T.J., 2005. Neutrophils contribute to
Am. J. Physiol. Regul. Integr. Comp. Physiol. 299, R832R842. muscle injury and impair its resolution after lengthening contractions in mice.
Mathur, S.K., Schwantes, E.A., Jarjour, N.N., Busse, W.W., 2008. Age-related changes J. Physiol. 562, 899913.
in eosinophil function in human subjects. Chest 133, 412419. Przybyla, B., Gurley, C., Harvey, J.F., Bearden, E., Kortebein, P., Evans, W.J., Sullivan,
Merly, F., Lescaudron, L., Rouaud, T., Crossin, F., Gardahaut, M.F., 1999. D.H., Peterson, C.A., Dennis, R.A., 2006. Aging alters macrophage properties in
Macrophages enhance muscle satellite cell proliferation and delay their human skeletal muscle both at rest and in response to acute resistance
differentiation. Muscle Nerve 22, 724732. exercise. Exp. Gerontol. 41, 320327.
Mertens, A., Holvoet, P., 2001. Oxidized LDL and HDL: antagonists in Qian, F., Wang, X., Zhang, L., Chen, S., Piecychna, M., Allore, H., Bockenstedt, L.,
atherothrombosis. FASEB J. 15, 20732084. Malawista, S., Bucala, R., Shaw, A.C., Fikrig, E., Montgomery, R.R., 2012.
Montoya-Ortiz, G., 2013. Immunosenescence, aging, and systemic lupus Age-associated elevation in TLR5 leads to increased inammatory responses in
erythematous. Autoimmun. Dis. 2013, 267078. the elderly. Aging Cell 11, 104110.
Mounier, R., Theret, M., Arnold, L., Cuvellier, S., Bultot, L., Goransson, O., Sanz, N., Qian, F., Wang, X., Zhang, L., Lin, A., Zhao, H., Fikrig, E., Montgomery, R.R., 2011.
Ferry, A., Sakamoto, K., Foretz, M., Viollet, B., Chazaud, B., 2013. AMPKalpha1 Impaired interferon signaling in dendritic cells from older donors infected
regulates macrophage skewing at the time of resolution of inammation in vitro with West Nile virus. J. Infect. Dis. 203, 14151424.
during skeletal muscle regeneration. Cell Metab. 18, 251264. Radley, H.G., Grounds, M.D., 2006. Cromolyn administration (to block mast cell
Mourkioti, F., Rosenthal, N., 2008. NF-kappaB signaling in skeletal muscle: degranulation) reduces necrosis of dystrophic muscle in mdx mice. Neurobiol.
prospects for intervention in muscle diseases. J. Mol. Med. (Berl.) 86, 747759. Dis. 23, 387397.
Murton, A.J., Constantin, D., Greenhaff, P.L., 2008. The involvement of the ubiquitin Ramprasad, M.P., Fischer, W., Witztum, J.L., Sambrano, G.R., Quehenberger, O.,
proteasome system in human skeletal muscle remodelling and atrophy. Steinberg, D., 1995. The 94- to 97-kDa mouse macrophage membrane protein
Biochim. Biophys. Acta 1782, 730743. that recognizes oxidized low density lipoprotein and phosphatidylserine-rich
Myburgh, K.H., Kruger, M.J., Smith, C., 2012. Accelerated skeletal muscle recovery liposomes is identical to macrosialin, the mouse homologue of human CD68.
after in vivo polyphenol administration. J. Nutr. Biochem. 23, 10721079. Proc. Natl. Acad. Sci. U. S. A. 92, 95809584.
Nguyen, H.X., Lusis, A.J., Tidball, J.G., 2005. Null mutation of myeloperoxidase in Renault, V., Piron-Hamelin, G., Forestier, C., DiDonna, S., Decary, S., Hentati, F.,
mice prevents mechanical activation of neutrophil lysis of muscle cell Saillant, G., Butler-Browne, G.S., Mouly, V., 2000. Skeletal muscle regeneration
membranes in vitro and in vivo. J. Physiol. 565, 403413. and the mitotic clock. Exp. Gerontol. 35, 711719.
Nguyen, H.X., Tidball, J.G., 2003a. Interactions between neutrophils and Rigamonti, E., Touvier, T., Clementi, E., Manfredi, A.A., Brunelli, S., Rovere-Querini,
macrophages promote macrophage killing of rat muscle cells in vitro. J. P., 2013. Requirement of inducible nitric oxide synthase for skeletal muscle
Physiol. 547, 125132. regeneration after acute damage. J. Immunol. 190, 17671777.
Nguyen, H.X., Tidball, J.G., 2003b. Null mutation of gp91phox reduces muscle Rigamonti, E., Zordan, P., Sciorati, C., Rovere-Querini, P., Brunelli, S., 2014.
membrane lysis during muscle inammation in mice. J. Physiol. 553, 833841. Macrophage plasticity in skeletal muscle repair. BioMed. Res. Int. 2014,
Niwa, Y., Kasama, T., Miyachi, Y., Kanoh, T., 1989. Neutrophil chemotaxis, 560629.
phagocytosis and parameters of reactive oxygen species in human aging: Robertson, T.A., Maley, M.A., Grounds, M.D., Papadimitriou, J.M., 1993. The role of
cross-sectional and longitudinal studies. Life Sci. 44, 16551664. macrophages in skeletal muscle regeneration with particular reference to
Nyugen, J., Agrawal, S., Gollapudi, S., Gupta, S., 2010. Impaired functions of chemotaxis. Exp. Cell Res. 207, 321331.
peripheral blood monocyte subpopulations in aged humans. J. Clin. Immunol. Ruffell, D., Mourkioti, F., Gambardella, A., Kirstetter, P., Lopez, R.G., Rosenthal, N.,
30, 806813. Nerlov, C., 2009. A CREB-C/EBPbeta cascade induces M2 macrophage-specic
Ochoa, O., Sun, D., Reyes-Reyna, S.M., Waite, L.L., Michalek, J.E., McManus, L.M., gene expression and promotes muscle injury repair. Proc. Natl. Acad. Sci. U. S.
Shireman, P.K., 2007. Delayed angiogenesis and VEGF production in CCR2-/- A. 106, 1747517480.
mice during impaired skeletal muscle regeneration. Am. J. Physiol. Regul. Saclier, M., Cuvellier, S., Magnan, M., Mounier, R., Chazaud, B., 2013a.
Integr. Comp. Physiol. 293, R651661. Monocyte/macrophage interactions with myogenic precursor cells during
Ottnad, E., Parthasarathy, S., Sambrano, G.R., Ramprasad, M.P., Quehenberger, O., skeletal muscle regeneration. FEBS J. 280, 41184130.
Kondratenko, N., Green, S., Steinberg, D., 1995. A macrophage receptor for Saclier, M., Yacoub-Youssef, H., Mackey, A.L., Arnold, L., Ardjoune, H., Magnan, M.,
oxidized low density lipoprotein distinct from the receptor for acetyl low Sailhan, F., Chelly, J., Pavlath, G.K., Mounier, R., Kjaer, M., Chazaud, B., 2013b.
density lipoprotein: partial purication and role in recognition of oxidatively Differentially activated macrophages orchestrate myogenic precursor cell fate
damaged cells. Proc. Natl. Acad. Sci. U. S. A. 92, 13911395. during human skeletal muscle regeneration. Stem Cells 31, 384396.
Owens, D.J., Sharples, A.P., Polydorou, I., Alwan, N., Donovan, T.F., Tang, J., Cooper, Sag, D., Carling, D., Stout, R.D., Suttles, J., 2008. Adenosine
R.G., Fraser, W.D., Morton, J.P., Stewart, C., Close, G.L., 2015. A systems based 5 -monophosphate-activated protein kinase promotes macrophage
investigation into vitamin D and skeletal muscle repair, regeneration and polarization to an anti-inammatory functional phenotype. J. Immunol. 181,
hypertrophy. Am. J. Physiol. Endocrinol. Metab. 309 (12), E1019E1031. 86338641.
Panda, A., Arjona, A., Sapey, E., Bai, F., Fikrig, E., Montgomery, R.R., Lord, J.M., Shaw, Serrano, A.L., Baeza-Raja, B., Perdiguero, E., Jardi, M., Munoz-Canoves, P., 2008.
A.C., 2009. Human innate immunosenescence: causes and consequences for Interleukin-6 is an essential regulator of satellite cell-mediated skeletal muscle
immunity in old age. Trends Immunol. 30, 325333. hypertrophy. Cell Metab. 7, 3344.
Panda, A., Qian, F., Mohanty, S., van Duin, D., Newman, F.K., Zhang, L., Chen, S., Shaw, A.C., Goldstein, D.R., Montgomery, R.R., 2013. Age-dependent dysregulation
Towle, V., Belshe, R.B., Fikrig, E., Allore, H.G., Montgomery, R.R., Shaw, A.C., of innate immunity. Nat. Rev. Immunol. 13, 875887.
2010. Age-associated decrease in TLR function in primary human dendritic Shefer, G., Van de Mark, D.P., Richardson, J.B., Yablonka-Reuveni, Z., 2006.
cells predicts inuenza vaccine response. J. Immunol. 184, 25182527. Satellite-cell pool size does matter: dening the myogenic potency of aging
Paulsen, G., Mikkelsen, U.R., Raastad, T., Peake, J.M., 2012. Leucocytes, cytokines skeletal muscle. Dev. Biol. 294, 5066.
and satellite cells: what role do they play in muscle damage and regeneration Shepherd, V.L., Hoidal, J.R., 1990. Clearance of neutrophil-derived myeloperoxidase
following eccentric exercise? Exerc. Immunol. Rev. 18, 4297. by the macrophage mannose receptor. Am. J. Respir. Cell Mol. Biol. 2, 335340.
Payne, G.W., 2006. Effect of inammation on the aging microcirculation: impact on Sonnet, C., Lafuste, P., Arnold, L., Brigitte, M., Poron, F., Authier, F.J., Chretien, F.,
skeletal muscle blood ow control. Microcirculation 13, 343352. Gherardi, R.K., Chazaud, B., 2006. Human macrophages rescue myoblasts and
Pedersen, B.K., 2011. Muscles and their myokines. J. Exp. Biol. 214, 337346. myotubes from apoptosis through a set of adhesion molecular systems. J. Cell
Pedersen, B.K., Febbraio, M.A., 2008. Muscle as an endocrine organ: focus on Sci. 119, 24972507.
muscle-derived interleukin-6. Physiol. Rev. 88, 13791406. Spalding, K.L., Bhardwaj, R.D., Buchholz, B.A., Druid, H., Frisen, J., 2005.
Pedersen, B.K., Febbraio, M.A., 2012. Muscles, exercise and obesity: skeletal muscle Retrospective birth dating of cells in humans. Cell 122, 133143.
as a secretory organ. Nat. Rev. Endocrinol. 8, 457465. Spencer, M.J., Walsh, C.M., Dorshkind, K.A., Rodriguez, E.M., Tidball, J.G., 1997.
Perdiguero, E., Sousa-Victor, P., Ruiz-Bonilla, V., Jardi, M., Caelles, C., Serrano, A.L., Myonuclear apoptosis in dystrophic mdx muscle occurs by perforin-mediated
Munoz-Canoves, P., 2011. p38/MKP-1-regulated AKT coordinates macrophage cytotoxicity. J. Clin. Investig. 99, 27452751.
transitions and resolution of inammation during tissue repair. J. Cell Biol. 195, Srikuea, R., Zhang, X., Park-Sarge, O.K., Esser, K.A., 2012. VDR and CYP27B1 are
307322. expressed in C2C12 cells and regenerating skeletal muscle: potential role in
Pereira, M.G., Baptista, I.L., Carlassara, E.O., Moriscot, A.S., Aoki, M.S., Miyabara, suppression of myoblast proliferation. Am. J. Physiol. Cell Physiol. 303,
E.H., 2014a. Leucine supplementation improves skeletal muscle regeneration C396C405.
after cryolesion in rats. PloS One 9, e85283. Stratos, I., Li, Z., Herlyn, P., Rotter, R., Behrendt, A.K., Mittlmeier, T., Vollmar, B.,
Pereira, M.G., Silva, M.T., Carlassara, E.O., Goncalves, D.A., Abrahamsohn, P.A., 2013. Vitamin D increases cellular turnover and functionally restores the
Kettelhut, I.C., Moriscot, A.S., Aoki, M.S., Miyabara, E.H., 2014b. Leucine skeletal muscle after crush injury in rats. Am. J. Pathol. 182, 895904.
supplementation accelerates connective tissue repair of injured tibialis Teixeira, C.F., Zamuner, S.R., Zuliani, J.P., Fernandes, C.M., Cruz-Hoing, M.A.,
anterior muscle. Nutrients 6, 39814001. Fernandes, I., Chaves, F., Gutierrez, J.M., 2003. Neutrophils do not contribute to
Philippidis, P., Mason, J.C., Evans, B.J., Nadra, I., Taylor, K.M., Haskard, D.O., Landis, local tissue damage, but play a key role in skeletal muscle regeneration, in
R.C., 2004. Hemoglobin scavenger receptor CD163 mediates interleukin-10 mice injected with Bothrops asper snake venom. Muscle Nerve 28, 449459.
release and heme oxygenase-1 synthesis: antiinammatory
36 C. Domingues-Faria et al. / Ageing Research Reviews 26 (2016) 2236

Tidball, J.G., Berchenko, E., Frenette, J., 1999. Macrophage invasion does not Warren, G.L., Hulderman, T., Mishra, D., Gao, X., Millecchia, L., OFarrell, L., Kuziel,
contribute to muscle membrane injury during inammation. J. Leukoc. Biol. 65, W.A., Simeonova, P.P., 2005. Chemokine receptor CCR2 involvement in skeletal
492498. muscle regeneration. FASEB J. 19, 413415.
Tidball, J.G., Villalta, S.A., 2010. Regulatory interactions between muscle and the Warren, G.L., OFarrell, L., Summan, M., Hulderman, T., Mishra, D., Luster, M.I.,
immune system during muscle regeneration. Am. J. Physiol. Regul. Integr. Kuziel, W.A., Simeonova, P.P., 2004. Role of CC chemokines in skeletal muscle
Comp. Physiol. 298, R1173R1187. functional restoration after injury. Am. J. Physiol. Cell Physiol. 286,
Tidball, J.G., Wehling-Henricks, M., 2004. Evolving therapeutic strategies for C1031C1036.
Duchenne muscular dystrophy: targeting downstream events. Pediatr. Res. 56, Wehling-Henricks, M., Sokolow, S., Lee, J.J., Myung, K.H., Villalta, S.A., Tidball, J.G.,
831841. 2008. Major basic protein-1 promotes brosis of dystrophic muscle and
Tidball, J.G., Wehling-Henricks, M., 2007. Macrophages promote muscle attenuates the cellular immune response in muscular dystrophy. Hum. Mol.
membrane repair and muscle bre growth and regeneration during modied Genet. 17, 22802292.
muscle loading in mice in vivo. J. Physiol. 578, 327336. Welle, S., 2002. Cellular and molecular basis of age-related sarcopenia. Can. J. Appl.
Torrente, Y., El Fahime, E., Caron, N.J., Del Bo, R., Belicchi, M., Pisati, F., Tremblay, Physiol. Revue Can. Physiol. Appl. 27, 1941.
J.P., Bresolin, N., 2003. Tumor necrosis factor-alpha (TNF-alpha) stimulates Wenisch, C., Patruta, S., Daxbock, F., Krause, R., Horl, W., 2000. Effect of age on
chemotactic response in mouse myogenic cells. Cell Transplant. 12, 91100. human neutrophil function. J. Leukoc. Biol. 67, 4045.
van Duin, D., Mohanty, S., Thomas, V., Ginter, S., Montgomery, R.R., Fikrig, E., Allore, Woo, M., Isganaitis, E., Cerletti, M., Fitzpatrick, C., Wagers, A.J., Jimenez-Chillaron,
H.G., Medzhitov, R., Shaw, A.C., 2007. Age-associated defect in human TLR-1/2 J., Patti, M.E., 2011. Early life nutrition modulates muscle stem cell number:
function. J. Immunol. 178, 970975. implications for muscle mass and repair. Stem Cells Dev. 20, 17631769.
Vasson, M.P., Farges, M.C., Goncalves-Mendes, N., Talvas, J., Ribalta, J., Wu, Z., Woodring, P.J., Bhakta, K.S., Tamura, K., Wen, F., Feramisco, J.R., Karin, M.,
Winklhofer-Roob, B., Rock, E., Rossary, A., 2013. Does aging affect the immune Wang, J.Y., Puri, P.L., 2000. p38 and extracellular signal-regulated kinases
status? A comparative analysis in 300 healthy volunteers from France, Austria regulate the myogenic program at multiple steps. Mol. Cell. Biol. 20,
and Spain. Immun. Ageing: I & A 10, 38. 39513964.
Verdijk, L.B., Snijders, T., Drost, M., Delhaas, T., Kadi, F., van Loon, L.J., 2014. Satellite Yang, Y., Xu, Y., Li, W., Wang, G., Song, Y., Yang, G., Han, X., Du, Z., Sun, L., Ma, K.,
cells in human skeletal muscle; from birth to old age. Age (Dordr.) 36, 545557. 2009. STAT3 induces muscle stem cell differentiation by interaction with
Villalta, S.A., Deng, B., Rinaldi, C., Wehling-Henricks, M., Tidball, J.G., 2011a. myoD. Cytokine 46, 137141.
IFN-gamma promotes muscle damage in the mdx mouse model of Duchenne Yin, H., Price, F., Rudnicki, M.A., 2013. Satellite cells and the muscle stem cell niche.
muscular dystrophy by suppressing M2 macrophage activation and inhibiting Physiol. Rev. 93, 2367.
muscle cell proliferation. J. Immunol. 187, 54195428. Zador, E., Mendler, L., Takacs, V., de Bleecker, J., Wuytack, F., 2001. Regenerating
Villalta, S.A., Nguyen, H.X., Deng, B., Gotoh, T., Tidball, J.G., 2009. Shifts in soleus and extensor digitorum longus muscles of the rat show elevated levels
macrophage phenotypes and macrophage competition for arginine of TNF-alpha and its receptors, TNFR-60 and TNFR-80. Muscle Nerve 24,
metabolism affect the severity of muscle pathology in muscular dystrophy. 10581067.
Hum. Mol. Genet. 18, 482496. Zerria, K., Jerbi, E., Hammami, S., Maarou, A., Boubaker, S., Xiong, J.P., Arnaout,
Villalta, S.A., Rinaldi, C., Deng, B., Liu, G., Fedor, B., Tidball, J.G., 2011b. M.A., Fathallah, D.M., 2006. Recombinant integrin CD11b A-domain blocks
Interleukin-10 reduces the pathology of mdx muscular dystrophy by polymorphonuclear cells recruitment and protects against skeletal muscle
deactivating M1 macrophages and modulating macrophage phenotype. Hum. inammatory injury in the rat. Immunology 119, 431440.
Mol. Genet. 20, 790805. Zhang, C., Li, Y., Wu, Y., Wang, L., Wang, X., Du, J., 2013. Interleukin-6/signal
Villalta, S.A., Rosenthal, W., Martinez, L., Kaur, A., Sparwasser, T., Tidball, J.G., transducer and activator of transcription 3 (STAT3) pathway is essential for
Margeta, M., Spencer, M.J., Bluestone, J.A., 2014. Regulatory T cells suppress macrophage inltration and myoblast proliferation during muscle
muscle inammation and injury in muscular dystrophy. Sci. Transl. Med. 6, regeneration. J. Biol. Chem. 288, 14891499.
258ra142. Zhang, J., Xiao, Z., Qu, C., Cui, W., Wang, X., Du, J., 2014. CD8 T cells are involved in
Walrand, S., Guillet, C., Salles, J., Cano, N., Boirie, Y., 2011. Physiopathological skeletal muscle regeneration through facilitating MCP-1 secretion and
mechanism of sarcopenia. Clin. Geriatr. Med. 27, 365385. Gr1(high) macrophage inltration. J. Immunol. 193, 51495160.

Das könnte Ihnen auch gefallen